Clinical genomics of renal epithelial tumors

Clinical genomics of renal epithelial tumors

Cancer Genetics 204 (2011) 285e297 REVIEW Clinical genomics of renal epithelial tumors Jill M. Hagenkord a,b, Zoran Gatalica c, Eric Jonasch d, Fede...

678KB Sizes 0 Downloads 50 Views

Cancer Genetics 204 (2011) 285e297

REVIEW

Clinical genomics of renal epithelial tumors Jill M. Hagenkord a,b, Zoran Gatalica c, Eric Jonasch d, Federico A. Monzon e,f,* a b

Department of Pathology, Creighton University School of Medicine, Omaha, NE, USA; iKaryos Diagnostics, Inc., Palo Alto, CA, USA; c Caris Life Sciences, Inc., Phoenix, AZ; d Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; e Department of Pathology and Laboratory Medicine, The Methodist Hospital and The Methodist Hospital Research Institute, Houston, TX, USA; f Department of Pathology, Weill-Cornell Medical College, New York, NY, USA Kidney and upper urinary tract cancers account for approximately 54,000 cases every year in the United States, and represent about 3.7% of adult malignancies, with more than 13,000 annual deaths. Classification of renal tumors is typically based on histomorphologic characteristics but, on occasion, morphologic characteristics are not sufficient. Each of the most common histologic subtypes harbors specific recurrent genetic abnormalities, such as deletion of 3p in conventional clear cell carcinoma, trisomy 7 and 17 in papillary renal cell carcinoma, multiple monosomies in chromophobe renal cell carcinoma, and a nearly diploid genome in benign oncocytomas. Knowledge of this information can provide diagnostic support and prognostic refinement in renal epithelial tumors. Identification of the specific subtype of a renal tumor is critical in guiding surveillance for recurrence and the appropriate use of targeted therapies. Cytogenomic arrays are increasingly being used as a clinical tool for genome-wide assessment of copy number and loss of heterozygosity in renal tumors. In addition, the improved understanding of the hereditary causes of renal tumors and their role in sporadic malignancies has led to the development of more effective targeted therapies. This review summarizes the genetic and genomic changes in the most common types of renal epithelial tumors and highlights the clinical implications of these aberrations. Keywords Renal cell carcinoma, diagnosis, prognosis, genetics, chromosomal imbalances, cytogenomic array, cytogenetics, SNP array, virtual karyotype ª 2011 Elsevier Inc. All rights reserved.

Kidney and upper urinary tract cancers account for approximately 54,000 cases every year in the United States, and represent about 3.7% of adult malignancies, with more than 13,000 annual deaths (1,2). Renal cell carcinoma (RCC) is the most common renal malignancy, with three common subtypes representing about 95% of all renal tumors: clear cell (ccRCC, 75% of RCC), papillary (pRCC, 10%), and chromophobe (chRCC, 5%). The fourth most common renal epithelial tumor is oncocytoma (OC, 5%), a benign neoplasm (Figure 1) (3). Although the majority of renal tumors occur in a sporadic fashion, approximately 2e4% occur in the setting of hereditary predisposition syndromes (4). There are four main hereditary cancer syndromes involving the renal epithelium: von Hippel Lindau (VHL) syndrome, which predisposes to development of ccRCC; hereditary papillary  syndrome renal cell carcinoma (HPRCC); Birt-Hogg-Dube

Received June 9, 2011; accepted June 10, 2011. * Corresponding author. E-mail address: [email protected] 2210-7762/$ - see front matter ª 2011 Elsevier Inc. All rights reserved. doi:10.1016/j.cancergen.2011.06.001

(BHDS), which predisposes to development of chRCC and OC; and hereditary leiomyomatosis and renal cell carcinoma (HLRCC), which predispose to pRCC and collecting duct carcinomas (3). Categorization of renal tumors can typically be done on the basis of histomorphologic characteristics. The correct determination of histologic subtype is critical because each has a distinct biologic behavior and therapeutic indications (5e7). However, morphologically challenging cases do occur. Even with generous sampling of large resection specimens, some tumors may have non-specific characteristics or overlap in morphologic features between tumor types. These morphologically challenging tumors are usually termed “unclassified renal cell carcinomas,” “renal cell carcinoma not otherwise specified,” or “eosinophilic renal carcinoma” in surgical pathology reports (3). Non-specific diagnoses confound efforts of the clinical team to predict tumor behavior, define appropriate follow up strategies, and guide therapeutic decisions. Difficulty in the morphologic assessment of renal tumors is compounded when evaluating scant tissue obtained from biopsy, or when architectural

286

J.M. Hagenkord et al.

Figure 1 Morphology and genomic profiles for the most common renal epithelial tumors. Each renal epithelial tumor has morphologic (left column) and chromosomal copy number profiles (right column) that are characteristic to each subtype (red, loss; blue, gain; red stripes, aUPD). (A) Clear cell RCC, n Z 130, with characteristic loss of 3p and frequent imbalances in chromosomes 5, 7, 9, and 14. (B) Papillary RCC, n Z 26, with characteristic gain of chromosomes 7 and 17 and frequent imbalances in chromosomes 3 (including aUPD), 12, 16, and 20. (C) Chromophobe RCC, n Z 18, note hypodiploid complement with frequent losses of chromosomes 1, 2, 6, 10, 13, 17, and 21. (D) Oncocytoma, n Z 30, with majority of tumors showing normal chromosomal complement and frequent complete or partial loss of chromosome 1.

context is lost, as in fine needle core biopsy specimens (8,9). Immunohistochemical (IHC) profiles can help categorize morphologically challenging tumors (10), but sometimes overlapping phenotypes or non-specific staining profiles render IHC unsuccessful in resolving the differential diagnostic dilemmas. Other laboratory studies, such as conventional cytogenetics, cytogenomic arrays, fluorescence in situ hybridization (FISH), or microsatellite polymerase chain reaction to assess loss of heterozygosity (LOH) can further aid in classification. These assays are able to assess copy number changes directly or by inference (LOH), either in a global or targeted approach. They are useful for classification of renal epithelial tumors because each of the most common subtypes harbors specific recurrent genomic abnormalities, as described below (Table 1, Figure 1) (11). Each assay has inherent strengths and weaknesses that should be considered when evaluating published data and when selecting the appropriate methods for clinical diagnostics (12). Cytogenomic arrays, such as array comparative genomic hybridization (aCGH) and single-

nucleotide polymorphism (SNP) arrays, have been used to classify RCC on the basis of the specific genomic profiles for each subtype (Figure 1) (13,14). Cytogenomic arrays can provide not only diagnostic support, but also identify additional genomic changes, some of which are associated with outcome in specific subtypes. Although most patients with renal epithelial tumors have an excellent prognosis, 30% of patients with initially organconfined ccRCC will develop metastases. In addition, due to the paucity of overt clinical manifestations of localized disease, up to 30% of patients will present with metastases at the time of initial diagnosis (2). Treatment for advanced or metastatic kidney cancer is a formidable challenge with the traditional therapies currently available. Investigation of the Mendelian single-gene syndromes, such as von Hippel Lindau syndrome (VHL: VHL gene), hereditary papillary renal  (BHD: cell carcinoma (HPRCC: MET gene), Birt-Hogg-Dube BHD gene), and hereditary leiomyomatosis renal cell carcinoma (HLRCC: FH gene), however, have provided an opportunity to develop pathway-specific therapies (6).

Clinical genomics of renal tumors Table 1

287

Frequency of classic chromosomal aberrations in renal epithelial neoplasms

Type of renal tumor

Classic cytogenetic findings

Clear cell RCC

del(3)(p): 3p14, 3p21, 3p25-p26

Papillary RCC

Trisomy 7 and/or 17

Chromophobe RCC

Loss of 1, 2, 6, 10, 13, 17 and/or 21

Mucinous tubular and spindle cell carcinoma Oncocytoma

Loss of 1, 14, and 15 Chr 1 loss or normal

% Cases with chromosomal abnormality 98 98 81 100 100 67/43 100/38 100 100/50 95 74 100 100 100 100 100

N

Platform

52 118 26 11 98 19/20 9/16 6 19 10 19 4 12 6

LOH CG aCGH FISH SNP array FISH CGH FISH SNP array LOH FISH aCGH SNP array SNP array

10 15

FISH SNP array

Subtype

Low /high grade Type 1/type 2 Type 1/type 2

Reference (25) (26) (29) (28) (27) (119) (120) (28) (27) (25) (73) (29) (27) (27) (73) (27)

Abbreviations: N, number; CG, cytogenetics.

The aim of this review is to summarize our current knowledge on the genetics of the most frequent sporadic and hereditary renal epithelial tumors and to describe how evaluation of these abnormalities can help refine diagnoses, indicate prognoses, and potentially tailor therapy in patients with these tumors.

Clear cell renal cell carcinoma Histologic features of clear cell renal carcinoma ccRCC is the most common renal malignancy. It is believed to originate from cells in the proximal tubule of the nephron. Histologically, ccRCC may have a solid, alveolar, or acinar cell architectural pattern, and they typically contain a regular network of thin-walled blood vessels (Figure 1A) (3). The cytoplasm is commonly filled with lipids and glycogen, which dissolve upon histologic processing and create a clear cytoplasm surrounded by distinct cell membranes, which gives this tumor its name. There may also be a population of cells with eosinophilic cytoplasm, particularly in higher-grade tumors, which can cause a histopathologic dilemma about proper classification.

and is the leading cause of mortality. Approximately 80% of individuals with VHL syndrome have an affected parent, and about 20% have de novo gene mutations. VHL mutations are highly penetrant, and almost all individuals who have a mutation express disease-related symptoms by age 65. The manifestations and severity are highly variable both within and between families, even among those with the same mutation. Approximately 72% of the inherited mutations are point mutations or small deletions/insertions that can be detected by sequence analysis of all three exons of the VHL gene, and the remaining 28% are partial or complete gene deletions that can be detected by duplication/deletion analysis (16,17). The second hit can result from wild-type VHL gene deletion (i.e., 3p loss in the tumors), point mutations, or hypermethylation of the VHL promoter (18). Acquired mutations in the VHL gene are commonly found in sporadic ccRCC tumors (19,20). Non-VHL related familial ccRCC has also been reported. Some of these cases have been explained by recurrent chromosomal translocations involving 3p, while a few others arise in patients with BHDS (see below; (21e24)). However, a large number of patients with familial ccRCC have no discernible genetic cause (22).

Hereditary clear cell renal cell carcinoma

Genetics of sporadic clear cell renal cell carcinoma

Von Hippel Lindau disease (VHL) is an autosomal dominant cancer predisposition syndrome due to a germline mutation in the VHL tumor suppressor gene at chromosome 3p25-p26 (chr3:10,183,319-10,193,744/hg19). Patients with VHL have an autosomal dominant predisposition to develop ccRCCs, as well as hemangioblastomas of the brain, spinal cord, and retina, pheochromocytomas, pancreatic cysts, and endolymphatic sac tumors of the inner ear (3,15). Clear cell renal cell carcinoma occurs in about 40% of individuals with VHL

Like the VHL-related ccRCC, the vast majority of sporadic ccRCCs have a loss of chromosomal material on the short arm of chromosome 3 (Table 1) (25e29). The deletion of 3p can encompass the entire chromosomal arm or a minimally deleted region from 3p21 to 3p25, which includes the VHL tumor suppressor gene. The VHL protein targets hypoxiainducible factors (HIF) for ubiquitin-mediated degradation (7). When VHL is mutated, HIF accumulates in the nucleus, leading to increased transcription of a number of

288

J.M. Hagenkord et al.

downstream pathway genes that promote angiogenesis, growth, and survival, such as vascular endothelial growth factor (VEGF), the glucose transporter (GLUT1) and transforming growth factor-beta (TGFB1) (6). Other recurrent chromosomal imbalances also occur in these tumors, with variable frequencies; some of the more common are trisomy 5 or 5q gain, trisomy 7, 9p loss, and 14q loss (Figure 1). Mutations in other genes located in 3p have been reported recently in sporadic tumors. PBRM1 (3p21.1, chr3:52,579,36852,713,739/hg19) appears to be mutated in approximately 40% of ccRCCs, while SETD2 (3p21.31, chr3:47,057,90047,205,467/hg19) seems to be mutated in approximately 3% (30,31). Both genes are involved in chromatin remodeling at the level of histone H3 modification and recognition. The biologic and clinical significance of these mutations is currently unknown.

Clinical relevance of genomics in clear cell renal cell carcinoma Genomic information can be used for diagnostic, prognostic, and predictive purposes in ccRCC. In morphologically ambiguous cases, identification of a deletion of 3p can help to clarify the diagnosis (27). However, monosomy and acquired uniparental disomy (aUPD) of chromosome 3 occasionally can be seen in pRCC and chRCC (Figure 1), so 3p loss should be evaluated in the context of morphology and, preferably, the complete genomic profile. Surgical excision is curative therapy in the majority of ccRCC cases, but up to 30% of patients with organ-confined tumors (stage pT1-2) develop metastases after nephrectomy (32). Nuclear grade has been shown to be the strongest morphologic predictor of poor outcome in ccRCC, but some studies indicate that it is not independent from overall stage (33e35). Specific chromosomal imbalances in ccRCC have Table 2

Genetic and genomic features associated with prognosis in clear cell renal cell carcinoma

Prognosis

Prognostic finding

Good

5q gain ccA gene expression profile 8q gain 9p loss

Poor

been associated with favorable outcomes or poor prognosis (Table 2). Gain of 5q has been reported as a good prognostic factor, while 8p gain, 9p loss, and 14q loss have been associated with poor outcome (25e27,29,36e40). Most of these associations, however, have been reported in small cohorts, except for 9p loss, whose association with poor outcome has been repeatedly observed in multiple studies (25,26,36,38,39). It is possible that these chromosomal imbalances might identify specific subsets of ccRCC patients, as it was recently shown that 9p loss appears to be specifically associated with poor outcome in patients with localized, small renal masses, and 14q loss seems to confer poor prognosis in patients with non-metastatic tumors (39,40). Prognostic categories based on gene expression or cytogenomic array profiling have also been reported. Brannon and coworkers described two subtypes of ccRCC (ccA/ ccB) based on gene expression profiling, with markedly different outcomes (41). In addition, Arai and coworkers reported that hierarchical clustering of ccRCCs based on patterns of chromosomal imbalances can identify two groups of patients with distinctly different recurrence-free survival and overall survival (OS; (42)). Understanding the genetic alterations in ccRCC and their impact on the VHL-HIF pathway has led to the development of targeted drugs for ccRCC patients with metastatic disease. Many of the HIF-regulated genes are targets for new therapeutic approaches for ccRCC, such as bevacizumab, sunitinib, sorafenib, and pazopanib (6,7). Bevacizumab, an antibody targeting VEGF-A, was shown to improve progression-free survival (PFS) in two randomized studies comparing bevacizumab plus interferon-alpha (IFN) to IFN monotherapy (43). Sorafenib, an oral small-molecule protein kinase inhibitor effective against the VEGF receptor (VEGFR), the platelet-derived growth factor receptor, and others, was approved in 2005 by the U.S. Food and Drug Administration (FDA) for treatment of metastatic ccRCC (44). It was closely

% Cases with feature

N

Platform

Reference

Association

(26) (41) (40) (26) (36) (25) (38)

Better overall survival (P Z 0.001) Better disease specific survival (P Z 0.002) Decreased overall survival (P < 0.0001) METs at Dx (P Z 0.006) 5y-CSS 43 vs. 88 (P < 0.001) Low vs. high stage Decreased survival (P Z 0.033 in multivariate analysis) Shorter recurrenceefree survival (P < 0.01) and higher likelihood of recurrence in pT1a tumors (P Z 0.01) Low vs. high stage Low vs. high grade (P Z 0.0143) Higher risk for recurrence (P Z 0.002) and shorter overall survival (P Z 0.030) in non-metastatic ccRCC Worse disease specific survival (P Z 0.002)

56.8 46

118 177

13 21 18 23 16

85 118 73 43 282

CG Gene expression microarray SNP array CG FISH LOH CG

13.8

703

CG / FISH

(39)

LOH aCGH SNP array

(25,29) (25,29) (40)

Gene expression microarray

(41)

14q loss

31 38 55

52 26 85

ccB gene expression profile

34

177

Abbreviations: N, number; CG, cytogenetics; Dx, diagnosis; CSS, cancerespecific survival.

Clinical genomics of renal tumors followed by sunitinib, which was approved in early 2006 (45). Pazopanib, another multiple kinase inhibitor, was approved in 2009 (46). These agents all provide a significant prolongation of PFS when compared to IFN therapy or to placebo, but it has been harder to confirm an OS advantage, possibly due to the crossover (change to a new therapy) to other effective agents on most of these clinical trials. The challenge at this point in time is to define tumor and host features responsible for innate and acquired resistance to anti-VEGF therapy, since no reliable predictive biomarkers have been identified to date. Our group recently reported that 8q gain seems to separate patients treated with anti-angiogenic agents into different subgroups with markedly different OS (40).

Papillary renal cell carcinoma Histologic features of papillary renal cell carcinoma Papillary renal cell carcinomas comprise approximately 10% of renal cell carcinomas (3). From a histological standpoint, pRCCs are characterized by tumor epithelial cells forming various proportions of papillae and tubules (Figure 1). The tumor papillae contain a delicate, fibrovascular core and can show aggregates of foamy macrophages (3). Papillary renal cell tumors have been morphologically divided into small (type 1) and large (type 2) cell tumors. Type 1 pRCC have papillae covered by small cells with scanty cytoplasm, arranged in a single layer on the papillary basement membrane. Type 2 pRCC tumor cells are typically of a higher grade with eosinophilic cytoplasm and pseudo-stratified nuclei on papillary cores (Figure 1) (3).

Hereditary papillary renal cell carcinoma Hereditary papillary renal cell carcinoma (HPRCC) is a highly penetrant hereditary renal cancer syndrome in which affected individuals are at risk for the development of bilateral, multifocal, type 1 pRCC (6,47). The kidney is the only organ known to be affected in HPRCC patients, and although the tumors are usually well differentiated, they are malignant and can metastasize. Unlike VHL, HPRCC involves the activation of a protooncogene, not inactivation of a tumor suppressor gene. HPRCC is due to missense mutations of the MET protooncogene at 7q31.2 (chr7:116,312,459-116,438,439/hg19), which results in the constitutive activation of the tyrosine kinase domain (18). The MET gene encodes the cell surface receptor for hepatocyte growth factor (HGF), also belonging to the tyrosine kinase class of receptor proteins (48), and MET gain-of-function mutations trigger cell proliferation, neovascularization, and increased cell motility (18). Molecular targeting approaches are being developed to inhibit the interaction of HGF and its receptor, and suppression of the downstream signaling cascade of activated c-MET (49). Type I pRCC can also be seen in BHDS (described further below). Hereditary leiomyomatosis renal cell carcinoma (HLRCC) is another hereditary cancer syndrome that may manifest with pRCC. HLRCC is an autosomal dominant disorder characterized by cutaneous leiomyomata, uterine leiomyomata in females, and/or a single renal tumor. Most renal

289 tumors are type 2 pRCC, but other types of aggressive renal carcinomas can be seen (50,51). Penetrance for pRCC is lower than for the cutaneous and uterine manifestations, with only 20e35% of patients developing pRCC (6). Affected individuals are typically born with a heterozygous mutation in the FH gene at 1q42-q44 (chr1:241660906-241683054/ hg19), encoding the enzyme fumarate hydratase (52). The majority of mutations in FH are point mutations or small deletions/insertions (50,51), although exonic and whole-gene deletions have been reported (53,54). FH is an essential enzyme for the conversion of fumarate to malate in the Krebs cycle. The loss of FH function impairs the Krebs cycle, giving way to glycolitic metabolism, cellular pseudo-hypoxia, and up-regulation of HIF and HIF-inducible transcripts (6,55). The diagnosis of HLRCC is clinical, based on the presence of multiple cutaneous leiomyomas or by a single cutaneous leiomyoma in the presence of a positive family history of HLRCC-related tumors. The diagnosis is confirmed by testing of fumarate hydratase enzyme activity in cultured skin fibroblasts or lymphoblastoid cells showing reduced activity (60%), or by molecular genetic testing (56). Studies of the tricarboxylic acid cycle and the VHL-HIF pathways have provided the foundation for metabolic therapeutic approaches in patients with HLRCC-associated kidney cancer as well as other hereditary and sporadic forms of RCC (7). In an interesting parallel to the HLRCC kindreds described above, germline mutations in subunits of the succinate dehydrogenase (SDH ) gene, another Krebs cycle enzyme, can predispose to early-onset kidney cancers in addition to paragangliomas and carry implications for medical surveillance (57,58). There have been reports of chromophobe RCC in patients with germline mutations in SDHB (hereditary paraganglioma/pheochromocytoma) (7,57,59).

Genetics of sporadic papillary renal cell carcinoma Mutations of MET have also been identified in a subset of tumors from patients with sporadic type 1 pRCC (7). The genomic profile of pRCC displays whole chromosomal gains, characteristically trisomy 7, which includes the HGF and MET genes, and trisomy 17. Gains of chromosomes 12, 16, and 20 are also frequently seen (Figure 1) (27,60). It has been proposed that renal papillary adenomas have only trisomy 7 and 17, while carcinomas are marked by additional numerical chromosomal changes (60). Trisomy 7 and 17 occur in both type 1 and type 2 pRCC (27,61), and debates persist in the literature regarding the validity of dividing pRCC into cytomorphologic subtypes. Type 1 pRCC, however, has been shown to have significantly better survival than type 2 (18,33,61).

Clinical relevance of genomics in papillary renal cell carcinoma RCC with papillary features and extensive clear cell changes may be difficult to classify as either ccRCC or pRCC. Xp11.2 translocation carcinoma (see below) also shows papillary architecture with clear cell features and is therefore usually within the differential diagnosis. Positive IHC staining for alpha-methylacyl-CoA-racemace and/or the presence of 7/17 gains can help make the diagnosis of pRCC in these

290 cases (62). Subtyping of pRCC is clinically important because tumor type is an independent predictor of outcome and the 5-year cancer-specific survival of pRCC is considerably better than that of ccRCC in the non-metastatic setting. In addition, the presence of 1p gain in pRCC has been reported as a poor prognostic factor (63). Unfortunately, once metastatic, the prognosis of pRCC is poor, and several studies suggest it is inferior to ccRCC (64). This is due, in part, to the lack of effective systemic therapies for this histologic subtype. Targeting the consequences of HIF up-regulation and reversing or modulating the inactivation of FH are potential molecular therapeutic targets for treatment of metastatic pRCC arising in the context of HLRCC (6). For patients with MET mutations or patients with sporadic pRCC, clinical trials targeting the MET pathway are currently underway. Other clinical trials have demonstrated that the mammalian target of rapamycin inhibition may be of some value (65), as is the epidermal growth factor blockade (66). Linkage between specific mutations or chromosomal patterns and response to specific agents may help refine therapy for these patients in the future (7).

Chromophobe renal cell carcinoma Histologic features of chromophobe renal cell carcinoma Chromophobe renal cell carcinoma (chRCC) accounts for approximately 5% of surgically removed renal epithelial tumors (3). From a histological standpoint, chRCC shows solid growth pattern with focal calcifications and broad fibrotic septae. The tumor cells are large, polygonal, with clear cytoplasm and prominent cell membranes. These cells are commonly admixed with smaller cells with granular, eosinophilc cytoplasm. In contrast to ccRCC, the blood vessels tend to be thick-walled and eccentrically hyalinized (3). Oncocytic or eosinophilic variants of chRCC are not uncommon and represent a diagnostic challenge by morphology (67).

Hereditary chromophobe renal cell carcinoma Birt-Hogg-Dube syndrome (BHDS) is an autosomal dominant condition characterized clinically by benign cutaneous tumors (fibrofolliculomas, trichodiscomas, and acrochordons), pulmonary cysts resulting in recurrent spontaneous pneumothoraces, and multifocal, bilateral renal cancer of varying histologies (68,69). BHDS shows clinical heterogeneity, and patients do not always have the characteristic phenotypical triad (70,71). The most common renal cell tumor seen in BHDS is the so-called oncocytic hybrid tumor (50%), which is a hybrid of morphologic features between oncocytoma and chromophobe histologic cell types. Chromophobe RCC presents in 33% of BHDS patients, and renal oncocytoma in 5% (6). Other types of renal tumors reported with lower frequency include ccRCC and pRCC (3). Mutations in the folliculin gene (FLCN, chr17:17115529-17140502/hg19) are considered causative (72). FLCN mutations have been reported in 88% of BHDS family members and can be found in several exons of the gene, although there is a mutational hotspot in

J.M. Hagenkord et al. exon 11 (70). A common 3.5-megabase deletion of 17p11.2 is present in approximately 70% of renal tumors from individuals affected with BHDS, supporting the tumor suppressor gene function of FLCN (7,18).

Genetics of somatic chromophobe renal cell carcinoma Chromophobe RCC typically has a hypodiploid genome, with whole chromosomal losses of Y, 1, 2, 6, 10, 13q, 17, and 21 (Figure 1) (27,73). These chromosomal losses have been demonstrated by metaphase cytogenetics, FISH, CGH, SNP array analysis, and microsatellite LOH (27,73e75). To date, no recurrent single gene mutational events have been reported for sporadic chRCC (76).

Clinical relevance of genomics in chromophobe renal cell carcinoma ChRCC are rarely aggressive so correct diagnosis is critical for appropriate patient management. Most chRCC are stage T1 and T2 (86%), and only 10% show extension through the renal capsule (3). Chromophobe RCC demonstrates morphologic overlap with ccRCC, especially when showing eosinophilic cytoplasm (77). The characteristic pattern of chromosomal losses is helpful in establishing the correct diagnosis (67,73). ChRCC and hybrid chRCC/oncocytoma tumors can be seen in BHDS. The function of the FLCN gene product, folliculin, is not well-characterized, but it has been linked to regulation of the mTOR, AMPK, LKB1, and TSC1/TSC2 pathways (7,78). The finding that mutations of two additional Krebs cycle enzymes (FH and SDH ), and disruption of the nutrient sensing pathway via mutation of FLCN all cause renal cell carcinoma provide important clues to the pathogenesis of RCC and the role of metabolism in carcinogenesis. Future work on targeting and modulating the glycolytic pathway in RCC will likely pay dividends in the discovery of new agents for the treatment of chRCC, and possibly also ccRCC.

Oncocytoma Histologic features of renal oncocytomas Renal oncocytomas (OC) are benign neoplasms which account for about 3e7% of all renal tumors. Oncocytoma tumor cells have abundant, granular eosinophilic cytoplasm (reflecting an abundance of mitocondria) arranged in nests, tubulocystic, solid, or trabecular patterns (3). One of the most prominent microscopic features of oncocytoma is the edematous, myxomatous, or hyalinized stroma that surrounds the tumor cells (Figure 1D). The nuclei are typically round and centrally located, but it is common to observe cells with large, bizarre nuclei. In addition, nuclear halos (a feature usually associated with chRCC) may be seen in OC, sometimes making distinction from chRCC difficult (79).

Clinical genomics of renal tumors

Hereditary renal oncocytomas As discussed above, renal oncocytomas can occur in patients with BHDS. Renal oncocytosis is a disease in which patients have a propensity to develop multiple synchronous and metachronous oncocytic tumors in the kidneys. This presentation has been observed in the presence of BHDS, but it is also seen in cases without BHDS and in cases with chronic renal failure (80,81).

Genetics of somatic renal oncocytomas The majority of renal oncocytomas (50e60%) show a normal chromosomal complement (Figure 1) (27,73,75,82). Approximately 40% of oncocytomas show complete or partial loss of chromosome 1. Other frequent changes include loss of Y (15%) and monosomy 14 (15%), which are usually seen in conjunction with chromosome 1 loss. Trisomy 7 and structural rearrangements involving 11q12-q13 have been reported in a small percentage of OC. Chromosomal abnormalities seem to be more frequent in sporadic tumors, while familial OC appears to have a higher frequency of cytogenetically normal tumors (83). To date, no recurrent single-gene mutational events have been reported for sporadic OC.

Clinical relevance of genomics of renal oncocytomas Renal oncocytomas behave in a benign fashion. Although one case of metastatic oncocytoma has been reported, this tumor was characterized only by IHC and no molecular analyses were performed (84). Thus, the true nature of this “metastatic OC” has not been established with certainty, and might represent misclassified chRCC or the eosinophilic variant of ccRCC. The distinction of OC from chRCC is one of the diagnostic pitfalls frequently encountered with renal epithelial tumors (77). Genomic profiles can be useful in correctly distinguishing between benign and malignant renal tumors. For example, many studies have reported that chromophobe RCC shows complex simultaneous losses of chromosomes 1, 2, 6, 10, 13, 17, and 21 (27,73); and, although occasional losses of each of these chromosomes have been reported in OC, the simultaneous loss of all these chromosomes is only seen in chRCC.

Rare renal epithelial tumors Xp11.2 translocation renal cell carcinomas Renal cell tumors associated with an Xp11.2 translocation typically occur in children or young adults (3). The architecture of these tumors shows characteristic morphologic features, with cells presenting abundant clear cytoplasm arranged in nests, papillary or pseudopapillary structures, and frequent psammoma bodies (calcium deposits). These tumors are characterized by the presence of a chromosomal rearrangement in Xp11.2. This translocation involves a breakpoint at the TFE3 gene on Xp11.2 and various fusion partners, including PRCC (1q21), ASPL (17q25), PSF (1p34), CLTC (17q23), and NonO (Xq12) (18,85). Positive

291 nuclear immunostaining for TFE3 is a surrogate marker for an Xp11 translocation RCC when observed in pediatric tumors, but the sensitivity of this stain in adult tumors has not been established (86,87). These tumors have an unpredictable behavior, with some patients presenting with widely metastatic disease and others showing more indolent clinical course. Lymph node status is currently the best prognostic marker for this entity (88,89). It is expected that knowledge of the molecular pathways affected by TFE3 overexpression may help guide therapy, because it appears that at least some Xp11.2 translocation tumors are associated with defects in mitotic checkpoint control. This might render them more responsive to chemotherapeutic agents such as vincrinstine and paclitaxel (18,90).

Mucinous tubular spindle cell carcinoma Mucinous tubular and spindle cell carcinomas (MTSCC) are an uncommon, low-grade renal epithelial tumors characterized morphologically by small, elongated tubules lined by cuboidal cells and/or cords of spindled cells separated by mucinous stroma (91). Not all MTSCC have classic morphology, however, and some show overlaping features with pRCC (92,93). Genetically, these tumors are characterized by multiple chromosomal losses, but the exact losses described have varied between method and investigator (91). Several groups have reported complete or partial losses of 1, 4, 6, 8, 13, 14, 15, and 22 when using genome-wide methods (27,94,95). Compared with MTSCCs, other RCCs with spindle cell features (sarcomatoid change) behave aggressively, thus underscoring the importance of correct categorization of these tumor types. It has been suggested that MTSCC is a variant of papillary renal cell carcinoma, based on morphological and immunohistochemical similarities (91,96), although the chromosomal profiles are markedly different (27,97).

Discussion Genetic and genomic information can assist in the diagnosis, prognosis, and therapeutic management of renal epithelial tumors. Often, the diagnosis is straightforward for renal tumors, based on their classic histologic characteristics. A small but significant percentage of tumors, however, have ambiguous morphologic features, and approximately 5% are reported as “unclassified” due to overlapping morphologic characteristics, even in well-sampled nephrectomy specimens (3,98). Proper diagnosis is critical to ensure appropriate patient management because each subtype of renal epithelial tumor has a specific recurrence risk and treatment regimen. The 5-year survival and disease-free progression of renal cell carcinomas varies by subtype: chRCC (100 and 94%), pRCC (86 and 88%), ccRCC (76 and 70%), RCC unclassified (24 and 18%) (5). Oncocytomas are benign neoplasms with no risk of metastasis, but they can morphologically mimic some renal cell carcinomas and are often part of the differential diagnosis. Cytogenomic profiles obtained from array-based studies can be useful in the evaluation of diagnostically challenging renal epithelial tumors, even in cases where FISH and IHC yield ambiguous results (27,99).

292 Kim et al. reported that SNP arrays could resolve 94% of morphologically challenging renal tumors (27). In addition, Viera et al. reported that the genetic diagnosis of renal tumors by comparative genomic hybridization on FNA biopsies can improve differential diagnosis (98). Core biopsies (CB) or fine-needle aspirates (FNA) are now frequently used for diagnostic purposes and to guide appropriate intervention. CB/FNA sampling poses an additional diagnostic challenge due to scant tissue, sampling error, and loss of architectural context (8,9). These diagnostic modalities are being used more frequently because of the increased detection of incidental kidney tumors with abdominal CT and/ or ultrasonography from the evaluation of an unrelated problem (98). Surgical resection is generally indicated in patients with a solid renal mass to determine if the mass is malignant (100). Surgery, if performed, provides not only diagnostic tissue, but in most cases also definitive therapy. For the elderly and others at high surgical risk, surgery may be deferred if the tumor is radiologically benign-appearing or percutaneous radiofrequency ablation or cryotherapy may be an alternative. In these cases, CB or FNA are important to confirm the diagnosis prior to treatment (101). In this context, it is expected that evaluation of genomic features of renal epithelial tumors will assist in providing diagnostic and prognostic information in samples with limited tissue. In addition to assisting in correct classification, the ability to identify tumors within each subtype that are most likely to recur would allow management strategies to be further tailored to each patient’s risk. For ccRCC and pRCC, the strongest predictors of survival are tumor stage, nuclear grade, and necrosis (33,102,103). However, there is currently no reliable biomarker that can predict metastatic or local recurrence in patients with organ confined tumors (stages I and II), and the significance of histologic subtype for prognosis is not independent of stage (102,104). Cytogenomic profiles can assess the diagnostic genomic changes as well as genomic changes associated with prognosis. For example, detection of a 3p deletion in a morphologically challenging renal tumor would support the diagnosis of ccRCC because nearly 100% of ccRCC demonstrate this feature (Figure 1). In addition, a cytogenomic profile could detect the deletion of 9p or 14q, both associated with poor prognosis (Table 2) (26,29,36,37,105). This information could allow surgeons and oncologists to design appropriate monitoring and interventions for those tumors that are more likely to recur, such as changing monitoring practices or the use of adjuvant treatments. The emergence of targeted therapies, such as antiangiogenic drugs and mTOR inhibitors, has dramatically improved the progression-free survival (PFS) of patients affected by ccRCC, and is gradually improving OS. Understanding of the VHL-HIF-VEGF cellular hypoxia pathway has provided the foundation for the development of treatment strategies based on tyrosine kinase inhibitors (TKIs) with activity against VEGF receptors, such as sunitinib and sorafenib (Table 3) (7). Likewise, MET mutations are seen in hereditary and sporadic pRCC, and clinical trials targeting the MET pathway are ongoing (7). Preclinical studies are underway targeting the BHD gene pathway for BHDS and sporadic chRCC (7). At this moment, there is no evidence that mutational status on VHL or MET genes modifies a tumor’s response to targeted agents. However, it is possible that as

J.M. Hagenkord et al. more specific targeted drugs are developed, the evaluation of mutational status might be necessary to qualify for therapy. Assessing the cytogenomic profiles of renal epithelial tumors allows for diagnostic support, refined prognostic information, and can guide appropriate choice of therapy. There are several techniques in use in clinical and research laboratories to assess chromosome copy number and/or LOH status, including conventional cytogenetics, FISH analysis, microsatellite LOH analysis, and array-based genomic analysis (aCGH or SNP arrays). The great majority of diagnostic and prognostic alterations can be detected with any of these approaches. Each technique has strengths and limitations, however, and these differences may explain some of the discrepancies reported in the literature regarding frequency of chromosomal imbalances, and should be kept in mind when guiding clinical testing strategies and evaluating diagnostic results. Conventional cytogenetics is the method of choice when assessing for non-recurrent balanced translocations, such as those present in familial non-VHL ccRCC. FISH can be used to detect the presence of recurrent chromosomal translocations, such as those present in pediatric tumors involving the Xp11.2 locus. Array-based genomic analysis is quickly becoming the method of choice for comprehensive evaluation of chromosomal imbalances in kidney and other solid tumors. Variability in the frequency of chromosome copy number and LOH status in renal tumors reported in the literature may be due to limitations in different assays (Table 1). In addition, variability may also arise from improper data normalization in tumors with hyperdiploid or hypodiploid genomes when using array-based techniques. Most software programs for arraybased copy number analysis use a mean or median normalization step to adjust for differences in signal intensity between different samples. This approach assumes that the overall ploidy of the test genome is close to two. When the test genome is hypo- or hyperdiploid, the normalization process results in the test genome log2 ratios being adjusted by an incorrect factor. In the case of MTSCC or chRCC, the genome is hypodiploid and the default normalization will make the diploid regions appear to be triploid. Without corrections to the normalization process, CGH and array-based genomic analysis may yield false chromosomal gains (106). The rapid development of NGS technologies may allow the simultaneous analysis of mutational status, translocation events, and chromosome copy number changes with a single technology in the near future (107,108). Although not specifically applied to renal tumors, promising advances in copy number analysis with NGS have been shown to be useful in paraffin-embedded tissues and, thus, could enable the routine clinical utilization of this approach (109).

Conclusions The correct categorization of renal tumors has an immediate impact on diagnosis, prognosis, therapy, and post-surgical surveillance. Although the chromosomal lesions that characterize each subtype of renal epithelial neoplasms have been known for some time, this knowledge has not been incorporated in the routine diagnostic evaluation of these tumors. The advent of new technologies, such as cytogenomic arrays, enables the use of cytogenomic profiles in the

Relationship between genetic alterations and novel targeted approaches for treatment of renal cell carcinoma Targeted agents in clinical trials

Syndrome

Phenotype

Gene/location

Pathways affected

FDA-approved compounds

Von Hippel Lindau Disease (110)

Kidney  Renal cysts  Mulifocal ccRCC Endocrine  Pheochromcytomas CNS  Retinal angiomas  Cerebellar and spinal hemangioblastomas Pancreatic  Pancreatic cysts  Pancreatic neuroendocrine tumors Kidney  Multifocal papillary RCC Skin  Fibrofolliculomas  Trichodiscomas  Achrocordons Lung  Lung cysts Kidney  Oncocytomas  Chromophobe RCC  ccRCC Kidney  Papillary type II RCC

VHL/3p25(111)

HIF regulation Primary cilium Collagen IV homeostasis Fibonectin homesostasis

Bevacizumab (43) Sunitinib (45) Sorafenib (44) Pazopanib (46)

Axitinib Tivozanib

None

XL184 GSK1363089 None

Hereditary papillary RCC (7) Birt Hogg Dube Syndrome (113)

Hereditary leiomyomatosis RCC syndrome (114) Hereditary paraganglioma syndrome Tuberous sclerosis

Endocrine organs  Paragangliomas Kidney  ccRCC CNS  Cortical tubers  Subependymal giant cell astrocytomas (SEGA) Skin  Ash leaf spots  Shagreen patch Kidney  Renal angiomyolipomas  ccRCC

MET/7q31(112) Folliculin/17p11.2(72)

Nutrient Sensing AMPK signaling/ PI3K pathway via mTOR

None

Fumarate hydratase/ 1q42-3(54)

Krebs cycle HIF regulation

None

Bevacizumabþ erlotinib (NCT01130519)

Succinate dehydrogenase D/ 1p36(115)

Krebs cycle

None

None

Hamartin/9q34(116) Tuberin/16p13(117)

PI3 Kinase pathway/mTOR homeostasis

Everolimus for SEGA(118)

Everolimus (multiple studies) Sirolimus Temsirolimus

Preclinical models Rodent knockout models

Clinical genomics of renal tumors

Table 3

Rodent knockout models

Rodent knockout models

Eker Rat Murine knockout models

293

294 clinical evaluation of renal epithelial tumors, even in technically challenging samples, such as FFPE and scant biopsies. Defining the prognosis for a patient with renal cell carcinoma (RCC) is currently more art than science, and is based largely upon algorithms that evaluate clinical features and reflect overall patient health status rather than tumor biology. The recently reported associations of mutation status, gene expression profiles, and chromosomal imbalances with prognosis in ccRCC and pRCC patients suggest that molecular classification of renal epithelial tumors will be an important component of clinical management in these patients. Similarly, despite the fact that therapy for RCC was launched from discoveries in the basic cancer biology of this disease, few reliable biomarkers exist to predict response to targeted therapy. The emergence of targeted therapies directed to specific genetic alterations and the association of specific genetic events with response to therapy would likely reinforce the need for molecular characterization of tumors for therapeutic selection. In addition, novel technologies, such as next-generation sequencing (NGS), might allow for a refined molecular classification of renal tumors, which should inform the development of more targeted agents.

References 1. Cancer facts & figures 2008. Atlanta: American Cancer Society; 2008. 2. DeVita VT, Hellman S, Rosenberg SA. Cancer, principles and practice of oncology. Philadelphia: Lippincott, Williams & Wilkins; 2001. 3. Eble JN, Sauter G, Epstein J, et al. Pathology and Genetics of Tumours of the Urinary System and Male Genital Organs. Lyon: IARC; 2004. 4. Axwijk PH, Kluijt I, de Jong D, et al. Hereditary causes of kidney tumours. Eur J Clin Invest 2010;40:433e439. 5. 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;26:281e291. 6. Rosner I, Bratslavsky G, Pinto PA, et al. The clinical implications of the genetics of renal cell carcinoma. Urol Oncol 2009; 27:131e136. 7. Linehan WM, Pinto PA, Bratslavsky G, et al. Hereditary kidney cancer: unique opportunity for disease-based therapy. Cancer 2009;115:2252e2261. 8. Volpe A, Kachura JR, Geddie WR, et al. Techniques, safety and accuracy of sampling of renal tumors by fine needle aspiration and core biopsy. J Urol 2007;178:379e386. 9. Lebret T, Poulain JE, Molinie V, et al. Percutaneous core biopsy for renal masses: indications, accuracy and results. J Urol 2007;178:1184e1188. 10. Allory Y, Bazille C, Vieillefond A, et al. Profiling and classification tree applied to renal epithelial tumours. Histopathology 2008;52:158e166. 11. van den Berg E, Dijkhuizen T, Oosterhuis JW, et al. Cytogenetic classification of renal cell cancer. Cancer Genet Cytogenet 1997;95:103e107. 12. Bridge JA, Cushman-Vokoun AM. Molecular diagnostics of soft tissue tumors. Arch Pathol Lab Med 2011;135:588e601. 13. Wilhelm M, Veltman JA, Olshen AB, et al. Array-based comparative genomic hybridization for the differential diagnosis of renal cell cancer. Cancer Res 2002;62:957e960. 14. Monzon FA, Hagenkord J, Lyons-Weiler M, et al. Whole genome SNP arrays as a potential diagnostic tool for the detection of characteristic chromosomal aberrations in renal epithelial tumors. Mod Pathol 2008;21:599e608.

J.M. Hagenkord et al. 15. Uchida K, Miyao N, Masumori N, et al. Recurrence of renal cell carcinoma more than 5 years after nephrectomy. Int J Urol 2002;9:19e23. 16. Stolle C, Glenn G, Zbar B, et al. Improved detection of germline mutations in the von Hippel-Lindau disease tumor suppressor gene. Hum Mutat 1998;12:417e423. 17. Banks RE, Tirukonda P, Taylor C, et al. Genetic and epigenetic analysis of von Hippel-Lindau (VHL) gene alterations and relationship with clinical variables in sporadic renal cancer. Cancer Res 2006;66:2000e2011. 18. Cheng L, Zhang S, MacLennan GT, et al. Molecular and cytogenetic insights into the pathogenesis, classification, differential diagnosis, and prognosis of renal epithelial neoplasms. Hum Pathol 2009;40:10e29. 19. Iliopoulos O. von Hippel-Lindau disease: genetic and clinical observations. Front Horm Res 2001;28:131e166. 20. Kim WY, Kaelin WG. Role of VHL gene mutation in human cancer. J Clin Oncol 2004;22:4991e5004. 21. Poland KS, Azim M, Folsom M, et al. A constitutional balanced t(3;8)(p14;q24.1) translocation results in disruption of the TRC8 gene and predisposition to clear cell renal cell carcinoma. Genes Chromosomes Cancer 2007;46:805e812. 22. Woodward ER, Ricketts C, Killick P, et al. Familial non-VHL clear cell (conventional) renal cell carcinoma: clinical features, segregation analysis, and mutation analysis of FLCN. Clin Cancer Res 2008;14:5925e5930. 23. McKay L, Frydenberg M, Lipton L, et al. Case report: renal cell carcinoma segregating with a t(2;3)(q37.3;q13.2) chromosomal translocation in an Ashkenazi Jewish family. Fam Cancer 2010: 1e5. 24. Bodmer Dl, van den Hurk W, van Groningen JJM, et al. Understanding familial and non-familial renal cell cancer. Hum Mol Genet 2002;11:2489e2498. 25. Bugert P, Kovacs G. Molecular differential diagnosis of renal cell carcinomas by microsatellite analysis. Am J Pathol 1996; 149:2081e2088. 26. Gunawan B, Huber W, Holtrup M, et al. Prognostic impacts of cytogenetic findings in clear cell renal cell carcinoma: gain of 5q31-qter predicts a distinct clinical phenotype with favorable prognosis. Cancer Res 2001;61:7731e7738. 27. Kim HJ, Shen SS, Ayala AG, et al. Virtual-karyotyping with SNP microarrays in morphologically challenging renal cell neoplasms: a practical and useful diagnostic modality. Am J Surg Pathol 2009;33:1276e1286. 28. Receveur AO, Couturier J, Molinie V, et al. Characterization of quantitative chromosomal abnormalities in renal cell carcinomas by interphase four-color fluorescence in situ hybridization. Cancer Genet Cytogenet 2005;158:110e118. 29. Yoshimoto T, Matsuura K, Karnan S, et al. High-resolution analysis of DNA copy number alterations and gene expression in renal clear cell carcinoma. J Pathol 2007;213:392e401. 30. Varela I, Tarpey P, Raine K, et al. Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma. Nature 2011;469:539e542. 31. Dalgliesh GL, Furge K, Greenman C, et al. Systematic sequencing of renal carcinoma reveals inactivation of histone modifying genes. Nature 2010;463:360e363. 32. Motzer RJ, Bander NH, Nanus DM. Renal-cell carcinoma. N Engl J Med 1996;335:865e875. 33. Delahunt B, Bethwaite PB, Nacey JN. Outcome prediction for renal cell carcinoma: evaluation of prognostic factors for tumours divided according to histological subtype. Pathology 2007;39:459e465. 34. Patard J-J, Leray E, Rioux-Leclercq N, et al. Prognostic value of histologic subtypes in renal cell carcinoma: a multicenter experience. J Clin Oncol 2005;23:2763e2771. 35. Kim H, Cho NH, Kim D-S, et al. Renal cell carcinoma in South Korea: a multicenter study. Hum Pathol 2004;35:1556e1563.

Clinical genomics of renal tumors 36. Brunelli M, Eccher A, Gobbo S, et al. Loss of chromosome 9p is an independent prognostic factor in patients with clear cell renal cell carcinoma. Mod Pathol 2008;21:1e6. 37. Kardas I, Mrozek K, Babinska M, et al. Cytogenetic and molecular findings in 75 clear cell renal cell carcinomas. Oncol Rep 2005;13:949e956. 38. Klatte T, Rao PN, de Martino M, et al. Cytogenetic Profile Predicts Prognosis of Patients With Clear Cell Renal Cell Carcinoma. J Clin Oncol 2009;27:746e753. 39. La Rochelle J, Klatte T, Dastane A, et al. Chromosome 9p deletions identify an aggressive phenotype of clear cell renal cell carcinoma. Cancer 2010;116:4696e4702. 40. Monzon FA, Alvarez K, Peterson LE, et al. Chromosome 14q loss defines a molecular subtype of clear cell renal cell carcinoma associated with poor prognosis. Mod Pathol, in press. 41. Brannon AR, Reddy A, Seiler M, et al. Molecular stratification of clear cell renal cell carcinoma by consensus clustering reveals distinct subtypes and survival patterns. Genes Cancer 2010;1: 152e163. 42. Arai E, Ushijima S, Tsuda H, et al. Genetic clustering of clear cell renal cell carcinoma based on array-comparative genomic hybridization: its association with DNA methylation alteration and patient outcome. Clin Cancer Res 2008;14:5531e5539. 43. Escudier B, Pluzanska A, Koralewski P, et al. Bevacizumab plus interferon alpha-2a for treatment of metastatic renal cell carcinoma: a randomised, double-blind phase III trial. Lancet 2007;370:2103e2111. 44. Escudier B, Eisen T, Stadler WM, et al. Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med 2007;356: 125e134. 45. Motzer RJ, Hutson TE, Tomczak P, et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med 2007;356:115e124. 46. Sternberg CN, Davis ID, Mardiak J, et al. Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial. J Clin Oncol 2010;28:1061e1068. 47. Zbar B, Tory K, Merino M, et al. Hereditary papillary renal cell carcinoma. J Urol 1994;151:561e566. 48. Bottaro DP, Rubin JS, Faletto DL, et al. Identification of the hepatocyte growth factor receptor as the c-met proto-oncogene product. Science 1991;251:802e804. 49. Linehan WM, Vasselli J, Srinivasan R, et al. Genetic basis of cancer of the kidney: disease-specific approaches to therapy. Clin Cancer Res 2004;10:6282Se6289S. 50. Toro JR, Nickerson ML, Wei MH, et al. Mutations in the fumarate hydratase gene cause hereditary leiomyomatosis and renal cell cancer in families in North America. Am J Hum Genet 2003;73:95e106. 51. Wei MH, Toure O, Glenn GM, et al. Novel mutations in FH and expansion of the spectrum of phenotypes expressed in families with hereditary leiomyomatosis and renal cell cancer. J Med Genet 2006;43:18e27. 52. Lehtonen HJ, Kiuru M, Ylisaukko-Oja SK, et al. Increased risk of cancer in patients with fumarate hydratase germline mutation. J Med Genet 2006;43:523e526. 53. Smit DL, Mensenkamp AR, Badeloe S, et al. Hereditary leiomyomatosis and renal cell cancer in families referred for fumarate hydratase germline mutation analysis. Clin Genet 2011;79:49e59. 54. 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; 30:406e410. 55. Isaacs JS, Jung YJ, Mole DR, et al. HIF overexpression correlates with biallelic loss of fumarate hydratase in renal cancer: novel role of fumarate in regulation of HIF stability. Cancer Cell 2005;8:143e153.

295 56. Pithukpakorn M, Wei MH, Toure O, et al. Fumarate hydratase enzyme activity in lymphoblastoid cells and fibroblasts of individuals in families with hereditary leiomyomatosis and renal cell cancer. J Med Genet 2006;43:755e762. 57. Vanharanta S, Buchta M, McWhinney SR, et al. Early-onset renal cell carcinoma as a novel extraparaganglial component of SDHB-associated heritable paraganglioma. Am J Hum Genet 2004;74:153e159. 58. Neumann HP, Pawlu C, Peczkowska M, et al. Distinct clinical features of paraganglioma syndromes associated with SDHB and SDHD gene mutations. J Amer Med Assoc 2004;292: 943e951. 59. Ricketts C, Woodward ER, Killick P, et al. Germline SDHB mutations and familial renal cell carcinoma. J Natl Cancer Inst 2008;100:1260e1262. 60. Kovacs G, Fuzesi L, Emanual A, et al. Cytogenetics of papillary renal cell tumors. Genes Chromosomes Cancer 1991;3: 249e255. 61. Balint I, Szponar A, Jauch A, et al. Trisomy 7 and 17 mark papillary renal cell tumours irrespectively of variation of the phenotype. J Clin Pathol 2009;62:892e895. 62. Haudebourg J, Hoch B, Fabas T, et al. Strength of molecular cytogenetic analyses for adjusting the diagnosis of renal cell carcinomas with both clear cells and papillary features: a study of three cases. Virchows Arch 2010;457:397e404. 63. Szponar A, Zubakov D, Pawlak J, et al. Three genetic developmental stages of papillary renal cell tumors: duplication of chromosome 1q marks fatal progression. Int J Cancer 2009; 124:2071e2076. 64. Ronnen EA, Kondagunta GV, Ishill N, et al. Treatment outcome for metastatic papillary renal cell carcinoma patients. Cancer 2006;107:2617e2621. 65. Dutcher JP, de Souza P, McDermott D, et al. Effect of temsirolimus versus interferon-alpha on outcome of patients with advanced renal cell carcinoma of different tumor histologies. Med Oncol 2009;26:202e209. 66. Gordon MS, Hussey M, Nagle RB, et al. Phase II study of erlotinib in patients with locally advanced or metastatic papillary histology renal cell cancer: SWOG S0317. J Clin Oncol 2009; 27:5788e5793. 67. Monzon F, Shen S, Kemp B, et al. Oncocytic Renal Neoplasms. A molecular approach to a common diagnostic challenge. Pathol Case Rev 2010;15:20e24. 68. Birt AR, Hogg GR, Dube WJ. Hereditary multiple fibrofolliculomas with trichodiscomas and acrochordons. Arch Dermatol 1977;113:1674e1677. 69. Toro JR, Pautler SE, Stewart L, et al. Lung cysts, spontaneous pneumothorax, and genetic associations in 89 families with Birt-Hogg-Dube syndrome. Am J Respir Crit Care Med 2007; 175:1044e1053. 70. Toro JR, Wei MH, Glenn GM, et al. BHD mutations, clinical and molecular genetic investigations of Birt-Hogg-Dube syndrome: a new series of 50 families and a review of published reports. J Med Genet 2008;45:321e331. 71. Kunogi M, Kurihara M, Ikegami TS, et al. Clinical and genetic spectrum of Birt-Hogg-Dube syndrome patients in whom pneumothorax and/or multiple lung cysts are the presenting feature. J Med Genet 2010;47:281e287. 72. 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;2:157e164. 73. Brunelli M, Eble JN, Zhang S, et al. Eosinophilic and classic chromophobe renal cell carcinomas have similar frequent losses of multiple chromosomes from among chromosomes 1, 2, 6, 10, and 17, and this pattern of genetic abnormality is not present in renal oncocytoma. Mod Pathol 2005;18: 161e169.

296 74. Jones TD, Eble JN, Cheng L. Application of molecular diagnostic techniques to renal epithelial neoplasms. Clin Lab Med 2005;25:279e303. 75. Speicher MR, Schoell B, du Manoir S, et al. Specific loss of chromosomes 1, 2, 6, 10, 13, 17, and 21 in chromophobe renal cell carcinomas revealed by comparative genomic hybridization. Am J Pathol 1994;145:356e364. 76. Petersson F, Gatalica Z, Grossmann P, et al. Sporadic hybrid oncocytic/chromophobe tumor of the kidney: a clinicopathologic, histomorphologic, immunohistochemical, ultrastructural, and molecular cytogenetic study of 14 cases. Virchows Arch 2010;456:355e365. 77. Abrahams NA, Tamboli P. Oncocytic Renal neoplasms: diagnostic considerations. Clin Lab Med 2005;25:317e339. 78. Maher ER. Genetics of familial renal cancers. Nephron 2011; 118:e21ee26. 79. Petersson F, Gatalic Z, Grossmann P, et al. Sporadic hybrid oncocytic/chromophobe tumor of the kidney: a clinicopathologic, histomorphologic, immunohistochemical, ultrastructural, and molecular cytogenetic study of 14 cases. Virchows Arch 2010;456:355e365. 80. Al-Saleem T, Cairns P, Dulaimi EA, et al. The genetics of renal oncocytosis: a possible model for neoplastic progression. Cancer Genet Cytogenet 2004;152:23e28. 81. Tickoo SK, Reuter VE, Amin MB, et al. Renal oncocytosis: a morphologic study of fourteen cases. Am J Surg Pathol 1999; 23:1094e1101. 82. Brunelli M, Gobbo S, Cossu-Rocca P, et al. Chromosomal gains in the sarcomatoid transformation of chromophobe renal cell carcinoma. Mod Pathol 2007;20:303e309. 83. Junker K, Weirich G, Moravek P, et al. Familial and Sporadic Renal Oncocytomas. A comparative molecular and genetic analysis. Eur Urol 2001;40:330e336. 84. Oxley JD, Sullivan J, Mitchelmore A, et al. Metastatic renal oncocytoma. J Clin Pathol 2007;60:720e722. 85. Argani P. The evolving story of renal translocation carcinomas. Am J Clin Pathol 2006;126:332e334. 86. 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;26:1553e1566. 87. Mir MC, Trilla E, De Torres IM, et al. Altered transcription factor E3 expression in unclassified adult renal cell carcinoma indicates adverse pathological features and poor outcome. BJU Int [Epub ahead of print]. doi: 10.1111/j.1464e410X.2010.09818.x. 88. Malouf GG, Camparo P, Molinie V, et al. Transcription factor E3 and transcription factor EB renal cell carcinomas: clinical features, biological behavior and prognostic factors. J Urol 2011;185:24e29. 89. Ross H, Argani P. Xp11 translocation renal cell carcinoma. Pathology 2010;42:369e373. 90. Weterman MA, van Groningen JJ, Tertoolen L, et al. Impairment of MAD2B-PRCC interaction in mitotic checkpoint defective t(X;1)-positive renal cell carcinomas. Proc Natl Acad Sci USA 2001;98:13808e13813. 91. 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;11:13e21. 92. Fine SW, Argani P, DeMarzo AM, et al. Expanding the histologic spectrum of mucinous tubular and spindle cell carcinoma of the kidney. Am J Surg Pathol 2006;30:1554e1560. 93. Argani P, Netto GJ, Parwani AV. Papillary renal cell carcinoma with low-grade spindle cell foci: a mimic of mucinous tubular and spindle cell carcinoma. Am J Surg Pathol 2008;32:1353e1359. 94. Rakozy C, Schmahl GE, Bogner S, et al. Low-grade tubularmucinous renal neoplasms: morphologic, immunohistochemical, and genetic features. Mod Pathol 2002;15:1162e1171.

J.M. Hagenkord et al. 95. Brandal P, Lie AK, Bassarova A, et al. Genomic aberrations in mucinous tubular and spindle cell renal cell carcinomas. Mod Pathol 2006;19:186e194. 96. 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;30:13e19. 97. 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;19:488e493. 98. Vieira J, Henrique R, Ribeiro FR, et al. Feasibility of differential diagnosis of kidney tumors by comparative genomic hybridization of fine needle aspiration biopsies. Genes Chromosomes Cancer 2010;49:935e947. 99. Hagenkord JM, Parwani AV, Lyons-Weiler MA, et al. Virtual karyotyping with SNP microarrays reduces uncertainty in the diagnosis of renal epithelial tumors. Diagn Pathol 2008;3:44. 100. Bosniak MA, Birnbaum BA, Krinsky GA, et al. Small renal parenchymal neoplasms: further observations on growth. Radiology 1995;197:589e597. 101. Abouassaly R, Lane BR, Novick AC. Active surveillance of renal masses in elderly patients. J Urol 2008;180:505e508. 102. Lam JS, Shvarts O, Leppert JT, et al. Renal cell carcinoma 2005: new frontiers in staging, prognostication and targeted molecular therapy. J Urol 2005;173:1853e1862. 103. Motzer RJ, Bacik J, Mazumdar M. Prognostic factors for survival of patients with stage IV renal cell carcinoma: memorial sloan-kettering cancer center experience. Clin Cancer Res 2004;10:6302Se6303S. 104. Amin MB, Tamboli P. Impact of histologic subtyping of renal epithelial neoplasms: authors’ reply. Am J Surg Pathol 2003; 27:1022e1024. 105. Moch H, Presti JC Jr, Sauter G, et al. Genetic aberrations detected by comparative genomic hybridization are associated with clinical outcome in renal cell carcinoma. Cancer Res 1996;56:27e30. 106. Staaf J, Jonsson G, Ringner M, et al. Normalization of arrayCGH data: influence of copy number imbalances. BMC genomics 2007;8:382. 107. Ding L, Wendl MC, Koboldt DC, et al. Analysis of nextgeneration genomic data in cancer: accomplishments and challenges. Hum Mol Genet 2010;19:R188eR196. 108. Talkowski ME, Ernst C, Heilbut A, et al. Next-generation sequencing strategies enable routine detection of balanced chromosome rearrangements for clinical diagnostics and genetic research. Am J Hum Genet 2011;88:469e481. 109. Wood HM, Belvedere O, Conway C, et al. Using nextgeneration sequencing for high resolution multiplex analysis of copy number variation from nanogram quantities of DNA from formalin-fixed paraffin-embedded specimens. Nucleic Acids Res 2010;38:e151. 110. Lonser RR, Glenn GM, Walther M, et al. von Hippel-Lindau disease. Lancet 2003;361:2059e2067. 111. Latif F, Tory K, Gnarra J, et al. Identification of the von HippelLindau disease tumor suppressor gene. Science 1993;260: 1317e1320. 112. Schmidt L, Duh FM, Chen F, et al. Germline and somatic mutations in the tyrosine kinase domain of the MET protooncogene in papillary renal carcinomas. Nat Genet 1997;16: 68e73. 113. Menko FH, van Steensel MA, Giraud S, et al. Birt-Hogg-Dube syndrome: diagnosis and management. Lancet Oncol 2009;10: 1199e1206. 114. Sudarshan S, Pinto PA, Neckers L, et al. Mechanisms of disease: hereditary leiomyomatosis and renal cell cancerea distinct form of hereditary kidney cancer. Nat Clin Pract Urol 2007;4:104e110.

Clinical genomics of renal tumors 115. Astuti D, Douglas F, Lennard TW, et al. Germline SDHD mutation in familial phaeochromocytoma. Lancet 2001;357: 1181e1182. 116. van Slegtenhorst M, de Hoogt R, Hermans C, et al. Identification of the tuberous sclerosis gene TSC1 on chromosome 9q34. Science 1997;277:805e808. 117. European Chromosome 16 Tuberous Sclerosis Consortium. Identification and characterization of the tuberous sclerosis gene on chromosome 16. Cell 1993;75:1305e1315.

297 118. Krueger DA, Care MM, Holland K, et al. Everolimus for subependymal giant-cellastrocytomas in tuberous sclerosis. N Engl J Med 2010;363:1801e1811. 119. Lager DJ, Huston BJ, Timmerman TG, et al. Papillary renal tumors. Morphologic, cytochemical, and genotypic features. Cancer 1995;76:669e673. 120. Jiang F, Richter J, Schraml P, et al. Chromosomal imbalances in papillary renal cell carcinoma: genetic differences between histological subtypes. Am J Pathol 1998;153:1467e1473.