Expert Review of Molecular Diagnostics Downloaded from informahealthcare.com by Yale Dermatologic Surgery on 07/20/15 For personal use only.

Review

BAP1, PBRM1 and SETD2 in clear-cell renal cell carcinoma: molecular diagnostics and possible targets for personalized therapies Expert Rev. Mol. Diagn. Early online, 1–10 (2015)

Francesco Piva‡1, Matteo Santoni‡2, Marc R Matrana3, Suma Satti3, Matteo Giulietti1, Giulia Occhipinti1, Francesco Massari4, Liang Cheng5, AntonioLopez-Beltran6, Marina Scarpelli7, Giovanni Principato1, Stefano Cascinu2 and Rodolfo Montironi*7

Several novel recurrent mutations of histone modifying and chromatin remodeling genes have been identified in renal cell carcinoma. These mutations cause loss of function of several genes located in close proximity to VHL and include PBRM1, BAP1 and SETD2. PBRM1 encodes for BAF180, a component of the SWI/SNF chromatin remodeling complex, and is inactivated in, on average, 36% of clear cell renal cell carcinoma (ccRCC). Mutations of BAP1 encode for the histone deubiquitinase BRCA1 associated protein-1, and are present in 10% of ccRCCs. They are largely mutually exclusive with PBRM1 mutations. Mutations to SETD2, a histone methyltransferase, occur in 10% of ccRCC. BAP1- or SETD2-mutated ccRCCs have been associated with poor overall survival, while PBRM1 mutations seem to identify a favorable group of ccRCC tumors. This review describes the roles of PBRM1, BAP1 and SETD2 in the development and progression of ccRCC and their potential for future personalized approaches. KEYWORDS: BRCA1-associated protein-1 . clear cell renal cell carcinoma . molecular diagnostics . Polybromo-1 .

personalized therapies . SETD2

1

Department of Specialistic Clinical and Odontostomatological Sciences, Polytechnic University of Marche, Ancona, Italy 2 Medical Oncology, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy, 3 Ochsner Cancer Institute, New Orleans, LA, USA, 4 Medical Oncology, Azienda Ospedaliera Universitaria Integrata, University of Verona, Verona, Italy, 5 Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA, 6 Department of Pathology and Surgery, Faculty of Medicine, Cordoba, Spain, 7 Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy *Author for correspondence: [email protected]

Authors equally contributed

informahealthcare.com

Renal cell carcinoma (RCC) includes sporadic or hereditary carcinomas arising from cells of the proximal renal tubular epithelium. Clear cell renal carcinomas (ccRCCs) represent the majority of kidney tumors, accounting for the 70% of RCC. They constitute a heterogeneous group that varies in histology, molecular alterations in tumor-suppressor gene and oncogenes, clinical courses and responses to treatment. In the last years, much research has focused on identifying clinical and pathological features associated with the prognosis of ccRCC patients, especially in the context of relatively new targeted therapies. These agents have led to major advances in the management of RCC patients and are directed toward VEGF [1,2] or its receptor [3–6] and mammalian target of rapamycin (mTOR) [7,8]. However, despite recent success, responses of primary ccRCC tumors and metastases to these agents are

10.1586/14737159.2015.1068122

variable, and complete responses are relatively uncommon [9]. Indeed, the increasing availability of molecularly targeted drugs for patients with ccRCC stresses the need for reliable biomarkers to predict tumor behavior. The inactivation of the von Hippel–Lindau (VHL) tumor-suppressor gene constitutes a crucial step in ccRCC carcinogenesis; however, additional genetic and epigenetic alterations are required for the malignant transformation to ccRCC [10]. In ccRCC, one VHL allele is damaged from an intragenic mutation, and then the loss of the second allele due to a large deletion follows. This event causes a loss of heterozygosity, but it is not sufficient to cause cancer [11]. Therefore, VHL is thought to be a weak tumor-suppressor gene. In fact, it has been shown that VHL inactivation alone causes a senescent-like phenotype [11]. As deletions along VHL gene are usually large (several megabases) and located in 3p25 chromosomal

 2015 Informa UK Ltd

ISSN 1473-7159

1

Expert Review of Molecular Diagnostics Downloaded from informahealthcare.com by Yale Dermatologic Surgery on 07/20/15 For personal use only.

Review

Piva, Santoni, Matrana, et al.

region, additional near tumor-suppressor genes, such as BRCA1-associated protein-1 (BAP1), Polybromo-1 (PBRM1) and Set domain-containing 2 (SETD2), are lost (FIGURE 1). Subsequently, the loss of the remaining PBRM1 or BAP1 allele can be associated with different ccRCC grades and aggressiveness [12]. Patients with BAP1 or SETD2 mutant ccRCCs have been associated with poor overall survival, while PBRM1 mutations seem to identify a favorable group of ccRCC tumors [13,14]. The roles of these genes as therapeutic targets have not been elucidated yet. This review describes the functions and role of PBRM1, BAP1 and SETD2 in the development and progression of ccRCC, as well as their potential for future personalized approaches in the diagnosis and treatment of these patients. BAP1

The BAP1 gene is constituted by 17 exons. The protein is a deubiquitinating enzyme that binds to breast cancer type 1 susceptibility protein (BRCA1) and BRCA1-associated RING domain protein 1 (BARD1), inhibiting their capability to mediate ubiquitination and autoubiquitination and acting as a tumor suppressor [15]. The BAP1 region that interacts with BRCA1 is located between amino acids 596 and 721, while the region that interacts with BARD1 is from 182 to 365. However, a previous study did not detect the deubiquitylation of BRCA1 [16], maybe due to a different method to flag NH2 terminus of BAP1, the region where the catalytic domain should be. Thus, further data are necessary to support these properties. Moreover, BAP1 modulates histone H2A and HCFC1 deubiquitination and regulates transcription, cell cycle and growth, response to DNA damage and chromatin dynamics. In particular, BAP1 deubiquitinates H2A and makes E2F1 target gene promoters accessible. BAP1 is mainly localized in the nucleus, bound to chromatin and retained in the cytoplasm when monoubiquitinated from the E2/E3 hybrid ubiquitin-protein ligase (FIGURE 2). In the mouse, VHL is on a different chromosome than BAP1, PBRM1 or SETD2. While the loss of 3p in humans simultaneously deletes one copy of BAP1, loss of heterozygosity in the mouse corresponding VHL region does not affect BAP1 (FIGURE 1). These results explain why VHL heterozygous humans, but not mice, develop ccRCC and why a mouse model of ccRCC has been lacking [17]. BAP1 regulates G1/S phase transition and is highly expressed in testis, placenta and ovary, and present in breast tissue. Mutant BAP1 does not act as dominant negative, because the wild type allele compensates for the mutated allele. Therefore, BAP1 acts as a two-hit tumor suppressor in ccRCC. Although both copies of BAP1 can be lost, the inactivation of both BAP1 alleles through a point mutation has not been observed [12]. Although BAP1 is a tumor suppressor when overexpressed, and its mutants are associated with the development of metastases in uveal melanoma [18], its knockdown seems to have conflicting effects. In fact, its loss has been associated with the doi: 10.1586/14737159.2015.1068122

tumorigenesis of 92.1 uveal melanoma cells [19]. On the other hand, its knockdown down-regulated E2F1 target genes, resulting in the blockade of S phase initiation, cell growth and invasiveness in OCM1 and OM431 uveal melanoma cells [20]. Depletion of BAP1 expression in cutaneous melanoma cells reduced proliferation and induced apoptosis, inhibiting tumor growth in vivo, but its overexpression in melanocytes suppressed proliferation [21]. In mesothelioma cells, BAP1 knockdown has been shown to inhibit growth by the down-regulation of the E2F-responsive growth-related genes Cyclin A2, E2F1, p107 (RBL1) and CDC25C. This could be due to the post-translational inactivation of HCF-1. To explain this paradox, it has been hypothesized that BAP1 does not promote tumorigenesis by accelerating cell growth, but allows a slowed down but tolerant G1/S checkpoint, leading to slower but uncontrolled tumor growth [22]. Molecular effects of BAP1 mutations

The functionality of some BAP1 missense mutant proteins (Ser63Cys, Phe81Val, Cys91Trp and Ala95Asp) was assessed by in vitro enzymatic assay. Phe81Val, Cys91Trp and Ala95Asp showed decreased levels of ubiquitin cleavage activity, whereas Ser63Cys showed an increased level compared with wild-type BAP1. However, both over- and under-ubiquitination of target histones led to genomic instability [22]. The influence of mutated BAP1 was investigated on binding capacity with its most important interactors showing that HCF-1 levels did not differ between BAP1-deficient and reconstituted 769-P cells. Impairing the HCF-1 binding to BAP1 prevented the BAP1 inhibitory effect on cell proliferation but did not alter its ability to deubiquitinate H2A. The interaction of BAP1 with HCF-1 was analyzed in tumor grafts and the results were similar to those in cell lines. BAP1 bound to HCF-1 and BAP1 mutations did not correlate with ubiquitinated H2A (H2Aub1) levels. In other words, BAP1 binding to HCF-1, but not H2Aub1 deubiquitination, is involved in RCC tumor-suppression function [12]. PBRM1

The PBRM1 (also PB1 or BAF180) gene gives rise to at least nine splicing isoforms. It is a subunit of the SWI/SNF transcription-modulating chromatin remodeling complex that controls DNA accessibility for transcription [23]. The complex contains an ATPase subunit to supply energy for chromatin remodeling operations. PBRM1 protein harbors six bromodomains that bind acetylated residues on histone tails. The bromodomains are not equivalent in specificity as they have different target affinity. This suggests that they may ‘read and interpret’ the histone code. The bromodomains and bromoadjacent homology domains are frequently hit by mutations in ccRCC. In breast cancers where PBRM1 was found to be mutated, it was demonstrated that PBRM1 is a regulator of p21 gene promoter accessibility, since PBRM1 induces p21 expression that, in turn, inhibits cell proliferation [24]. PBRM1 seems to be required for cellular senescence, a Expert Rev. Mol. Diagn.

BAP1, PBRM1 & SETD2 in ccRCC

Human chromosomes 1 2 3

Expert Review of Molecular Diagnostics Downloaded from informahealthcare.com by Yale Dermatologic Surgery on 07/20/15 For personal use only.

4

Review

Mouse chromosomes 1

VHL (10, 141, 635–10, 153, 670; 3p25.3) BAP1 (52, 401, 004-52, 410, 105; 3p21.1) PBRM1 (52, 545, 352-52, 685, 850; 3p21.1) SETD2 (47, 016, 408-47, 163, 977; 3p21.31)

2 3 4

5

5

6

6

7

7

8

8

9

9

10

10

11

11

12

12

13

13

14

14

15

15

16

16

17

17

18

18

19

19

20

X

21

Y

VHL (113, 624, 021-113, 631, 633; 6qE3)

SETD2 (110, 532, 597-110, 618, 633; 9qF2)

BAP1 (31, 251, 489-31, 259, 929; 14qB) PBRM1 (31, 019, 138-31, 121, 592; 14qB)

22 X Y

Figure 1. Different chromosome location of VHL, BAP1, PBRM1 and SETD2 between human and mouse.

tumor-suppressor mechanism, by regulating p53-mediated transcription of specific genes [25]. PBRM1 is important for transcriptional silencing induced by DNA double-strand breaks (DSBs) and promotes repair at damaged sites, and since PBRM1 mutants failed to perform these functions, it supports its tumor-suppressor activity [26]. Besides the role in transcriptional regulation, PBRM1 prevents tumorigenesis by promoting centromeric cohesion and genome stability [27]. Moreover, genes involved in chromosomal instability and cellular proliferation result in up-regulation following PBRM1 knockdown. In particular, RNAi-mediated depletion of PBRM1 in ccRCC cell lines has been shown to increase proliferation, colony formation and cell migration, consistent with a role as a tumor suppressor [28]. This role is also supported by the evidence that PBRM1 is involved in other malignancies, such as pancreatic cancer [29]. SETD2

SETD2 produces at least three alternative splicing transcripts and its products are histone methyltransferases. SETD2 has been found to act as tumor-suppressor gene in patients with breast cancer and leukemias [30,31]. Mutations in SETD2 are associated with increased loss of DNA methylation at nonpromoter regions. This is probably due to reductions of histone

informahealthcare.com

H3 (H3K36) trimethylation (H3K36me3) that is involved in the maintenance of a heterochromatic state [32]. A study that compared 42 primary ccRCC tumors versus normal kidney samples identified thousands of genomic regions with differences in chromatin accessibility. In particular, SETD2-mutated tumors were associated with nucleosome depletion, that is, an increased chromatin accessibility, and, most likely, this was due to H3K36me3 deficiency. Genes harboring alterations in nucleosome positioning and histone modifications may show aberrancies in splicing (mainly including exon skipping and intron retention) and alternative transcription start and termination sites [33]. SETD2 plays an important role also in DNA damage repair. In fact, cells lacking SETD2 show microsatellite instability and an elevated spontaneous mutation rate, since H3K36me3 is required to recruit the mismatch recognition protein hMutSa onto chromatin [34]. Moreover, H3K36me3 is required for ATM and p53-mediated checkpoint activations upon DNA DSBs [35] and homologous recombination repair of DSBs, as it facilitates the recruitment of repairing proteins [35,36]. Frequency of BAP1, PBRM1 & SETD2 mutations

Several studies have estimated the prevalence of VHL, BAP1, PBRM1 and SETD2 mutations. We have collected these data

doi: 10.1586/14737159.2015.1068122

Review

Piva, Santoni, Matrana, et al.

samples expressing PBRM1 had the wildtype gene [12]. As expected, most of the truncating mutations affected PBRM1 production. In the same samples, only in 15 out of 348 ccRCCs SETD2 was mutated. Interestingly, the inactivation of PBRM1 and BAP1 is less common in papillary RCC, chromophobe RCC (chRCC) and oncocytoma than in ccRCC tumors. Ub In particular, loss of PBRM1 protein Ub BAP1 expression occurred in only 3, 6 and 0% BRCA1 of papillary RCC, chRCC and oncocytoUb BARD1 Ub mas, whereas BAP1 loss was not observed BAP1 in any of these three tumors [41]. A recent Cancer study on a cohort of papillary RCCs and their matched normal samples highlighted BRCA1 BAP1 that SETD2 was significantly mutated [42]. UBE2O Interestingly, Gerlinger et al. analyzed BARD1 distinct subclones within primary tumors Nucleus Tumor suppression Intracellular or their metastases of 10 sporadic ccRCCs to assess the intratumor heteroFigure 2. BAP1 activity in ccRCC. BAP1 protein is a deubiquitinating enzyme that geneity. They showed that ccRCCs binds to breast cancer type 1 susceptibility protein (BRCA1) and BRCA1-associated RING adopted a branched rather than linear domain protein 1 (BARD1), acting as a tumor suppressor. BAP1 is mainly localized in the evolution. Moreover, VHL and PBRM1 nucleus and secondly retained in the cytoplasm when monoubiquitinated from E2/E3 hybrid ubiquitin-protein ligase (UBE2O). mutations were on the trunks of the phylogenetic trees, supporting that the in TABLE 1. However, it is important to assess how severe a muta- VHL and PBRM1 inactivating events occur early in tumor tion is in terms of alteration of protein function or protein development [43]. loss. Not only can destruction of the classical 5’ and 3’ splice sites alter pre-mRNA splicing and give rise to partly or totally Correlations among BAP1, PBRM1 & SETD2 mutated exon skipping or intron retention, but all mutations lying in genes exons or introns, both synonymous and missense, can cause Several studies have searched for favored or disfavored correlasplicing alterations [37]. Splicing alterations can cause severe tions between mutated genes. Surprisingly, although loss of effects, provoking frameshift errors and, consequently, the crea- BAP1 or PBRM1 was observed in 45% of ccRCC, concomition of premature termination codons. This event triggers a tant BAP1 and PBRM1 loss was reported only in 3% of surveillance mechanism called ‘nonsense mediated decay’ caus- ccRCC cases [32]. In particular, few tumors had both ing transcript degradation and protein loss. Although some BAP1 and PBRM1 loss, and this simultaneous loss in ccRCC softwares are able to predict if a mutation can alter splicing [38], may be disadvantageous, although the reasons are such softwares are unable to show the resulting splicing pattern unclear [12,44]. These results, confirmed in other investigain terms of sequences lost or retained. tions [45], suggest that contemporary mutations in BAP1 and In VHL, BAP1, PBRM1 and SETD2, there are base substitu- PBRM1 are negatively selected in ccRCC; in other words tions, insertions or deletions that could cause frameshift and tumors may segregate into BAP1 or PBRM1-deficient subnonsense mutations. The latter two are truncating mutations types. Instead, in a study on 185 ccRCC tumors, mutual that can directly cause loss of the protein or its function. exclusivity between PBRM1 and BAP1 mutations was not A study that found 49.2, 29.2, 7.6 and 5.9% of tumors har- statistically significant [39]; according to another study, loss of bored mutated VHL, PBRM1, SETD2 and BAP1 and also PBRM1 expression did not correlate with VHL mutations [40]. highlighted that truncating mutations represent 67, 82.5, However, it is important to note that studies with small 78.50 and 36% of mutations in VHL, PBRM1, SETD2 and sample size might not have enough power to detect an BAP1 respectively [39]. association. Furthermore, some authors showed that PBRM1 protein Concerning PBRM1 and SETD2 genes, a meta-analysis across expression is present only in about 30% of ccRCC [40]; corre- ccRCC studies confirmed that the frequency of tumors lating sequencing with immunohistochemistry data showed that with both PBRM1 and SETD2 mutated exceeded the frequency about 90% of samples that did not express PBRM1 had a expected by chance alone [13]. Therefore, PBRM1 and mutation in the corresponding gene, and about 90% of the SETD2 can cooperate in renal tumorigenesis [44].

Expert Review of Molecular Diagnostics Downloaded from informahealthcare.com by Yale Dermatologic Surgery on 07/20/15 For personal use only.

Extracellular

doi: 10.1586/14737159.2015.1068122

Expert Rev. Mol. Diagn.

informahealthcare.com

45 N/A 11 N/A N/A 2 The percentage is the sum of synonymous and non-synonymous mutations. N/A: Not assessed data.

52 N/A 14 Selected case cohort 176

81

[69]

41 10 10 5 1 1 41 11 10 Selected case cohort 240

82

[69]

11 3 4 4 0 0 26 11 8 Selected case cohort 106

41

[68]

7 1 2 3 0 0 42 8 17 55 Selected case cohort 98

[39]

19 6 3 3 1 1 29 8 6 Selected case cohort 185

49

[13]

20 6 4 4 1 1 30 7 6 Selected case cohort 188

50

[32]

22 6 1 6 1 1 34 13 11 Selected case cohort 417

56

Ref VHL (%) Study design

The promoter hypermethylation has been hypothesized to explain why about 10% of ccRCC are negative for PBRM1 or BAP1 proteins, according to immunohistochemistry, but they do not show evidence of point mutations. In fact, CpG hypermethylation represents another way to inactivate one allele of a gene, alternative to mutations. To sustain this hypothesis, the methylation status of CpG loci near the transcription start site of BAP1, PBRM1 and SETD2 was assessed in 50 primary ccRCCs. These genes were not or rarely methylated in both tumor and normal specimens [55]. Instead, in clinical samples of ccRCC, the identification of chromatin accessibility decreases in PBRM1 and SETD2 genes, suggesting the importance of epigenetic changes in this tumor and supporting the manipulation of the epigenome as a potential therapeutic strategy [56]. Thus, further efforts are required to identify the mechanisms underlying such events.

Cohort

Epigenetics

BAP1 (%)

SETD2 (%)

PBRM1 (%)

BAP1 + PBRM1 (%)

BAP1 + SETD2 (%)

SETD2 + PBRM1 (%)

VHL + BAP1 (%)

VHL + SETD2 (%)

VHL + PBRM1 (%)

A germline BAP1 mutation G121A (Gly41Ser) was identified in one individual, not having a germline VHL mutation, who had two and one first and second-degree relatives, respectively, with RCC [12]. The mutation A277G (Thr93Ala) in BAP1 has been shown to alter the RNA splicing pattern, which gives rise to 80% of aberrant mature transcripts that lead to a frameshift, whereas approximately 20% corresponded to the missense mutation. In TABLE 1, we summarize the BAP1 mutations identified by the same authors and associated with a tumor predisposition syndrome, including uveal melanoma, malignant pleural mesothelioma and cutaneous melanoma [46]. BAP1 sequencing in 83 unrelated probands with familial RCC revealed that a new germline mutation, T41A (L14H), predisposed to early onset of ccRCC. This was supported by several findings, including: this variant co-segregates with the RCC phenotype; it targets the catalytic domain of BAP1; the wild type amino acid L14 is highly conserved across species suggesting its irreplaceableness; the mutation is associated with LOH [47]. In the same study, other two mutations were discovered, although they have been likely considered as benign (TABLE 2). Germline mutations leading to BAP1 hereditary cancer predisposition syndrome can also cause uveal and cutaneous melanoma, mesothelioma [48–51] and basal cell carcinoma [52]. Notably, patients with the same BAP1 mutational status may develop different cancer types depending on the type of carcinogen exposure and, naturally, on the presence of other gene mutations [53]. BAP1 germline mutations have been found in several tissues. However, not all these tissues harboring such mutations develop cancer, suggesting that the mechanisms underlying such tolerance to mutations are still unrevealed. Moreover, Wang et al. have presented the preliminary The Cancer Genome Atlas exome sequencing data from 499 RCC cases [54]. The final result from this study on the loss of heterozygosity patterns to assess the interactions between germline and somatic mutations are awaited.

Table 1. ccRCC marker frequencies.

Expert Review of Molecular Diagnostics Downloaded from informahealthcare.com by Yale Dermatologic Surgery on 07/20/15 For personal use only.

Are germinal BAP1 mutations associated with RCC predisposition?

[12]

Review

BAP1, PBRM1 & SETD2 in ccRCC

doi: 10.1586/14737159.2015.1068122

Review

Piva, Santoni, Matrana, et al.

Expert Review of Molecular Diagnostics Downloaded from informahealthcare.com by Yale Dermatologic Surgery on 07/20/15 For personal use only.

Table 2. Germline mutations in BAP1 gene causing renal cell carcinoma. Gene

Protein

Predicted effects

Ref.

37+1 delG

N-A

Probably affecting splicing

[46]

T41A

Leu14His

Missense (or splice) exon 1

[47]

C45G

N-A

3’UTR

[48]

78_79 delGG

Val27Ala

Truncating

[46]

G121A

Gly41Ser

Probably benign

[47]

256_277 and A277G

Ile87Met and Thr93Ala

In/del exon 5

[46]

A277G

Thr93Ala

Affecting splicing and truncating

[46]

437+1 G>A

N-A

Truncating

[46]

629 dupT

Met211His

Truncating

[46]

660-11 T>A

N-A

Probably affecting splicing

[46]

A869G

Asn290Ser

probably benign

[47]

932-58_59 delTG

N-A

Probably affecting splicing

[48]

C1182G

Tyr394*

Unknown

[48]

1654 delG

Asp552Ile

Truncating

[46]

1882_1885 del TCAC

Ser628Pro

Unknown

[48]

C2050T

Gln684*

Unknown

[48]

2057-22A>C

N-A

Probably affecting splicing

[48]

2057-4G>T

N-A

Probably affecting splicing

[48]

Prognostic significance of BAP1, PBRM1 & SETD2 mutations

Several studies focused on the prognostic significance of BAP1, PBRM1 and SETD2 mutational status. An analysis on 176 tumors showed that BAP1 loss correlated with high Fuhrman nuclear grade and mTORC1 activation [12]. Other authors confirmed a strong correlation between BAP1 mutations and higher Fuhrman nuclear grades [39]. Moreover, BAP1 loss in ccRCC may be associated with poor prognosis [14,32,39,45]. In particular, a retrospective analysis of 145 patients validated using a further independent cohort of 327 patients confirmed that tumors with mutations of either PBRM1 or BAP1 were more likely to present with higher tumor stage 3–4. They found that BAP1 mutations, but no VHL or PBRM1 mutations, were significantly associated with worse cancer-specific survival. They also reported a poor median overall survival (4.6 years) in patients with BAP1-mutated ccRCC compared to patients with PBRM1-mutated tumors (10.6 years) [14]. A study of BAP1 protein expression in 1479 patients with clinically localized ccRCC showed that BAP1 loss correlated with the most aggressive forms of ccRCC, even among low-risk patients. However, in this study, not all high-grade tumors showed loss of BAP1 expression [57]. A potential explanation of its negative prognostic role may be represented by mTORC1 activation following BAP1 inactivation, suggesting doi: 10.1586/14737159.2015.1068122

that these tumors could be more responsive to mTORC1 inhibitors. Interestingly, BAP1 truncating, compared with missense mutations, presented worse prognosis, but this was not maintained for BAP1 and SETD2 in a different cohort. Moreover, SETD2 mutations were related to the time to recurrence [13]. BAP1- and PBRM1-mutant tumors were also associated with different gene expression signatures. The former had alterations of genes implicated in growth-factor signaling and the latter involved genes implicated in the cytoskeleton and tissue architecture. This suggests that ccRCC can be classified into at least two distinct entities, biologically and clinically [14]. SETD2 mutations were related with little worse prognosis [13], whereas SETD2 frameshift mutations were associated with advanced tumors stages [39]. A retrospective study that investigated the associations between computed tomography diagnostic imaging features and RCC mutations highlighted that VHL-mutated tumors were associated with well-defined tumor margins, nodular tumor enhancement and gross appearance of intratumor vascularity. BAP1-mutated tumors were associated with renal vein invasion, whereas SETD2-mutated tumors were not associated with any computed tomography feature studied. Moreover, multicystic ccRCC did not harbor mutations of SETD2 and BAP1 [58]. Expert Rev. Mol. Diagn.

Expert Review of Molecular Diagnostics Downloaded from informahealthcare.com by Yale Dermatologic Surgery on 07/20/15 For personal use only.

BAP1, PBRM1 & SETD2 in ccRCC

As for PBRM1 loss, it was associated neither with high-grade nor with low-grade tumor, but there was a correlation between tumors that had exclusively lost PBRM1 and low tumor grade [12]. Another study showed that loss of PBRM1 expression correlated with tumor T stage. Indeed, in this study, approximately half of the pT1/pT2 and only about 20% of the pT3/ pT4 tumors expressed PBRM1. Late-stage, high-grade and poorly differentiated tumors were mainly PBRM1-negative and independent from VHL mutation status. PBRM1-positive tumors correlated with better prognosis than patients with PBRM1 absent tumors [40]. In addition, PBRM1 and SETD2 mutations were not associated with differences in overall survival between patients with one or both mutated genes [44]. Finally, ccRCC with mutations either in PBRM1 or BAP1 or SETD2 correlated with higher stage but not with tumor size, supporting the idea that chromatin alteration are involved in disease progression. Moreover, specific PBRM1, SETD2, BAP1 mutations are associated advanced tumor stage and grade [39]. BAP1 & treatment sensitivity

From a therapeutic point of view, it is important to assess if distinct tumor subtypes differ in treatment response to different therapies, for example, ionizing radiation or drugs. It is known that stresses causing DNA DSBs, such as ionizing radiation, trigger BAP1 phosphorylation, assembly of the homologous recombination factors BRCA1 and RAD51, recruiting chromatin in the proximity of damaged sites [59,60]. It has been shown that BAP1-deficient cells were more vulnerable to g-radiation [12], suggesting greater sensitivity to radiotherapy in BAP1-deficient RCC than RCC wild-type for BAP1. In a previous study, mesothelioma cell lines with both wild-type and mutated or deleted BAP1 were exposed to 10 Gy of ionizing radiation, and the formation of RAD51 or BRCA1 complexes was assessed. No differences were found between BAP1 wild-type and deficient cell lines, highlighting that BAP1 may not have an important role in the formation of DNA repair complexes [22]. Regarding drugs, since BAP1 loss determines ubiquitinated H2A accumulation, masking various gene promoter transcripts, histone deacetylase (HDAC) inhibitors may reverse this effect. Indeed, valproic acid decreased H2A-ub levels and reduced the fraction of viable 92.1 human uveal melanoma cells, showing that BAP1 made these cells vulnerable to HDAC inhibitors [19]. At present, several HDAC inhibitors are under evaluation in patients with metastatic RCC. LBH589 (panobinostat), a panDAC inhibitor, was tested in a Phase II trial enrolling patients previously treated with tyrosine kinase inhibitors and mTOR inhibitors. They did not report objective responses, as all patients progressed or stopped treatment prior to the 16-week revaluation [61]. Similar results in terms of efficacy were obtained in another Phase II trial of FK228 (depsipeptide), another HDAC inhibitor [62]. The list of HDAC inhibitors also includes vorinostat, MS-275, belinostat and entinostat (in combination with IL-2), which are currently under study in mRCC patients [63–66], respectively. informahealthcare.com

Review

Furthermore, BAP1 loss sensitized cells to the PARP inhibitor olaparib [12]. Currently, a Phase I study is evaluating the combination of olaparib and AZD5363, an AKT inhibitor, in patients with advanced solid tumors including RCC refractory to standard therapy [67]. Expert commentary

Greater understanding of the molecular drivers of ccRCC has led to revolutionary new targeted therapies for patients with metastatic or otherwise inoperable ccRCC over the last decade. These newer targeted therapies, including VEGF-targeted tyrosine kinase inhibitors mTOR inhibitors and a monoclonal antibody, have virtually supplanted the older immunotherapies for ccRCC in most patients, offering a higher response rate and much less toxicity. But, despite the therapy advances made, there are virtually no complete responses in metastatic ccRCC to targeted therapy, and nearly all patients with metastatic ccRCC treated with targeted therapy alone eventually succumb to this disease [9]. Thus, even greater understanding of the molecular and genetic drivers of ccRCC is necessary to identify novel therapeutic targets in hope of eliciting cures. Likewise, identifying biomarkers in ccRCC is critical for prognostication and stratifying patients in order to properly pair them with the most appropriate therapy. BAP1, PBRM1 and SETD2 represent three candidate genes that have strong potential implications as possible biomarkers for prognostication in ccRCC and may represent possible therapeutic targets. Several studies have shown that BAP1 mutations are associated with more aggressive tumors including higher Fuhrman grade, later stage at presentation and upregulation of the mTOR pathway [12–14,39,45]. This also means that BAP1 presents itself as a potential therapeutic target, and as discussed above, trials of HDAC inhibitors in ccRCC are ongoing. Likewise, there is evidence to suggest that BAP1 mutation may be predictive of mTOR inhibitor sensitivity, but there is no data to support this either way [12]. BAP1 mutations may be associated with great sensitivity to radiotherapy, which may be an important predictive implication in ccRCC, which is a relatively radio-insensitive tumor, but more clinical research is necessary to confirm this [12]. PBRM1 also has potential prognostic implications as mutations in this gene have been associated with lower tumor grade and earlier stage disease at presentation. And, while there has been less translational and clinical research into the implications of SETD2 mutations, these histone-modifying mutations may also hold important clinical implications. Obviously, much more research is needed to further define the roles of these three genes as they relate to defining prognosis in ccRCC and as potential therapeutic targets. Five-year view

In the next years, the assessment of BAP1, PBRM1 and SETD2 mutations should be integrated with the clinicopathological features of ccRCC patients, such as prognostic groups. Furthermore, the predictive role of these mutations should be doi: 10.1586/14737159.2015.1068122

Expert Review of Molecular Diagnostics Downloaded from informahealthcare.com by Yale Dermatologic Surgery on 07/20/15 For personal use only.

Review

Piva, Santoni, Matrana, et al.

investigated in patients treated with targeted agents. Moreover, research should be focused on the potential correlation between the risk of relapsing after partial or complete nephrectomy and BAP1, PBRM1 and SETD2 mutations. New targeted immunotherapy, including PD-1 and PD-L1 targeted agents, are being developed and are expected to be approved in ccRCC in the near future. Appropriately sequencing and/or combining these new drugs with current targeted therapies or other agents will be essential in maximizing patient outcomes. Potential biomarkers, such as BAP1, PBRM1 and SETD2, and others will be all the more important in order to personalize therapies for patients as our understanding of the molecular heterogeneity of ccRCC expands and the number of available treatment options for

this disease continues to grow. And, it is likely that diseasespecific panels of important genes such as VHL, BAP1, PBRM1 and SETD2 will one day help clinicians to better define prognosis and personalize targeted treatments plans for patients with ccRCC. Financial & competing interests disclosure

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties. No writing assistance was utilized in the production of this manuscript.

Key issues .

Mutations to BAP1, PBRM1 and SETD2, in close proximity to VHL gene, are frequent in ccRCC patients.

.

In the mouse, VHL is on a different chromosome than BAP1, PBRM1 or SETD2, thus explaining why VHL heterozygous humans, but not mice, develop.

.

The inactivation of PBRM1 and BAP1 is less common in papillary, chromophobe RCC and oncocytoma than in ccRCC.

.

BAP1 mutation may be predictive of the sensitivity to mTOR inhibitors and radiotherapy.

.

Patients with BAP1- or SETD2-mutant ccRCC have been associated with poor overall survival, while PBRM1 mutations seem to identify a favorable group of ccRCC tumors.

.

Potential biomarkers, such as BAP1, PBRM1 and SETD2, and others will be all the more important in order to personalize therapies for ccRCC patients.

.

The roles of these genes as therapeutic targets in ccRCC patients have not been elucidated yet.

.

Research should be also focused on the potential correlation between the risk of relapsing after partial or complete nephrectomy and BAP1, PBRM1 and SETD2 mutations.

Papers of special note have been highlighted as: . of interest .. of considerable interest 1.

2.

3.

4.

5.

Escudier B, Pluzanska A, Koralewski P, et al. Bevacizumab plus interferon alfa-2a for treatment of metastatic renal cell carcinoma: a randomised, double-blind phase III trial. Lancet 2007;370:2103-11 Rini BI, Halabi S, Rosenberg JE, et al. Bevacizumab plus interferon alfa compared with interferon alfa monotherapy in patients with metastatic renal cell carcinoma: CALGB 90206. J Clin Oncol 2008;26: 5422-8 Escudier B, Eisen T, Stadler WM, et al. Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med 2007;356:125-34

6.

doi: 10.1586/14737159.2015.1068122

Rini BI, Escudier B, Tomczak P, et al. Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomised phase 3 trial. Lancet 2011;378:1931-9

7.

Hudes G, Carducci M, Tomczak P, et al. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med 2007;356:2271-81

8.

Motzer RJ, Escudier B, Oudard S, et al. Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet 2008;372:449-56

9.

Iacovelli R, Alesini D, Palazzo A, et al. Targeted therapies and complete responses in first line treatment of metastatic renal cell carcinoma. A meta-analysis of published trials. Cancer Treat Rev 2014;40:271-5

Motzer RJ, Hutson TE, Tomczak P, et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med 2007;356:115-24 Sternberg CN, Davis ID, Mardiak J, et al. Pazopanib in locally advanced or metastatic

patients treated with different first line therapies for advanced renal cell carcinoma (RCC).

renal cell carcinoma: results of a randomized phase III trial. J Clin Oncol 2010;28: 1061-8

References

.

This meta-analysis focused on the frequency of complete responses in

10.

Brugarolas J. Molecular genetics of clear-cell renal cell carcinoma. J Clin Oncol 2014;32: 1968-76

.

He described the molecular background of clear cell RCC.

11.

Young AP, Schlisio S, Minamishima YA, et al. VHL loss actuates a HIF-independent senescence programme mediated by Rb and p400. Nat Cell Biol 2008;10:361-9

12.

Pena-Llopis S, Vega-Rubin-de-Celis S, Liao A, et al. BAP1 loss defines a new class of renal cell carcinoma. Nat Genet 2012;44: 751-9

..

They markedly contributed to our knowledge on the role of BAP1 loss in clear cell RCC.

13.

Hakimi AA, Ostrovnaya I, Reva B, et al. Adverse outcomes in clear cell renal cell carcinoma with mutations of 3p21 epigenetic regulators BAP1 and SETD2: a report by MSKCC and the

Expert Rev. Mol. Diagn.

BAP1, PBRM1 & SETD2 in ccRCC

KIRC TCGA research network. Clin Cancer Res 2013;19:3259-67 14.

Expert Review of Molecular Diagnostics Downloaded from informahealthcare.com by Yale Dermatologic Surgery on 07/20/15 For personal use only.

..

15.

16.

17.

18.

19.

20.

21.

22.

23.

24.

25.

Kapur P, Pena-Llopis S, Christie A, et al. Effects on survival of BAP1 and PBRM1 mutations in sporadic clear-cell renal-cell carcinoma: a retrospective analysis with independent validation. Lancet Oncol 2013;14:159-67 They showed the prognostic of BAP1 and PBMR1 mutations in RCC patients.

factor components BRD7 and BAF180 are critical regulators of p53 required for induction of replicative senescence. Proc Natl Acad Sci USA 2010;107:14280-5 26.

27.

Kakarougkas A, Ismail A, Chambers AL, et al. Requirement for PBAF in transcriptional repression and repair at DNA breaks in actively transcribed regions of chromatin. Mol Cell 2014;55:723-32 Brownlee PM, Chambers AL, Cloney R, et al. BAF180 promotes cohesion and prevents genome instability and aneuploidy. Cell Rep 2014;6:973-81

Nishikawa H, Wu W, Koike A, et al. BRCA1-associated protein 1 interferes with BRCA1/BARD1 RING heterodimer activity. Cancer Res 2009;69:111-19

28.

Mallery DL, Vandenberg CJ, Hiom K. Activation of the E3 ligase function of the BRCA1/BARD1 complex by polyubiquitin chains. EMBO J 2002;21:6755-62

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;469:539-42

29.

Shain AH, Giacomini CP, Matsukuma K, et al. Convergent structural alterations define SWItch/Sucrose NonFermentable (SWI/SNF) chromatin remodeler as a central tumor suppressive complex in pancreatic cancer. Proc Natl Acad Sci USA 2012;109:E252-9

Wang SS, Gu YF, Wolff N, et al. Bap1 is essential for kidney function and cooperates with Vhl in renal tumorigenesis. Proc Natl Acad Sci USA 2014;111:16538-43 Harbour JW, Onken MD, Roberson ED, et al. Frequent mutation of BAP1 in metastasizing uveal melanomas. Science 2010;330:1410-13 Landreville S, Agapova OA, Matatall KA, et al. Histone deacetylase inhibitors induce growth arrest and differentiation in uveal melanoma. Clin Cancer Res 2012;18: 408-16 Pan H, Jia R, Zhang L, et al. BAP1 regulates cell cycle progression through E2F1 target genes and mediates transcriptional silencing via H2A monoubiquitination in uveal melanoma cells. Int J Biochem Cell Biol 2015;60:176-84 Kumar R, Taylor M, Miao B, et al. BAP1 Has a Survival Role in Cutaneous Melanoma. J Invest Dermatol 2015;135: 1089-97 Bott M, Brevet M, Taylor BS, et al. The nuclear deubiquitinase BAP1 is commonly inactivated by somatic mutations and 3p21.1 losses in malignant pleural mesothelioma. Nat Genet 2011;43:668-72

30.

31.

32.

Zhang J, Ding L, Holmfeldt L, et al. The genetic basis of early T-cell precursor acute lymphoblastic leukaemia. Nature 2012;481: 157-63 Creighton CJ, Morgan M, Gunaratne PH, et al. Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature 2013;499:43-9

..

They identified 19 significantly mutated genes underlying the development of clear cell RCC.

33.

Simon JM, Hacker KE, Singh D, et al. Variation in chromatin accessibility in human kidney cancer links H3K36 methyltransferase loss with widespread RNA processing defects. Genome Res 2014;24:241-50

34.

Su D, Singer EA, Srinivasan R. Molecular pathways in renal cell carcinoma: recent advances in genetics and molecular biology. Curr Opin Oncol 2015;27:217-23 Xia W, Nagase S, Montia AG, et al. BAF180 is a critical regulator of p21 induction and a tumor suppressor mutated in breast cancer. Cancer Res 2008;68:1667-74

35.

Burrows AE, Smogorzewska A, Elledge SJ. Polybromo-associated BRG1-associated

36.

informahealthcare.com

Al Sarakbi W, Sasi W, Jiang WG, et al. The mRNA expression of SETD2 in human breast cancer: correlation with clinico-pathological parameters. BMC Cancer 2009;9:290

Li F, Mao G, Tong D, et al. The histone mark H3K36me3 regulates human DNA mismatch repair through its interaction with MutSalpha. Cell 2013;153: 590-600 Carvalho S, Vitor AC, Sridhara SC, et al. SETD2 is required for DNA double-strand break repair and activation of the p53-mediated checkpoint. Elife 2014;3: e02482 Pfister SX, Ahrabi S, Zalmas LP, et al. SETD2-dependent histone H3K36 trimethylation is required for

Review

homologous recombination repair and genome stability. Cell Rep 2014;7:2006-18 37.

Raponi M, Douglas AG, Tammaro C, et al. Evolutionary constraint helps unmask a splicing regulatory region in BRCA1 exon 11. PLoS One 2012;7:e37255

38.

Piva F, Giulietti M, Burini AB, Principato G. SpliceAid 2: a database of human splicing factors expression data and RNA target motifs. Hum Mutat 2012;33: 81-5

39.

Hakimi AA, Chen YB, Wren J, et al. Clinical and pathologic impact of select chromatin-modulating tumor suppressors in clear cell renal cell carcinoma. Eur Urol 2013;63:848-54

40.

Pawlowski R, Muhl SM, Sulser T, et al. Loss of PBRM1 expression is associated with renal cell carcinoma progression. Int J Cancer 2013;132:E11-17

41.

Ho TH, Kapur P, Joseph RW, et al. Loss of PBRM1 and BAP1 expression is less common in non-clear cell renal cell carcinoma than in clear cell renal cell carcinoma. Urol Oncol 2015;33:23 e9-14

42.

Durinck S, Stawiski EW, Pavia-Jimenez A, et al. Spectrum of diverse genomic alterations define non-clear cell renal carcinoma subtypes. Nat Genet 2015;47: 13-21

43.

Gerlinger M, Horswell S, Larkin J, et al. Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat Genet 2014;46:225-33

.

They focused on the phylogenesis and tumor heterogeneity of clear cell RCC.

44.

Pena-Llopis S, Christie A, Xie XJ, Brugarolas J. Cooperation and antagonism among cancer genes: the renal cancer paradigm. Cancer Res 2013;73:4173-9

45.

Kandoth C, McLellan MD, Vandin F, et al. Mutational landscape and significance across 12 major cancer types. Nature 2013;502: 333-9

46.

Popova T, Hebert L, Jacquemin V, et al. Germline BAP1 mutations predispose to renal cell carcinomas. Am J Hum Genet 2013;92:974-80

47.

Farley MN, Schmidt LS, Mester JL, et al. A novel germline mutation in BAP1 predisposes to familial clear-cell renal cell carcinoma. Mol Cancer Res 2013;11: 1061-71

48.

Abdel-Rahman MH, Pilarski R, Cebulla CM, et al. Germline BAP1 mutation predisposes to uveal melanoma, lung adenocarcinoma,

doi: 10.1586/14737159.2015.1068122

Review

Piva, Santoni, Matrana, et al.

meningioma, and other cancers. J Med Genet 2011;48:856-9

Buck MJ, Raaijmakers LM, Ramakrishnan S, et al. Alterations in chromatin accessibility and DNA methylation in clear cell renal cell carcinoma. Oncogene 2014;33:4961-5

Testa JR, Cheung M, Pei J, et al. Germline BAP1 mutations predispose to malignant mesothelioma. Nat Genet 2011;43:1022-5

57.

Njauw CN, Kim I, Piris A, et al. Germline BAP1 inactivation is preferentially associated with metastatic ocular melanoma and cutaneous-ocular melanoma families. PLoS ONE 2012;7:e35295

Joseph RW, Kapur P, Serie DJ, et al. Loss of BAP1 protein expression is an independent marker of poor prognosis in patients with low-risk clear cell renal cell carcinoma. Cancer 2014;120:1059-67

58.

51.

Aoude LG, Vajdic CM, Kricker A, et al. Prevalence of germline BAP1 mutation in a population-based sample of uveal melanoma cases. Pigment Cell Melanoma Res 2013;26: 278-9

Karlo CA, Di Paolo PL, Chaim J, et al. Radiogenomics of clear cell renal cell carcinoma: associations between CT imaging features and mutations. Radiology 2014;270:464-71

59.

52.

de la Fouchardiere A, Cabaret O, Savin L, et al. Germline BAP1 mutations predispose also to multiple basal cell carcinomas. Clin Genet 2014

Eletr ZM, Yin L, Wilkinson KD. BAP1 is phosphorylated at serine 592 in S-phase following DNA damage. FEBS Lett 2013;587:3906-11

60.

Yu H, Pak H, Hammond-Martel I, et al. Tumor suppressor and deubiquitinase BAP1 promotes DNA double-strand break repair. Proc Natl Acad Sci USA 2014;111: 285-90

49.

50.

Expert Review of Molecular Diagnostics Downloaded from informahealthcare.com by Yale Dermatologic Surgery on 07/20/15 For personal use only.

56.

53.

Betti M, Casalone E, Ferrante D, et al. Inference on germline BAP1 mutations and asbestos exposure from the analysis of familial and sporadic mesothelioma in a high-risk area. Genes Chromosomes Cancer 2015;54:51-62

54.

Jiayin W CL, Mingchao X, et al. Integrated analysis of germline and somatic variants in renal clear cell carcinoma. AACR Annual Meeting 2014; Cancer Res, San Diego, CA, 2014

55.

Ibragimova I, Maradeo ME, Dulaimi E, Cairns P. Aberrant promoter hypermethylation of PBRM1, BAP1, SETD2, KDM6A and other chromatin-modifying genes is absent or rare in clear cell RCC. Epigenetics 2013;8: 486-93

doi: 10.1586/14737159.2015.1068122

61.

62.

63.

Hainsworth JD, Infante JR, Spigel DR, et al. A phase II trial of panobinostat, a histone deacetylase inhibitor, in the treatment of patients with refractory metastatic renal cell carcinoma. Cancer Invest 2011;29:451-5 Stadler WM, Margolin K, Ferber S, et al. A phase II study of depsipeptide in refractory metastatic renal cell cancer. Clin Genitourin Cancer 2006;5:57-60 Clinical trials.gov, A service of the U.S. National Institutes of Health, Vorinostat in Treating Patients With Kidney Cancer.

Available from: https://clinicaltrials.gov/ct2/ show/NCT00278395 64.

Clinical trials.gov, A service of the U.S. National Institutes of Health, MS-275 in Treating Patients With Advanced Solid Tumors or Lymphoma. Available from: https://clinicaltrials.gov/ct2/show/ NCT00020579

65.

Clinical trials.gov, A service of the U.S. National Institutes of Health, Study of Oral PXD101 in Patients With Advanced Solid Tumors or Lymphoma. Available from: https://clinicaltrials.gov/ct2/show/ NCT00413075

66.

Clinical trials.gov, A service of the U.S. National Institutes of Health, Entinostat in Combination With Aldesleukin in Treating Patients With Metastatic Kidney Cancer. Available from: https://clinicaltrials.gov/ct2/ show/NCT01038778

67.

Clinical trials.gov, A service of the U.S. National Institutes of Health, Trial of Olaparib in Combination With AZD5363 (ComPAKT). Available from: https:// clinicaltrials.gov/ct2/show/NCT02338622

68.

Guo G, Gui Y, Gao S, et al. Frequent mutations of genes encoding ubiquitin-mediated proteolysis pathway components in clear cell renal cell carcinoma. Nat Genet 2012;44:17-19

69.

Sato Y, Yoshizato T, Shiraishi Y, et al. Integrated molecular analysis of clear-cell renal cell carcinoma. Nat Genet 2013;45: 860-7

Expert Rev. Mol. Diagn.

BAP1, PBRM1 and SETD2 in clear-cell renal cell carcinoma: molecular diagnostics and possible targets for personalized therapies.

Several novel recurrent mutations of histone modifying and chromatin remodeling genes have been identified in renal cell carcinoma. These mutations ca...
618KB Sizes 0 Downloads 13 Views