Context

Tumors of the genitourinary tract can be diagnostically challenging, particularly in core biopsies and cystoscopic biopsies with limited material. Immunohistochemistry is a valuable tool to use when morphology alone is insufficient for diagnosis.

Objectives

To review tumors and benign lesions of the kidney, urinary bladder, prostate gland, testis, and paratesticular structures with an emphasis on difficult differential diagnoses, as well as staining patterns in normal tissue. Recommended immunohistochemical stain panels are discussed that can assist in the diagnostic workup.

Data Sources

Review of current literature.

Conclusions

Immunohistochemistry is a valuable tool, assisting in the diagnosis of problematic tumors and benign lesions of the genitourinary tract.

This article will review immunohistochemical markers commonly used as adjuncts in diagnostic problems of tumors and benign lesions of the genitourinary system. This review will focus on the use of markers in differential problems, particularly those encountered in small core biopsy, cystoscopic, and transurethral specimens. Normal structures and tumors of the kidney are discussed, including the crucial role immunohistochemistry (IHC) plays in subtyping of renal epithelial tumors. Problematic flat lesions of the urinary bladder and histologic variants of urothelial carcinoma are examined. Immunohistochemistry of normal and benign and neoplastic changes in the prostate gland are covered, as well as applications of newer diagnostic markers. Immunohistochemistry used in the differential of germ cell tumors and sex cord–stromal tumors in the testis are presented. In addition, the differential diagnoses of tumors of paratesticular structures are discussed. Data in the tables are represented by symbols as defined in Table 1. Antibody abbreviations used in the data tables are compiled in Table 2 for easy reference.

Table 1.

Data Interpretation

Data Interpretation
Data Interpretation
Table 2.

Antibody Abbreviations in the Data Tables

Antibody Abbreviations in the Data Tables
Antibody Abbreviations in the Data Tables

Most renal tumors are epithelial neoplasms; clear cell renal cell carcinoma (CRCC), papillary renal cell carcinoma (PRCC), chromophobe renal cell carcinoma (ChRCC), and oncocytoma account for more than 90% of this group of tumors.13  Other uncommon and rare renal epithelial neoplasms include clear cell papillary renal cell carcinoma (CPRCC), mucinous tubular and spindle cell carcinoma, renal medullary carcinoma, collecting duct carcinoma (CDC), tubulocystic renal cell carcinoma, and Xp11 translocation renal cell carcinoma (TRCC).13  Upper urinary tract urothelial carcinoma (UUC) is usually included in the diagnostic consideration. Diagnosis of a renal epithelial neoplasm tends to be straightforward, based on histologic features alone, especially in a nephrectomy specimen. However, histologic subtyping of these renal epithelial tumors and the distinction of an epithelial tumor from a nonepithelial tumor or a metastasis in a limited sample, such as a needle core biopsy or a fine-needle aspiration specimen, can be challenging. In these scenarios, IHC stains may play a crucial role in reaching a definitive diagnosis.

When working on a renal biopsy specimen obtained from a renal mass, in our opinion the following questions should be raised and appropriate diagnostic approaches should be taken: (1) Is this a low-grade renal epithelial neoplasm or a reactive condition, such as reactive tubules in xanthogranulomatous pyelonephritis, interstitial nephritis due to viral infection, malakoplakia, or other benign condition? (2) If this is a renal epithelial tumor, how should one further subtype this tumor? and (3) If this is a neoplastic process, can one exclude a metastasis or a nonepithelial neoplasm? In this portion of the article, we will review and update the most useful immunomarkers and IHC panels, on the basis of the literature and our experience, in attempting to resolve these diagnostic conundrums, with a specific focus on how to classify a renal epithelial neoplasm and how to avoid overutilization or underutilization of the massive number of biomarkers. It should be emphasized here that there are overwhelming numbers of biomarkers reported in the literature, and investigators appear to have their own preference and experience using certain sets of biomarkers.465  As a result, the International Society of Urological Pathology4  reached no absolute consensus regarding an individual antibody or panel of antibodies to be used for classifying renal tumors or confirming renal metastases.

Low-Grade Epithelial Neoplasm Versus Benign/Reactive Condition

The first question to ask when evaluating a biopsy sample from a renal mass is whether it represents a benign or malignant process. Particularly, is this a low-grade renal epithelial neoplasm or a reactive condition? As a general rule, histologic features such as clear cells, papillary structures, and vascular-rich tissues are clues to suggest a neoplastic process. In contrast, mixed acute and chronic inflammation, atrophic tubules and glomeruli, fibrosis, and granulomas are suggestive of a benign/reactive process. Numerous renal epithelial tumor–associated immunomarkers have been reported in the literature in differentiating subtypes of renal epithelial neoplasms.47  Most of these markers, such as cluster of differentiation (CD) 10, renal cell carcinoma marker (RCCma), α-methylacyl-CoA racemase (P504S), epithelial membrane antigen (EMA), paired box gene (PAX) 8, PAX2, S100A1, and kidney-specific cadherin (ksp-cad), are also expressed in proximal and/or distal renal tubules as well. Therefore, most of these markers have little value in the distinction of a benign process from a renal epithelial tumor. However, vimentin, carbonic anhydrase IX (CAIX), CD117, and kidney injury molecule–1 (KIM-1) are not usually expressed in normal renal tubules, with the exception of KIM-1. In the condition of acute tubular injury, KIM-1 is usually expressed in renal proximal tubules. Therefore, if a low-grade CRCC is suspected, staining with CAIX, KIM-1, and vimentin can be performed, and positive staining for these markers will support a diagnosis of malignancy. Table 3 lists the expression of frequently used immunomarkers in 20 cases of benign renal tissues from Geisinger Medical Center, Danville, Pennsylvania.

Table 3.

Expression of Immunomarkers in Proximal Tubules, Distal Normal Tubules, and Glomerulia,b

Expression of Immunomarkers in Proximal Tubules, Distal Normal Tubules, and Glomerulia,b
Expression of Immunomarkers in Proximal Tubules, Distal Normal Tubules, and Glomerulia,b

Differential Diagnosis of Subtypes of Renal Epithelial Neoplasms

Based on the histologic features, renal epithelial neoplasms can be classified into 2 main groups: low nuclear grade (Fuhrman nuclear grades 1 and 2) and high nuclear grade (Fuhrman nuclear grades 3 and 4 or sarcomatoid features). The group of low-grade neoplasms can be further divided into (1) tumors with clear cell/granular cell features; (2) tumors with both clear cell and papillary features; and (3) tumors with papillary features. The specific immunoprofile for each entity is summarized in Figure 1. The common differential diagnosis and the frequently used immunomarkers are summarized in Table 4.

Figure 1.

Summary of the specific immunoprofile for each renal neoplasm. Abbreviations: AML, angiomyolipoma; Ber-EP4, epithelial cell adhesion molecule; CAIX, carbonic anhydrase IX; CD, cluster of differentiation; CDC, collecting duct carcinoma; ChRCC, chromophobe renal cell carcinoma; CK, cytokeratin; CPRCC, clear cell papillary renal cell carcinoma; CRCC, clear cell renal cell carcinoma; EMA, epithelial membrane antigen; GATA3, GATA-binding protein 3; HMB-45, human melanoma black 45; INI-1, integrase interactor 1; KIM-1, kidney injury molecule–1; ksp-cad, kidney-specific cadherin; MA, metanephric adenoma; Mart-1, melanoma-associated antigen recognized by T cells 1; MTSCC, mucinous tubular and spindle cell carcinoma; ND, no data; OCT4, octamer-binding transcription factor 4; PAX, paired box gene; pCEA, polyclonal carcinoembryonic antigen; PRCC, papillary renal cell carcinoma; pVHL, von Hippel–Lindau tumor suppressor; P504S, α-methylacyl-CoA racemase; RCC, renal cell carcinoma; RCCma, renal cell carcinoma marker; RMC, renal medullary carcinoma; SMA, smooth muscle actin; S100P, placental S100; TFE3, transcription factor E3; UUC, upper urinary tract urothelial carcinoma; WT1, Wilms tumor 1; Xp11 TRCC, Xp11 translocation renal cell carcinoma.

Figure 1.

Summary of the specific immunoprofile for each renal neoplasm. Abbreviations: AML, angiomyolipoma; Ber-EP4, epithelial cell adhesion molecule; CAIX, carbonic anhydrase IX; CD, cluster of differentiation; CDC, collecting duct carcinoma; ChRCC, chromophobe renal cell carcinoma; CK, cytokeratin; CPRCC, clear cell papillary renal cell carcinoma; CRCC, clear cell renal cell carcinoma; EMA, epithelial membrane antigen; GATA3, GATA-binding protein 3; HMB-45, human melanoma black 45; INI-1, integrase interactor 1; KIM-1, kidney injury molecule–1; ksp-cad, kidney-specific cadherin; MA, metanephric adenoma; Mart-1, melanoma-associated antigen recognized by T cells 1; MTSCC, mucinous tubular and spindle cell carcinoma; ND, no data; OCT4, octamer-binding transcription factor 4; PAX, paired box gene; pCEA, polyclonal carcinoembryonic antigen; PRCC, papillary renal cell carcinoma; pVHL, von Hippel–Lindau tumor suppressor; P504S, α-methylacyl-CoA racemase; RCC, renal cell carcinoma; RCCma, renal cell carcinoma marker; RMC, renal medullary carcinoma; SMA, smooth muscle actin; S100P, placental S100; TFE3, transcription factor E3; UUC, upper urinary tract urothelial carcinoma; WT1, Wilms tumor 1; Xp11 TRCC, Xp11 translocation renal cell carcinoma.

Close modal
Table 4.

Useful Immunomarkers in the Diagnosis of Renal Epithelial Neoplasmsa

Useful Immunomarkers in the Diagnosis of Renal Epithelial Neoplasmsa
Useful Immunomarkers in the Diagnosis of Renal Epithelial Neoplasmsa

Renal Epithelial Tumor With Clear Cell/Granular Cell Features

Three main entities are included in this group: low-grade CRCC, ChRCC, and oncocytoma. The useful immunomarkers for the distinction of these 3 entities are listed in Table 5. The staining for cytokeratin (CK) 7 tends to be diffuse (>50% of tumor cells stained) in ChRCC, focal and patchy (<10% of tumor cells stained) in oncocytoma, and negative in CRCC. If a CRCC is diffusely positive for CK7, then a CPRCC should be considered. The nuclear and cytoplasmic staining for S100A1 in oncocytoma can be focal, but it is rarely observed in ChRCC, even focally. Caution should be taken, as S100A1 positivity is frequently seen in CRCC. In our experience, CK7, epithelial cell adhesion molecule (Ber-EP4), S100A1, and CD15 can serve as the initial IHC panel to differentiate a ChRCC from an oncocytoma. If this first IHC panel is inconclusive, additional markers, such as CD82, claudin 7, and claudin 8, can be used. In contrast, coexpression of EMA and vimentin and positivity for CAIX, RCCma, and KIM-1 are the diagnostic panel for CRCCs. Epithelioid angiomyolipoma (AML) with clear cytoplasm is a rare entity and sometimes may present as a mimic. Angiomyolipoma is positive for human melanoma black 45 (HMB-45), melanoma-associated antigen recognized by T cells 1 (Mart-1), and smooth muscle actin (SMA) and negative for CK and other renal cell carcinoma–associated markers such as PAX8/PAX2, CAIX, and RCCma.

Table 5.

Differential Diagnosis of Low-Grade Renal Cell Tumor With Clear Cell/Granular Cell Featuresa

Differential Diagnosis of Low-Grade Renal Cell Tumor With Clear Cell/Granular Cell Featuresa
Differential Diagnosis of Low-Grade Renal Cell Tumor With Clear Cell/Granular Cell Featuresa

Renal Epithelial Tumors With Both Clear Cell and Papillary Features

The main differential diagnosis for a renal cell tumor with both clear cell and papillary features includes CPRCC, CRCC with papillary features, PRCC (type I) with clear cell features, and TRCC. The useful immunomarkers in differentiating these tumors are summarized in Table 6.

Table 6.

Differential Diagnosis of Renal Cell Tumors With Both Clear Cell and Papillary Featuresa

Differential Diagnosis of Renal Cell Tumors With Both Clear Cell and Papillary Featuresa
Differential Diagnosis of Renal Cell Tumors With Both Clear Cell and Papillary Featuresa

Clear cell papillary renal cell carcinoma is a recently recognized, distinct renal epithelial neoplasm that is more frequently seen in patients with end-stage renal disease.54,55,66  The key histologic features include papillary structures, tubules, solid and cystic components, all lined by tumor cells with clear cytoplasm, and usually low Fuhrman nuclear grade.54,55,66  The nuclei tend to be located away from the basement membranes and toward the luminal aspect of the tumor cells, creating subnuclear vacuolization similar to secretory endometrium.5456,66  These tumors lack the characteristic molecular features of PRCC and CRCC, including trisomies of chromosomes 7 and 17, deletions of 3p25, von Hippel–Lindau gene (VHL) mutation, and VHL promoter hypermethylation.54  Immunohistochemically, these tumors are almost always positive for CK7 and CAIX but negative for P504S, CD10, RCCma, and transcription factor E3 (TFE3).54  High-molecular-weight CK (34BE12, CK903) was reported to show positivity in 56% of cases54  but usually showed negativity in CRCC, PRCC, and TRCC. Interestingly, parafibromin tends to be absent or only weakly expressed in parathyroid carcinoma but showed positivity in 100% of CPRCCs and was also expressed in 7% and 19% of CRCCs and PRCCs, respectively.54  To complicate this matter further, Williamson et al53  reported 12 cases of CPRCC-like tumors in patients with VHL disease sharing histologic features of sporadic CPRCC; however, 82% of these cases lacked the characteristic immunohistochemical profile of sporadic CPRCC. These tumors were frequently negative or only focally positive for CK7 and showed diffuse CD10 positivity in 64% of the cases and strong P504S staining in 27% of the cases.53 

In contrast to the IHC profile for CPRCC, CRCC with papillary features is usually positive for EMA, vimentin, CAIX, CD10, and RCCma, and negative for CK7, parafibromin, and 34BE12. In our experience, P504S expression can be seen in approximately 50% of CRCCs.19  The staining pattern of CAIX in CRCC is circumferential membrane staining of tumor cells, whereas the staining pattern in CPRCC tends to spare the apical surface of the tumor cells and outline the basolateral membranes.4  Papillary renal cell carcinoma with clear cell features, especially type I, is another key differential diagnosis, which typically shows a low nuclear grade and frequent clear cell changes, in comparison to type II PRCC, with high nuclear grade, an abundant amount of eosinophilic cytoplasm, nuclear stratification, and less frequent clear cell changes. Type I PRCC is usually diffusely positive for CK7, P504S, and focally positive for CAIX and CD10. An example of type I PRCC with clear cell features on a small-needle core biopsy specimen is shown in Figure 2, A through D. A reliable IHC panel in the distinction of type I PRCC from type II PRCC is not available yet; however, type II PRCC is frequently negative or only focally positive for CK7, strongly and diffusely positive for P504S, and can be positive for CK20.

Figure 2.

An example of type I papillary renal cell carcinoma with clear cell features on a small needle core biopsy. A, Hematoxylin-eosin–stained section. B, Diffuse and strong positivity for CK7. C, Diffuse and strong positivity for P504S. D, Focal positivity for CAIX (original magnification ×10 [A through D]).

Figure 2.

An example of type I papillary renal cell carcinoma with clear cell features on a small needle core biopsy. A, Hematoxylin-eosin–stained section. B, Diffuse and strong positivity for CK7. C, Diffuse and strong positivity for P504S. D, Focal positivity for CAIX (original magnification ×10 [A through D]).

Close modal

The TRCC group of rare renal neoplasms predominately arises in pediatric patients with chromosomal translocation involving the TFE3 transcription factor gene located at 11p11.2 locus or the TFEB gene at 6p21.6,7,55,58  Based on a study of 443 consecutive nephrectomies from a single institution, the incidence of TRCC in adults has been reported as 1.6%.57  The typical morphologic features include papillary and nested structures lined by clear to granular cells with voluminous cytoplasm, hyaline material, and psammoma bodies.58  Immunohistochemically, the tumor cells are positive for TFE3, cathepsin K, HMB-45, Mart-1, P504S, RCCma, and PAX8, only focally positive for CAIX, and usually negative for epithelial markers including cytokeratin AE1/3 (AE1/3), EMA, and CK7.55 

Renal Epithelial Tumors With Papillary Features

There is significant overlapping of the immunostaining profiles of PRCC and mucinous tubular and spindle cell carcinoma.6  The distinction between these 2 entities should primarily rely on histologic features. In contrast, metanephric adenoma is usually positive for S100, Wilms tumor 1 (WT1), and CD57 and negative for renal cell carcinoma–associated markers, such as P504S and RCCma.20,33  The useful immunomarkers for distinguishing these 3 entities are summarized in Table 7.

Table 7.

Differential Diagnosis of Renal Cell Tumors With Papillary Featuresa

Differential Diagnosis of Renal Cell Tumors With Papillary Featuresa
Differential Diagnosis of Renal Cell Tumors With Papillary Featuresa

Differential Diagnosis of High-Grade Renal Epithelial Tumors

The main diagnostic considerations for a high-grade renal epithelial neoplasm include high-grade CRCC, high-grade PRCC (type II), sarcomatoid renal cell carcinoma, UUC, CDC, renal medullary carcinoma, TRCC, and metastasis. The useful markers for this differential diagnosis are summarized in Tables 8 and 9. Carvalho and colleagues67  suggested that p63, CK7, PAX8, and integrase interactor 1 (INI-1) comprised an optimal IHC panel to differentiate poorly differentiated urothelial carcinoma from other high-grade renal epithelial tumors. Their study demonstrated that (1) all urothelial carcinomas (N = 18) were positive for p63, 15 of 18 (83%) were negative for PAX8, 94% were positive for both CK7 and 34BE12, and all were positive for INI-1; and (2) all CDC cases were negative for p63, and 5 of 6 were positive for PAX8.67  The study by Albadine et al67  concluded that 100% of CDCs (N = 21) were positive for PAX8, and only 14% were positive for p63; whereas 97% of UUCs (N = 34) were positive for p63, and only 8.8% were positive for PAX8. Loss of INI-1 expression in nearly 100% of renal medullary carcinoma cases has been well documented in many studies.61,62,67  Interestingly, octamer-binding transcription factor 3/4 (OCT3/4) expression has been demonstrated in 10 of 14 renal medullary carcinoma cases (71%) but was negative in 5 of 5 CDC cases and 10 of 10 UUC cases.60  Chang et al59  reported PAX8 expression in 69% (31 of 45) of sarcomatoid renal cell carcinomas and only in 4% (2 of 45) of sarcomatoid urothelial carcinomas of the bladder, whereas 18% (2 of 11) of sarcomatoid urothelial carcinomas of the upper urinary tract were positive for PAX8, showing some overlapping staining features with sarcomatoid renal cell carcinoma.59 

Table 8.

Differential Diagnosis of High-Grade Renal Epithelial Tumorsa

Differential Diagnosis of High-Grade Renal Epithelial Tumorsa
Differential Diagnosis of High-Grade Renal Epithelial Tumorsa
Table 9.

Common Metastases in the Kidneya,b

Common Metastases in the Kidneya,b
Common Metastases in the Kidneya,b

Epithelial Neoplasm Versus Nonepithelial Neoplasm

Angiomyolipoma is the most common nonepithelial renal neoplasm, and a classic one that can be easily diagnosed by its histologic features of mixed components of spindle cells, adipose tissue, and hyalinized blood vessels. An epithelioid angiomyolipoma with clear cell features may potentially pose a diagnostic challenge; however, a panel of IHC markers such as SMA, HMB-45, Mart-1, and CK is helpful in reaching a definitive diagnosis. Small round cell neoplasms can present as a primary renal tumor, including neuroendocrine carcinoma, Ewing sarcoma/primitive neuroectodermal tumor, neuroblastoma, lymphoma, plasmacytomas, and myeloid sarcoma. We recently encountered a case of primary precursor B-cell lymphoblastic lymphoma with a focal bone metastasis in a 4-year-old boy. The tumor mimicked Ewing sarcoma/primitive neuroectodermal tumor both morphologically and immunohistochemically. Figure 3, A through F, demonstrates the tumor cells to be positive for CD99, Friend leukemia virus integration 1 (Fli-1), PAX5, CD10, and terminal deoxynucleotidyl transferase (TdT) and to have a markedly increased mindbomb homolog 1 (MIB-1/Ki-67) proliferative index (not shown).

Figure 3.

Primary precursor B-cell lymphoma of the kidney in a 4-year-old boy. A, Hematoxylin-eosin–stained section. B, Membranous staining for CD99. C, Nuclear staining for Fli-1. D, Nuclear staining for PAX5. E, Nuclear staining for TdT. F, Membranous staining for CD10 (original magnification ×10 [A through F]).

Figure 3.

Primary precursor B-cell lymphoma of the kidney in a 4-year-old boy. A, Hematoxylin-eosin–stained section. B, Membranous staining for CD99. C, Nuclear staining for Fli-1. D, Nuclear staining for PAX5. E, Nuclear staining for TdT. F, Membranous staining for CD10 (original magnification ×10 [A through F]).

Close modal

Primary Renal Epithelial Tumor Versus Metastasis

Metastasis in the kidney is rare; the most common site of origin of a metastatic carcinoma to the kidney is lung, including adenocarcinoma, squamous cell carcinoma, and small cell carcinoma. Other sites of origin may include colon, breast, gastrointestinal tract, pancreatobiliary tract, melanoma, and ovary. Adrenal cortical carcinoma may be a direct invasion or a metastasis. Steroidogenic factor 1 (SF-1) is a recently described, orphan member of the nuclear hormone receptor superfamily, which plays a crucial role in the differentiation of steroidogenic tissue. SF-1 has been reported to show positivity in 100% of adrenal cortical adenomas and 90% of adrenal cortical carcinomas and negativity in PRCC, ChRCC, and oncocytoma.64  Only 3 of 20 CRCC cases showed focal positivity, with less than 10% of tumor cells stained.64  All adrenal cortical neoplasms were negative for EMA, whereas most renal cell carcinomas were positive.64  Another study by Sangoi and colleagues65  demonstrated that SF-1 expression was seen in 86% of adrenal cortical neoplasms (N = 54) and none of 185 metastatic CRCCs. Expression of KIM-1, PAX8, and EMA was observed in 83%, 83%, and 78% of CRCCs, respectively.65  In contrast, all 54 adrenal cortical neoplasms were negative for these 3 markers.65  A small percentage of CRCCs can be positive for calretinin, inhibin-α, and Melan-A; whereas a small percentage of adrenal cortical neoplasms can be positive for CD10, CK, and CAIX.65  SF-1, EMA, PAX8/or KIM-1/or pVHL are the optimal initial IHC panel to differentiate these 2 entities.46,64,65  The useful diagnostic markers for metastasis in the kidney are summarized in Table 9.

In this section, we focus on lesions that may pose a diagnostic dilemma in cystoscopic biopsy material that is often scant, superficial, fragmented, or compromised by cautery artifact. Immunohistochemistry can be a valuable aid; however, it does not replace careful evaluation of morphology, including any normal urothelium that may be present.

Normal Urothelium and Flat Lesions

Evaluation of urothelial lesions presents a unique problem, as common immunohistochemical markers exhibit staining patterns specific to particular compartments within normal urothelium. Familiarity with staining in normal histology is key to understanding how changes in the normal staining pattern correlate to various conditions in the morphologic spectrum of changes seen in reactive processes, hyperplasia, dysplasia, and carcinoma. Normal urothelium is 4 to 7 cells in thickness and consists of a layer of basal cells, 2 to 5 layers of intermediate cells, and a layer of superficial umbrella cells. Cystoscopic biopsies of a flat urothelial lesion can be problematic and immunohistochemistry can be quite helpful if morphologic evaluation alone is insufficient. We recommend a panel that includes CK20, CD44, p53, and Ki-67. Compartmental staining patterns of normal urothelium and flat lesions are summarized in Table 1068–83 and illustrated in Figure 4, A through F.

Table 10.

Recommended Panel for Evaluation of Flat Urothelial Lesionsa

Recommended Panel for Evaluation of Flat Urothelial Lesionsa
Recommended Panel for Evaluation of Flat Urothelial Lesionsa
Figure 4.

A, CD44 stain in normal urothelium shows faint staining in the basal cell layer. B, CD44 stain in polypoid cystitis stains all cell layers of the urothelium. C, Ki-67 stains a few basal and deep intermediate cells in normal urothelium. D, Ki-67 expression may be increased in polypoid cystitis but will still be confined predominantly to basal cells. E, p53 may show weak staining of a few cells in normal urothelium. F, p53 may stain more cells and with slightly more intensity in reactive processes such as polypoid cystitis (original magnifications ×10 [A, C, and E], ×5 [B, D, and F]).

Figure 4.

A, CD44 stain in normal urothelium shows faint staining in the basal cell layer. B, CD44 stain in polypoid cystitis stains all cell layers of the urothelium. C, Ki-67 stains a few basal and deep intermediate cells in normal urothelium. D, Ki-67 expression may be increased in polypoid cystitis but will still be confined predominantly to basal cells. E, p53 may show weak staining of a few cells in normal urothelium. F, p53 may stain more cells and with slightly more intensity in reactive processes such as polypoid cystitis (original magnifications ×10 [A, C, and E], ×5 [B, D, and F]).

Close modal

Nephrogenic Adenoma and Its Mimickers

Several lesions can have an exophytic papillary or polypoid cystoscopic appearance, including papillary urothelial neoplasms, polypoid cystitis, and nephrogenic adenoma. Although nephrogenic adenomas occur most commonly in the bladder, they may also develop in the renal pelvis, ureters, and urethra. If the lesion has a polypoid or papillary growth pattern, the differential diagnoses may include papillary urothelial neoplasms and polypoid cystitis; however, nephrogenic adenomas may also exhibit tubular, cystic, and solid histologic pattern, broadening the differential diagnosis to include prostatic adenocarcinoma, ectopic prostatic tissue, and clear cell adenocarcinoma of the bladder. Immunohistochemistry can help differentiate between these various lesions, and the pertinent markers are summarized in Table 11.82,8496 

Table 11.

Useful Markers in the Differential Diagnoses of Nephrogenic Adenoma (NA)a

Useful Markers in the Differential Diagnoses of Nephrogenic Adenoma (NA)a
Useful Markers in the Differential Diagnoses of Nephrogenic Adenoma (NA)a

Invasive Urothelial Carcinoma With Variant Histology

Variants of invasive urothelial carcinoma present a challenge, as many cystoscopic biopsies contain very limited material and may not show urothelial dysplasia or an invasive carcinoma with a more classic appearance. Invasive urothelial carcinoma may show a micropapillary pattern similar to that seen in carcinomas from other primary sites, as well as epithelioid mesothelioma. Table 12 summarizes markers useful in this differential.50,97107  Similarly, the plasmacytoid variant of invasive urothelial carcinoma can be confused with plasmacytoma, adenocarcinomas, and other tumors. Table 13 summarizes the markers we recommend for this morphologic pattern.50,108114  The nested variant of invasive urothelial carcinoma can be difficult to distinguish from a benign proliferation of von Brunn nests, as well as tumors exhibiting a nested pattern such as paraganglioma, carcinoid, and melanoma. There is great overlap in the immunohistochemical staining pattern of these entities, particularly between paraganglioma and carcinoid tumors where presence or absence of S100-positive sustentacular cells may be the only clue. These stains are not particularly helpful to distinguish between urothelial carcinoma and von Brunn nests, unless the lesion is a carcinoma and diffuse positivity for CK20 and increased Ki-67 expression is present; otherwise, negative or weak CK20 staining and low Ki-67 expression can be indicative of either entity. Markers for tumors with a nested morphology are summarized in Table 14.115125 

Table 12.

Recommended Markers for Tumors With Micropapillary Morphologya

Recommended Markers for Tumors With Micropapillary Morphologya
Recommended Markers for Tumors With Micropapillary Morphologya
Table 13.

Recommended Markers for Tumors With Plasmacytoid Morphologya

Recommended Markers for Tumors With Plasmacytoid Morphologya
Recommended Markers for Tumors With Plasmacytoid Morphologya
Table 14.

Recommended Markers for Tumors With Nested Morphologya

Recommended Markers for Tumors With Nested Morphologya
Recommended Markers for Tumors With Nested Morphologya

Adenocarcinoma in the Bladder

The primary question when an adenocarcinoma is discovered in the bladder is whether or not it is a primary neoplasm, or has spread to the bladder either by direct extension or metastasis. Table 15 offers a basic immunohistochemical panel to differentiate primary bladder adenocarcinoma from other adenocarcinomas that commonly spread to the bladder, as well as conventional urothelial carcinoma and clear cell renal cell carcinoma.* Nephrogenic adenoma may also be included in the differential diagnosis and is covered in Table 11.

Table 15.

Markers in the Differential of Urothelial Adenocarcinomaa

Markers in the Differential of Urothelial Adenocarcinomaa
Markers in the Differential of Urothelial Adenocarcinomaa

Spindle Cell Neoplasms in the Bladder

Spindle cell lesions are uncommon in the bladder; however, they present a difficult diagnostic group in cystoscopic biopsies where the primary consideration is a sarcomatoid urothelial carcinoma versus a mesenchymal tumor. The differential diagnosis of mesenchymal neoplasms is further clouded by lack of a standard classification scheme in the urothelial tract, in part because of their rarity. The markers useful in the differential of spindle cell neoplasms are summarized in Table 16.72,82,139150  We prefer the term inflammatory myofibroblastic tumor, which encompasses a variety of terms in the literature, including inflammatory pseudotumor, pseudosarcomatous myofibroblastic tumor, inflammatory pseudosarcomatous fibromyxoid tumor, and fibromyxoid pseudotumor; this term is reflective of the low-grade nature of this lesion and does not incite the same alarm and confusion as the terms that include pseudosarcoma.

Table 16.

Markers in Spindle Cell Neoplasms of the Bladdera,b

Markers in Spindle Cell Neoplasms of the Bladdera,b
Markers in Spindle Cell Neoplasms of the Bladdera,b

The diagnosis of prostate carcinoma is largely achieved by histomorphologic assessment on routine hematoxylin-eosin sections of prostate surgical specimens. One of the key diagnostic criteria for malignancy is the absence of basal cells. The identification of basal cells on a hematoxylin-eosin section of core needle biopsies is not always straightforward; some artifacts may make this task challenging.151  Immunohistochemistry using antibodies against basal cells is an objective and reliable tool to demonstrate the presence or absence of basal cells. However, the lack of basal cells in isolation is not diagnostic of malignancy, since benign mimics can show false-negative reactivity. A number of studies have identified biomarkers that are overexpressed in prostate carcinoma and their precursor. The application of cancer overexpressed biomarkers in conjunction with basal cell markers provide additional confidence in making a definitive diagnosis of prostate cancer in challenging cases. In addition, the differential considerations between urothelial and prostatic origin arise occasionally, especially in transurethral resection specimens. Under those circumstances, immunohistochemistry becomes a valuable aid in reaching an accurate diagnosis. The commonly used immunomarkers in prostate surgical pathology include ones aiding in the diagnosis of malignancy and others defining prostate organ specificity.

Immunomarkers That Aid in the Diagnosis of Malignancy

Two groups of biomarkers are used to aid in the diagnosis of prostate carcinoma on core needle biopsy specimens. The first group is composed of basal cell markers in which a negative immunoreaction indicates malignancy. The second group consists of upregulated biomarkers in which a positive immunoreaction suggests malignancy.

Basal Cell Markers

p63, a cloned homologue of the p53 tumor suppressor gene, is a nuclear protein involved in the regulation of growth and development in epithelium of various tissues including prostate.152  Anti-p63 labels the p63 protein in the nuclei of basal or progenitor cells in a variety of epithelia, serving as a basal cell marker with a nuclear staining pattern in prostate surgical pathology.152  Many studies concluded that the absence of p63 stain indicates prostate carcinoma.153157  Thus, the application of p63 immunostain is essential when confronting challenging cases. However, aberrant diffuse p63 expression in prostatic carcinomas had been reported.158160  In the study of Osunkoya et al,158  21 cases of prostatic carcinomas revealed aberrant diffuse expression of p63 (100% of cancer cells in 19 of 21; 75% in 2 of 21); all were negative for high-molecular-weight cytokeratin (HMWCK, or CK903), positive for prostate-specific antigen (PSA) and P504S. A few reports161,162  also documented false-positive p63 staining in prostate cancers with a basal cell distribution. Although aberrant expression of p63 is rare, caution should be exercised when working on cases with equivocal features.

HMWCK (CK903, clone 34βE12) is a well-known basal cell marker. Gown and Vogel163  investigated the expression of monoclonal antibodies generated against different human intermediate filament proteins in a variety of human normal and neoplastic tissue in 1984; they first described the staining pattern of antibody 34βE12 in prostate as being basal glandular epithelial cells only in distribution. Thereafter, CK903 was studied extensively in normal and neoplastic prostatic tissues, validating its diagnostic utility as a differentiating immunomarker between benign and malignant process, with the lack of basal cell staining being an indicator of malignancy.163167  However, lack of basal cell staining is also noted in 5% to 23% of benign prostatic glands.151,154,165,168  Googe et al166  and Yang et al167  reported focal patchy immunoreactivity (nonbasal cell distribution) for basal cell markers (especially HMWCK) in high-grade prostatic carcinomas. Thus, Epstein151  advised that negative basal cell staining in small glandular (and other) prostatic lesions is supportive of a diagnosis of prostatic carcinoma only in the appropriate hematoxylin-eosin context. CK5/6, another HMWCK, is expressed in basal cells. A few studies evaluated CK5/6 expression in various prostate normal, benign, and neoplastic tissues, suggesting superior sensitivity and reliability of CK5/6 compared with CK903.169,170 

Markers Overexpressed in Prostate Cancer

P504S, the gene encoding α-methylacyl-coenzyme A racemase (AMACR), was first reported by Xu et al171  in 2000. By using complementary DNA (cDNA) library subtraction in conjunction with high-throughput microarray screening techniques, they found that P504S is overexpressed in prostate carcinomas, while low or undetectable in normal prostatic tissues. In 2001, Jiang et al172  found 100% cases of prostate carcinomas expressing P504S regardless of Gleason score, with positive staining defined as continuous, dark cytoplasmic staining or apical granular staining that can be observed easily at low-power magnification. In contrast, 88% of the benign prostatic tissue was negative for P504; only 12% showed focal, weak reactivity. Subsequently, P504S expression was evaluated in a variety of benign and neoplastic prostatic tissues,173179  demonstrating that P504S is overexpressed in prostate carcinoma and high-grade prostatic intraepithelial neoplasia (HGPIN), with a reported sensitivity in the range of 62% to 100%; the staining patterns were variable, from diffuse and strong (most reports) to focal and weak.162  By comparison, variants of prostate carcinoma showed lower sensitivity, including 62% to 68%180  and 72%179  for foamy gland carcinoma; 83%181  and 70% to 77%180  for pseudohyperplastic carcinoma; and 67% for atrophic carcinoma.182  A few studies181,183,184  investigated P504S expression in postradiation-therapy prostate carcinomas and reported a positive rate of 80% to 100%. The benign atypical glands in postradiation-therapy prostate were P504S negative. Their findings suggested the diagnostic utility of P504S immunostaining in distinction between postradiation prostate carcinoma and radiation-induced atypia in benign prostatic epithelium.

Although benign prostate glands usually lack P504S expression, positive immunoreactivity for P504S was described by several investigators in partial atrophy, atypical adenomatous hyperplasia, and nephrogenic adenoma (NA). Partial atrophy is the most common benign mimicker of prostate carcinoma on core needle biopsy. Wang et al185  evaluated P504S and basal cell expression in 198 cases of partial atrophy and found P504S expression in 69.2% of cases, and positive basal cells in 68.7% that showed a focal patchy staining pattern. Herawi et al186  reported patchy basal staining in 87% of partial atrophy foci and P504S positivity in 79% of cases. Atypical adenomatous hyperplasia is another well-known mimicker of prostate carcinoma. Yang et al187  reported P504S expression in 17.5% cases of atypical adenomatous hyperplasia, while Skinnider et al86  reported 14%. The reported expression of P504S in nephrogenic adenoma ranges from 58% to 100%, and most nephrogenic adenomas were also CK903 negative.86,188,189  The similarity in certain morphologic features and the P504S+/CK903− immunoprofile of nephrogenic adenoma make the distinction between prostate carcinoma from nephrogenic adenoma challenging. Additional immunomarkers, especially PSA, PAX2 (or PAX8), and CK7 can help to differentiate nephrogenic adenoma from prostate carcinoma. Nephrogenic adenoma is usually negative, or only focally and weakly reactive to PSA, and positive for CK7 as well as PAX2,95,173,190  while prostate carcinoma expresses PSA and is nonreactive to CK7 and PAX2 (see Figure 5, A through H).

Figure 5.

A, Nephrogenic adenoma. B, Nuclear staining for PAX-2, nephrogenic adenoma. C, Diffuse CK7 staining, nephrogenic adenoma. D, Lack of prostate-specific antigen (PSA) expression, nephrogenic adenoma. E, Prostate adenocarcinoma. F, Lack of expression for PAX-2, prostate adenocarcinoma. G, Negative for CK7, prostate adenocarcinoma. H, Strong and diffuse staining for PSA, prostate adenocarcinoma (hematoxylin-eosin, original magnification ×5 [A and E]; original magnification ×5 [B through D and F through H]).

Figure 5.

A, Nephrogenic adenoma. B, Nuclear staining for PAX-2, nephrogenic adenoma. C, Diffuse CK7 staining, nephrogenic adenoma. D, Lack of prostate-specific antigen (PSA) expression, nephrogenic adenoma. E, Prostate adenocarcinoma. F, Lack of expression for PAX-2, prostate adenocarcinoma. G, Negative for CK7, prostate adenocarcinoma. H, Strong and diffuse staining for PSA, prostate adenocarcinoma (hematoxylin-eosin, original magnification ×5 [A and E]; original magnification ×5 [B through D and F through H]).

Close modal

P504S is upregulated in prostate carcinomas and HGPIN, and can serve as a positive tissue marker; however, by itself, P504S positivity is not specific as it may also be expressed in benign mimickers such as atypical adenomatous hyperplasia and nephrogenic adenoma. Jiang et al179  suggested using a positive P504S marker along with a negative basal cell–specific marker, such as CK903 and/or p63, to help confirm the diagnosis when small atypical glands are seen. Zhou et al156  reported that a basal cell cocktail (CK903 plus p63) improves the detection of prostate basal cells. Browne et al191  prospectively stained 123 cases with a basal cell cocktail and P504s; the immunostain results contributed to rendering a final diagnosis in 70% of cases. They suggest that combining P504s and the basal cell cocktail on a single slide would be superior to using either marker separately. Currently, an antibody cocktail composed of P504S, CK903, and p63 is routinely used.

ERG, a transcription factor in the erythroblastosis virus E26 transforming sequence (ETS) family known to be expressed in endothelial cells, is overexpressed in subsets of prostate carcinoma, acute myeloid leukemia, and Ewing sarcoma. A number of studies evaluated ERG expression by immunohistochemistry in benign and neoplastic prostatic tissues, reporting a positive rate of 38% to 45% in prostate carcinoma, 22% to 29% in HGPIN, and rare expression in benign glands.192196  In nonprostatic tumors, ERG expression is exclusively identified in vascular tumors, 70% of extramedullary myeloid tumors, 7% of Ewing sarcomas, and rare large cell undifferentiated pulmonary carcinoma, as well as mesothelioma.192,194  Those data suggest that ERG is highly specific for prostate carcinoma, in addition to vascular tumors, extramedullary myeloid tumors, and rare Ewing sarcoma, but has low sensitivity. Its utility in the differential between benign mimics and prostate carcinoma on core needle biopsies may be limited. However, the high specificity for prostate carcinoma may prove to be of value in the identification of prostate primary cancer when working on tumors of unknown origin.

Immunomarkers Used in Identification of Prostate Primary Tumor

Prostate-specific antigen and prostate-specific acid phosphatase (PSAP) are 2 well-known prostate tissue–specific immunomarkers, and both are highly sensitive and relatively specific; however, their expression in nonprostatic tissues has been reported, including melanoma, breast carcinoma, and salivary ductal carcinoma.162,197202  It had been documented that poorly differentiated prostate carcinomas express less PSA and PSAP than benign prostatic tissue and low-grade prostate carcinomas. The reported sensitivities range from 25% to 35% when using monoclonal antibody and approximately 95% when using polyclonal antibody.133,162,203,204  We evaluated PSA expression (using both monoclonal and polyclonal antibodies) in 133 cases of prostate carcinoma (low-intermediate grade: 97; high grade: 36) and 909 cases of nonprostatic tumors from a broad spectrum of human tissues and demonstrated the following: (1) PSA expression is identified in 100% cases of prostate carcinoma with both antibodies; the staining intensity is stronger for polyclonal antibody than monoclonal antibody; (2) No difference can be appreciated when comparing different grades of prostate carcinoma; (3) PSA expression in nonprostatic tumors is 1.3% (12 of 909) when using polyclonal antibody and 1.1% when using monoclonal antibody. The positive cases are 1 of 20 papillary renal cell carcinomas (focal, weak), 1 of 74 breast invasive ductal carcinomas (focal, moderate), and 10 of 93 for polyclonal antibody and 8 of 93 for monoclonal antibody in melanoma (focal, weak). Our data showed a PSA sensitivity of 100%, and specificity of 98.7%.194 

P501S, or prostein, is a 553-amino-acid protein identified by cDNA library subtraction followed by high-throughput microarray screening technique.205  Prostein (P501S) messenger RNA and protein expression are prostate specific, restricted to both normal and malignant prostate tissue; all other nonprostatic normal or malignant tissues lack prostein expression.205,206  Sheridan et al207  immunohistochemically evaluated prostein expression in 20 cases of primary prostate carcinoma, 20 cases of normal prostate tissue, and 69 cases of metastatic prostate carcinoma, finding prostein expression in 100% of primary prostate carcinomas and normal prostate tissue, and 99% of metastatic prostate carcinomas. They described the staining pattern as perinuclear cytoplasmic (Golgi) in distribution. Yin et al208  found that 100% of benign prostate tissue, 94.1% of primary prostate carcinomas, and 87% of metastatic prostate carcinomas were immunoreactive to prostein. Both studies also examined PSA expression, showing similar staining results. The data, although limited, suggest the potential utility of prostein as a prostate-specific marker in the detection of metastatic prostate carcinoma.

NKX3.1 is a prostate-specific homeobox gene located on chromosome band 8p21. In human normal and neoplastic tissues, NKX3.1 expression is highly specific, restricted to prostate, testis, and breast with a nuclear staining pattern as illustrated in Figure 6, A and B.209−211 Gelmann et al210  evaluated the expression of NKX3.1 in 4061 samples encompassing a broad spectrum of human cancers and normal tissues and found that NKX3.1 is expressed primarily in benign and malignant prostatic epithelial cells, but also normal testis, 5% to 9% of breast invasive ductal carcinomas, and 26% to 27% of breast invasive lobular carcinomas. Chuang et al133  evaluated the sensitivity of prostate-specific immunomarkers, including PSA, prostein (P501S), prostate-specific membrane antigen (PSMA), NKX3.1, and proPSA (pPSA) on 38 poorly differentiated prostate carcinomas and 35 high-grade invasive urothelial carcinomas. They found the sensitivities for labeling prostate carcinoma were 97.4% for PSA, 100% for P501S, 92.1% for PSMA, 94.7% for NKX3.1, and 94.7% for pPSA. Thus, they suggested using PSA as the first screening marker in differentiating high-grade prostate carcinoma from high-grade urothelial carcinoma; other markers are useful when PSA shows negative or equivocal staining. Gurel et al209  evaluated the expression of NKX3.1, PAS, and PSAP in 69 cases of metastatic prostate carcinomas and 349 cases representing a variety of nonprostatic tumors. The sensitivity for identifying metastatic prostatic adenocarcinomas overall was 98.6% for NKX3.1, 94.2% for PSA, and 98.6% for PSAP. The specificity of NKX3.1 was 99.7% (1 of 349 nonprostatic tumors positive). The sole positive nonprostatic cancer case was an invasive lobular carcinoma of the breast. The available limited data suggest that NKX3.1 is a highly sensitive and relatively specific tissue marker for prostate carcinoma and may have utility in identification of prostate origin when working on tumors of unknown primary.

Figure 6.

A, Prostate adenocarcinoma. B, NKX2.1 expression, nuclear pattern, prostate adenocarcinoma (hematoxylin-eosin, original magnification ×10 [A]; original magnification ×10 [B]).

Figure 6.

A, Prostate adenocarcinoma. B, NKX2.1 expression, nuclear pattern, prostate adenocarcinoma (hematoxylin-eosin, original magnification ×10 [A]; original magnification ×10 [B]).

Close modal

Testicular tumors account for only 1% of all tumors, so they are relatively rare for many pathologists and may present a significant diagnostic challenge. Most testicular tumors occurring at any age are germ cell tumors (GCTs). Approximately 50% of GCTs in adults are pure classic seminomas, but approximately 30% will be mixed GCTs. Each component in mixed GCTs may exhibit multiple morphologic patterns, creating additional challenges. The histologic patterns of GCTs often overlap with those of sex cord–stromal tumors (SCSTs), and some tumors may have both germ cell and sex cord–stromal components to further complicate matters. Identifying all components of a testicular tumor is critical for patient management, as the various components may or may not respond to radiation or chemotherapy.1,212  The newer stem cell markers, such as SALL4, OCT4, and Nanog, are particularly useful in identification of GCTs. Immunomarkers useful in the differential identification of GCTs are presented in Table 17213–255 and illustrated in Figure 7, A through F. Markers that are helpful in distinguishing between GCTs and SCSTs are presented in Table 18.212,232,252,256  We recommend an initial panel consisting of sal-like 4 (SALL4), placental-like alkaline phosphatase (PLAP), inhibin α, and calretinin to differentiate between GCTs and SCSTs; if all 4 markers show negativity, then other neoplasms such as adenocarcinomas, melanoma, or lymphoma should be considered. Markers useful in the identification of SCSTs are included in Table 19217,256–280 and a typical pattern of SF-1 expression is illustrated in Figure 8.

Table 17.

Markers Recommended in the Differential of Germ Cell Tumorsa,b

Markers Recommended in the Differential of Germ Cell Tumorsa,b
Markers Recommended in the Differential of Germ Cell Tumorsa,b
Figure 7.

A, PLAP stains membranes of intratubular germ cell neoplasia (ITGCN). B, OCT-4 stains nuclei of ITGCN. C, Inhibin-α stains the Sertoli cells within the seminiferous tubule, as well as the Leydig cells in the interstitium, but not the neoplastic germ cells (ITGCN). D, CAM 5.2 stains the epithelium of the rete testis, but not the ITGCN that is showing a pagetoid pattern. E, PAX8 will also stain the nuclei of the rete testis epithelium, but not ITGCN. F, Glypican-3 highlights the syncytiotrophoblast in this yolk sac tumor (original magnification ×5 [A through F]).

Figure 7.

A, PLAP stains membranes of intratubular germ cell neoplasia (ITGCN). B, OCT-4 stains nuclei of ITGCN. C, Inhibin-α stains the Sertoli cells within the seminiferous tubule, as well as the Leydig cells in the interstitium, but not the neoplastic germ cells (ITGCN). D, CAM 5.2 stains the epithelium of the rete testis, but not the ITGCN that is showing a pagetoid pattern. E, PAX8 will also stain the nuclei of the rete testis epithelium, but not ITGCN. F, Glypican-3 highlights the syncytiotrophoblast in this yolk sac tumor (original magnification ×5 [A through F]).

Close modal
Table 18.

Markers Useful to Distinguish Between Germ Cell Tumors and Sex Cord/Stromal Tumorsa,b

Markers Useful to Distinguish Between Germ Cell Tumors and Sex Cord/Stromal Tumorsa,b
Markers Useful to Distinguish Between Germ Cell Tumors and Sex Cord/Stromal Tumorsa,b
Table 19.

Markers Recommended in the Differential of Sex Cord/Stromal Tumorsa

Markers Recommended in the Differential of Sex Cord/Stromal Tumorsa
Markers Recommended in the Differential of Sex Cord/Stromal Tumorsa
Figure 8.

Nuclei of Sertoli cells (red arrow) within seminiferous tubules stain for SF-1, as do Leydig cells (green arrow) in the interstitium. Note that spermatocytic precursors (black arrow) within the seminiferous tubules do not stain (original magnification ×10).

Figure 8.

Nuclei of Sertoli cells (red arrow) within seminiferous tubules stain for SF-1, as do Leydig cells (green arrow) in the interstitium. Note that spermatocytic precursors (black arrow) within the seminiferous tubules do not stain (original magnification ×10).

Close modal

Tumors of the paratesticular structures are rare and it can be difficult to distinguish the primary site of origin and whether or not the tumor is primary to the paratesticular structures, or metastatic from the abdomen or pelvis. The distinction between epithelial or mesothelial origin is also problematic in these tumors. Markers that may help in the distinction of primary epithelial neoplasms of the rete testis and epididymis, as well as mesothelial neoplasms, are presented in Table 20.217,256,281302  If the tumor in this region appears to be metastatic, Table 21 lists markers that may help in the identification of adenocarcinomas occurring in the male pelvis.

Table 20.

Markers Useful in the Diagnosis of Paratesticular Tumorsa,b

Markers Useful in the Diagnosis of Paratesticular Tumorsa,b
Markers Useful in the Diagnosis of Paratesticular Tumorsa,b
Table 21.

Markers Useful in the Differential of Adenocarcinoma in the Male Pelvisa

Markers Useful in the Differential of Adenocarcinoma in the Male Pelvisa
Markers Useful in the Differential of Adenocarcinoma in the Male Pelvisa

Immunohistochemisty has broad diagnostic applications in genitourinary pathology and is a useful adjunct to morphology in difficult cases. It can be applied to benign and malignant neoplasms, but this must be done in the context of comparing to expression of these markers in normal tissues.

1
Eble
JN
,
Sauter
G
,
Epstein
JI
,
Sesterhenn
IA
.
Pathology and Genetics of Tumours of the Urinary System and Male Genital Organs
.
Lyon, France
:
IARC Press;
2004
.
World Health Organization Classification of Tumours; vol 7
.
2
Murphy
WM
,
Grignon
DJ
,
Perlman
EJ
.
Tumors of the Kidney, Bladder, and Related Urinary Structures
.
Washington, DC
:
American Registry of Pathology;
2004
.
AFIP Atlas of Tumor Pathology; 4th series, fascicle 1
.
3
MacLennan
GT
,
Cheng
L
.
Neoplasms of the kidney
.
In
:
Bostwick
DG
,
Chen
L
,
eds
.
Urologic Surgical Pathology. 2nd ed
.
Philadelphia, PA
:
Elsevier;
2008
:
77
172
.
4
Tan
PH
,
Cheng
L
,
Rioux-Leclercq
N
,
et al
.
Renal tumors: diagnostic and prognostic biomarkers
.
Am J Surg Pathol
.
2013
;
37
(
10
):
1518
1531
.
5
Lin
F
,
Yang
XJ
.
Kidney
.
In
:
Lin
F
,
Prichard
JW
,
Liu
H
,
Wilkerson
ML
,
Schuerch
C
,
eds
.
Handbook of Practical Immunohistochemistry: Frequently Asked Questions
.
New York, NY
:
Springer;
2011
:
335
354
.
6
Truong
LD
,
Shen
SS
.
Immunohistochemical diagnosis of renal neoplasms
.
Arch Pathol Lab Med
.
2011
;
135
(
1
):
92
109
.
7
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
(
4
):
467
480
.
8
Skinnider
BF
,
Amin
MB
.
An immunohistochemical approach to the differential diagnosis of renal tumors
.
Semin Diagn Pathol
.
2005
;
22
(
1
):
51
68
.
9
Kim
MK
,
Kim
S
.
Immunohistochemical profile of common epithelial neoplasms arising in the kidney
.
Appl Immunohistochem Mol Morphol
.
2002
;
10
(
4
):
332
338
.
10
Skinnider
BF
,
Folpe
AL
,
Hennigar
RA
,
et al
.
Distribution of cytokeratins and vimentin in adult renal neoplasms and normal renal tissue: potential utility of a cytokeratin antibody panel in the differential diagnosis of renal tumors
.
Am J Surg Pathol
.
2005
;
29
(
6
):
747
754
.
11
Kobayashi
N
,
Matsuzaki
O
,
Shirai
S
,
Aoki
I
,
Yao
M
,
Nagashima
Y
.
Collecting duct carcinoma of the kidney: an immunohistochemical evaluation of the use of antibodies for differential diagnosis
.
Hum Pathol
.
2008
;
39
(
9
):
1350
1359
.
12
Haitel
A
,
Susani
M
,
Wick
N
,
Mazal
PR
,
Wrba F. c-kit overexpression in chromophobe renal cell carcinoma is not associated with c-kit mutation of exons 9 and 11
.
Am J Surg Pathol
.
2005
;
29
(
6
):
842
.
13
Huo
L
,
Sugimura
J
,
Tretiakova
MS
,
et al
.
C-kit expression in renal oncocytomas and chromophobe renal cell carcinomas
.
Hum Pathol
.
2005
;
36
(
3
):
262
268
.
14
Memeo
L
,
Jhang
J
,
Assaad
AM
,
et al
.
Immunohistochemical analysis for cytokeratin 7, KIT, and PAX2: value in the differential diagnosis of chromophobe cell carcinoma
.
Am J Clin Pathol
.
2007
;
127
(
2
):
225
229
.
15
Pan
CC
,
Chen
PC
,
Chiang
H
.
Overexpression of KIT (CD117) in chromophobe renal cell carcinoma and renal oncocytoma
.
Am J Clin Pathol
.
2004
;
121
(
6
):
878
883
.
16
Went
P
,
Dirnhofer
S
,
Salvisberg
T
,
et al
.
Expression of epithelial cell adhesion molecule (EpCam) in renal epithelial tumors
.
Am J Surg Pathol
.
2005
;
29
(
1
):
83
88
.
17
Lin
F
,
Zhang
PL
,
Yang
XJ
,
et al
.
Human kidney injury molecule-1 (hKIM-1): a useful immunohistochemical marker for diagnosing renal cell carcinoma and ovarian clear cell carcinoma
.
Am J Surg Pathol
.
2007
;
31
(
3
):
371
381
.
18
Jiang
Z
,
Fanger
GR
,
Woda
BA
,
et al
.
Expression of alpha-methylacyl-CoA racemase (P504s) in various malignant neoplasms and normal tissues: a study of 761 cases
.
Hum Pathol
.
2003
;
34
(
8
):
792
796
.
19
Lin
F
,
Brown
RE
,
Shen
T
,
Yang
XJ
,
Schuerch
C
.
Immunohistochemical detection of P504S in primary and metastatic renal cell carcinomas
.
Appl Immunohistochem Mol Morphol
.
2004
;
12
(
2
):
153
159
.
20
Azabdaftari
G
,
Alroy
J
,
Banner
BF
,
Ucci
A
,
Bhan
I
,
Cheville
JC
.
S100 protein expression distinguishes metanephric adenomas from other renal neoplasms
.
Pathol Res Pract
.
2008
;
204
(
10
):
719
723
.
21
Li
G
,
Barthelemy
A
,
Feng
G
,
et al
.
S100A1: a powerful marker to differentiate chromophobe renal cell carcinoma from renal oncocytoma
.
Histopathology
.
2007
;
50
(
5
):
642
647
.
22
Rocca
PC
,
Brunelli
M
,
Gobbo
S
,
et al
.
Diagnostic utility of S100A1 expression in renal cell neoplasms: an immunohistochemical and quantitative RT-PCR study
.
Mod Pathol
.
2007
;
20
(
7
):
722
728
.
23
Daniel
L
,
Lechevallier
E
,
Giorgi
R
,
et al
.
Pax-2 expression in adult renal tumors
.
Hum Pathol
.
2001
;
32
(
3
):
282
287
.
24
Gokden
N
,
Gokden
M
,
Phan
DC
,
McKenney
JK
.
The utility of PAX-2 in distinguishing metastatic clear cell renal cell carcinoma from its morphologic mimics: an immunohistochemical study with comparison to renal cell carcinoma marker
.
Am J Surg Pathol
.
2008
;
32
(
10
):
1462
1467
.
25
Mazal
PR
,
Stichenwirth
M
,
Koller
A
,
Blach
S
,
Haitel
A
,
Susani
M
.
Expression of aquaporins and PAX-2 compared to CD10 and cytokeratin 7 in renal neoplasms: a tissue microarray study
.
Mod Pathol
.
2005
;
18
(
4
):
535
540
.
26
Dorai
T
,
Sawczuk
IS
,
Pastorek
J
,
Wiernik
PH
,
Dutcher
JP
.
The role of carbonic anhydrase IX overexpression in kidney cancer
.
Eur J Cancer
.
2005
;
41
(
18
):
2935
2947
.
27
Avery
AK
,
Beckstead
J
,
Renshaw
AA
,
Corless
CL
.
Use of antibodies to RCC and CD10 in the differential diagnosis of renal neoplasms
.
Am J Surg Pathol
.
2000
;
24
(
2
):
203
210
.
28
Camparo
P
,
Vasiliu
V
,
Molinie
V
,
et al
.
Renal translocation carcinomas: clinicopathologic, immunohistochemical, and gene expression profiling analysis of 31 cases with a review of the literature
.
Am J Surg Pathol
.
2008
;
32
(
5
):
656
670
.
29
Chu
P
,
Arber
DA
.
Paraffin-section detection of CD10 in 505 nonhematopoietic neoplasms: frequent expression in renal cell carcinoma and endometrial stromal sarcoma
.
Am J Clin Pathol
.
2000
;
113
(
3
):
374
382
.
30
Pan
CC
,
Chen
PC
,
Ho
DM
.
The diagnostic utility of MOC31, BerEP4, RCC marker and CD10 in the classification of renal cell carcinoma and renal oncocytoma: an immunohistochemical analysis of 328 cases
.
Histopathology
.
2004
;
45
(
5
):
452
459
.
31
Paner
GP
,
Srigley
JR
,
Radhakrishnan
A
,
et al
.
Immunohistochemical analysis of mucinous tubular and spindle cell carcinoma and papillary renal cell carcinoma of the kidney: significant immunophenotypic overlap warrants diagnostic caution
.
Am J Surg Pathol
.
2006
;
30
(
1
):
13
19
.
32
Shen
SS
,
Ro
JY
,
Tamboli
P
,
et al
.
Mucinous tubular and spindle cell carcinoma of kidney is probably a variant of papillary renal cell carcinoma with spindle cell features
.
Ann Diagn Pathol
.
2007
;
11
(
1
):
13
21
.
33
Olgac
S
,
Hutchinson
B
,
Tickoo
SK
,
Reuter
VE
.
Alpha-methylacyl-CoA racemase as a marker in the differential diagnosis of metanephric adenoma
.
Mod Pathol
.
2006
;
19
(
2
):
218
224
.
34
Zhou
M
,
Roma
A
,
Magi-Galluzzi
C
.
The usefulness of immunohistochemical markers in the differential diagnosis of renal neoplasms
.
Clin Lab Med
.
2005
;
25
(
2
):
247
257
.
35
McGregor
DK
,
Khurana
KK
,
Cao
C
,
et al
.
Diagnosing primary and metastatic renal cell carcinoma: the use of the monoclonal antibody 'Renal Cell Carcinoma Marker'
.
Am J Surg Pathol
.
2001
;
25
(
12
):
1485
1492
.
36
Liu
L
,
Qian
J
,
Singh
H
,
Meiers
I
,
Zhou
X
,
Bostwick
DG
.
Immunohistochemical analysis of chromophobe renal cell carcinoma, renal oncocytoma, and clear cell carcinoma: an optimal and practical panel for differential diagnosis
.
Arch Pathol Lab Med
.
2007
;
131
(
8
):
1290
1297
.
37
Adley
BP
,
Papavero
V
,
Sugimura
J
,
Teh
BT
,
Yang
XJ
.
Diagnostic value of cytokeratin 7 and parvalbumin in differentiating chromophobe renal cell carcinoma from renal oncocytoma
.
Anal Quant Cytol Histol
.
2006
;
28
(
4
):
228
236
.
38
Martignoni
G
,
Pea
M
,
Chilosi
M
,
et al
.
Parvalbumin is constantly expressed in chromophobe renal carcinoma
.
Mod Pathol
.
2001
;
14
(
8
):
760
767
.
39
Young
AN
,
Amin
MB
,
Moreno
CS
,
et al
.
Expression profiling of renal epithelial neoplasms: a method for tumor classification and discovery of diagnostic molecular markers
.
Am J Pathol
.
2001
;
158
(
5
):
1639
1651
.
40
Shen
SS
,
Krishna
B
,
Chirala
R
,
Amato
RJ
,
Truong
LD
.
Kidney-specific cadherin, a specific marker for the distal portion of the nephron and related renal neoplasms
.
Mod Pathol
.
2005
;
18
(
7
):
933
940
.
41
Mazal
PR
,
Exner
M
,
Haitel
A
,
et al
.
Expression of kidney-specific cadherin distinguishes chromophobe renal cell carcinoma from renal oncocytoma
.
Hum Pathol
.
2005
;
36
(
1
):
22
28
.
42
Adley
BP
,
Gupta
A
,
Lin
F
,
Luan
C
,
Teh
BT
,
Yang
XJ
.
Expression of kidney-specific cadherin in chromophobe renal cell carcinoma and renal oncocytoma
.
Am J Clin Pathol
.
2006
;
126
(
1
):
79
85
.
43
Young
AN
,
de Oliveira Salles
PG
,
Lim
SD
,
et al
.
Beta defensin-1, parvalbumin, and vimentin: a panel of diagnostic immunohistochemical markers for renal tumors derived from gene expression profiling studies using cDNA microarrays
.
Am J Surg Pathol
.
2003
;
27
(
2
):
199
205
.
44
Javed
R
,
Zhai
QJ
,
Shen
SS
,
Krishman
B
,
Ro
JY
,
Truong
LD
.
PAX-8 is a specific marker for renal neoplasms: comparison with PAX-2, renal cell carcinoma marker antigen (RCCM) and kidney specific cadherin (KSP)
.
Mod Pathol
.
2009
;
22
(
1S
):
174A
.
45
Osunkoya
AO
,
Cohen
C
,
Lawson
D
,
Picken
MM
,
Amin
MB
,
Young
AN
.
Claudin-7 and claudin-8: immunohistochemical markers for the differential diagnosis of chromophobe renal cell carcinoma and renal oncocytoma
.
Hum Pathol
.
2009
;
40
(
2
):
206
210
.
46
Lin
F
,
Shi
J
,
Liu
H
,
et al
.
Immunohistochemical detection of the von Hippel-Lindau gene product (pVHL) in human tissues and tumors: a useful marker for metastatic renal cell carcinoma and clear cell carcinoma of the ovary and uterus
.
Am J Clin Pathol
.
2008
;
129
(
4
):
592
605
.
47
Kim
SS
,
Choi
YD
,
Jin
XM
,
et al
.
Immunohistochemical stain for cytokeratin 7, S100A1 and claudin 8 is valuable in differential diagnosis of chromophobe renal cell carcinoma from renal oncocytoma
.
Histopathology
.
2009
;
54
(
5
):
633
635
.
48
Lin
F
,
Shi
J
,
Yang
XJ
,
Zhang
PL
,
Dupree
W
.
A useful panel of immunohistochemical markers in differentiating papillary renal cell carcinoma from papillary urothelial carcinoma
.
Mod Pathol
.
2008
;
21
(
1S
):
166A
.
49
Shen
SS
,
Truong
LD
,
Scarpelli
M
,
Lopez-Beltran
A
.
Role of immunohistochemistry in diagnosing renal neoplasms: when is it really useful?
Arch Pathol Lab Med
.
2012
;
136
(
4
):
410
417
.
50
Liu
H
,
Shi
J
,
Wilkerson
ML
,
Lin
F
.
Immunohistochemical evaluation of GATA3 expression in tumors and normal tissues: a useful immunomarker for breast and urothelial carcinomas
.
Am J Clin Pathol
.
2012
;
138
(
1
):
57
64
.
51
Yang
XJ
,
Zhou
M
,
Hes
O
,
et al
.
Tubulocystic carcinoma of the kidney: clinicopathologic and molecular characterization
.
Am J Surg Pathol
.
2008
;
32
(
2
):
177
187
.
52
Zhou
M
,
Yang
XJ
,
Lopez
JI
,
et al
.
Renal tubulocystic carcinoma is closely related to papillary renal cell carcinoma: implications for pathologic classification
.
Am J Surg Pathol
.
2009
;
33
(
12
):
1840
1849
.
53
Williamson
SR
,
Zhang
S
,
Eble
JN
,
et al
.
Clear cell papillary renal cell carcinoma-like tumors in patients with von Hippel-Lindau disease are unrelated to sporadic clear cell papillary renal cell carcinoma
.
Am J Surg Pathol
.
2013
;
37
(
8
):
1131
1139
.
54
Rohan
SM
,
Xiao
Y
,
Liang
Y
,
et al
.
Clear-cell papillary renal cell carcinoma: molecular and immunohistochemical analysis with emphasis on the von Hippel-Lindau gene and hypoxia-inducible factor pathway-related proteins
.
Mod Pathol
.
2011
;
24
(
9
):
1207
1220
.
55
Ross
H
,
Martignoni
G
,
Argani
P
.
Renal cell carcinoma with clear cell and papillary features
.
Arch Pathol Lab Med
.
2012
;
136
(
4
):
391
399
.
56
Cui
C
,
Ziober
A
,
Bing
Z
.
Expression of parafibromin in clear cell papillary renal cell carcinoma
.
Appl Immunohistochem Mol Morphol
.
2013
;
21
(
4
):
322
325
.
57
Komai
Y
,
Fujiwara
M
,
Fujii
Y
,
et al
.
Adult Xp11 translocation renal cell carcinoma diagnosed by cytogenetics and immunohistochemistry
.
Clin Cancer Res
.
2009
;
15
(
4
):
1170
1176
.
58
Argani
P
,
Antonescu
CR
,
Illei
PB
,
et al
.
Primary renal neoplasms with the ASPL-TFE3 gene fusion of alveolar soft part sarcoma: a distinctive tumor entity previously included among renal cell carcinomas of children and adolescents
.
Am J Pathol
.
2001
;
159
(
1
):
179
192
.
59
Chang
A
,
Brimo
F
,
Montgomery
EA
,
Epstein
JI
.
Use of PAX8 and GATA3 in diagnosing sarcomatoid renal cell carcinoma and sarcomatoid urothelial carcinoma
.
Hum Pathol
.
2013
;
44
(
8
):
1563
1568
.
60
Rao
P
,
Tannir
NM
,
Tamboli
P
.
Expression of OCT3/4 in renal medullary carcinoma represents a potential diagnostic pitfall
.
Am J Surg Pathol
.
2012
;
36
(
4
):
583
588
.
61
Liu
Q
,
Galli
S
,
Srinivasan
R
,
Linehan
WM
,
Tsokos
M
,
Merino
MJ
.
Renal medullary carcinoma: molecular, immunohistochemistry, and morphologic correlation
.
Am J Surg Pathol
.
2013
;
37
(
3
):
368
374
.
62
Hollmann
TJ
,
Hornick
JL
.
INI1-deficient tumors: diagnostic features and molecular genetics
.
Am J Surg Pathol
.
2011
;
35
(
10
):
e47
e63
.
63
Albadine
R
,
Schultz
L
,
Illei
P
,
et al
.
PAX8 (+)/p63 (-) immunostaining pattern in renal collecting duct carcinoma (CDC): a useful immunoprofile in the differential diagnosis of CDC versus urothelial carcinoma of upper urinary tract
.
Am J Surg Pathol
.
2010
;
34
(
7
):
965
969
.
64
Enriquez
ML
,
Lal
P
,
Ziober
A
,
Wang
L
,
Tomaszewski
JE
,
Bing
Z
.
The use of immunohistochemical expression of SF-1 and EMA in distinguishing adrenocortical tumors from renal neoplasms
.
Appl Immunohistochem Mol Morphol
.
2012
;
20
(
2
):
141
145
.
65
Sangoi
AR
,
Fujiwara
M
,
West
RB
,
et al
.
Immunohistochemical distinction of primary adrenal cortical lesions from metastatic clear cell renal cell carcinoma: a study of 248 cases
.
Am J Surg Pathol
.
2011
;
35
(
5
):
678
686
.
66
Williamson
SR
,
Eble
JN
,
Cheng
L
,
Grignon
DJ
.
Clear cell papillary renal cell carcinoma: differential diagnosis and extended immunohistochemical profile
.
Mod Pathol
.
2013
;
26
(
5
):
697
708
.
67
Carvalho
JC
,
Thomas
DG
,
McHugh
JB
,
Shah
RB
,
Kunju
LP
.
p63, CK7, PAX8 and INI-1: an optimal immunohistochemical panel to distinguish poorly differentiated urothelial cell carcinoma from high-grade tumours of the renal collecting system
.
Histopathology
.
2012
;
60
(
4
):
597
608
.
68
Mallofre
C
,
Castillo
M
,
Morente
V
,
Sole
M
.
Immunohistochemical expression of CK20, p53, and Ki-67 as objective markers of urothelial dysplasia
.
Mod Pathol
.
2003
;
16
(
3
):
187
191
.
69
Nakopoulou
L
,
Vourlakou
C
,
Zervas
A
,
Tzonou
A
,
Gakiopoulou
H
,
Dimopoulos
MA
.
The prevalence of bcl-2, p53, and Ki-67 immunoreactivity in transitional cell bladder carcinomas and their clinicopathologic correlates
.
Hum Pathol
.
1998
;
29
(
2
):
146
154
.
70
Southgate
J
,
Harnden
P
,
Trejdosiewicz
LK
.
Cytokeratin expression patterns in normal and malignant urothelium: a review of the biological and diagnostic implications
.
Histol Histopathol
.
1999
;
14
(
2
):
657
664
.
71
McKenney
JK
,
Desai
S
,
Cohen
C
,
Amin
MB
.
Discriminatory immunohistochemical staining of urothelial carcinoma in situ and non-neoplastic urothelium: an analysis of cytokeratin 20, p53, and CD44 antigens
.
Am J Surg Pathol
.
2001
;
25
(
8
):
1074
1078
.
72
McKenney
JK
,
Amin
MB
.
The role of immunohistochemistry in the diagnosis of urinary bladder neoplasms
.
Semin Diagn Pathol
.
2005
;
22
(
1
):
69
87
.
73
Cina
SJ
,
Lancaster-Weiss
KJ
,
Lecksell
K
,
Epstein
JI
.
Correlation of Ki-67 and p53 with the new World Health Organization/International Society of Urological Pathology Classification System for Urothelial Neoplasia
.
Arch Pathol Lab Med
.
2001
;
125
(
5
):
646
651
.
74
Laguna
P
,
Smedts
F
,
Nordling
J
,
et al
.
Keratin expression profiling of transitional epithelium in the painful bladder syndrome/interstitial cystitis
.
Am J Clin Pathol
.
2006
;
125
(
1
):
105
110
.
75
Sarkis
AS
,
Dalbagni
G
,
Cordon-Cardo
C
,
et al
.
Association of P53 nuclear overexpression and tumor progression in carcinoma in situ of the bladder
.
J Urol
.
1994
;
152
(
2, pt 1
):
388
392
.
76
Yin
H
,
Leong
AS
.
Histologic grading of noninvasive papillary urothelial tumors: validation of the 1998 WHO/ISUP system by immunophenotyping and follow-up
.
Am J Clin Pathol
.
2004
;
121
(
5
):
679
687
.
77
Harnden
P
,
Eardley
I
,
Joyce
AD
,
Southgate
J
.
Cytokeratin 20 as an objective marker of urothelial dysplasia
.
Br J Urol
.
1996
;
78
(
6
):
870
875
.
78
Petraki
CD
,
Sfikas
CP
.
Review: non-papillary urothelial lesions of the urinary bladder: morphological classification and immunohistochemical markers
.
In Vivo
.
2008
;
22
(
4
):
493
501
.
79
Soini
Y
,
Turpeenniemi-Hujanen
T
,
Kamel
D
,
et al
.
p53 immunohistochemistry in transitional cell carcinoma and dysplasia of the urinary bladder correlates with disease progression
.
Br J Cancer
.
1993
;
68
(
5
):
1029
1035
.
80
Sun
W
,
Zhang
PL
,
Herrera
GA
.
p53 protein and Ki-67 overexpression in urothelial dysplasia of bladder
.
Appl Immunohistochem Mol Morphol
.
2002
;
10
(
4
):
327
331
.
81
Yin
H
,
He
Q
,
Li
T
,
Leong
AS
.
Cytokeratin 20 and Ki-67 to distinguish carcinoma in situ from flat non-neoplastic urothelium
.
Appl Immunohistochem Mol Morphol
.
2006
;
14
(
3
):
260
265
.
82
Emerson
RE
,
Cheng
L
.
Immunohistochemical markers in the evaluation of tumors of the urinary bladder: a review
.
Anal Quant Cytol Histol
.
2005
;
27
(
6
):
301
316
.
83
Hodges
KB
,
Lopez-Beltran
A
,
Davidson
DD
,
Montironi
R
,
Cheng
L
.
Urothelial dysplasia and other flat lesions of the urinary bladder: clinicopathologic and molecular features
.
Hum Pathol
.
2010
;
41
(
2
):
155
162
.
84
Oliva
E
,
Amin
MB
,
Jimenez
R
,
Young
RH
.
Clear cell carcinoma of the urinary bladder: a report and comparison of four tumors of mullerian origin and nine of probable urothelial origin with discussion of histogenesis and diagnostic problems
.
Am J Surg Pathol
.
2002
;
26
(
2
):
190
197
.
85
Torenbeek
R
,
Lagendijk
JH
,
Van Diest
PJ
,
Bril
H
,
van de Molengraft
FJ
,
Meijer
CJ
.
Value of a panel of antibodies to identify the primary origin of adenocarcinomas presenting as bladder carcinoma
.
Histopathology
.
1998
;
32
(
1
):
20
27
.
86
Skinnider
BF
,
Oliva
E
,
Young
RH
,
Amin
MB
.
Expression of alpha-methylacyl-CoA racemase (P504S) in nephrogenic adenoma: a significant immunohistochemical pitfall compounding the differential diagnosis with prostatic adenocarcinoma
.
Am J Surg Pathol
.
2004
;
28
(
6
):
701
705
.
87
Xiao
GQ
,
Burstein
DE
,
Miller
LK
,
Unger
PD
.
Nephrogenic adenoma: immunohistochemical evaluation for its etiology and differentiation from prostatic adenocarcinoma
.
Arch Pathol Lab Med
.
2006
;
130
(
6
):
805
810
.
88
Gilcrease
MZ
,
Delgado
R
,
Vuitch
F
,
Albores-Saavedra
J
.
Clear cell adenocarcinoma and nephrogenic adenoma of the urethra and urinary bladder: a histopathologic and immunohistochemical comparison
.
Hum Pathol
.
1998
;
29
(
12
):
1451
1456
.
89
McCluggage
WG
,
Ganesan
R
,
Hirschowitz
L
,
Miller
K
,
Rollason
TP
.
Ectopic prostatic tissue in the uterine cervix and vagina: report of a series with a detailed immunohistochemical analysis
.
Am J Surg Pathol
.
2006
;
30
(
2
):
209
215
.
90
Halat
S
,
Eble
JN
,
Grignon
DJ
,
et al
.
Ectopic prostatic tissue: histogenesis and histopathological characteristics
.
Histopathology
.
2011
;
58
(
5
):
750
758
.
91
Fromont
G
,
Barcat
L
,
Gaudin
J
,
Irani
J
.
Revisiting the immunophenotype of nephrogenic adenoma
.
Am J Surg Pathol
.
2009
;
33
(
11
):
1654
1658
.
92
Herawi
M
,
Drew
PA
,
Pan
CC
,
Epstein
JI
.
Clear cell adenocarcinoma of the bladder and urethra: cases diffusely mimicking nephrogenic adenoma
.
Hum Pathol
.
2010
;
41
(
4
):
594
601
.
93
Tong
GX
,
Melamed
J
,
Mansukhani
M
,
et al
.
PAX2: a reliable marker for nephrogenic adenoma
.
Mod Pathol
.
2006
;
19
(
3
):
356
363
.
94
Kunju
LP
,
Mehra
R
,
Snyder
M
,
Shah
RB
.
Prostate-specific antigen, high-molecular-weight cytokeratin (clone 34betaE12), and/or p63: an optimal immunohistochemical panel to distinguish poorly differentiated prostate adenocarcinoma from urothelial carcinoma
.
Am J Clin Pathol
.
2006
;
125
(
5
):
675
681
.
95
Allan
CH
,
Epstein
JI
.
Nephrogenic adenoma of the prostatic urethra: a mimicker of prostate adenocarcinoma
.
Am J Surg Pathol
.
2001
;
25
(
6
):
802
808
.
96
Tong
GX
,
Weeden
EM
,
Hamele-Bena
D
,
et al
.
Expression of PAX8 in nephrogenic adenoma and clear cell adenocarcinoma of the lower urinary tract: evidence of related histogenesis?
Am J Surg Pathol
.
2008
;
32
(
9
):
1380
1387
.
97
Lopez-Beltran
A
,
Montironi
R
,
Blanca
A
,
Cheng
L
.
Invasive micropapillary urothelial carcinoma of the bladder
.
Hum Pathol
.
2010
;
41
(
8
):
1159
1164
.
98
Lotan
TL
,
Ye
H
,
Melamed
J
,
Wu
XR
,
Shih
I
,
Epstein
JI
.
Immunohistochemical panel to identify the primary site of invasive micropapillary carcinoma
.
Am J Surg Pathol
.
2009
;
33
(
7
):
1037
1041
.
99
Johansson
SL
,
Borghede
G
,
Holmang
S
.
Micropapillary bladder carcinoma: a clinicopathological study of 20 cases
.
J Urol
.
1999
;
161
(
6
):
1798
1802
.
100
Tacha
D
,
Zhou
D
,
Cheng
L
.
Expression of PAX8 in normal and neoplastic tissues: a comprehensive immunohistochemical study
.
Appl Immunohistochem Mol Morphol
.
2011
;
19
(
4
):
293
299
.
101
Kadivar
M
,
Boozari
B
.
Applications and limitations of immunohistochemical expression of “Napsin-A” in distinguishing lung adenocarcinoma from adenocarcinomas of other organs
.
Appl Immunohistochem Mol Morphol
.
2013
;
21
(
3
):
191
195
.
102
Turner
BM
,
Cagle
PT
,
Sainz
IM
,
Fukuoka
J
,
Shen
SS
,
Jagirdar
J
.
Napsin
A
,
a new marker for lung adenocarcinoma, is complementary and more sensitive and specific than thyroid transcription factor 1 in the differential diagnosis of primary pulmonary carcinoma: evaluation of 1674 cases by tissue microarray
.
Arch Pathol Lab Med
.
2012
;
136
(
2
):
163
171
.
103
Bishop
JA
,
Sharma
R
,
Illei
PB
.
Napsin
A
and thyroid transcription factor-1 expression in carcinomas of the lung, breast, pancreas, colon, kidney, thyroid, and malignant mesothelioma
.
Hum Pathol
.
2010
;
41
(
1
):
20
25
.
104
Anttila
S
.
Epithelioid lesions of the serosa
.
Arch Pathol Lab Med
.
2012
;
136
(
3
):
241
252
.
105
Tot
T
.
The value of cytokeratins 20 and 7 in discriminating metastatic adenocarcinomas from pleural mesotheliomas
.
Cancer
.
2001
;
92
(
10
):
2727
2732
.
106
Ordonez
NG
.
Application of immunohistochemistry in the diagnosis of epithelioid mesothelioma: a review and update
.
Hum Pathol
.
2013
;
44
(
1
):
1
19
.
107
Chu
AY
,
Litzky
LA
,
Pasha
TL
,
Acs
G
,
Zhang
PJ
.
Utility of D2-40, a novel mesothelial marker, in the diagnosis of malignant mesothelioma
.
Mod Pathol
.
2005
;
18
(
1
):
105
110
.
108
Chu
PG
,
Weiss
LM
.
Immunohistochemical characterization of signet-ring cell carcinomas of the stomach, breast, and colon
.
Am J Clin Pathol
.
2004
;
121
(
6
):
884
892
.
109
Raspollini
MR
,
Sardi
I
,
Giunti
L
,
et al
.
Plasmacytoid urothelial carcinoma of the urinary bladder: clinicopathologic, immunohistochemical, ultrastructural, and molecular analysis of a case series
.
Hum Pathol
.
2011
;
42
(
8
):
1149
1158
.
110
Nigwekar
P
,
Tamboli
P
,
Amin
MB
,
Osunkoya
AO
,
Ben-Dor
D
,
Amin
MB
.
Plasmacytoid urothelial carcinoma: detailed analysis of morphology with clinicopathologic correlation in 17 cases
.
Am J Surg Pathol
.
2009
;
33
(
3
):
417
424
.
111
Lim
MG
,
Adsay
NV
,
Grignon
DJ
,
Osunkoya
AO
.
E-cadherin expression in plasmacytoid, signet ring cell and micropapillary variants of urothelial carcinoma: comparison with usual-type high-grade urothelial carcinoma
.
Mod Pathol
.
2011
;
24
(
2
):
241
247
.
112
Fritsche
HM
,
Burger
M
,
Denzinger
S
,
Legal
W
,
Goebell
PJ
,
Hartmann
A
.
Plasmacytoid urothelial carcinoma of the bladder: histological and clinical features of 5 cases
.
J Urol
.
2008
;
180
(
5
):
1923
1927
.
113
Lopez-Beltran
A
,
Requena
MJ
,
Montironi
R
,
Blanca
A
,
Cheng
L
.
Plasmacytoid urothelial carcinoma of the bladder
.
Hum Pathol
.
2009
;
40
(
7
):
1023
1028
.
114
Yeh
IT
,
Mies
C
.
Application of immunohistochemistry to breast lesions
.
Arch Pathol Lab Med
.
2008
;
132
(
3
):
349
358
.
115
Jones
TD
,
Zhang
S
,
Lopez-Beltran
A
,
et al
.
Urothelial carcinoma with an inverted growth pattern can be distinguished from inverted papilloma by fluorescence in situ hybridization, immunohistochemistry, and morphologic analysis
.
Am J Surg Pathol
.
2007
;
31
(
12
):
1861
1867
.
116
Volmar
KE
,
Chan
TY
,
De Marzo
AM
,
Epstein
JI
.
Florid von Brunn nests mimicking urothelial carcinoma: a morphologic and immunohistochemical comparison to the nested variant of urothelial carcinoma
.
Am J Surg Pathol
.
2003
;
27
(
9
):
1243
1252
.
117
Martignoni
G
,
Eble
JN
.
Carcinoid tumors of the urinary bladder: immunohistochemical study of 2 cases and review of the literature
.
Arch Pathol Lab Med
.
2003
;
127
(
1
):
e22
e24
.
118
Lin
O
,
Cardillo
M
,
Dalbagni
G
,
Linkov
I
,
Hutchinson
B
,
Reuter
VE
.
Nested variant of urothelial carcinoma: a clinicopathologic and immunohistochemical study of 12 cases
.
Mod Pathol
.
2003
;
16
(
12
):
1289
1298
.
119
Cintorino
M
,
Del Vecchio
MT
,
Bugnoli
M
,
Petracca
R
,
Leoncini
P
.
Cytokeratin pattern in normal and pathological bladder urothelium: immunohistochemical investigation using monoclonal antibodies
.
J Urol
.
1988
;
139
(
2
):
428
432
.
120
Cheng
L
,
Leibovich
BC
,
Cheville
JC
,
et al
.
Paraganglioma of the urinary bladder: can biologic potential be predicted?
Cancer
.
2000
;
88
(
4
):
844
852
.
121
Kato
H
,
Suzuki
M
,
Mukai
M
,
Aizawa
S
.
Clinicopathological study of pheochromocytoma of the urinary bladder: immunohistochemical, flow cytometric and ultrastructural findings with review of the literature
.
Pathol Int
.
1999
;
49
(
12
):
1093
1099
.
122
Kovacs
K
,
Bell
D
,
Gardiner
GW
,
Honey
RJ
,
Goguen
J
,
Rotondo
F
.
Malignant paraganglioma of the urinary bladder: immunohistochemical study of prognostic indicators
.
Endocr Pathol
.
2005
;
16
(
4
):
363
369
.
123
Grignon
DJ
,
Ro
JY
,
Mackay
B
,
et al
.
Paraganglioma of the urinary bladder: immunohistochemical, ultrastructural, and DNA flow cytometric studies
.
Hum Pathol
.
1991
;
22
(
11
):
1162
1169
.
124
Moyana
TN
,
Kontozoglou
T
.
Urinary bladder paragangliomas: an immunohistochemical study
.
Arch Pathol Lab Med
.
1988
;
112
(
1
):
70
72
.
125
Wasco
MJ
,
Daignault
S
,
Bradley
D
,
Shah
RB
.
Nested variant of urothelial carcinoma: a clinicopathologic and immunohistochemical study of 30 pure and mixed cases
.
Hum Pathol
.
2010
;
41
(
2
):
163
171
.
126
Parker
DC
,
Folpe
AL
,
Bell
J
,
et al
.
Potential utility of uroplakin III, thrombomodulin, high molecular weight cytokeratin, and cytokeratin 20 in noninvasive, invasive, and metastatic urothelial (transitional cell) carcinomas
.
Am J Surg Pathol
.
2003
;
27
(
1
):
1
10
.
127
Wang
HL
,
Lu
DW
,
Yerian
LM
,
et al
.
Immunohistochemical distinction between primary adenocarcinoma of the bladder and secondary colorectal adenocarcinoma
.
Am J Surg Pathol
.
2001
;
25
(
11
):
1380
1387
.
128
Werling
RW
,
Yaziji
H
,
Bacchi
CE
,
Gown
AM
.
CDX2, a highly sensitive and specific marker of adenocarcinomas of intestinal origin: an immunohistochemical survey of 476 primary and metastatic carcinomas
.
Am J Surg Pathol
.
2003
;
27
(
3
):
303
310
.
129
Bates
AW
,
Baithun
SI
.
Secondary neoplasms of the bladder are histological mimics of nontransitional cell primary tumours: clinicopathological and histological features of 282 cases
.
Histopathology
.
2000
;
36
(
1
):
32
40
.
130
Grignon
DJ
,
Ro
JY
,
Ayala
AG
,
Johnson
DE
,
Ordonez
NG
.
Primary adenocarcinoma of the urinary bladder: a clinicopathologic analysis of 72 cases
.
Cancer
.
1991
;
67
(
8
):
2165
2172
.
131
Tamboli
P
,
Mohsin
SK
,
Hailemariam
S
,
Amin
MB
.
Colonic adenocarcinoma metastatic to the urinary tract versus primary tumors of the urinary tract with glandular differentiation: a report of 7 cases and investigation using a limited immunohistochemical panel
.
Arch Pathol Lab Med
.
2002
;
126
(
9
):
1057
1063
.
132
Sim
SJ
,
Ro
JY
,
Ordonez
NG
,
Park
YW
,
Kee
KH
,
Ayala
AG
.
Metastatic renal cell carcinoma to the bladder: a clinicopathologic and immunohistochemical study
.
Mod Pathol
.
1999
;
12
(
4
):
351
355
.
133
Chuang
AY
,
DeMarzo
AM
,
Veltri
RW
,
Sharma
RB
,
Bieberich
CJ
,
Epstein
JI
.
Immunohistochemical differentiation of high-grade prostate carcinoma from urothelial carcinoma
.
Am J Surg Pathol
.
2007
;
31
(
8
):
1246
1255
.
134
Kaimaktchiev
V
,
Terracciano
L
,
Tornillo
L
,
et al
.
The homeobox intestinal differentiation factor CDX2 is selectively expressed in gastrointestinal adenocarcinomas
.
Mod Pathol
.
2004
;
17
(
11
):
1392
1399
.
135
Epstein
JI
,
Kuhajda
FP
,
Lieberman
PH
.
Prostate-specific acid phosphatase immunoreactivity in adenocarcinomas of the urinary bladder
.
Hum Pathol
.
1986
;
17
:
939
942
.
136
Chu
PG
,
Chung
L
,
Weiss
LM
,
Lau
SK
.
Determining the site of origin of mucinous adenocarcinoma: an immunohistochemical study of 175 cases
.
Am J Surg Pathol
.
2011
;
35
(
9
):
1830
1836
.
137
Rao
Q
,
Williamson
SR
,
Lopez-Beltran
A
,
et al
.
Distinguishing primary adenocarcinoma of the urinary bladder from secondary involvement by colorectal adenocarcinoma: extended immunohistochemical profiles emphasizing novel markers
.
Mod Pathol
.
2013
;
26
(
5
):
725
732
.
138
Chang
A
,
Amin
A
,
Gabrielson
E
,
et al
.
Utility of GATA3 immunohistochemistry in differentiating urothelial carcinoma from prostate adenocarcinoma and squamous cell carcinomas of the uterine cervix, anus, and lung
.
Am J Surg Pathol
.
2012
;
36
(
10
):
1472
1476
.
139
Ikegami
H
,
Iwasaki
H
,
Ohjimi
Y
,
Takeuchi
T
,
Ariyoshi
A
,
Kikuchi
M
.
Sarcomatoid carcinoma of the urinary bladder: a clinicopathologic and immunohistochemical analysis of 14 patients
.
Hum Pathol
.
2000
;
31
(
3
):
332
340
.
140
Westfall
DE
,
Folpe
AL
,
Paner
GP
,
et al
.
Utility of a comprehensive immunohistochemical panel in the differential diagnosis of spindle cell lesions of the urinary bladder
.
Am J Surg Pathol
.
2009
;
33
(
1
):
99
105
.
141
Torenbeek
R
,
Blomjous
CE
,
de Bruin
PC
,
Newling
DW
,
Meijer
CJ
.
Sarcomatoid carcinoma of the urinary bladder: clinicopathologic analysis of 18 cases with immunohistochemical and electron microscopic findings
.
Am J Surg Pathol
.
1994
;
18
(
3
):
241
249
.
142
Freeman
A
,
Geddes
N
,
Munson
P
,
et al
.
Anaplastic lymphoma kinase (ALK 1) staining and molecular analysis in inflammatory myofibroblastic tumours of the bladder: a preliminary clinicopathological study of nine cases and review of the literature
.
Mod Pathol
.
2004
;
17
(
7
):
765
771
.
143
Montgomery
EA
,
Shuster
DD
,
Burkart
AL
,
et al
.
Inflammatory myofibroblastic tumors of the urinary tract: a clinicopathologic study of 46 cases, including a malignant example inflammatory fibrosarcoma and a subset associated with high-grade urothelial carcinoma
.
Am J Surg Pathol
.
2006
;
30
(
12
):
1502
1512
.
144
Cessna
MH
,
Zhou
H
,
Sanger
WG
,
et al
.
Expression of ALK1 and p80 in inflammatory myofibroblastic tumor and its mesenchymal mimics: a study of 135 cases
.
Mod Pathol
.
2002
;
15
:
931
938
.
145
Alquati
S
,
Gira
FA
,
Bartoli
V
,
Contini
S
,
Corradi
D
.
Low-grade myofibroblastic proliferations of the urinary bladder
.
Arch Pathol Lab Med
.
2013
;
137
(
8
):
1117
1128
.
146
Bauer
JL
,
Miklos
AZ
,
Thompson
LD
.
Parotid gland solitary fibrous tumor: a case report and clinicopathologic review of 22 cases from the literature
.
Head Neck Pathol
.
2012
;
6
(
1
):
21
31
.
147
Martin
SE
,
Temm
CJ
,
Goheen
MP
,
Ulbright
TM
,
Hattab
EM
.
Cytoplasmic p63 immunohistochemistry is a useful marker for muscle differentiation: an immunohistochemical and immunoelectron microscopic study
.
Mod Pathol
.
2011
;
24
(
10
):
1320
1326
.
148
Heerema-McKenney
A
,
Wijnaendts
LC
,
Pulliam
JF
,
et al
.
Diffuse myogenin expression by immunohistochemistry is an independent marker of poor survival in pediatric rhabdomyosarcoma: a tissue microarray study of 71 primary tumors including correlation with molecular phenotype
.
Am J Surg Pathol
.
2008
;
32
(
10
):
1513
1522
.
149
Folpe
AL
.
MyoD1 and myogenin expression in human neoplasia: a review and update
.
Adv Anat Pathol
.
2002
;
9
(
3
):
198
203
.
150
Stock
N
,
Chibon
F
,
Binh
MB
,
et al
.
Adult-type rhabdomyosarcoma: analysis of 57 cases with clinicopathologic description, identification of 3 morphologic patterns and prognosis
.
Am J Surg Pathol
.
2009
;
33
(
12
):
1850
1859
.
151
Epstein
JI
.
Diagnosis of limited adenocarcinoma of the prostate
.
Histopathology
.
2012
;
60
(
1
):
28
40
.
152
Signoretti
S
,
Waltregny
D
,
Dilks
J
,
et al
.
p63 is a prostate basal cell marker and is required for prostate development
.
Am J Pathol
.
2000
;
157
:
1769
1775
.
153
Parsons
JK
,
Gage
WR
,
Nelson
WG
,
De Marzo
AM
.
p63 protein expression is rare in prostate adenocarcinoma: implications for cancer diagnosis and carcinogenesis
.
Urology
.
2001
;
58
(
4
):
619
624
.
154
Hameed
O
,
Humphrey
PA
.
Immunohistochemistry in diagnostic surgical pathology of the prostate
.
Semin Diagn Pathol
.
2005
;
22
(
1
):
88
104
.
155
Weinstein
MH
,
Signoretti
S
,
Loda
M
.
Diagnostic utility of immunohistochemical staining for p63, a sensitive marker of prostatic basal cells
.
Mod Pathol
.
2002
;
15
(
12
):
1302
1308
.
156
Zhou
M
,
Shah
R
,
Shen
R
,
Rubin
MA
.
Basal cell cocktail (34betaE12 + p63) improves the detection of prostate basal cells
.
Am J Surg Pathol
.
2003
;
27
(
3
):
365
371
.
157
Paner
GP
,
Luthringer
DJ
,
Amin
MB
.
Best practice in diagnostic immunohistochemistry: prostate carcinoma and its mimics in needle core biopsies
.
Arch Pathol Lab Med
.
2008
;
132
(
9
):
1388
1396
.
158
Osunkoya
AO
,
Hansel
DE
,
Sun
X
,
Netto
GJ
,
Epstein
JI
.
Aberrant diffuse expression of p63 in adenocarcinoma of the prostate on needle biopsy and radical prostatectomy: report of 21 cases
.
Am J Surg Pathol
.
2008
;
32
(
3
):
461
467
.
159
Baydar
DE
,
Kulac
I
,
Gurel
B
,
De Marzo
A
.
A case of prostatic adenocarcinoma with aberrant p63 expression: presentation with detailed immunohistochemical study and FISH analysis
.
Int J Surg Pathol
.
2011
;
19
(
1
):
131
136
.
160
Giannico
GA
,
Ross
HM
,
Lotan
T
,
Epstein
JI
.
Aberrant expression of p63 in adenocarcinoma of the prostate: a radical prostatectomy study
.
Am J Surg Pathol
.
2013
;
37
(
9
):
1401
1406
.
161
Oliai
BR
,
Kahane
H
,
Epstein
JI
.
Can basal cells be seen in adenocarcinoma of the prostate: an immunohistochemical study using high molecular weight cytokeratin (clone 34betaE12) antibody
.
Am J Surg Pathol
.
2002
;
26
(
9
):
1151
1160
.
162
Varma
M
,
Jasani
B
.
Diagnostic utility of immunohistochemistry in morphologically difficult prostate cancer: review of current literature
.
Histopathology
.
2005
;
47
(
1
):
1
16
.
163
Gown
AM
,
Vogel
AM
.
Monoclonal antibodies to human intermediate filament proteins, II: distribution of filament proteins in normal human tissues
.
Am J Pathol
.
1984
;
114
(
2
):
309
321
.
164
Brawer
MK
,
Peehl
DM
,
Stamey
TA
,
Bostwick
DG
.
Keratin immunoreactivity in the benign and neoplastic human prostate
.
Cancer Res
.
1985
;
45
(
8
):
3663
3667
.
165
Hedrick
L
,
Epstein
JI
.
Use of keratin 903 as an adjunct in the diagnosis of prostate carcinoma
.
Am J Surg Pathol
.
1989
;
13
(
5
):
389
396
.
166
Googe
PB
,
McGinley
KM
,
Fitzgibbon
JF
.
Anticytokeratin antibody 34 beta E12 staining in prostate carcinoma
.
Am J Clin Pathol
.
1997
;
107
(
2
):
219
223
.
167
Yang
XJ
,
Lecksell
K
,
Gaudin
P
,
Epstein
JI
.
Rare expression of high-molecular-weight cytokeratin in adenocarcinoma of the prostate gland: a study of 100 cases of metastatic and locally advanced prostate cancer
.
Am J Surg Pathol
.
1999
;
23
(
2
):
147
152
.
168
Wojno
KJ
,
Epstein
JI
.
The utility of basal cell-specific anti-cytokeratin antibody (34 beta E12) in the diagnosis of prostate cancer: a review of 228 cases
.
Am J Surg Pathol
.
1995
;
19
(
3
):
251
260
.
169
Abrahams
NA
,
Ormsby
AH
,
Brainard
J
.
Validation of cytokeratin 5/6 as an effective substitute for keratin 903 in the differentiation of benign from malignant glands in prostate needle biopsies
.
Histopathology
.
2002
;
41
:
35
41
.
170
Abrahams
NA
,
Bostwick
DG
,
Ormsby
AH
,
Qian
J
,
Brainard
JA
.
Distinguishing atrophy and high-grade prostatic intraepithelial neoplasia from prostatic adenocarcinoma with and without previous adjuvant hormone therapy with the aid of cytokeratin 5/6
.
Am J Clin Pathol
.
2003
;
120
(
3
):
368
376
.
171
Xu
J
,
Stolk
JA
,
Zhang
X
,
et al
.
Identification of differentially expressed genes in human prostate cancer using subtraction and microarray
.
Cancer Res
.
2000
;
60
(
6
):
1677
1682
.
172
Jiang
Z
,
Woda
BA
,
Rock
KL
,
et al
.
P504S: a new molecular marker for the detection of prostate carcinoma
.
Am J Surg Pathol
.
2001
;
25
(
11
):
1397
1404
.
173
Liu
H
,
Lin
F
,
Zhai
Q
.
Prostate gland
.
In
:
Lin
F
,
Prichard
JW
,
Liu
H
,
Wilkerson
ML
,
Schuerch
C
,
eds
.
Handbook of Practical Immunohistochemistry: Frequently Asked Questions
.
New York, NY
:
Springer;
2011
:
299
319
.
174
Luo
J
,
Zha
S
,
Gage
WR
,
et al
.
Alpha-methylacyl-CoA racemase: a new molecular marker for prostate cancer
.
Cancer Res
.
2002
;
62
(
8
):
2220
2226
.
175
Rubin
MA
,
Zhou
M
,
Dhanasekaran
SM
,
et al
.
alpha-Methylacyl coenzyme A racemase as a tissue biomarker for prostate cancer
.
JAMA
.
2002
;
287
(
13
):
1662
1670
.
176
Zhou
M
,
Chinnaiyan
AM
,
Kleer
CG
,
Lucas
PC
,
Rubin
MA
.
Alpha-Methylacyl-CoA racemase: a novel tumor marker over-expressed in several human cancers and their precursor lesions
.
Am J Surg Pathol
.
2002
;
26
(
7
):
926
931
.
177
Evans
AJ
.
Alpha-methylacyl CoA racemase (P504S): overview and potential uses in diagnostic pathology as applied to prostate needle biopsies
.
J Clin Pathol
.
2003
;
56
(
12
):
892
897
.
178
Zhou
M
,
Aydin
H
,
Kanane
H
,
Epstein
JI
.
How often does alpha-methylacyl-CoA-racemase contribute to resolving an atypical diagnosis on prostate needle biopsy beyond that provided by basal cell markers?
Am J Surg Pathol
.
2004
;
28
(
2
):
239
243
.
179
Jiang
Z
,
Woda
BA
,
Wu
CL
,
Yang
XJ
.
Discovery and clinical application of a novel prostate cancer marker: alpha-methylacyl CoA racemase (P504S)
.
Am J Clin Pathol
.
2004
;
122
(
2
):
275
289
.
180
Zhou
M
,
Jiang
Z
,
Epstein
JI
.
Expression and diagnostic utility of alpha-methylacyl-CoA-racemase (P504S) in foamy gland and pseudohyperplastic prostate cancer
.
Am J Surg Pathol
.
2003
;
27
(
6
):
772
778
.
181
Beach
R
,
Gown
AM
,
De Peralta-Venturina
MN
,
et al
.
P504S immunohistochemical detection in 405 prostatic specimens including 376 18-gauge needle biopsies
.
Am J Surg Pathol
.
2002
;
26
(
12
):
1588
1596
.
182
Farinola
MA
,
Epstein
JI
.
Utility of immunohistochemistry for alpha-methylacyl-CoA racemase in distinguishing atrophic prostate cancer from benign atrophy
.
Hum Pathol
.
2004
;
35
(
10
):
1272
1278
.
183
Amin
MB
,
Beach
R
,
Gown
AM
.
Use of a novel immunohistochemical (IHC) panel (P504S, p63 and 34betaE12) in the diagnosis of post-radiation therapy (PRT) prostate cancer (PCa)
.
Mod Pathol
.
2003
;
16
(
1S
):
139A
.
184
Yang
XJ
,
Laven
B
,
Tretiakova
M
,
et al
.
Detection of alpha-methylacyl-coenzyme A racemase in postradiation prostatic adenocarcinoma
.
Urology
.
2003
;
62
(
2
):
282
286
.
185
Wang
W
,
Sun
X
,
Epstein
JI
.
Partial atrophy on prostate needle biopsy cores: a morphologic and immunohistochemical study
.
Am J Surg Pathol
.
2008
;
32
(
6
):
851
857
.
186
Herawi
M
,
Parwani
AV
,
Irie
J
,
Epstein
JI
.
Small glandular proliferations on needle biopsies: most common benign mimickers of prostatic adenocarcinoma sent in for expert second opinion
.
Am J Surg Pathol
.
2005
;
29
(
7
):
874
880
.
187
Yang
XJ
,
Wu
CL
,
Woda
BA
,
et al
.
Expression of alpha-methylacyl-CoA racemase (P504S) in atypical adenomatous hyperplasia of the prostate
.
Am J Surg Pathol
.
2002
;
26
(
7
):
921
925
.
188
Gupta
A
,
Wang
HL
,
Policarpio-Nicolas
ML
,
et al
.
Expression of alpha-methylacyl-coenzyme A racemase in nephrogenic adenoma
.
Am J Surg Pathol
.
2004
;
28
(
9
):
1224
1229
.
189
Cossu-Rocca
P
,
Contini
M
,
Brunelli
M
,
et al
.
S-100A1 is a reliable marker in distinguishing nephrogenic adenoma from prostatic adenocarcinoma
.
Am J Surg Pathol
.
2009
;
33
(
7
):
1031
1036
.
190
Alexiev
BA
,
LeVea
CM
.
Nephrogenic adenoma of the urinary tract: a review
.
Int J Surg Pathol
.
2012
;
20
(
2
):
123
131
.
191
Browne
TJ
,
Hirsch
MS
,
Brodsky
G
,
Welch
WR
,
Loda
MF
,
Rubin
MA
.
Prospective evaluation of AMACR (P504S) and basal cell markers in the assessment of routine prostate needle biopsy specimens
.
Hum Pathol
.
2004
;
35
(
12
):
1462
1468
.
192
Miettinen
M
,
Wang
ZF
,
Paetau
A
,
et al
.
ERG transcription factor as an immunohistochemical marker for vascular endothelial tumors and prostatic carcinoma
.
Am J Surg Pathol
.
2011
;
35
(
3
):
432
441
.
193
Furusato
B
,
Tan
SH
,
Young
D
,
et al
.
ERG oncoprotein expression in prostate cancer: clonal progression of ERG-positive tumor cells and potential for ERG-based stratification
.
Prostate Cancer Prostatic Dis
.
2010
;
13
(
3
):
228
237
.
194
Liu
H
,
Shi
J
,
Wilkerson
M
,
Yang
XJ
,
Lin
F
.
Immunohistochemical evaluation of ERG expression in various benign and malignant tissues
.
Ann Clin Lab Sci
.
2013
;
43
(
1
):
3
9
.
195
Chaux
A
,
Albadine
R
,
Toubaji
A
,
et al
.
Immunohistochemistry for ERG expression as a surrogate for TMPRSS2-ERG fusion detection in prostatic adenocarcinomas
.
Am J Surg Pathol
.
2011
;
35
(
7
):
1014
1020
.
196
Yaskiv
O
,
Zhang
X
,
Simmerman
K
,
et al
.
The utility of ERG/P63 double immunohistochemical staining in the diagnosis of limited cancer in prostate needle biopsies
.
Am J Surg Pathol
.
2011
;
35
(
7
):
1062
1068
.
197
Scheble
VJ
,
Braun
M
,
Beroukhim
R
,
et al
.
ERG rearrangement is specific to prostate cancer and does not occur in any other common tumor
.
Mod Pathol
.
2010
;
23
(
8
):
1061
1067
.
198
Bodey
B
,
Bodey
B
Jr,
Kaiser
HE
.
Immunocytochemical detection of prostate specific antigen expression in human primary and metastatic melanomas
.
Anticancer Res
.
1997
;
17
(
3C
):
2343
2346
.
199
Bodey
B
,
Bodey
B
Jr,
Kaiser
HE
.
Immunocytochemical detection of prostate specific antigen expression in human breast carcinoma cells
.
Anticancer Res
.
1997
;
17
(
4A
):
2577
2581
.
200
Hall
RE
,
Clements
JA
,
Birrell
SN
,
Tilley
WD
.
Prostate-specific antigen and gross cystic disease fluid protein-15 are co-expressed in androgen receptor-positive breast tumours
.
Br J Cancer
.
1998
;
78
(
3
):
360
365
.
201
James
GK
,
Pudek
M
,
Berean
KW
,
Diamandis
EP
,
Archibald
BL
.
Salivary duct carcinoma secreting prostate-specific antigen
.
Am J Clin Pathol
.
1996
;
106
(
2
):
242
247
.
202
Fan
CY
,
Wang
J
,
Barnes
EL
.
Expression of androgen receptor and prostatic specific markers in salivary duct carcinoma: an immunohistochemical analysis of 13 cases and review of the literature
.
Am J Surg Pathol
.
2000
;
24
(
4
):
579
586
.
203
Varma
M
,
Morgan
M
,
Jasani
B
,
Tamboli
P
,
Amin
MB
.
Polyclonal anti-PSA is more sensitive but less specific than monoclonal anti-PSA: implications for diagnostic prostatic pathology
.
Am J Clin Pathol
.
2002
;
118
(
2
):
202
207
.
204
Goldstein
NS
.
Immunophenotypic characterization of 225 prostate adenocarcinomas with intermediate or high Gleason scores
.
Am J Clin Pathol
.
2002
;
117
(
3
):
471
477
.
205
Xu
J
,
Kalos
M
,
Stolk
JA
,
et al
.
Identification and characterization of prostein, a novel prostate-specific protein
.
Cancer Res
.
2001
;
61
(
4
):
1563
1568
.
206
Kalos
M
,
Askaa
J
,
Hylander
BL
,
et al
.
Prostein expression is highly restricted to normal and malignant prostate tissues
.
Prostate
.
2004
;
60
(
3
):
246
256
.
207
Sheridan
T
,
Herawi
M
,
Epstein
JI
,
Illei
PB
.
The role of P501S and PSA in the diagnosis of metastatic adenocarcinoma of the prostate
.
Am J Surg Pathol
.
2007
;
31
(
9
):
1351
1355
.
208
Yin
M
,
Dhir
R
,
Parwani
AV
.
Diagnostic utility of p501s (prostein) in comparison to prostate specific antigen (PSA) for the detection of metastatic prostatic adenocarcinoma
.
Diagn Pathol
.
2007
;
2
(
1
):
41
.
209
Gurel
B
,
Ali
TZ
,
Montgomery
EA
,
et al
.
NKX3.1 as a marker of prostatic origin in metastatic tumors
.
Am J Surg Pathol
.
2010
;
34
(
8
):
1097
1105
.
210
Gelmann
EP
,
Bowen
C
,
Bubendorf
L
.
Expression of NKX3.1 in normal and malignant tissues
.
Prostate
.
2003
;
55
(
2
):
111
117
.
211
Ornstein
DK
,
Cinquanta
M
,
Weiler
S
,
et al
.
Expression studies and mutational analysis of the androgen regulated homeobox gene NKX3.1 in benign and malignant prostate epithelium
.
J Urol
.
2001
;
165
(
4
):
1329
1334
.
212
Ye
H
,
Ulbright
TM
.
Difficult differential diagnoses in testicular pathology
.
Arch Pathol Lab Med
.
2012
;
136
(
4
):
435
446
.
213
Wilkerson
M
,
Lin
F
,
Shi
J
.
NANOG immunohistochemical expression in tumors
.
Mod Pathol
.
2012
;
25
(
S2
):
251A
.
214
Bahrami
A
,
Ro
JY
,
Ayala
AG
.
An overview of testicular germ cell tumors
.
Arch Pathol Lab Med
.
2007
;
131
(
8
):
1267
1280
.
215
Cao
D
,
Li
J
,
Guo
CC
,
Allan
RW
,
Humphrey
PA
.
SALL4 is a novel diagnostic marker for testicular germ cell tumors
.
Am J Surg Pathol
.
2009
;
33
(
7
):
1065
1077
.
216
Dekker
I
,
Rozeboom
T
,
Delemarre
J
,
Dam
A
,
Oosterhuis
JW
.
Placental-like alkaline phosphatase and DNA flow cytometry in spermatocytic seminoma
.
Cancer
.
1992
;
69
(
4
):
993
996
.
217
Emerson
RE
,
Ulbright
TM
.
Morphological approach to tumours of the testis and paratestis
.
J Clin Pathol
.
2007
;
60
(
8
):
866
880
.
218
Gopalan
A
,
Dhall
D
,
Olgac
S
,
et al
.
Testicular mixed germ cell tumors: a morphological and immunohistochemical study using stem cell markers, OCT3/4, SOX2 and GDF3, with emphasis on morphologically difficult-to-classify areas
.
Mod Pathol
.
2009
;
22
(
8
):
1066
1074
.
219
Jones
TD
,
Ulbright
TM
,
Eble
JN
,
Cheng
L
.
OCT4: a sensitive and specific biomarker for intratubular germ cell neoplasia of the testis
.
Clin Cancer Res
.
2004
;
10
(
24
):
8544
8547
.
220
Mostofi
FK
,
Sesterhenn
IA
,
Davis
CJ
Jr.
Immunopathology of germ cell tumors of the testis
.
Semin Diagn Pathol
.
1987
;
4
(
4
):
320
341
.
221
Tickoo
SK
,
Hutchinson
B
,
Bacik
J
,
et al
.
Testicular seminoma: a clinicopathologic and immunohistochemical study of 105 cases with special reference to seminomas with atypical features
.
Int J Surg Pathol
.
2002
;
10
(
1
):
23
32
.
222
de Jong
J
,
Stoop
H
,
Dohle
GR
,
et al
.
Diagnostic value of OCT3/4 for pre-invasive and invasive testicular germ cell tumours
.
J Pathol
.
2005
;
206
(
2
):
242
249
.
223
Caillaud
JM
,
Bellet
D
,
Carlu
C
,
Droz
JP
.
Immunohistochemistry of germ cell tumors of the testis: study of beta HCG and AFP
.
Prog Clin Biol Res
.
1985
;
203
:
139
140
.
224
Burke
AP
,
Mostofi
FK
.
Placental alkaline phosphatase immunohistochemistry of intratubular malignant germ cells and associated testicular germ cell tumors
.
Hum Pathol
.
1988
;
19
(
6
):
663
670
.
225
Burke
AP
,
Mostofi
FK
.
Intratubular malignant germ cells in testicular biopsies: clinical course and identification by staining for placental alkaline phosphatase
.
Mod Pathol
.
1988
;
1
(
6
):
475
479
.
226
Bailey
D
,
Marks
A
,
Stratis
M
,
Baumal
R
.
Immunohistochemical staining of germ cell tumors and intratubular malignant germ cells of the testis using antibody to placental alkaline phosphatase and a monoclonal anti-seminoma antibody
.
Mod Pathol
.
1991
;
4
(
2
):
167
171
.
227
Bosman
FT
,
Giard
RW
,
Nieuwenhuijen Kruseman AC, Knijnenburg G, Spaander PJ. Human chorionic gonadotrophin and alpha-fetoprotein in testicular germ cell tumours: a retrospective immunohistochemical study
.
Histopathology
.
1980
;
4
(
6
):
673
684
.
228
Bomeisl
PE
,
MacLennan
GT
.
Spermatocytic seminoma
.
J Urol
.
2007
;
177
(
2
):
734
.
229
Battifora
H
,
Sheibani
K
,
Tubbs
RR
,
Kopinski
MI
,
Sun
TT
.
Antikeratin antibodies in tumor diagnosis: distinction between seminoma and embryonal carcinoma
.
Cancer
.
1984
;
54
(
5
):
843
848
.
230
Bartkova
J
,
Rejthar
A
,
Bartek
J
,
Kovarik
J
.
Differentiation patterns of testicular germ-cell tumours as revealed by a panel of monoclonal antibodies
.
Tumour Biol
.
1987
;
8
(
1
):
45
56
.
231
Wittekind
C
,
Wichmann
T
,
Von Kleist
S
.
Immunohistological localization of AFP and HCG in uniformly classified testis tumors
.
Anticancer Res
.
1983
;
3
(
5
):
327
330
.
232
Wick
MR
,
Swanson
PE
,
Manivel
JC
.
Placental-like alkaline phosphatase reactivity in human tumors: an immunohistochemical study of 520 cases
.
Hum Pathol
.
1987
;
18
(
9
):
946
954
.
233
Rajpert-De Meyts
E
,
Kvist
M
,
Skakkebaek
NE
.
Heterogeneity of expression of immunohistochemical tumour markers in testicular carcinoma in situ: pathogenetic relevance
.
Virchows Arch
.
1996
;
428
(
3
):
133
139
.
234
Pallesen
G
,
Hamilton-Dutoit
SJ
.
Ki-1 (CD30) antigen is regularly expressed by tumor cells of embryonal carcinoma
.
Am J Pathol
.
1988
;
133
(
3
):
446
450
.
235
Niehans
GA
,
Manivel
JC
,
Copland
GT
,
Scheithauer
BW
,
Wick
MR
.
Immunohistochemistry of germ cell and trophoblastic neoplasms
.
Cancer
.
1988
;
62
(
6
):
1113
1123
.
236
Manivel
JC
,
Jessurun
J
,
Wick
MR
,
Dehner
LP
.
Placental alkaline phosphatase immunoreactivity in testicular germ-cell neoplasms
.
Am J Surg Pathol
.
1987
;
11
(
1
):
21
29
.
237
Manivel
JC
,
Simonton
S
,
Wold
LE
,
Dehner
LP
.
Absence of intratubular germ cell neoplasia in testicular yolk sac tumors in children: a histochemical and immunohistochemical study
.
Arch Pathol Lab Med
.
1988
;
112
(
6
):
641
645
.
238
Eglen
DE
,
Ulbright
TM
.
The differential diagnosis of yolk sac tumor and seminoma: usefulness of cytokeratin, alpha-fetoprotein, and alpha-1-antitrypsin immunoperoxidase reactions
.
Am J Clin Pathol
.
1987
;
88
(
3
):
328
332
.
239
Leroy
X
,
Augusto
D
,
Leteurtre
E
,
Gosselin
B
.
CD30 and CD117 (c-kit) used in combination are useful for distinguishing embryonal carcinoma from seminoma
.
J Histochem Cytochem
.
2002
;
50
(
2
):
283
285
.
240
Kraggerud
SM
,
Berner
A
,
Bryne
M
,
Pettersen
EO
,
Fossa
SD
.
Spermatocytic seminoma as compared to classical seminoma: an immunohistochemical and DNA flow cytometric study
.
APMIS
.
1999
;
107
(
3
):
297
302
.
241
Koshida
K
,
Wahren
B
.
Placental-like alkaline phosphatase in seminoma
.
Urol Res
.
1990
;
18
(
2
):
87
92
.
242
Cummings
OW
,
Ulbright
TM
,
Eble
JN
,
Roth
LM
.
Spermatocytic seminoma: an immunohistochemical study
.
Hum Pathol
.
1994
;
25
(
1
):
54
59
.
243
Jacobsen
GK
,
Norgaard-Pedersen
B
.
Placental alkaline phosphatase in testicular germ cell tumours and in carcinoma-in-situ of the testis: an immunohistochemical study
.
Acta Pathol Microbiol Immunol Scand A
.
1984
;
92
(
5
):
323
329
.
244
Hustin
J
,
Collette
J
,
Franchimont
P
.
Immunohistochemical demonstration of placental alkaline phosphatase in various states of testicular development and in germ cell tumours
.
Int J Androl
.
1987
;
10
(
1
):
29
35
.
245
Hittmair
A
,
Rogatsch
H
,
Hobisch
A
,
Mikuz
G
,
Feichtinger
H
.
CD30 expression in seminoma
.
Hum Pathol
.
1996
;
27
(
11
):
1166
1171
.
246
Kalhor
N
,
Ramirez
PT
,
Deavers
MT
,
Malpica
A
,
Silva
EG
.
Immunohistochemical studies of trophoblastic tumors
.
Am J Surg Pathol
.
2009
;
33
(
4
):
633
638
.
247
Ota
S
,
Hishinuma
M
,
Yamauchi
N
,
et al
.
Oncofetal protein glypican-3 in testicular germ-cell tumor
.
Virchows Arch
.
2006
;
449
(
3
):
308
314
.
248
Visfeldt
J
,
Jorgensen
N
,
Muller
J
,
Moller
H
,
Skakkebaek
NE
.
Testicular germ cell tumours of childhood in Denmark, 1943–1989: incidence and evaluation of histology using immunohistochemical techniques
.
J Pathol
.
1994
;
174
(
1
):
39
47
.
249
Kersemaekers
AM
,
Honecker
F
,
Stoop
H
,
et al
.
Identification of germ cells at risk for neoplastic transformation in gonadoblastoma: an immunohistochemical study for OCT3/4 and TSPY
.
Hum Pathol
.
2005
;
36
(
5
):
512
521
.
250
Zynger
DL
,
McCallum
JC
,
Luan
C
,
Chou
PM
,
Yang
XJ
.
Glypican 3 has a higher sensitivity than alpha-fetoprotein for testicular and ovarian yolk sac tumour: immunohistochemical investigation with analysis of histological growth patterns
.
Histopathology
.
2010
;
56
(
6
):
750
757
.
251
Looijenga
LH
,
Stoop
H
,
de Leeuw
HP
,
et al
.
POU5F1 (OCT3/4) identifies cells with pluripotent potential in human germ cell tumors
.
Cancer Res
.
2003
;
63
(
9
):
2244
2250
.
252
Hoei-Hansen
CE
,
Almstrup
K
,
Nielsen
JE
,
et al
.
Stem cell pluripotency factor NANOG is expressed in human fetal gonocytes, testicular carcinoma in situ and germ cell tumours
.
Histopathology
.
2005
;
47
(
1
):
48
56
.
253
Hart
AH
,
Hartley
L
,
Parker
K
,
et al
.
The pluripotency homeobox gene NANOG is expressed in human germ cell tumors
.
Cancer
.
2005
;
104
(
10
):
2092
2098
.
254
Zynger
DL
,
Dimov
ND
,
Luan
C
,
Teh
BT
,
Yang
XJ
.
Glypican 3: a novel marker in testicular germ cell tumors
.
Am J Surg Pathol
.
2006
;
30
(
12
):
1570
1575
.
255
Ezeh
UI
,
Turek
PJ
,
Reijo
RA
,
Clark
AT
.
Human embryonic stem cell genes OCT4, NANOG, STELLAR, and GDF3 are expressed in both seminoma and breast carcinoma
.
Cancer
.
2005
;
104
(
10
):
2255
2265
.
256
Emerson
RE
,
Ulbright
TM
.
The use of immunohistochemistry in the differential diagnosis of tumors of the testis and paratestis
.
Semin Diagn Pathol
.
2005
;
22
(
1
):
33
50
.
257
Wilkerson
M
,
Lin
F
,
Shi
J
.
SF-1 immunohistochemical expressio in tumors
.
Mod Pathol
.
2013
;
26
(
S2
):
230A
.
258
Devouassoux-Shisheboran
M
,
Deschildre
C
,
Mauduit
C
,
et al
.
Expression of galectin-3 in gonads and gonadal sex cord stromal and germ cell tumors
.
Oncol Rep
.
2006
;
16
(
2
):
335
340
.
259
Iczkowski
KA
,
Bostwick
DG
,
Roche
PC
,
Cheville
JC
.
Inhibin A is a sensitive and specific marker for testicular sex cord-stromal tumors
.
Mod Pathol
.
1998
;
11
(
8
):
774
779
.
260
Groisman
GM
,
Dische
MR
,
Fine
EM
,
Unger
PD
.
Juvenile granulosa cell tumor of the testis: a comparative immunohistochemical study with normal infantile gonads
.
Pediatr Pathol
.
1993
;
13
(
4
):
389
400
.
261
McCluggage
WG
,
Shanks
JH
,
Whiteside
C
,
Maxwell
P
,
Banerjee
SS
,
Biggart
JD
.
Immunohistochemical study of testicular sex cord-stromal tumors, including staining with anti-inhibin antibody
.
Am J Surg Pathol
.
1998
;
22
(
5
):
615
619
.
262
Nielsen
K
,
Jacobsen
GK
.
Malignant Sertoli cell tumour of the testis: an immunohistochemical study and a review of the literature
.
APMIS
.
1988
;
96
(
8
):
755
760
.
263
Sasano
H
,
Nakashima
N
,
Matsuzaki
O
,
et al
.
Testicular sex cord-stromal lesions: immunohistochemical analysis of cytokeratin, vimentin and steroidogenic enzymes
.
Virchows Arch A Pathol Anat Histopathol
.
1992
;
421
(
2
):
163
169
.
264
Sato
K
,
Ueda
Y
,
Sakurai
A
,
et al
.
Large cell calcifying Sertoli cell tumor of the testis: comparative immunohistochemical study with Leydig cell tumor
.
Pathol Int
.
2005
;
55
(
6
):
366
371
.
265
Kommoss
F
,
Oliva
E
,
Bittinger
F
,
et al
.
Inhibin-alpha CD99, HEA125, PLAP, and chromogranin immunoreactivity in testicular neoplasms and the androgen insensitivity syndrome
.
Hum Pathol
.
2000
;
31
(
9
):
1055
1061
.
266
Cao
QJ
,
Jones
JG
,
Li
M
.
Expression of calretinin in human ovary, testis, and ovarian sex cord-stromal tumors
.
Int J Gynecol Pathol
.
2001
;
20
(
4
):
346
352
.
267
Al-Agha
OM
,
Axiotis
CA
.
An in-depth look at Leydig cell tumor of the testis
.
Arch Pathol Lab Med
.
2007
;
131
(
2
):
311
317
.
268
Jimenez-Quintero
LP
,
Ro
JY
,
Zavala-Pompa
A
,
et al
.
Granulosa cell tumor of the adult testis: a clinicopathologic study of seven cases and a review of the literature
.
Hum Pathol
.
1993
;
24
(
10
):
1120
1125
.
269
Al-Bozom
IA
,
El-Faqih
SR
,
Hassan
SH
,
El-Tiraifi
AE
,
Talic
RF
.
Granulosa cell tumor of the adult type: a case report and review of the literature of a very rare testicular tumor
.
Arch Pathol Lab Med
.
2000
;
124
(
10
):
1525
1528
.
270
Ventura
T
,
Discepoli
S
,
Coletti
G
,
et al
.
Light microscopic, immunocytochemical and ultrastructural study of a case of Sertoli cell tumor of the testis
.
Tumori
.
1987
;
73
(
6
):
649
653
.
271
Perez-Atayde
AR
,
Joste
N
,
Mulhern
H
.
Juvenile granulosa cell tumor of the infantile testis: evidence of a dual epithelial-smooth muscle differentiation
.
Am J Surg Pathol
.
1996
;
20
(
1
):
72
79
.
272
Nistal
M
,
Lazaro
R
,
Garcia
J
,
Paniagua
R
.
Testicular granulosa cell tumor of the adult type
.
Arch Pathol Lab Med
.
1992
;
116
(
3
):
284
287
.
273
Morgan
DR
,
Brame
KG
.
Granulosa cell tumour of the testis displaying immunoreactivity for inhibin
.
BJU Int
.
1999
;
83
(
6
):
731
732
.
274
Matias-Guiu
X
,
Pons
C
,
Prat
J
.
Mullerian inhibiting substance, alpha-inhibin, and CD99 expression in sex cord-stromal tumors and endometrioid ovarian carcinomas resembling sex cord-stromal tumors
.
Hum Pathol
.
1998
;
29
(
8
):
840
845
.
275
Kratzer
SS
,
Ulbright
TM
,
Talerman
A
,
et al
.
Large cell calcifying Sertoli cell tumor of the testis: contrasting features of six malignant and six benign tumors and a review of the literature
.
Am J Surg Pathol
.
1997
;
21
(
11
):
1271
1280
.
276
Henley
JD
,
Young
RH
,
Ulbright
TM
.
Malignant Sertoli cell tumors of the testis: a study of 13 examples of a neoplasm frequently misinterpreted as seminoma
.
Am J Surg Pathol
.
2002
;
26
(
5
):
541
550
.
277
Harms
D
,
Kock
LR
.
Testicular juvenile granulosa cell and Sertoli cell tumours: a clinicopathological study of 29 cases from the Kiel Paediatric Tumour Registry
.
Virchows Arch
.
1997
;
430
(
4
):
301
309
.
278
Plata
C
,
Algaba
F
,
Andujar
M
,
et al
.
Large cell calcifying Sertoli cell tumour of the testis
.
Histopathology
.
1995
;
26
(
3
):
255
259
.
279
Hammerich
KH
,
Hille
S
,
Ayala
GE
,
et al
.
Malignant advanced granulosa cell tumor of the adult testis: case report and review of the literature
.
Hum Pathol
.
2008
;
39
(
5
):
701
709
.
280
Petersson
F
,
Bulimbasic
S
,
Sima
R
,
et al
.
Large cell calcifying Sertoli cell tumor: a clinicopathologic study of 1 malignant and 3 benign tumors using histomorphology, immunohistochemistry, ultrastructure, comparative genomic hybridization, and polymerase chain reaction analysis of the PRKAR1A gene
.
Hum Pathol
.
2010
;
41
(
4
):
552
559
.
281
Amin
MB
.
Selected other problematic testicular and paratesticular lesions: rete testis neoplasms and pseudotumors, mesothelial lesions and secondary tumors
.
Mod Pathol
.
2005
;
18
(
suppl 2
):
S131
S145
.
282
Delahunt
B
,
Eble
JN
,
King
D
,
Bethwaite
PB
,
Nacey
JN
,
Thornton
A
.
Immunohistochemical evidence for mesothelial origin of paratesticular adenomatoid tumour
.
Histopathology
.
2000
;
36
(
2
):
109
115
.
283
Ballotta
MR
,
Borghi
L
,
Barucchello
G
.
Adenocarcinoma of the rete testis: report of two cases
.
Adv Clin Pathol
.
2000
;
4
(
4
):
169
173
.
284
Detassis
C
,
Pusiol
T
,
Piscioli
F
,
Luciani
L
.
Adenomatoid tumor of the epididymis: immunohistochemical study of 8 cases
.
Urol Int
.
1986
;
41
(
3
):
232
234
.
285
Crisp-Lindgren
N
,
Travers
H
,
Wells
MM
,
Cawley
LP
.
Papillary adenocarcinoma of rete testis: autopsy findings, histochemistry, immunohistochemistry, ultrastructure, and clinical correlations
.
Am J Surg Pathol
.
1988
;
12
(
6
):
492
501
.
286
Jones
MA
,
Young
RH
,
Srigley
JR
,
Scully
RE
.
Paratesticular serous papillary carcinoma: a report of six cases
.
Am J Surg Pathol
.
1995
;
19
(
12
):
1359
1365
.
287
Kamiya
M
,
Eimoto
T
.
Malignant mesothelioma of the tunica vaginalis
.
Pathol Res Pract
.
1990
;
186
(
5
):
680
684
.
288
Menon
PK
,
Vasudevarao, Sabhiki A, Kudesia S, Joshi DP, Mathur UB. A case of carcinoma rete testis: histomorphological, immunohistochemical and ultrastructural findings and review of literature
.
Indian J Cancer
.
2002
;
39
(
3
):
106
111
.
289
Perez-Ordonez
B
,
Srigley
JR
.
Mesothelial lesions of the paratesticular region
.
Semin Diagn Pathol
.
2000
;
17
(
4
):
294
306
.
290
Winstanley
AM
,
Landon
G
,
Berney
D
,
Minhas
S
,
Fisher
C
,
Parkinson
MC
.
The immunohistochemical profile of malignant mesotheliomas of the tunica vaginalis: a study of 20 cases
.
Am J Surg Pathol
.
2006
;
30
(
1
):
1
6
.
291
Wachter
DL
,
Wunsch
PH
,
Hartmann
A
,
Agaimy
A
.
Adenomatoid tumors of the female and male genital tract: a comparative clinicopathologic and immunohistochemical analysis of 47 cases emphasizing their site-specific morphologic diversity
.
Virchows Arch
.
2011
;
458
(
5
):
593
602
.
292
Al-Salam
S
,
Hammad
FT
,
Salman
MA
,
AlAshari
M
.
Expression of Wilms tumor-1 protein and CD 138 in malignant mesothelioma of the tunica vaginalis
.
Pathol Res Pract
.
2009
;
205
(
11
):
797
800
.
293
Jones
MA
,
Young
RH
,
Scully
RE
.
Malignant mesothelioma of the tunica vaginalis: a clinicopathologic analysis of 11 cases with review of the literature
.
Am J Surg Pathol
.
1995
;
19
(
7
):
815
825
.
294
Kurihara
K
,
Oka
A
,
Mannami
M
,
Iwata
Y
.
Papillary adenocarcinoma of the epididymis
.
Acta Pathol Jpn
.
1993
;
43
(
7–8
):
440
443
.
295
Kim
JY
,
Lee
YT
,
Kang
HJ
,
Lee
CH
.
Primary mucinous cystadenoma of the spermatic cord within the inguinal canal
.
Diagn Pathol
.
2012
;
7
:
139
.
296
Sanchez-Chapado
M
,
Angulo
JC
,
Haas
GP
.
Adenocarcinoma of the rete testis
.
Urology
.
1995
;
46
(
4
):
468
475
.
297
Ganem
JP
,
Jhaveri
FM
,
Marroum
MC
.
Primary adenocarcinoma of the epididymis: case report and review of the literature
.
Urology
.
1998
;
52
(
5
):
904
908
.
298
Anchala
PR
,
Dhir
R
,
Parwani
AV
,
Zynger
DL
.
Immunohistochemical profile of paratesticular serous papillary adenocarcinoma and tunica vaginalis facilitates distinction from malignant mesothelioma
.
Int J Surg Pathol
.
2011
;
19
(
5
):
692
698
.
299
McCluggage
WG
,
Shah
V
,
Nott
C
,
Clements
B
,
Wilson
B
,
Hill
CM
.
Cystadenoma of spermatic cord resembling ovarian serous epithelial tumour of low malignant potential: immunohistochemical study suggesting Mullerian differentiation
.
Histopathology
.
1996
;
28
(
1
):
77
80
.
300
Bremmer
F
,
Schweyer
S
,
Behnes
CL
,
Blech
M
,
Radzun
HJ
.
Sertoliform cystadenoma: a rare benign tumour of the rete testis
.
Diagn Pathol
.
2013
;
8
:
23
.
301
Kacar
A
,
Senel
E
,
Caliskan
D
,
Demirel
F
,
Tiryaki
T
.
Sertoliform cystadenoma: a case with overlapping features
.
Pediatr Dev Pathol
.
2011
;
14
(
2
):
138
143
.
302
Nistal
M
,
Revestido
R
,
Paniagua
R
.
Bilateral mucinous cystadenocarcinoma of the testis and epididymis
.
Arch Pathol Lab Med
.
1992
;
116
(
12
):
1360
1363
.
303
Thiel
R
,
Effert
P
.
Primary adenocarcinoma of the seminal vesicles
.
J Urol
.
2002
;
168
(
5
):
1891
1896
.
304
Ormsby
AH
,
Haskell
R
,
Jones
D
,
Goldblum
JR
.
Primary seminal vesicle carcinoma: an immunohistochemical analysis of four cases
.
Mod Pathol
.
2000
;
13
(
1
):
46
51
.
305
Ohmori
T
,
Okada
K
,
Tabei
R
,
et al
.
CA125-producing adenocarcinoma of the seminal vesicle
.
Pathol Int
.
1994
;
44
(
4
):
333
337
.
306
Tong
GX
,
Memeo
L
,
Colarossi
C
,
et al
.
PAX8 and PAX2 immunostaining facilitates the diagnosis of primary epithelial neoplasms of the male genital tract
.
Am J Surg Pathol
.
2011
;
35
(
10
):
1473
1483
.
307
Zhao
C
,
Barner
R
,
Vinh
TN
,
McManus
K
,
Dabbs
D
,
Vang
R
.
SF-1 is a diagnostically useful immunohistochemical marker and comparable to other sex cord-stromal tumor markers for the differential diagnosis of ovarian sertoli cell tumor
.
Int J Gynecol Pathol
.
2008
;
27
(
4
):
507
514
.
308
Michal
M
,
Kazakov
DV
,
Kacerovska
D
,
et al
.
Paratesticular cystadenomas with ovarian stroma, metaplastic serous Mullerian epithelium, and male adnexal tumor of probable wolffian origin: a series of 5 hitherto poorly recognized testicular tumors
.
Ann Diagn Pathol
.
2013
;
17
(
2
):
151
158
.
* 

References 50, 72, 82, 85, 100, 126–138.

References 271, 274, 280, 288, 292, 293, 295, 298, 303–308.

Author notes

The authors have no relevant financial interest in the products or companies described in this article.