Context.—

Molecular testing is increasingly playing a key role in the diagnosis, prognosis, and treatment of neoplasms of the genitourinary system.

Objective.—

To provide a general overview of the clinically relevant molecular tests available for neoplasms of the genitourinary tract.

Data Sources.—

Relevant medical literature indexed on PubMed.

Conclusions.—

Understanding of the molecular oncology of genitourinary neoplasms is rapidly advancing, and the pathologist must be aware of the practical implications of molecular testing. While many genomic abnormalities are not yet clinically relevant, there is an increasing library of ancillary tests that may guide diagnosis, prognosis, and/or treatment of many neoplasms. Recurrent genomic abnormalities have been identified in many types of renal cell carcinoma, and some types of renal cell carcinoma are specifically defined by the molecular abnormality. Two major routes of developing urothelial carcinoma have been molecularly described. Recurrent translocations involving ETS family genes are found in approximately half of prostate cancer cases. Testicular germ cell tumors typically harbor i(12p). Penile neoplasms are often high-risk human papillomavirus–driven cancers. Nonetheless, even as genitourinary neoplasms are increasingly better understood at the molecular level, further research with eventual clinical validation is needed for optimal diagnosis, prognosis, and treatment of aggressive malignancies in the genitourinary tract.

Seemingly every day, novel genomic alterations are uncovered, or a new drug is developed to target a specific molecular abnormality. Indeed, it is now possible to perform next-generation sequencing on clinical specimens. However, these additional tests and drugs come at a price, and the clinical utility of many of these remains to be demonstrated. Here, we aim to review most of the clinically relevant molecular assays currently available that may aid in the diagnosis, prognosis, and treatment of neoplasia in the genitourinary system (Table). Certainly, there are molecular abnormalities that have been reported in the literature to be prognostically or therapeutically useful but that have been omitted in this review owing to a paucity of evidence of clinical utility and/or lack of widespread clinical availability.1  Additionally, it should be noted that as next-generation sequencing becomes more viable for clinical use, a cancer gene panel by this method should likely be considered for prognostically important or therapeutically targetable mutations.

Clinically Relevant Molecular Testing of Urologic Neoplasms

Clinically Relevant Molecular Testing of Urologic Neoplasms
Clinically Relevant Molecular Testing of Urologic Neoplasms

Renal cell carcinoma (RCC) is the most common primary kidney malignancy, and, as our understanding of the molecular underpinnings of RCC are better understood, many types of RCC are associated with, and even defined by, recurrent genomic abnormalities.

The most common type of RCC is clear cell RCC (CCRCC), which accounts for approximately 65% to 70% of all renal cancers.1  Classically, CCRCC is composed of neoplastic cells with clear to eosinophilic cytoplasm arranged in nests that are surrounded by a delicately anastomosing vasculature with nuclei of variable grade. CCRCC usually occurs sporadically but may occasionally occur in association with a familial syndrome.24  Sporadic CCRCC harbors genomic deletions and loss of heterozygosity on chromosome arm 3p, on which the VHL tumor suppressor gene is located.5  Although CCRCC can usually be diagnosed with routine histologic evaluation, possibly combined with immunohistochemistry (IHC), a fluorescence in situ hybridization (FISH) assay for 3p deletions is clinically available, which may be useful, albeit rarely, in challenging cases such as a biopsy with scant material.6  Notably, FISH analysis is unable to detect some cases of VHL loss, and molecular analysis is needed to identify copy number deletion, missense, and truncating mutations in VHL, which commonly occur in CCRCC.7 

The second most common type of RCC is papillary RCC (PRCC), accounting for nearly 20% of RCCs.8  Papillary RCC is also further subdivided into type 1 and type 2 PRCC. Type 1 PRCC is characterized by fibrovascular cores lined by a single layer of neoplastic epithelium containing scant, basophilic cytoplasm, and usually possessing low-grade nuclei. In contrast, type 2 PRCC is characterized by pseudostratified layers of neoplastic cells lining fibrovascular cores, often with more abundant cytoplasm and higher-grade nuclei.1  The difference between type 1 and type 2 PRCC is not only histologic, but also prognostic and genomic, as patients with type 2 PRCC typically have worse outcomes than patients with type 1 PRCC. Furthermore, type 1 PRCC is frequently associated with trisomy of chromosomes 7 and 17 and loss of chromosome Y,912  whereas recent studies have shown that type 2 PRCC is characterized by other genetic alterations, including alterations in different chromosomes than observed in type 1 PRCC.13,14  Indeed, type 2 PRCC may not even be a truly distinct entity, evidenced by the identification of other types of RCC in tumors previously diagnosed as type 2 PRCC. Nonetheless, there is currently no clinically available molecular assay to aid in the diagnosis of type 2 PRCC, but FISH for trisomy 7 and/or 17 could be used in cases in which type 1 PRCC is a consideration.15  Notably, type 1 PRCC can have considerable morphologic overlap with mucinous tubular and spindle cell carcinoma and clear cell papillary RCC, both of which lack trisomy 7 and 17; thus, FISH for trisomy 7 and/or 17 may be especially useful in this differential diagnosis.1618 

Translocation-associated RCC (tRCC) is a subtype of RCC defined by a translocation involving the TFE3 gene on chromosome band Xp11 with multiple gene partners (Figure 1, A and B) or the translocation t(6;11) creating the Alpha-TFEB gene fusion (Figure 2, A and B).1921  Morphologic features such as solid and papillary growth, voluminous cytoplasm, psammomatous calcifications, and extracellular basement membrane material are morphologic features that can aid in the diagnosis of tRCC.19,20  Even so, there is considerable morphologic overlap between tRCC and other RCC subtypes, such as CCRCC and PRCC.22,23  Immunohistochemistry is often useful to help differentiate tRCC from other subtypes of RCC, such as melanocytic markers or TFE3, but IHC is not entirely sensitive or specific for tRCC.21,24,25  FISH analysis for TFE3 and TFEB gene rearrangements is highly sensitive and specific for tRCC and is extremely useful to confirm the diagnosis of tRCC.23,2527 

Figure 1

MiT family translocation renal cell carcinoma with a translocation involving the TFE3 gene is often morphologically heterogeneous with both solid and papillary architectural patterns composed of neoplastic cells with voluminous cytoplasm (A). Because the TFE3 gene has multiple potential fusion partners, break-apart FISH (fluorescence in situ hybridization) probes are typically used to assess for a TFE3 gene translocation. Thus, a positive result occurs when the red (yellow arrow) and green (white arrow) probes are separated (B) (hematoxylin-eosin, original magnification ×100 [A]; original magnification ×1000 [B]).

Figure 2 The translocation t(6;11) creating the Alpha-TFEB fusion gene occurs in a subset of translocation-associated renal cell carcinomas. Morphologically, these tumors are often biphasic with smaller neoplastic cells surrounding extracellular basement membrane-type material admixed with larger neoplastic epithelial cells (A). The fusion partner of the TFEB gene is consistently the Alpha gene, and so dual-fusion FISH (fluorescence in situ hybridization) probes are typically used. When the red and green probes are juxtaposed (yellow arrow), the result is considered positive (B) (hematoxylin-eosin, original magnification ×100 [A]; original magnification ×1000 [B]).

Figure 3 Succinate dehydrogenase (SDH)–deficient renal cell carcinoma is characterized by neoplastic cells with smooth nuclear contours with fine chromatin and eosinophilic cytoplasm, frequently with vacuoles or flocculent inclusions (A). Negative immunohistochemistry for a subunit of the SDH enzyme, SDHB, demonstrates loss of SDH expression and confirms the diagnosis (B) (hematoxylin-eosin, original magnification ×100 [A]; original magnification ×100 [B]).

Figure 1

MiT family translocation renal cell carcinoma with a translocation involving the TFE3 gene is often morphologically heterogeneous with both solid and papillary architectural patterns composed of neoplastic cells with voluminous cytoplasm (A). Because the TFE3 gene has multiple potential fusion partners, break-apart FISH (fluorescence in situ hybridization) probes are typically used to assess for a TFE3 gene translocation. Thus, a positive result occurs when the red (yellow arrow) and green (white arrow) probes are separated (B) (hematoxylin-eosin, original magnification ×100 [A]; original magnification ×1000 [B]).

Figure 2 The translocation t(6;11) creating the Alpha-TFEB fusion gene occurs in a subset of translocation-associated renal cell carcinomas. Morphologically, these tumors are often biphasic with smaller neoplastic cells surrounding extracellular basement membrane-type material admixed with larger neoplastic epithelial cells (A). The fusion partner of the TFEB gene is consistently the Alpha gene, and so dual-fusion FISH (fluorescence in situ hybridization) probes are typically used. When the red and green probes are juxtaposed (yellow arrow), the result is considered positive (B) (hematoxylin-eosin, original magnification ×100 [A]; original magnification ×1000 [B]).

Figure 3 Succinate dehydrogenase (SDH)–deficient renal cell carcinoma is characterized by neoplastic cells with smooth nuclear contours with fine chromatin and eosinophilic cytoplasm, frequently with vacuoles or flocculent inclusions (A). Negative immunohistochemistry for a subunit of the SDH enzyme, SDHB, demonstrates loss of SDH expression and confirms the diagnosis (B) (hematoxylin-eosin, original magnification ×100 [A]; original magnification ×100 [B]).

Close modal

An exceptionally rare subgroup of RCC, which is characterized by ALK gene rearrangement, is included in the World Health Organization classification of kidney tumors as a provisional entity. These tumors, so-called ALK rearrangement–associated RCC, are exceptionally rare and may occur in children or adults.28,29  They are often located in the renal medulla, and they are morphologically characterized by large polygonal or spindle cells with abundant eosinophilic cytoplasm and frequent intracytoplasmic lumina.28  In these tumors, the ALK gene can have a number of fusion partners, and a FISH break-apart assay or a molecular assay can aid in the diagnosis.3033 

Another type of RCC that is defined by a specific genetic abnormality is succinate dehydrogenase (SDH)–deficient RCC, in which there is a double-hit inactivation of 1 of the SDH genes, most commonly SDHB, but also including SDHA, SDHC, or SDHD.3436  Morphologically, SDH-deficient RCC is characterized by a well-circumscribed or pushing border that may entrap benign tubules with scattered cysts containing eosinophilic material. The neoplastic cells have smooth nuclear contours with fine chromatin, lacking nucleoli. Most characteristically, the cytoplasm contains vacuoles or flocculent inclusions that contain eosinophilic or pale material (Figure 3, A).34,37,38  Nevertheless, the classic morphologic features may be only focally present, and the diagnosis must be confirmed with IHC. Immunohistochemistry for SDHB is available for clinical use, and loss of SDHB expression as assessed by IHC confirms the diagnosis of SDH-deficient RCC (Figure 3, B). There are 2 important things to note in regard to SDHB IHC. First, SDHB IHC is not only lost in patients with loss of heterozygosity of SDHB but also in SDHC and SDHD, though some mutations in SDHD may result in weak expression of SDHB IHC, and mutations in SDHA may result in a positive SDHB IHC finding.3946  The risk of a positive SDHB IHC result in a patient with loss of heterozygosity of SDHA is mitigated by the relative rarity of SDHA mutations.45  Thus, loss of SDHB expression by IHC confirms inactivation of an SDH gene but is not necessarily diagnostic of inactivation of the SDHB gene. Furthermore, this also means that SDHB IHC has a relatively high sensitivity for detection of SDH-deficient RCC. The second important thing to note is that in tumors with abundant clear cytoplasm, SDHB IHC expression may be markedly decreased, but it is not negative, and care should be taken to not misdiagnose these cases as SDH-deficient RCC.38  Most patients with SDH-deficient RCC have a germline mutation in an SDH gene, and the second hit inactivating the SDH gene results in neoplasia.34,36,47  Indeed, an autosomal dominant syndrome consisting of paraganglioma/pheochromocytoma, gastrointestinal stromal tumor, SDH-deficient RCC, and pituitary adenoma is described, and patients should be offered genetic testing if an SDH-deficient RCC is diagnosed.48 

Hereditary leiomyomatosis and RCC (HLRCC) syndrome is characterized by hereditary leiomyomatosis and a unique subtype of RCC. HLRCC-associated RCC typically possesses neoplastic cells arranged in tubular, tubulocystic, papillary (Figure 4, A), and solid growth patterns with large nuclei containing prominent “viral-inclusion–like” nucleoli (Figure 4, B), which may be focal.49  HLRCC is caused by germline mutations in the fumarate hydratase (FH) gene.50  Loss of heterozygosity in the wild-type FH gene locus eliminates FH protein function in the neoplastic cells, resulting in a metabolic shift from oxidative phosphorylation to aerobic glycolysis.51,52  Immunohistochemistry for FH is available for clinical use and can aid in diagnosis of HLRCC-associated RCC.53  Immunohistochemistry for 2-succino-cysteine, which accumulates in the cytoplasm of HLRCC-associated RCC, is more sensitive and specific than FH IHC, but this antibody is not currently commercially available.54,55 

Figure 4

Hereditary leiomyomatosis and renal cell carcinoma (HLRCC)–associated RCC is characterized by neoplastic cells arranged in tubular, tubulocystic, papillary (A), and solid growth patterns with large nuclei containing prominent “viral-inclusion–like” nucleoli (B) (hematoxylin-eosin, original magnifications ×100 [A] and ×400 [B]).

Figure 5 Cytology specimens containing urothelial carcinoma (UC) demonstrate both singly dispersed and cohesive groups of atypical urothelial cells that are enlarged with high nucleus to cytoplasmic ratio, irregular nuclear contours, and hyperchromasia (A). In select cases, fluorescence in situ hybridization analysis with UroVysion may be helpful in monitoring patients with a history of UC or for stratification of patients with abnormal cytologic findings and no clinical evidence of a tumor. UroVysion assesses for aneuploidy of chromosomes 3, 7, 17, and 9p21 (B) (Papanicolaou, original magnification ×400 [A]; original magnification ×1000 [B]).

Figure 6TERT promoter mutations occur in most urothelial carcinomas. In difficult cases, analysis for TERT promoter mutation may aid diagnosis. Recently, it was demonstrated that small cell carcinomas of the urinary bladder harbor TERT promoter mutations, supporting the theory that these tumors derive from the urothelium (A). A positive result for TERT promoter analysis using a melting point method is demonstrated in (B) (hematoxylin-eosin, original magnification ×200 [A]).

Figure 4

Hereditary leiomyomatosis and renal cell carcinoma (HLRCC)–associated RCC is characterized by neoplastic cells arranged in tubular, tubulocystic, papillary (A), and solid growth patterns with large nuclei containing prominent “viral-inclusion–like” nucleoli (B) (hematoxylin-eosin, original magnifications ×100 [A] and ×400 [B]).

Figure 5 Cytology specimens containing urothelial carcinoma (UC) demonstrate both singly dispersed and cohesive groups of atypical urothelial cells that are enlarged with high nucleus to cytoplasmic ratio, irregular nuclear contours, and hyperchromasia (A). In select cases, fluorescence in situ hybridization analysis with UroVysion may be helpful in monitoring patients with a history of UC or for stratification of patients with abnormal cytologic findings and no clinical evidence of a tumor. UroVysion assesses for aneuploidy of chromosomes 3, 7, 17, and 9p21 (B) (Papanicolaou, original magnification ×400 [A]; original magnification ×1000 [B]).

Figure 6TERT promoter mutations occur in most urothelial carcinomas. In difficult cases, analysis for TERT promoter mutation may aid diagnosis. Recently, it was demonstrated that small cell carcinomas of the urinary bladder harbor TERT promoter mutations, supporting the theory that these tumors derive from the urothelium (A). A positive result for TERT promoter analysis using a melting point method is demonstrated in (B) (hematoxylin-eosin, original magnification ×200 [A]).

Close modal

Although not yet codified into the World Health Organization classification of kidney tumors, a recently described group of RCCs harbors recurrent TCEB1 gene mutations.56  These tumors characteristically are composed of neoplastic cells possessing voluminous clear cytoplasm with nested, tubular, and/or papillary architecture with admixed thick, fibromuscular bands transecting the tumor. Previously, these tumors were likely diagnosed as CCRCC or clear cell papillary RCC, both of which are the main differential diagnoses. To resolve this, IHC for cytokeratin 7 (CK7), CA-IX, and CD10 may be useful, as TCEB1-mutated RCC is expected to be positive for CA-IX, CK7, and CD10. Currently, there are no specific clinical assays available for confirmation of the diagnosis of TCEB1-mutated RCC.

In addition to HLRCC and SDH-deficient RCC, there are other familial RCC syndromes. These include von Hippel–Lindau syndrome (VHL gene on chromosome arm 3p), hereditary papillary RCC (MET gene on chromosome arm 7p), Birt-Hogg-Dubé syndrome (FCLN on chromosome arm 17p), and tuberous sclerosis (TSC1 gene on chromosome arm 9q and TSC2 gene on chromosome arm 16p). Patients with von Hippel–Lindau syndrome have an increased risk of developing CCRCC.4,57  Patients with hereditary papillary RCC often present with multiple, bilateral PRCCs.58  Patients with Birt-Hogg-Dubé syndrome usually develop multiple, bilateral kidney tumors, including a characteristic hybrid oncocytic tumor.59  Patients with tuberous sclerosis often develop angiomyolipomas, but they can also develop morphologically unique RCCs.12  Although familial RCC syndromes are much less common than sporadic RCC, when recognized by the pathologist, patients may be referred for genetic testing and counseling.5,60 

Urothelial carcinoma (UC) accounts for nearly all of the cancers of the urinary bladder, ureter, and renal pelvis. Although UC accounts for such a high proportion of neoplasia of the urinary tract, clinical outcome among patients with UC is highly variable, ranging from noninvasive low-grade papillary UC to deeply invasive and lethal high-grade UC. Accurate prognostic stratification and treatment decisions rely on pathologic examination of biopsy, resection, and cytology specimens. Nonetheless, molecular testing may occasionally aid morphologic evaluation and guide the therapeutic decision process.

Molecular oncology data suggest there are 2 separate molecular routes for UC, one resulting in superficial disease and the second resulting in deeply invasive disease.61,62  The first route, which is associated with noninvasive or superficially invasive (ie, invasion into lamina propria) papillary UC, is composed of early, activating point mutations in FGFR3 or RAS family genes. A small subset of tumors in this group (∼15%) subsequently develops loss of function of 1 or more tumor suppressor genes, such as TP53, RB1, and PTEN. Acquisition of mutations in these tumor suppressor genes results in the potential for progression to deeply invasive disease. Alternatively, TP53, RB1, and PTEN mutations can occur in the absence of FGFR3 or RAS family genes, and this second route is typically associated with flat UC.

Although a few molecular diagnostic assays are available for prognosticating UC, these are not currently widely used in clinical practice. Rather, the most widely used molecular assay in regard to UC is likely UroVysion (Vysis Inc). UroVysion is a FISH assay performed on exfoliated cells in urine (Figure 5, A) that assesses for aneuploidy of chromosomes 3, 7, and 17 and loss of chromosome band 9p21, all of which are abnormalities characteristic of UC (Figure 5, B). UroVysion has a relatively high sensitivity and specificity for the presence of UC, and it has been implemented into many bladder cancer screening programs.63  Additionally, UroVysion can be used to monitor patients with a history of UC or for stratification of patients with an abnormal cytology result and no clinical or cystoscopic evidence of a bladder tumor. However, more recent studies demonstrate that the sensitivity and positive predictive value of UroVysion, particularly for low-grade urothelial carcinoma, may not be as optimal as initially thought.6467 

Also commonly present in UC are mutations in the TERT promoter gene, with 60% to 80% of UCs harboring a point mutation (Figure 6, A and B).6870  These mutations appear to be early events in oncogenesis of UC, and they are present in both low-grade papillary UC and high-grade UC. However, TERT promoter mutations are not entirely specific for UC, as they are also present in benign urothelial neoplasms, such as urothelial papilloma, inverted papilloma, and papillary urothelial neoplasm of low malignant potential.71,72  Furthermore, they are also frequently present in neoplasms of other organs, such as glioblastoma and melanoma, but they are notably absent in prostate cancer.7376  Thus, the practical implications of TERT promoter mutations in UC are likely in 3 situations: (1) the differential diagnosis of UC versus nonneoplastic urothelium (eg, cystitis glandularis); (2) the differential diagnosis of UC versus prostatic cancer, particularly in the setting of divergent differentiation; and (3) screening urine for recurrent UC. Although histomorphologic features of UC are usually diagnostic, some cases may include reactive urothelium in the differential; in this setting, a positive TERT promoter mutation test finding would weigh in favor of a neoplastic process over a reactive process, as reactive urothelium lacks TERT promoter mutations.71  A negative TERT promoter test result, however, does not exclude the possibility of an urothelial neoplasm, as not all cases of UC harbor TERT promoter mutations. Because prostate cancer lacks TERT promoter mutations, a positive TERT promoter test result would weigh in favor of UC over prostate cancer; an example of this is small cell carcinoma of the bladder, which has been demonstrated to harbor TERT promoter mutations in approximately half of cases, as it is clonally related to UC.73,77,78  Finally, TERT promoter mutations have been identified in urine specimens, and it has been proposed that testing urine of patients with a history of UC for TERT promoter mutations may be an effective method of screening for recurrent UC.79,80 

As UC becomes better understood, immunotherapy is likely to increasingly become a treatment option, and IHC performed on biopsy or resection material may guide therapeutic decision making. An example is the use of programmed death receptor-1 (PD-1) and programmed death ligand-1 (PD-L1) checkpoint inhibitors; indeed, atezolizumab, a PD-L1 inhibitor, is US Food and Drug Administration (FDA) approved for treatment of patients with chemotherapy-resistant advanced or metastatic UC.81  Trials are underway for similar drugs, including durvalumab, pembrolizumab, avelumab, nivolumab, and ipilimumab.82  Thus, it is likely that testing for PD-L1 expression in biopsies and resections for UC may become more routine in the near future. Indeed, the FDA has already approved the use of several of these agents with IHC as a companion diagnostic test in select patients with UC.83 

Prostatic adenocarcinoma is the most common malignancy in men, excluding skin cancer, and the clinical course is highly variable, varying from indolent to aggressive, lethal disease in a relatively large subset of men.84  Clearly, it is necessary to conduct patient stratification on the basis of the expected clinicopathologic behavior of these tumors, and this is done by using pathologic grade (ie, Gleason score and, more recently, Grade Groups) and stage (ie, AJCC [American Joint Committee on Cancer] Cancer Staging Manual, 8th edition).85,86  Additionally, as the molecular underpinnings of prostatic adenocarcinoma are elucidated, molecular testing may play a role in the risk stratification and treatment of some of these patients.

The discovery of recurrent gene fusions involving the ETS family of transcription factors in nearly half of prostatic cancers was the first major breakthrough in understanding the molecular basis of these tumors.8789  TMPRSS2-ERG is the dominant ETS gene fusion found in prostatic adenocarcinoma, the result of which brings ERG expression under androgen control on the basis of androgen receptor (AR)–mediated TMPRSS2 transcriptional regulation. Thus, ETS gene fusions, particularly TMPRSS2-ERG, are the basis for the development of many diagnostic, prognostic, and therapeutic-driven assays. Most ETS prostatic adenocarcinoma rearrangements can be detected by FISH,90,91  and IHC using an anti-ERG antibody, which detects the ERG gene fusion product, is also highly sensitive and specific for ERG aberrations.92  Therefore, FISH or IHC for ERG may be occasionally useful in the diagnosis of prostatic adenocarcinoma versus a benign process (Figure 7, A through C). However, it should be noted that IHC for ERG may be positive in high-grade prostatic intraepithelial neoplasia (HGPIN), and overexpression of ERG is not pathognomonic of malignancy.93  Another complication of ERG IHC is that in the castration-resistant state, ERG protein may not be overexpressed, even in the presence of ERG aberrations at the genomic level. In this setting, ERG FISH may be more helpful, such as in prostatic small cell carcinoma, which typically harbors an ERG aberration but often lacks detectable ERG protein expression.94  Furthermore, it is also notable that approximately half of prostatic cancers lack an ERG rearrangement, and a negative ERG FISH or IHC result does not exclude the diagnosis of prostatic adenocarcinoma. Recently, Kunju et al95  described an RNA in situ hybridization strategy for identifying ETS fusions involving genes other than ERG, such as ETV1, ETV4, and ETV5. The molecular underpinnings of ETS fusion-negative prostatic adenocarcinoma are also being studied, and abnormalities in SPINK1,96  PTEN,97  AURKA, MYCN,98  SPOP, HOXB13, CHD1, and ADRB299102  have all been implicated in the pathogenesis of prostate cancer. At present, however, these abnormalities hold little clinical relevance.

Figure 7

Recurrent gene fusions involving the ETS family of transcription factors occur in nearly half of prostate cancers, and TMPRSS2-ERG is the dominant ETS gene fusion found in prostate cancer. In select cases, such as metastatic poorly differentiated carcinoma (A and B), fluorescence in situ hybridization (FISH) for ERG gene translocation may be useful. Because ERG has multiple fusion partners, break-apart FISH probes are typically used (yellow and white arrows in [C] indicate widely separated FISH probe signals) (hematoxylin-eosin, original magnifications ×40 [A] and ×100 [B]; original magnification ×1000 [C]).

Figure 8 Basal cell carcinoma (BCC) of the prostate is composed of basaloid cells forming infiltrative nests and tubules (A), and approximately half of prostatic BCCs have been shown to harbor MYB-NFIB gene fusion (yellow arrows in [B]). Basal cell hyperplasia or adenoma may closely mimic prostatic BCC morphologically (C), but these benign basal proliferations lack MYB-NFIB gene fusion. The fusion partner of MYB is consistently NFIB, so FISH (fluorescence in situ hybridization) fusion probes are used, and a positive result occurs when the red and green probes are juxtaposed (B), whereas a negative result occurs when they are split (D) (hematoxylin-eosin, original magnifications ×100 [A] and ×200 [C]; original magnification ×1000 [B and D]).

Figure 7

Recurrent gene fusions involving the ETS family of transcription factors occur in nearly half of prostate cancers, and TMPRSS2-ERG is the dominant ETS gene fusion found in prostate cancer. In select cases, such as metastatic poorly differentiated carcinoma (A and B), fluorescence in situ hybridization (FISH) for ERG gene translocation may be useful. Because ERG has multiple fusion partners, break-apart FISH probes are typically used (yellow and white arrows in [C] indicate widely separated FISH probe signals) (hematoxylin-eosin, original magnifications ×40 [A] and ×100 [B]; original magnification ×1000 [C]).

Figure 8 Basal cell carcinoma (BCC) of the prostate is composed of basaloid cells forming infiltrative nests and tubules (A), and approximately half of prostatic BCCs have been shown to harbor MYB-NFIB gene fusion (yellow arrows in [B]). Basal cell hyperplasia or adenoma may closely mimic prostatic BCC morphologically (C), but these benign basal proliferations lack MYB-NFIB gene fusion. The fusion partner of MYB is consistently NFIB, so FISH (fluorescence in situ hybridization) fusion probes are used, and a positive result occurs when the red and green probes are juxtaposed (B), whereas a negative result occurs when they are split (D) (hematoxylin-eosin, original magnifications ×100 [A] and ×200 [C]; original magnification ×1000 [B and D]).

Close modal

AR signaling status is an important factor in determining treatment for castration-resistant patients, as patients with active androgen signaling (ie, retained expression of AR and or AR-regulated genes, including ERG) are candidates for more aggressive androgen deprivation therapy, regardless of the genomic status of AR and TMPRSS2-ERG. Because tissue biopsies are often impractical to perform routinely for patients with castration-resistant prostate cancer, there has been a focus on the development of minimally invasive biomarkers from blood and urine.103106  The profiling of circulating tumor cells and cell-free DNA has revealed truncated AR splice variants, AR copy number gain, and AR point mutations, which are linked to castration resistance.103,104,107,108  Specifically, AR splice variant 7 messenger RNA (AR-V7) in circulating tumor cells and copy number increase and point mutations in circulating cell-free DNA are associated with resistance to enzalutamide and abiraterone.104 

Gene expression profiling using DNA microarrays is difficult to perform on formalin-fixed, paraffin-embedded tissue and remains a more useful research tool than it is useful clinically. Nonetheless, several clinical assays have been developed, including Myriad Diagnostic's Polaris, Genomic Health's Oncotype DX Prostate Cancer Assay, and GenomeDx's Decipher. These assays may have some usefulness in prognostication of patients, but their clinical utility is not well defined at this time, and the clinical standard of prognostication remains standard clinicopathologic data.109111 

While FISH and IHC for ERG may be relevant to tissue biopsy or resection, there is also interest in the development of better and more specific biomarkers for prostate cancer surveillance. While serum protein-specific antigen (PSA), the current standard for prostate cancer screening, is sensitive for prostate cancer, it is not specific, as benign processes (eg, prostatitis) may increase serum PSA, and it may be increased in the setting of low-grade, indolent tumors. Because of this, assays that detect additional biomarkers associated with prostatic adenocarcinoma that are present in urine have been developed. As already discussed, TMPRSS2-ERG is present in a large subset of prostate cancers. Additionally, PCA3 is a noncoding RNA that is expressed in prostate cancer and HGPIN.112117  Several molecular assays for use on urine specimens—using some combination of TMPRSS2-ERG, PCA3, and PSA—are available for clinical use, including Mi-Prostate Score, which provides a likelihood of a patient having prostate cancer as well as the chance of that cancer being aggressive,118  and Progensa, which has a high negative predictive value.119  However, as the use of MRI-guided prostatic biopsy increases, the utility of prebiopsy molecular assays is likely decreasing. In addition to urine tests, there is also continued interest in developing better serum biomarkers. One such assay, which is commercially available, is 4Kscore (OPKO Health Inc), which measures 4 PSA-related molecules—total PSA, free PSA, intact PSA, and kallikrein-related peptidase 2—and is reported to predict risk of cancer more accurately than PSA alone.120 

An interesting subtype of prostate cancer is basal cell carcinoma (BCC).1  Previously, it has been referred to as adenoid cystic carcinoma, owing to its morphologic similarity to adenoid cystic carcinoma of the salivary gland and other sites.121  Basal cell carcinoma of the prostate typically is composed of basaloid cells forming infiltrative nests and tubules, often with cribriform architecture, extracellular hyaline material, and perineural invasion (Figure 8, A). Recently, it was demonstrated that a subset of prostatic BCCs also mimics adenoid cystic carcinoma on the molecular level, harboring rearrangement of the MYB gene.122  A subsequent study confirmed the presence of MYB-NFIB gene fusion in approximately half of prostatic BCCs (Figure 8, B), while prostatic basal cell hyperplasia and adenoma were ubiquitously negative for MYB gene rearrangements (Figure 8, C and D).123  Thus, although clinical validation is still necessary, it is possible that FISH analysis may soon aid in the diagnosis of prostatic BCC.

Several germline mutations have been found to increase a man's risk of developing prostate cancer. Germline mutations in BRCA1 and BRCA2 not only predispose women to breast and ovarian cancer, but they also confer an increased risk of developing prostate cancer.124129  Additionally, men who harbor pathogenic germline BRCA2 mutations may have earlier onset of disease and decreased survival.130133  In addition to BRCA1/2 mutations, a germline mutation in HOXB13, although rare, increases the risk of prostate cancer.100  However, there are currently no recommendations specifically for prostate cancer screening in men with BRCA1/2 or HOXB13 mutations.134136  In the absence of formal societal recommendations, Cheng et al137  proposed a screening algorithm for such men.

Tumors of the testis can be broadly split into 2 categories: germ cell tumors (GCTs) and sex cord stromal tumors. Germ cell tumors account for most testicular neoplasms, particularly in young adult men. Although the morphologic spectrum of GCTs is broad, most GCTs (approximately 80%) harbor a copy number gain of chromosome arm 12p, usually present as isochromosome 12p [i(12p)],138  and the presence of 12p abnormalities can be identified with FISH for chromosome arm 12p or with next-generation sequencing.139,140  Although most testis GCTs can be diagnosed on the basis only of morphology with judicious use of IHC, there are rare situations in which ancillary testing to demonstrate a chromosome arm 12p abnormality may be useful. Situations in which this testing may be informative include resolving the differential diagnosis of prepubertal-type versus postpubertal-type teratoma (Figure 9, A and B); identifying a somatic-type malignancy as GCT derived (ie, secondary somatic malignancy; Figure 9, C and D)141143 ; diagnosis of metastatic GCT139 ; and early detection of germ cell neoplasia in semen samples.138,144  While most teratomas occurring in adult men are malignant, a subset of teratomas in adult men have recently been described that appear to behave indolently and lack i(12p).145  Thus, demonstration of i(12p) would preclude a diagnosis of prepubertal-type teratoma and would imply malignant potential. Secondary somatic-type malignancies occur in approximately 5% of testis GCTs and can manifest as sarcoma, carcinoma, primitive neuroectodermal tumor, or leukemia; demonstration of i(12p) in the somatic malignancy is indicative of GCT origin.141143 

Figure 9

Most testicular germ cell tumors harbor chromosome arm 12p abnormalities. Prepubertal teratoma is a benign tumor composed of both epithelial and mesenchymal elements (A), and it lacks chromosome arm 12p abnormalities (B). Occasionally, testis-derived germ cell tumors develop a secondary somatic malignancy such as adenocarcinoma (C), and fluorescence in situ hybridization for isochromosome 12p can aid in the diagnosis (yellow arrow in [D]) (hematoxylin-eosin, original magnifications ×100 [A] and ×200 [C]; original magnification ×1000 [B and D]).

Figure 10 A subset of penile squamous cell carcinomas (SCCs) is associated with high-risk human papillomavirus (HPV) infection. The warty subtype of penile SCC is characterized by complex papillae containing atypical squamous cells and koilocytes (A and B). In cases associated with high-risk HPV infections, HPV in situ hybridization is typically positive (C) (hematoxylin-eosin, original magnification ×40 [A and B]; original magnification ×400 [C]).

Figure 9

Most testicular germ cell tumors harbor chromosome arm 12p abnormalities. Prepubertal teratoma is a benign tumor composed of both epithelial and mesenchymal elements (A), and it lacks chromosome arm 12p abnormalities (B). Occasionally, testis-derived germ cell tumors develop a secondary somatic malignancy such as adenocarcinoma (C), and fluorescence in situ hybridization for isochromosome 12p can aid in the diagnosis (yellow arrow in [D]) (hematoxylin-eosin, original magnifications ×100 [A] and ×200 [C]; original magnification ×1000 [B and D]).

Figure 10 A subset of penile squamous cell carcinomas (SCCs) is associated with high-risk human papillomavirus (HPV) infection. The warty subtype of penile SCC is characterized by complex papillae containing atypical squamous cells and koilocytes (A and B). In cases associated with high-risk HPV infections, HPV in situ hybridization is typically positive (C) (hematoxylin-eosin, original magnification ×40 [A and B]; original magnification ×400 [C]).

Close modal

Most penile neoplasms are squamous cell carcinomas (SCCs), and, like SCC of the cervix and oropharynx, a subset of penile SCCs is associated with high-risk human papillomavirus (HPV) infection (Figure 10, A and B).146  High-risk HPV DNA is integrated into the host cell genome, leading to production of the viral proteins E6 and E7, which inactivate TP53 and RB1 and increase expression of p16. Thus, HPV-associated SCC is typically p53/p16+, whereas non-HPV–associated SCC is typically p53+/p16, and IHC for p16 or in situ hybridization for high-risk HPV can be used to assess for HPV status (Figure 10, C).147  While routine pathologic examination is the most reliable predictor of outcome,148  confirming the presence or absence of high-risk HPV may be useful in select situations. Interestingly, while p16+ penile SCC has generally been shown to have better prognosis than p16 penile SCC,149  basaloid penile SCC is thought to be HPV driven and confers a worse prognosis than conventional penile SCC.150 

Nonetheless, relative to malignancies of other sites, the molecular underpinnings of penile SCC are poorly understood and are still to be elucidated. Recently, next-generation sequencing revealed frequent alterations in TP53, CDKN2A, PIK3CA, MYC, HRAS, and SOX2,151  alterations similar to those found in SCC of various other sites.152  However, the clinical utility of these findings are currently limited. The penile SCC study found no significant associations between an individual gene's mutation status and tumor grade, stage, or histology.

The PD-1/PD-L1 immune checkpoint is a hot area of investigation as a potential target in malignancies of various sites (eg, lung carcinoma), and penile SCC is no exception. Indeed, it has been demonstrated that most non-HPV–related penile SCCs express PD-L1 as assessed with IHC, and this provides a rationale for targeted anti–PD-1 and anti–PD-L1 immunotherapy in penile SCC.153 

Immunotherapy has already been discussed in this review insofar that the PD-1/PD-L1 immune checkpoint may be clinically relevant in some genitourinary malignancies. Proper patient selection for immunotherapy is critical, as not all patients demonstrate response to these agents. Direct assessment for PD-1/PD-L1 expression via IHC for PD-L1 is discussed above and has been evaluated in many clinical trials.154  Alternative methods for determining patient selection for immunotherapy include tumor mutational burden (TMB) and microsatellite instability (MSI). Indeed, TMB has been demonstrated to be more significantly associated with response to anti–PD-1 and anti–PD-L1 immunotherapy than IHC for PD-1/PD-L1 expression, but TMB analysis is not currently widely available for clinical use.155,156  Microsatellite instability, on the other hand, is more readily available for clinical use than TMB, either through IHC for mismatch repair enzymes (ie, MLH1, PMS2, MSH2, and MSH6) or molecular analysis. Because MSI-high (MSI-H) tumors are unstable and hypermutational, they tend to express high levels of checkpoint proteins, such as PD-1 and PD-L1.157,158  Clinical trials in a variety of tumors have demonstrated that MSI-H tumors are associated with response to immunotherapy, and, in May 2017, the FDA broadly approved pembrolizumab for use in all pediatric and adult patients with MSI-H or mismatch repair-deficient solid tumors.157159  Thus, although MSI-H is a relatively rare event in genitourinary malignancies, there are instances in which testing for MSI may be clinically beneficial.155,160 

Clearly, molecular understanding of genitourinary neoplasms has advanced to the point of being able to incorporate some molecular assays into clinical use. Recurrent genomic abnormalities have been identified in many types of RCC, and some types of RCC are specifically defined by the molecular abnormality. Two major routes of developing UC have been molecularly described. Recurrent translocations involving ETS family genes are found in approximately half of prostate cancers. Testicular GCTs typically harbor i(12p). Penile neoplasms are often high risk and HPV driven. Nonetheless, even as genitourinary neoplasms are increasingly better understood at the molecular level, further research with eventual clinical validation is needed for optimal diagnosis, prognosis, and treatment of aggressive malignancies in the genitourinary tract.

1
Cheng
L
,
MacLennan
GT
,
Bostwick
DG
.
Urologic Surgical Pathology. 4th ed
.
Philadelphia, PA
:
Elsevier;
2019
.
2
Pavlovich
CP
,
Walther
MM
,
Eyler
RA
, et al.
Renal tumors in the Birt-Hogg-Dube syndrome
.
Am J Surg Pathol
.
2002
;
26
(
12
):
1542
1552
.
3
Bjornsson
J
,
Short
MP
,
Kwiatkowski
DJ
,
Henske
EP
.
Tuberous sclerosis-associated renal cell carcinoma: clinical, pathological, and genetic features
.
Am J Pathol
.
1996
;
149
(
4
):
1201
1208
.
4
Seizinger
BR
,
Rouleau
GA
,
Ozelius
LJ
, et al.
Von Hippel-Lindau disease maps to the region of chromosome 3 associated with renal cell carcinoma
.
Nature
.
1988
;
332
(
6161
):
268
269
.
5
Linehan
WM
,
Bratslavsky
G
,
Pinto
PA
, et al.
Molecular diagnosis and therapy of kidney cancer
.
Annu Rev Med
.
2010
;
61
:
329
343
.
6
Barocas
DA
,
Mathew
S
,
DelPizzo
JJ
, et al.
Renal cell carcinoma sub-typing by histopathology and fluorescence in situ hybridization on a needle-biopsy specimen
.
BJU Int
.
2007
;
99
(
2
):
290
295
.
7
Sato
Y
,
Yoshizato
T
,
Shiraishi
Y
, et al.
Integrated molecular analysis of clear-cell renal cell carcinoma
.
Nat Genet
.
2013
;
45
(
8
):
860
867
.
8
Amin
MB
,
Amin
MB
,
Tamboli
P
, et al.
Prognostic impact of histologic subtyping of adult renal epithelial neoplasms: an experience of 405 cases
.
Am J Surg Pathol
.
2002
;
26
(
3
):
281
291
.
9
Schraml
P
,
Muller
D
,
Bednar
R
, et al.
Allelic loss at the D9S171 locus on chromosome 9p13 is associated with progression of papillary renal cell carcinoma
.
J Pathol
.
2000
;
190
(
4
):
457
461
.
10
Kovacs
G
,
Fuzesi
L
,
Emanual
A
,
Kung
HF
.
Cytogenetics of papillary renal cell tumors
.
Genes Chromosomes Cancer
.
1991
;
3
(
4
):
249
255
.
11
Kattar
MM
,
Grignon
DJ
,
Wallis
T
, et al.
Clinicopathologic and interphase cytogenetic analysis of papillary (chromophilic) renal cell carcinoma
.
Mod Pathol
.
1997
;
10
(
11
):
1143
1150
.
12
Jiang
F
,
Richter
J
,
Schraml
P
, et al.
Chromosomal imbalances in papillary renal cell carcinoma: genetic differences between histological subtypes
.
Am J Pathol
.
1998
;
153
(
5
):
1467
1473
.
13
Antonelli
A
,
Tardanico
R
,
Balzarini
P
, et al.
Cytogenetic features, clinical significance and prognostic impact of type 1 and type 2 papillary renal cell carcinoma
.
Cancer Genet Cytogenet
.
2010
;
199
(
2
):
128
133
.
14
Cancer Genome Atlas Research Network
,
Linehan
WM
,
Spellman
PT
, et al
.
Comprehensive molecular characterization of papillary renal-cell carcinoma
.
N Engl J Med
.
2016
;
374
(
2
):
135
145
.
15
Hughson
MD
,
Dickman
K
,
Bigler
SA
,
Meloni
AM
,
Sandberg
AA
.
Clear-cell and papillary carcinoma of the kidney: an analysis of chromosome 3, 7, and 17 abnormalities by microsatellite amplification, cytogenetics, and fluorescence in situ hybridization
.
Cancer Genet Cytogenet
.
1998
;
106
(
2
):
93
104
.
16
Cossu-Rocca
P
,
Eble
JN
,
Delahunt
B
, et al.
Renal mucinous tubular and spindle carcinoma lacks the gains of chromosomes 7 and 17 and losses of chromosome Y that are prevalent in papillary renal cell carcinoma
.
Mod Pathol
.
2006
;
19
(
4
):
488
493
.
17
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
.
18
Fisher
KE
,
Yin-Goen
Q
,
Alexis
D
, et al.
Gene expression profiling of clear cell papillary renal cell carcinoma: comparison with clear cell renal cell carcinoma and papillary renal cell carcinoma
.
Mod Pathol
.
2014
;
27
(
2
):
222
230
.
19
Argani
P
,
Antonescu
CR
,
Couturier
J
, et al.
PRCC-TFE3 renal carcinomas: morphologic, immunohistochemical, ultrastructural, and molecular analysis of an entity associated with the t(X;1)(p11.2;q21)
.
Am J Surg Pathol
.
2002
;
26
(
12
):
1553
1566
.
20
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
.
21
Argani
P
,
Lae
M
,
Hutchinson
B
, et al.
Renal carcinomas with the t(6;11)(p21;q12): clinicopathologic features and demonstration of the specific alpha-TFEB gene fusion by immunohistochemistry, RT-PCR, and DNA PCR
.
Am J Surg Pathol
.
2005
;
29
(
2
):
230
240
.
22
Argani
P
,
Olgac
S
,
Tickoo
SK
, et al.
Xp11 translocation renal cell carcinoma in adults: expanded clinical, pathologic, and genetic spectrum
.
Am J Surg Pathol
.
2007
;
31
(
8
):
1149
1160
.
23
Green
WM
,
Yonescu
R
,
Morsberger
L
, et al.
Utilization of a TFE3 break-apart FISH assay in a renal tumor consultation service
.
Am J Surg Pathol
.
2013
;
37
(
8
):
1150
1163
.
24
Martignoni
G
,
Pea
M
,
Gobbo
S
, et al.
Cathepsin-K immunoreactivity distinguishes MiTF/TFE family renal translocation carcinomas from other renal carcinomas
.
Mod Pathol
.
2009
;
22
(
8
):
1016
1022
.
25
Rao
Q
,
Williamson
SR
,
Zhang
S
, et al.
TFE3 break-apart FISH has a higher sensitivity for Xp11.2 translocation-associated renal cell carcinoma compared with TFE3 or cathepsin K immunohistochemical staining alone: expanding the morphologic spectrum
.
Am J Surg Pathol
.
2013
;
37
(
6
):
804
815
.
26
Argani
P
,
Yonescu
R
,
Morsberger
L
, et al.
Molecular confirmation of t(6;11)(p21;q12) renal cell carcinoma in archival paraffin-embedded material using a break-apart TFEB FISH assay expands its clinicopathologic spectrum
.
Am J Surg Pathol
.
2012
;
36
(
10
):
1516
1526
.
27
Mosquera
JM
,
Dal Cin
P
,
Mertz
KD
, et al.
Validation of a TFE3 break-apart FISH assay for Xp11.2 translocation renal cell carcinomas
.
Diagn Mol Pathol
.
2011
;
20
(
3
):
129
137
.
28
Smith
NE
,
Deyrup
AT
,
Marino-Enriquez
A
, et al.
VCL-ALK renal cell carcinoma in children with sickle-cell trait: the eighth sickle-cell nephropathy?
Am J Surg Pathol
.
2014
;
38
(
6
):
858
863
.
29
Sukov
WR
,
Hodge
JC
,
Lohse
CM
, et al.
ALK alterations in adult renal cell carcinoma: frequency, clinicopathologic features and outcome in a large series of consecutively treated patients
.
Mod Pathol
.
2012
;
25
(
11
):
1516
1525
.
30
Gobbo
S
,
Eble
JN
,
Maclennan
GT
, et al.
Renal cell carcinomas with papillary architecture and clear cell components: the utility of immunohistochemical and cytogenetical analyses in differential diagnosis
.
Am J Surg Pathol
.
2008
;
32
(
12
):
1780
1786
.
31
Brunelli
M
,
Eble
JN
,
Zhang
S
, et al.
Metanephric adenoma lacks the gains of chromosomes 7 and 17 and loss of Y that are typical of papillary renal cell carcinoma and papillary adenoma
.
Mod Pathol
.
2003
;
16
(
10
):
1060
1063
.
32
Cheng
L
,
Gu
J
,
Eble
JN
, et al.
Molecular genetic evidence for different clonal origin of components of human renal angiomyolipomas
.
Am J Surg Pathol
.
2001
;
25
(
10
):
1231
1236
.
33
Jones
TD
,
Eble
JN
,
Wang
M
, et al.
Molecular genetic evidence for the independent origin of multifocal papillary tumors in patients with papillary renal cell carcinomas
.
Clin Cancer Res
.
2005
;
11
(
20
):
7226
7233
.
34
Gill
AJ
,
Hes
O
,
Papathomas
T
, et al.
Succinate dehydrogenase (SDH)-deficient renal carcinoma: a morphologically distinct entity: a clinicopathologic series of 36 tumors from 27 patients
.
Am J Surg Pathol
.
2014
;
38
(
12
):
1588
1602
.
35
Yakirevich
E
,
Ali
SM
,
Mega
A
, et al.
A novel SDHA-deficient renal cell carcinoma revealed by comprehensive genomic profiling
.
Am J Surg Pathol
.
2015
;
39
(
6
):
858
863
.
36
Papathomas
TG
,
Gaal
J
,
Corssmit
EP
, et al.
Non-pheochromocytoma (PCC)/paraganglioma (PGL) tumors in patients with succinate dehydrogenase-related PCC-PGL syndromes: a clinicopathological and molecular analysis
.
Eur J Endocrinol
.
2014
;
170
(
1
):
1
12
.
37
Gill
AJ
,
Pachter
NS
,
Chou
A
, et al.
Renal tumors associated with germline SDHB mutation show distinctive morphology
.
Am J Surg Pathol
.
2011
;
35
(
10
):
1578
1585
.
38
Gill
AJ
,
Pachter
NS
,
Clarkson
A
, et al.
Renal tumors and hereditary pheochromocytoma-paraganglioma syndrome type 4
.
N Engl J Med
.
2011
;
364
(
9
):
885
886
.
39
Gill
AJ
,
Benn
DE
,
Chou
A
, et al.
Immunohistochemistry for SDHB triages genetic testing of SDHB, SDHC, and SDHD in paraganglioma-pheochromocytoma syndromes
.
Hum Pathol
.
2010
;
41
(
6
):
805
814
.
40
Castelblanco
E
,
Santacana
M
,
Valls
J
, et al.
Usefulness of negative and weak-diffuse pattern of SDHB immunostaining in assessment of SDH mutations in paragangliomas and pheochromocytomas
.
Endocr Pathol
.
2013
;
24
(
4
):
199
205
.
41
Pai
R
,
Manipadam
MT
,
Singh
P
,
Ebenazer
A
,
Samuel
P
,
Rajaratnam
S.
Usefulness of succinate dehydrogenase B (SDHB) immunohistochemistry in guiding mutational screening among patients with pheochromocytoma-paraganglioma syndromes
.
APMIS
.
2014
;
122
(
11
):
1130
1135
.
42
Giubellino
A
,
Lara
K
,
Martucci
V
, et al.
urinary bladder paragangliomas: how immunohistochemistry can assist to identify patients with SDHB germline and somatic mutations
.
Am J Surg Pathol
.
2015
;
39
(
11
):
1488
1492
.
43
Menara
M
,
Oudijk
L
,
Badoual
C
, et al.
SDHD immunohistochemistry: a new tool to validate SDHx mutations in pheochromocytoma/paraganglioma
.
J Clin Endocrinol Metab
.
2015
;
100
(
2
):
E287
E291
.
44
van Nederveen
FH
,
Gaal
J
,
Favier
J
, et al.
An immunohistochemical procedure to detect patients with paraganglioma and phaeochromocytoma with germline SDHB, SDHC, or SDHD gene mutations: a retrospective and prospective analysis
.
Lancet Oncol
.
2009
;
10
(
8
):
764
771
.
45
Papathomas
TG
,
Oudijk
L
,
Persu
A
, et al.
SDHB/SDHA immunohistochemistry in pheochromocytomas and paragangliomas: a multicenter interobserver variation analysis using virtual microscopy: a Multinational Study of the European Network for the Study of Adrenal Tumors (ENS@T)
.
Mod Pathol
.
2015
;
28
(
6
):
807
821
.
46
Santi
R
,
Rapizzi
E
,
Canu
L
, et al.
Potential pitfalls of SDH immunohistochemical detection in paragangliomas and phaeochromocytomas harbouring germline SDHx gene mutation
.
Anticancer Res
.
2017
;
37
(
2
):
805
812
.
47
Evenepoel
L
,
Papathomas
TG
,
Krol
N
, et al.
Toward an improved definition of the genetic and tumor spectrum associated with SDH germ-line mutations
.
Genet Med
.
2015
;
17
(
8
):
610
620
.
48
Ricketts
CJ
,
Forman
JR
,
Rattenberry
E
, et al.
Tumor risks and genotype-phenotype-proteotype analysis in 358 patients with germline mutations in SDHB and SDHD
.
Hum Mutat
.
2010
;
31
(
1
):
41
51
.
49
Merino
MJ
,
Torres-Cabala
C
,
Pinto
P
,
Linehan
WM
.
The morphologic spectrum of kidney tumors in hereditary leiomyomatosis and renal cell carcinoma (HLRCC) syndrome
.
Am J Surg Pathol
.
2007
;
31
(
10
):
1578
1585
.
50
Tomlinson
IP
,
Alam
NA
,
Rowan
AJ
, et al.
Germline mutations in FH predispose to dominantly inherited uterine fibroids, skin leiomyomata and papillary renal cell cancer
.
Nat Genet
.
2002
;
30
(
4
):
406
410
.
51
Udager
AM
,
Alva
A
,
Chen
YB
, et al.
Hereditary leiomyomatosis and renal cell carcinoma (HLRCC): a rapid autopsy report of metastatic renal cell carcinoma
.
Am J Surg Pathol
.
2014
;
38
(
4
):
567
577
.
52
Linehan
WM
,
Rouault
TA
.
Molecular pathways: fumarate hydratase-deficient kidney cancer—targeting the Warburg effect in cancer
.
Clin Cancer Res
.
2013
;
19
(
13
):
3345
3352
.
53
Trpkov
K
,
Hes
O
,
Agaimy
A
, et al.
Fumarate hydratase-deficient renal cell carcinoma is strongly correlated with fumarate hydratase mutation and hereditary leiomyomatosis and renal cell carcinoma syndrome
.
Am J Surg Pathol
.
2016
;
40
(
7
):
865
875
.
54
Buelow
B
,
Cohen
J
,
Nagymanyoki
Z
, et al.
Immunohistochemistry for 2-succinocysteine (2SC) and fumarate hydratase (FH) in cutaneous leiomyomas may aid in identification of patients with HLRCC (hereditary leiomyomatosis and renal cell carcinoma syndrome)
.
Am J Surg Pathol
.
2016
;
40
(
7
):
982
988
.
55
Joseph
NM
,
Solomon
DA
,
Frizzell
N
,
Rabban
JT
,
Zaloudek
C
,
Garg
K.
Morphology and immunohistochemistry for 2SC and FH aid in detection of fumarate hydratase gene aberrations in uterine leiomyomas from young patients
.
Am J Surg Pathol
.
2015
;
39
(
11
):
1529
1539
.
56
Hakimi
AA
,
Tickoo
SK
,
Jacobsen
A
, et al.
TCEB1-mutated renal cell carcinoma: a distinct genomic and morphological subtype
.
Mod Pathol
.
2015
;
28
(
6
):
845
853
.
57
Montani
M
,
Heinimann
K
,
von Teichman
A
,
Rudolph
T
,
Perren
A
,
Moch
H.
VHL-gene deletion in single renal tubular epithelial cells and renal tubular cysts: further evidence for a cyst-dependent progression pathway of clear cell renal carcinoma in von Hippel-Lindau disease
.
Am J Surg Pathol
.
2010
;
34
(
6
):
806
815
.
58
Schmidt
L
,
Duh
FM
,
Chen
F
, et al.
Germline and somatic mutations in the tyrosine kinase domain of the MET proto-oncogene in papillary renal carcinomas
.
Nat Genet
.
1997
;
16
(
1
):
68
73
.
59
Nickerson
ML
,
Warren
MB
,
Toro
JR
, et al.
Mutations in a novel gene lead to kidney tumors, lung wall defects, and benign tumors of the hair follicle in patients with the Birt-Hogg-Dube syndrome
.
Cancer Cell
.
2002
;
2
(
2
):
157
164
.
60
Przybycin
CG
,
Magi-Galluzzi
C
,
McKenney
JK
.
Hereditary syndromes with associated renal neoplasia: a practical guide to histologic recognition in renal tumor resection specimens
.
Adv Anat Pathol
.
2013
;
20
(
4
):
245
263
.
61
Cheng
L
,
Davison
DD
,
Adams
J.
Biomarkers in bladder cancer: translational and clinical implications
.
Crit Rev Oncol Hematol
.
2014
;
89
(
1
):
73
111
.
62
Cheng
L
,
Zhang
S
,
MacLennan
GT
, et al.
Bladder cancer: translating molecular genetic insights into clinical practice
.
Hum Pathol
.
2011
;
42
(
4
):
455
481
.
63
Sokolova
IA
,
Halling
KC
,
Jenkins
RB
, et al.
The development of a multitarget, multicolor fluorescence in situ hybridization assay for the detection of urothelial carcinoma in urine
.
J Mol Diagn
.
2000
;
2
(
3
):
116
123
.
64
Dimashkieh
H
,
Wolff
DJ
,
Smith
TM
,
Houser
PM
,
Nietert
PJ
,
Yang
J.
Evaluation of urovysion and cytology for bladder cancer detection: a study of 1835 paired urine samples with clinical and histologic correlation
.
Cancer Cytopathol
.
2013
;
121
(
10
):
591
597
.
65
Caraway
NP
,
Khanna
A
,
Fernandez
RL
, et al.
Fluorescence in situ hybridization for detecting urothelial carcinoma: a clinicopathologic study
.
Cancer Cytopathol
.
2010
;
118
(
5
):
259
268
.
66
Ferra
S
,
Denley
R
,
Herr
H
,
Dalbagni
G
,
Jhanwar
S
,
Lin
O.
Reflex UroVysion testing in suspicious urine cytology cases
.
Cancer
.
2009
;
117
(
1
):
7
14
.
67
McHale
T
,
Ohori
NP
,
Cieply
KM
,
Sherer
C
,
Bastacky
SI
.
Comparison of urinary cytology and fluorescence in situ hybridization in the detection of urothelial neoplasia: an analysis of discordant results
.
Diagn Cytopathol
.
2019
;
47
(
4
):
282
288
.
68
Bell
RJ
,
Rube
HT
,
Xavier-Magalhaes
A
, et al.
Understanding TERT promoter mutations: a common path to immortality
.
Mol Cancer Res
.
2016
;
14
(
4
):
315
323
.
69
Kinde
I
,
Munari
E
,
Faraj
SF
, et al.
TERT promoter mutations occur early in urothelial neoplasia and are biomarkers of early disease and disease recurrence in urine
.
Cancer Res
.
2013
;
73
(
24
):
7162
7167
.
70
Gunes
C
,
Wezel
F
,
Southgate
J
,
Bolenz
C.
Implications of TERT promoter mutations and telomerase activity in urothelial carcinogenesis
.
Nat Rev Urol
.
2018
;
15
(
6
):
386
393
.
71
Cheng
L
,
Davidson
DD
,
Wang
M
, et al.
Telomerase reverse transcriptase (TERT) promoter mutation analysis of benign, malignant and reactive urothelial lesions reveals a subpopulation of inverted papilloma with immortalizing genetic change
.
Histopathology
.
2016
;
69
(
1
):
107
113
.
72
Cheng
L
,
Montironi
R
,
Lopez-Beltran
A.
TERT promoter mutations occur frequently in urothelial papilloma and papillary urothelial neoplasm of low malignant potential
.
Eur Urol
.
2017
;
71
(
3
):
497
498
.
73
Priemer
DS
,
Wang
M
,
Zhang
S
, et al.
Small-cell carcinomas of the urinary bladder and prostate: TERT promoter mutation status differentiates sites of malignancy and provides evidence of common clonality between small-cell carcinoma of the urinary bladder and urothelial carcinoma
.
Eur Urol Focus
.
2018
;
4
(
6
):
880
888
.
74
Killela
PJ
,
Reitman
ZJ
,
Jiao
Y
, et al.
TERT promoter mutations occur frequently in gliomas and a subset of tumors derived from cells with low rates of self-renewal
.
Proc Natl Acad Sci U S A
.
2013
;
110
(
15
):
6021
6026
.
75
Huang
FW
,
Hodis
E
,
Xu
MJ
,
Kryukov
GV
,
Chin
L
,
Garraway
LA
.
Highly recurrent TERT promoter mutations in human melanoma
.
Science
.
2013
;
339
(
6122
):
957
959
.
76
Horn
S
,
Figl
A
,
Rachakonda
PS
, et al.
TERT promoter mutations in familial and sporadic melanoma
.
Science
.
2013
;
339
(
6122
):
959
961
.
77
Zheng
X
,
Zhuge
J
,
Bezerra
SM
, et al.
High frequency of TERT promoter mutation in small cell carcinoma of bladder, but not in small cell carcinoma of other origins
.
J Hematol Oncol
.
2014
;
7
:
47
.
78
Zheng
X
,
Liu
D
,
Fallon
JT
,
Zhong
M.
Distinct genetic alterations in small cell carcinoma from different anatomic sites
.
Exp Hematol Oncol
.
2015
;
4
:
2
.
79
Dahmcke
CM
,
Steven
KE
,
Larsen
LK
, et al.
A prospective blinded evaluation of urine-DNA testing for detection of urothelial bladder carcinoma in patients with gross hematuria
.
Eur Urol
.
2016
;
70
(
6
):
916
919
.
80
Critelli
R
,
Fasanelli
F
,
Oderda
M
, et al.
Detection of multiple mutations in urinary exfoliated cells from male bladder cancer patients at diagnosis and during follow-up
.
Oncotarget
.
2016
;
7
(
41
):
67435
67448
.
81
Rosenberg
JE
,
Hoffman-Censits
J
,
Powles
T
, et al.
Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial
.
Lancet
.
2016
;
387
(
10031
):
1909
1920
.
82
Gupta
S
,
Gill
D
,
Poole
A
,
Agarwal
N.
Systemic immunotherapy for urothelial cancer: current trends and future directions
.
Cancers (Basel)
.
2017
;
9
(
2
).
83
US Food and Drug Administration
.
FDA updates prescribing information for Keytruda and Tecentriq
.
2018
. .
84
Bostwick
DG
,
Cheng
L.
Precursors of prostate cancer
.
Histopathology
.
2012
;
60
(
1
):
4
27
.
85
Amin
MB
,
American Joint Committee on Cancer, American Cancer Society
.
AJCC Cancer Staging Manual
.
8th ed.
Amin
MB
,
Edge
SB
,
Greene
FL
,
et al, eds
.
Chicago IL
:
American Joint Committee on Cancer, Springer;
2017
:
xvii.
86
Cheng
L
,
Montironi
R
,
Bostwick
DG
, et al.
Staging of prostate cancer
.
Histopathology
.
2012
;
60
(
1
):
87
117
.
87
Tomlins
SA
,
Rhodes
DR
,
Perner
S
, et al.
Recurrent fusion of TMPRSS2 and ETS transcription factor genes in prostate cancer
.
Science
.
2005
;
310
(
5748
):
644
648
.
88
Rubin
MA
,
Maher
CA
,
Chinnaiyan
AM
.
Common gene rearrangements in prostate cancer
.
J Clin Oncol
.
2011
;
29
(
27
):
3659
3668
.
89
Mehra
R
,
Tomlins
SA
,
Yu
J
, et al.
Characterization of TMPRSS2-ETS gene aberrations in androgen-independent metastatic prostate cancer
.
Cancer Res
.
2008
;
68
(
10
):
3584
3590
.
90
Mehra
R
,
Tomlins
SA
,
Shen
R
, et al.
Comprehensive assessment of TMPRSS2 and ETS family gene aberrations in clinically localized prostate cancer
.
Mod Pathol
.
2007
;
20
(
5
):
538
544
.
91
Tomlins
SA
,
Laxman
B
,
Dhanasekaran
SM
, et al.
Distinct classes of chromosomal rearrangements create oncogenic ETS gene fusions in prostate cancer
.
Nature
.
2007
;
448
(
7153
):
595
599
.
92
Williamson
SR
,
Zhang
S
,
Yao
JL
, et al.
ERG-TMPRSS2 rearrangement is shared by concurrent prostatic adenocarcinoma and prostatic small cell carcinoma and absent in small cell carcinoma of the urinary bladder: evidence supporting monoclonal origin
.
Mod Pathol
.
2012
(
8
):
1120
1127
.
93
Morais
CL
,
Guedes
LB
,
Hicks
J
,
Baras
AS
,
De Marzo
AM
,
Lotan
TL. ERG
and PTEN status of isolated high-grade PIN occurring in cystoprostatectomy specimens without invasive prostatic adenocarcinoma
.
Hum Pathol
.
2016
;
55
:
117
125
.
94
Schelling
LA
,
Williamson
SR
,
Zhang
S
, et al.
Frequent TMPRSS2-ERG rearrangement in prostatic small cell carcinoma detected by fluorescence in situ hybridization: the superiority of fluorescence in situ hybridization over ERG immunohistochemistry
.
Hum Pathol
.
2013
;
44
(
10
):
2227
2233
.
95
Kunju
LP
,
Carskadon
S
,
Siddiqui
J
,
Tomlins
SA
,
Chinnaiyan
AM
,
Palanisamy
N.
Novel RNA hybridization method for the in situ detection of ETV1, ETV4, and ETV5 gene fusions in prostate cancer
.
Appl Immunohistochem Mol Morphol
.
2014
;
22
(
8
):
e32
e40
.
96
Tomlins
SA
,
Rhodes
DR
,
Yu
J
, et al.
The role of SPINK1 in ETS rearrangement-negative prostate cancers
.
Cancer Cell
.
2008
;
13
(
6
):
519
528
.
97
Yoshimoto
M
,
Cunha
IW
,
Coudry
RA
, et al.
FISH analysis of 107 prostate cancers shows that PTEN genomic deletion is associated with poor clinical outcome
.
Br J Cancer
.
2007
;
97
(
5
):
678
685
.
98
Beltran
H
,
Rickman
DS
,
Park
K
, et al.
Molecular characterization of neuroendocrine prostate cancer and identification of new drug targets
.
Cancer Discov
.
2011
;
1
(
6
):
487
495
.
99
Barbieri
CE
,
Baca
SC
,
Lawrence
MS
, et al.
Exome sequencing identifies recurrent SPOP, FOXA1 and MED12 mutations in prostate cancer
.
Nat Genet
.
2012
;
44
(
6
):
685
689
.
100
Ewing
CM
,
Ray
AM
,
Lange
EM
, et al.
Germline mutations in HOXB13 and prostate-cancer risk
.
N Engl J Med
.
2012
;
366
(
2
):
141
149
.
101
Grasso
CS
,
Wu
YM
,
Robinson
DR
, et al.
The mutational landscape of lethal castration-resistant prostate cancer
.
Nature
.
2012
;
487
(
7406
):
239
243
.
102
Yu
J
,
Cao
Q
,
Mehra
R
, et al.
Integrative genomics analysis reveals silencing of beta-adrenergic signaling by polycomb in prostate cancer
.
Cancer Cell
.
2007
;
12
(
5
):
419
431
.
103
Carreira
S
,
Romanel
A
,
Goodall
J
, et al.
Tumor clone dynamics in lethal prostate cancer
.
Sci Transl Med
.
2014
;
6
(
254
):
254ra125
.
104
Antonarakis
ES
,
Lu
C
,
Wang
H
, et al.
AR-V7 and resistance to enzalutamide and abiraterone in prostate cancer
.
N Engl J Med
.
2014
;
371
(
11
):
1028
1038
.
105
Quek
SI
,
Wong
OM
,
Chen
A
, et al.
Processing of voided urine for prostate cancer RNA biomarker analysis
.
Prostate
.
2015
;
75
(
16
):
1886
1895
.
106
Dijkstra
S
,
Mulders
PF
,
Schalken
JA
.
Clinical use of novel urine and blood based prostate cancer biomarkers: a review
.
Clin Biochem
.
2014
;
47
(
10-11
):
889
896
.
107
Romanel
A
,
Gasi Tandefelt
D
,
Conteduca
V
, et al.
Plasma AR and abiraterone-resistant prostate cancer
.
Sci Transl Med
.
2015
;
7
(
312
)
:312re10.
108
Azad
AA
,
Volik
SV
,
Wyatt
AW
, et al.
Androgen receptor gene aberrations in circulating cell-free DNA: biomarkers of therapeutic resistance in castration-resistant prostate cancer
.
Clin Cancer Res
.
2015
;
21
(
10
):
2315
2324
.
109
Klein
EA
,
Haddad
Z
,
Yousefi
K
, et al.
Decipher genomic classifier measured on prostate biopsy predicts metastasis risk
.
Urology
.
2016
;
90
:
148
152
.
110
Cuzick
J
,
Berney
DM
,
Fisher
G
, et al.
Prognostic value of a cell cycle progression signature for prostate cancer death in a conservatively managed needle biopsy cohort
.
Br J Cancer
.
2012
;
106
(
6
):
1095
1099
.
111
Cuzick
J
,
Swanson
GP
,
Fisher
G
, et al.
Prognostic value of an RNA expression signature derived from cell cycle proliferation genes in patients with prostate cancer: a retrospective study
.
Lancet Oncol
.
2011
;
12
(
3
):
245
255
.
112
Bussemakers
MJ
,
van Bokhoven
A
,
Verhaegh
GW
, et al.
DD3: a new prostate-specific gene, highly overexpressed in prostate cancer
.
Cancer Res
.
1999
;
59
(
23
):
5975
5979
.
113
de Kok
JB
,
Verhaegh
GW
,
Roelofs
RW
, et al.
DD3(PCA3), a very sensitive and specific marker to detect prostate tumors
.
Cancer Res
.
2002
;
62
(
9
):
2695
2698
.
114
Schmidt
U
,
Fuessel
S
,
Koch
R
, et al.
Quantitative multi-gene expression profiling of primary prostate cancer
.
Prostate
.
2006
;
66
(
14
):
1521
1534
.
115
Popa
I
,
Fradet
Y
,
Beaudry
G
,
Hovington
H
,
Beaudry
G
,
Tetu
B.
Identification of PCA3 (DD3) in prostatic carcinoma by in situ hybridization
.
Mod Pathol
.
2007
;
20
(
11
):
1121
1127
.
116
Warrick
JI
,
Tomlins
SA
,
Carskadon
SL
, et al.
Evaluation of tissue PCA3 expression in prostate cancer by RNA in situ hybridization: a correlative study with urine PCA3 and TMPRSS2-ERG
.
Mod Pathol
.
2014
;
27
(
4
):
609
620
.
117
Salagierski
M
,
Schalken
JA
.
Molecular diagnosis of prostate cancer: PCA3 and TMPRSS2:ERG gene fusion
.
J Urol
.
2012
;
187
(
3
):
795
801
.
118
Tomlins
SA
,
Day
JR
,
Lonigro
RJ
, et al.
Urine TMPRSS2:ERG plus PCA3 for individualized prostate cancer risk assessment
.
Eur Urol
.
2016
;
70
(
1
):
45
53
.
119
Hessels
D
,
Klein Gunnewiek
JM
,
van Oort
I
, et al.
DD3(PCA3)-based molecular urine analysis for the diagnosis of prostate cancer [discussion in Eur Urol
.
2003
;
44
(
1
):
15
16
].
Eur Urol
.
2003
;
44
(
1
):
8
15
.
120
Vickers
A
,
Cronin
A
,
Roobol
M
, et al.
Reducing unnecessary biopsy during prostate cancer screening using a four-kallikrein panel: an independent replication
.
J Clin Oncol
.
2010
;
28
(
15
):
2493
2498
.
121
Frankel
K
,
Craig
JR
.
Adenoid cystic carcinoma of the prostate: report of a case
.
Am J Clin Pathol
.
1974
;
62
(
5
):
639
645
.
122
Bishop
JA
,
Yonescu
R
,
Epstein
JI
,
Westra
WH
.
A subset of prostatic basal cell carcinomas harbor the MYB rearrangement of adenoid cystic carcinoma
.
Hum Pathol
.
2015
;
46
(
8
):
1204
1208
.
123
Magers
MJ
,
Iczkowski
KA
,
Montironi
R
, et al.
MYB-NFIB gene fusion in prostatic basal cell carcinoma: clinicopathologic correlates and comparison with basal cell adenoma and florid basal cell hyperplasia
[published online June 12, 2019]
.
Mod Pathol
. doi:.
124
Kote-Jarai
Z
,
Leongamornlert
D
,
Saunders
E
, et al.
BRCA2 is a moderate penetrance gene contributing to young-onset prostate cancer: implications for genetic testing in prostate cancer patients
.
Br J Cancer
.
2011
;
105
(
8
):
1230
1234
.
125
Breast Cancer Linkage Consortium
.
Cancer risks in BRCA2 mutation carriers
.
J Natl Cancer Inst
.
1999
;
91
(
15
):
1310
1316
.
126
Vickers
AJ
,
Ulmert
D
,
Sjoberg
DD
, et al.
Strategy for detection of prostate cancer based on relation between prostate specific antigen at age 40-55 and long term risk of metastasis: case-control study
.
BMJ
.
2013
;
346:f2023.
127
Leongamornlert
D
,
Mahmud
N
,
Tymrakiewicz
M
, et al.
Germline BRCA1 mutations increase prostate cancer risk
.
Br J Cancer
.
2012
;
106
(
10
):
1697
1701
.
128
Thompson
D
,
Easton
DF
,
Breast Cancer Linkage Consortium
.
Cancer incidence in BRCA1 mutation carriers
.
J Natl Cancer Inst
.
2002
;
94
(
18
):
1358
1365
.
129
Loeb
S
,
Carter
HB
,
Catalona
WJ
,
Moul
JW
,
Schroder
FH
.
Baseline prostate-specific antigen testing at a young age
.
Eur Urol
.
2012
;
61
(
1
):
1
7
.
130
Gallagher
DJ
,
Gaudet
MM
,
Pal
P
, et al.
Germline BRCA mutations denote a clinicopathologic subset of prostate cancer
.
Clin Cancer Res
.
2010
;
16
(
7
):
2115
2121
.
131
Castro
E
,
Goh
C
,
Olmos
D
, et al.
Germline BRCA mutations are associated with higher risk of nodal involvement, distant metastasis, and poor survival outcomes in prostate cancer
.
J Clin Oncol
.
2013
;
31
(
14
):
1748
1757
.
132
Castro
E
,
Goh
C
,
Leongamornlert
D
, et al.
Effect of BRCA mutations on metastatic relapse and cause-specific survival after radical treatment for localised prostate cancer
.
Eur Urol
.
2015
;
68
(
2
):
186
193
.
133
Na
R
,
Zheng
SL
,
Han
M
, et al.
Germline mutations in ATM and BRCA1/2 distinguish risk for lethal and indolent prostate cancer and are associated with early age at death
.
Eur Urol
.
2017
;
71
(
5
):
740
747
.
134
Leão
RRN
,
Price
AJ
,
Hamilton
JR
.
Germline BRCA mutation in male carriers-ripe for precision oncology?
Prostate Cancer Prostatic Dis
.
2018
;
21
(
1
):
48
56
.
135
Annala
M
,
Struss
WJ
,
Warner
EW
, et al.
Treatment outcomes and tumor loss of heterozygosity in germline DNA repair-deficient prostate cancer
.
Eur Urol
.
2017
;
72
(
1
):
34
42
.
136
Das
S
,
Salami
SS
,
Spratt
DE
,
Kaffenberger
SD
,
Jacobs
MF
,
Morgan
TM
.
Bringing prostate cancer germline genetics into clinical practice [published online ahead of print February 5, 2019]
.
J Urol
. doi:
137
Cheng
HH
,
Pritchard
CC
,
Montgomery
B
,
Lin
DW
,
Nelson
PS
.
Prostate cancer screening in a new era of genetics
.
Clin Genitourin Cancer
.
2017
;
15
(
6
):
625
628
.
138
Cheng
L
,
Albers
P
,
Berney
DM
, et al.
Testicular cancer
.
Nat Rev Dis Primers
.
2018
;
5;
4
(
1
):
29
.
139
Kernek
KM
,
Brunelli
M
,
Ulbright
TM
, et al.
Fluorescence in situ hybridization analysis of chromosome 12p in paraffin-embedded tissue is useful for establishing germ cell origin of metastatic tumors
.
Mod Pathol
.
2004
;
17
(
11
):
1309
1313
.
140
Chovanec
M
,
Cheng
L.
Molecular characterization of testicular germ cell tumors: chasing the underlying pathways
.
Future Oncol
.
2019
;
15
(
3
):
227
229
.
141
Motzer
RJ
,
Amsterdam
A
,
Prieto
V
, et al.
Teratoma with malignant transformation: diverse malignant histologies arising in men with germ cell tumors
.
J Urol
.
1998
;
159
(
1
):
133
138
.
142
Kum
JB
,
Ulbright
TM
,
Williamson
SR
, et al.
Molecular genetic evidence supporting the origin of somatic-type malignancy and teratoma from the same progenitor cell
.
Am J Surg Pathol
.
2012
;
36
(
12
):
1849
1856
.
143
Mikuz
G
,
Colecchia
M.
Teratoma with somatic-type malignant components of the testis: a review and an update
.
Virchows Arch
.
2012
;
461
(
1
):
27
32
.
144
Cheng
L
,
Lyu
B
,
Roth
LM
.
Perspectives on testicular germ cell neoplasms
.
Hum Pathol
.
2017
;
59
(
1
):
10
25
.
145
Zhang
C
,
Berney
DM
,
Hirsch
MS
,
Cheng
L
,
Ulbright
TM
.
Evidence supporting the existence of benign teratomas of the postpubertal testis: a clinical, histopathologic, and molecular genetic analysis of 25 cases
.
Am J Surg Pathol
.
2013
;
37
(
6
):
827
835
.
146
Chaux
A
,
Cubilla
AL
.
The role of human papillomavirus infection in the pathogenesis of penile squamous cell carcinomas
.
Semin Diagn Pathol
.
2012
;
29
(
2
):
67
71
.
147
Mannweiler
S
,
Sygulla
S
,
Winter
E
,
Regauer
S.
Two major pathways of penile carcinogenesis: HPV-induced penile cancers overexpress p16ink4a, HPV-negative cancers associated with dermatoses express p53, but lack p16ink4a overexpression
.
J Am Acad Dermatol
.
2013
;
69
(
1
):
73
81
.
148
Chaux
A
,
Cubilla
AL
.
Stratification systems as prognostic tools for defining risk of lymph node metastasis in penile squamous cell carcinomas
.
Semin Diagn Pathol
.
2012
;
29
(
2
):
83
89
.
149
Bethune
G
,
Campbell
J
,
Rocker
A
,
Bell
D
,
Rendon
R
,
Merrimen
J.
Clinical and pathologic factors of prognostic significance in penile squamous cell carcinoma in a North American population
.
Urology
.
2012
;
79
(
5
):
1092
1097
.
150
Chaux
A
,
Reuter
V
,
Lezcano
C
,
Velazquez
EF
,
Torres
J
,
Cubilla
AL
.
Comparison of morphologic features and outcome of resected recurrent and nonrecurrent squamous cell carcinoma of the penis: a study of 81 cases
.
Am J Surg Pathol
.
2009
;
33
(
9
):
1299
1306
.
151
McDaniel
AS
,
Hovelson
DH
,
Cani
AK
, et al.
Genomic profiling of penile squamous cell carcinoma reveals new opportunities for targeted therapy
.
Cancer Res
.
2015
;
75
(
24
):
5219
5227
.
152
Hoadley
KA
,
Yau
C
,
Wolf
DM
, et al.
Multiplatform analysis of 12 cancer types reveals molecular classification within and across tissues of origin
.
Cell
.
2014
;
158
(
4
):
929
944
.
153
Udager
AM
,
Liu
TY
,
Skala
SL
, et al.
Frequent PD-L1 expression in primary and metastatic penile squamous cell carcinoma: potential opportunities for immunotherapeutic approaches
.
Ann Oncol
.
2016
;
27
(
9
):
1706
1712
.
154
Mann
SA
,
Lopez-Beltran
A
,
Massari
F
, et al.
Targeting the programmed cell death-1 pathway in genitourinary tumors: current progress and future perspectives
.
Curr Drug Metab
.
2017
;
18
(
8
):
700
711
.
155
Samstein
RM
,
Lee
CH
,
Shoushtari
AN
, et al.
Tumor mutational load predicts survival after immunotherapy across multiple cancer types
.
Nat Genet
.
2019
;
51
(
2
):
202
206
.
156
Wang
Z
,
Duan
J
,
Cai
S
, et al.
Assessment of blood tumor mutational burden as a potential biomarker for immunotherapy in patients with non-small cell lung cancer with use of a next-generation sequencing cancer gene panel [published online ahead of print February 28, 2019]
.
JAMA Oncol
. doi:
157
Dudley
JC
,
Lin
MT
,
Le
DT
,
Eshleman
JR
.
Microsatellite instability as a biomarker for PD-1 blockade
.
Clin Cancer Res
.
2016
;
22
(
4
):
813
820
.
158
Le
DT
,
Durham
JN
,
Smith
KN
, et al.
Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade
.
Science
.
2017
;
357
(
6349
):
409
413
.
159
US Food and Drug Administration
.
FDA News Release. FDA approves first cancer treatment for any solid tumor with a specific genetic feature
.
2017
. .
160
Abida
W
,
Cheng
ML
,
Armenia
J
, et al.
Analysis of the prevalence of microsatellite instability in prostate cancer and response to immune checkpoint blockade [published online ahead of print December 27, 2018]
.
JAMA Oncol
. doi:

Author notes

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

Presented in part at the 5th Princeton Integrated Pathology Symposium; April 15, 2018; Plainsboro, New Jersey.