Skip to main content
Proceedings of the National Academy of Sciences of the United States of America logoLink to Proceedings of the National Academy of Sciences of the United States of America
. 2016 Feb 10;113(8):2170–2175. doi: 10.1073/pnas.1525735113

Genomic characterization of sarcomatoid transformation in clear cell renal cell carcinoma

Mark Bi a,b, Siming Zhao a,b, Jonathan W Said c, Maria J Merino d, Adebowale J Adeniran e, Zuoquan Xie f, Cayce B Nawaf f, Jaehyuk Choi g, Arie S Belldegrun h, Allan J Pantuck h, Harriet M Kluger i, Kaya Bilgüvar a, Richard P Lifton a,b,1, Brian Shuch c,1
PMCID: PMC4776463  PMID: 26864202

Significance

Parts of clear cell renal cell carcinomas (ccRCCs) sometimes have histologic features characteristic of a sarcoma. So-called sarcomatoid tumors are more aggressive, difficult to treat, and associated with a poor prognosis. Their pathogenesis has been uncertain. Through separate exome sequencing of carcinomatous and sarcomatoid components, we show that these components share many somatic mutations, including many in genes characteristic of ccRCC. Sarcomatoid elements had significantly more new somatic mutations, particularly in cancer driver genes, than carcinomatous components. In particular, tumor protein p53, AT-rich interaction domain 1A, and BRCA1 associated protein 1 had sarcomatoid-specific homozygous mutation in 10 tumors and were all mutually exclusive, implicating these genes in sarcomatoid degeneration.

Keywords: kidney cancer, sarcomatoid, transformation, p53, dedifferentiation

Abstract

The presence of sarcomatoid features in clear cell renal cell carcinoma (ccRCC) confers a poor prognosis and is of unknown pathogenesis. We performed exome sequencing of matched normal-carcinomatous-sarcomatoid specimens from 21 subjects. Two tumors had hypermutation consistent with mismatch repair deficiency. In the remainder, sarcomatoid and carcinomatous elements shared 42% of somatic single-nucleotide variants (SSNVs). Sarcomatoid elements had a higher overall SSNV burden (mean 90 vs. 63 SSNVs, P = 4.0 × 10−4), increased frequency of nonsynonymous SSNVs in Pan-Cancer genes (mean 1.4 vs. 0.26, P = 0.002), and increased frequency of loss of heterozygosity (LOH) across the genome (median 913 vs. 460 Mb in LOH, P < 0.05), with significant recurrent LOH on chromosomes 1p, 9, 10, 14, 17p, 18, and 22. The most frequent SSNVs shared by carcinomatous and sarcomatoid elements were in known ccRCC genes including von Hippel–Lindau tumor suppressor (VHL), polybromo 1 (PBRM1), SET domain containing 2 (SETD2), phosphatase and tensin homolog (PTEN). Most interestingly, sarcomatoid elements acquired biallelic tumor protein p53 (TP53) mutations in 32% of tumors (P = 5.47 × 10−17); TP53 mutations were absent in carcinomatous elements in nonhypermutated tumors and rare in previously studied ccRCCs. Mutations in known cancer drivers AT-rich interaction domain 1A (ARID1A) and BRCA1 associated protein 1 (BAP1) were significantly mutated in sarcomatoid elements and were mutually exclusive with TP53 and each other. These findings provide evidence that sarcomatoid elements arise from dedifferentiation of carcinomatous ccRCCs and implicate specific genes in this process. These findings have implications for the treatment of patients with these poor-prognosis cancers.


Sarcomatoid transformation is common in various epithelial malignancies, featuring further loss of differentiation and acquisition of characteristics typical of a sarcoma. In renal cell carcinoma (RCC), sarcomatoid features are observed in 5% of tumors. However, among individuals with stage IV disease, it occurs in 15% (1, 2). Although once believed to represent a distinct subtype of RCC, it is now considered a specific histologic feature (3). Whereas sarcomatoid features are found in all forms of kidney cancer, >65% of cases are found with clear cell RCC (ccRCC) (4, 5). When sarcomatoid features are present, renal tumors generally are large (median size 10 cm), invasive (20%), and/or metastatic (50%) at presentation (4, 5). Whereas all such tumors are considered to be Fuhrman grade IV (6), their prognosis is significantly worse compared with other high-grade tumors (7). These tumors, when metastatic, have among the poorest survival of all genitourinary malignancies, with a median survival of only 6 mo (1, 2, 4). Even with resected localized disease, nearly 75% recur and have a median survival of <2 y (4, 8). The response to systemic therapy is poor, with rare durable responses occurring with any therapeutic strategy (912).

Although there have been major breakthroughs in the understanding of kidney cancer, progress in the characterization of the genetic events associated with sarcomatoid kidney tumors has been limited (9). Various theories have been proposed regarding the origins of sarcomatoid features in renal tumors. Given that they virtually always occur in conjunction with typical epithelial RCC elements, the terminology of a “mixed malignancy” appeared a half century ago (13). Proposals have included independent occurrences of tumor types in close proximity, as has been observed in various genitourinary malignancies (14), and the influence of tumor microenvironment (15). The current prevailing theory is that sarcomatoid features represents a subclonal dedifferentiation or transformation from an incident carcinomatous component (16). However, the current theory is based on limited evidence. Evidence of common cell of origin is suggested by the shared patterns of X chromosome inactivation (17). Although there is limited evidence that the epithelial component transforms into the sarcomatoid element, groups have considered sarcomatoid features to result from a final common dedifferentiation pathway in RCC (16). This hypothesis is based on the sarcomatoid component more frequently metastasizing, possessing higher tumor grade, increased proliferative index, and frequent reduced expression of epithelial adhesion molecules such as E-cadherin (1820). Because some tumors demonstrate increased expression of N-cadherin, it has been suggested that epithelial-mesenchymal transformation (EMT) may be involved in the development of sarcomatoid elements (21). However, what could be driving this process has escaped elucidation.

To address the molecular basis for sarcomatoid elements in RCC, we performed whole exome sequencing of distinct regions of clear cell and sarcomatoid morphology from the same tumors.

Materials and Methods

Patient and Specimen Acquisition.

From 1989 to 2010, all patients undergoing nephrectomy for presumed renal cancer at the University of California, Los Angeles (UCLA) had clinical information entered into an approved database. ccRCC tumors featuring sarcomatoid transformation were reviewed by a genitourinary (GU) pathologist (J.W.S.). The clinical data, tumor characteristics, and survival have been described (1, 4, 20). A second GU pathologist (M.J.M.) reviewed representative slides to confirm the presence of distinct morphologic regions that represented (i) ccRCC and (ii) a definitive region with sarcomatoid transformation. All living patients studied provided written informed consent for participation in this research. A waiver of consent was approved to study anonymized samples from deceased patients. The research protocol was approved by the UCLA and Yale Human Investigation Committees. All experiments were conducted according to the principles expressed in the Declaration of Helsinki.

DNA Extraction, Exome Capture, and Exome Sequencing.

Twenty-nine tumors had available formalin-fixed, paraffin-embedded blocks with adjacent normal kidney for genomic control. The blocks were reviewed by a third independent GU pathologist (A.J.A.), who confirmed the histology and identified distinct regions of normal kidney, clear cell, and sarcomatoid histology. From each of these regions, 1-mm punches were obtained and DNA extracted by using a described protocol (22). Exome capture was performed by using Nimblegen 2.1M Human Exome Array followed by 74 base paired-end DNA sequencing on the Illumina HiSeq instrument. Tumor regions were sequenced to greater depth of coverage than normal tissue to account for admixture of tumor and normal cells. High quality sequences were obtained for 21 matched sets of normal, carcinomatous, and sarcomatoid elements.

Sequence Analysis and Comparisons.

Sequences were aligned to the hg19 reference genome by using Burrow–Wheeler Aligner-MEM (BWA-MEM) (23). Somatic single nucleotide variant (SNV) calling was performed by using Mutect (24) and indel calling was performed by using Indelocator (https://www.broadinstitute.org/cancer/cga/indelocator). Additional somatic mutation calls were acquired by using a reported pipeline (25). Variant calls with less than a total of eight independent reads in any of the three sequenced samples from each tumor were discarded. Variants previously identified as germ-line variants in 1000 Genomes Project (26); National Heart, Lung, and Blood Institute Exome Sequencing Project; and the Yale University exome database were excluded. All somatic mutation calls were manually verified by visual inspection. Somatic mutations identified by Mutect in one component (e.g., the sarcomatoid but not carcinomatous elements) were called specific for that component when the variant was not called in the other component, had P ≤ 0.05 for two-tailed Fisher’s exact test of the difference in the distribution of reference and nonreference reads between the sarcomatoid and carcinomatous components, and MAF <10% or nonreference read count <3 in the component lacking the variant. Empirically, component-specific variants had median counts of 14 independent variant reads in the component in which they were called and 0 variant reads in the component in which they were absent. All other somatic mutations were called in both components or were not excluded and were classified as shared. Phylogenetic trees were constructed for each sample based on the component distributions of somatic mutations. In gene burden analysis, the significance threshold for gene mutations was P = 0.05 for known cancer driver genes (Pan-Cancer gene set; ref. 27) and P = 2.5 × 10−6 for all other genes to account for consideration of ∼20,000 different genes. Chromosomal segments with loss of heterozygosity (LOH) were identified from departure of the minor allele frequencies of heterozygous SNPs in tumor samples from the frequencies seen in matched normal samples (25). Chromosomal arms with elevated rates of LOH specific to sarcomatoid components were identified by using a binomial distribution with a false discovery rate < 0.25. Tumor purities were estimated by using the difference in allele frequencies between tumor and normal components in regions of LOH.

Results in these tumors were compared with sequencing results seen in 424 ccRCCs in The Cancer Genome Atlas (TCGA) ccRCC (28). Local pathology reports of TCGA samples were reviewed to identify samples with sarcomatoid features. Data from cBioPortal was used to determine the frequency of specific mutations, overall mutational burden, and concomitant LOH in these samples (29).

Results

Exome Sequencing of Carcinomatous and Sarcomatoid Elements of ccRCCs.

The clinical and pathologic features of the 21 patients with ccRCC with sarcomatoid transformation are shown in Table S1. Cancer-specific survival was poor, with a 1- and 2-y survival of 38 and 30%, respectively (Fig. S1). Similar to other cohorts, tumors were large (median 10 cm), were frequently associated with metastases (66.6%), and frequently showed local invasion (80.9% T3/T4; Table S1).

Table S1.

Clinical characteristics (n = 21)

Characteristic Classification Mean/median/count
Gender, n (%) Male 15 (71.4)
Female 6 (28.5)
Age, y Mean 60.57 ± 12.3
Median 61
Tumor size, cm Mean 11.27 ± 4.3
Median 10
Tumor stage, n (%) T1 2 (9.5)
T2 2 (9.5)
T3 15 (71.4)
T4 2 (9.5)
N stage, n (%) 0 6 (76.2)
1+ 15 (23.8)
M stage, n (%) 0 7 (33.3)
1 14 (66.6)
% Sarcomatoid Mean 46.9 ± 26.9
Median 50

Means are shown with SDs.

Fig. S1.

Fig. S1.

Patient disease-specific survival.

Carcinomatous and sarcomatoid elements were separately dissected from each tumor. Whole exome sequencing was separately performed on matched DNA samples comprising normal tissue, carcinomatous, and sarcomatoid components of each primary tumor. A summary of sequencing metrics is shown in Table S2. Normal, carcinomatous, and sarcomatoid components were sequenced to a mean depth of 135, 177, and 171 independent reads per targeted base in the exome. Median tumor purity was estimated at 62% (range 33–82%) for the sarcomatoid and 46% (range 18–75%) for the carcinomatous components; these values are similar to estimates in other ccRCC cohorts such as TCGA (median purity 54%, range 18–87%; ref. 28). There was no correlation between median read depth per sample and the number of somatic mutations detected in either carcinomatous or sarcomatoid regions (both r2 < 0.01, P > 0.9), consistent with high sensitivity for calling of somatic mutations.

Table S2.

Sequence summary statistics

Origin Normal Carcinomatous Sarcomatoid
No. of samples 21 21 21
No. of reads (M) 132 210 204
Median independent reads 109 150 142
Mean independent reads 135 177 171
% on genome 87.39 87.29 85.95
% on target 70.47 58.81 57.96
% of targeted bases ≥ 20 reads 94.08 95.56 94.76
Mean error rate, % 0.71 0.92 0.94

Landscape of Mutation Burden.

Somatic single nucleotide variants (SSNVs) and chromosome segments showing LOH were called in each tumor as described in Materials and Methods. In 19 tumors, the mean total number of SSNVs, including both shared and component-specific, was 108 ± 33 (range 41–163; Fig. 1A). The other two tumors were >5 SD outliers in both tumor components, with a total mutation burden of 597 in one tumor and 434 SSNVs in the other. These two tumors also had a mutational signature characteristic of mismatch repair (MMR) deficiency, with an abundance of C:G > T:A transitions and a paucity of A:T > C:G, A:T > T:A, and C:G > G:C transversions (30) (Fig. 1B). These tumors were considered to have hypermutation based on prior definitions (31, 32) and evidence of mismatch repair deficiency. Consistent with this classification, one hypermutated tumor had a heterozygous somatic truncating mutation at R389 in mutS homolog 2 (MSH2) in both carcinomatous and sarcomatoid elements with sarcomatoid-specific LOH at this locus and a sarcomatoid-specific heterozygous E1085K mutation in polymerase ε (POLE). Neither of these samples had a rare germ-line protein-altering variant (minor allele frequency <0.001) in MMR genes (MSH2-6, MLH1, MLH3, PMS1-2, PSMP3, POLE). In contrast, there were no examples of hypermutation in the ccRCCs studied by TCGA (no tumor with more than 128 SSNVs) (28, 32) (P = 0.002) (Fig. S2). Both hypermutated tumors had homozygous/hemizygous von Hippel–Lindau tumor suppressor (VHL) SSNVs, making misclassification unlikely, suggesting that these hypermutated tumors may be more likely to develop sarcomatoid features. Because these hypermutated tumors may differ from the others biologically, these tumors were separately analyzed. Cancer genes with somatic mutations in these tumors are shown in Fig. S3.

Fig. 1.

Fig. 1.

Somatic mutations in 21 renal tumors with sarcomatoid features. (A) Somatic mutation counts in 21 tumors by tumor component. Sample IDs labeled on bottom axis. (B) Somatic mutation pattern by single nucleotide change. (C) Presence of somatic mutations and LOH for significantly mutated and genes of interest. (D) Frequency of LOH events by chromosome region in the carcinomatous (green) and sarcomatoid (red) tumor components for 14 nonhypermutated tumors with complete genome-wide LOH data. (E) Presence of LOH in chromosomal segments with significant sarcomatoid-specific LOH.

Fig. S2.

Fig. S2.

Number of somatic mutations (log scale) in each cohort (TCGA Clear Cell/KIRC; ref. (27) and clear cell and sarcomatoid component of our tumor cohort. Note hypermutated outliers (samples 1 and 2), one of which had a shared and a sarcomatoid-specific MSH2 and POLE mutation, respectively. The median number of mutations was greater for both the carcinomatous and sarcomatoid components compared with the TCGA clear cell cohort (Mann–Whitney u test, *P = 0.005 and **P < 0.001).

Fig. S3.

Fig. S3.

Phylogenetic trees of 21 ccRCC tumors. Branch and trunk lengths correspond to the number of somatic mutations in each tumor component, including shared, carcinomatous-specific, and sarcomatoid-specific mutations. Mutations in previously described ccRCC genes, Pan-Cancer genes, other recurrently mutated genes (TSG101, RQCD1, LRIF1, PTK7, and FAT family), and MMR genes in hypermutated samples are shown. Sample IDs are labeled at top left of each phylogenetic tree.

Common Origin of Carcinomatous and Sarcomatoid Elements.

Among the nonhypermutated tumors, sarcomatoid and carcinomatous elements shared a mean of 45/108 (41.7%) SSNVs, providing unequivocal evidence that these elements arise from a common cell of origin that bears many somatic mutations. The known cancer genes (using the Pan-Cancer gene set; ref. 27) that most frequently shared somatic mutations in both carcinomatous and sarcomatous elements were VHL (SSNV+LOH in both elements in 11/19 tumors), polybromo 1 (PBRM1) (SSNV+LOH in both elements in 4/19 tumors), and SET domain containing 2 (SETD2) (SSNV+LOH in both elements in 4/19 tumors) (Fig. 1C and Table S3). Moreover, these three genes are linked to one another on chromosome 3p; this segment shows LOH in every tumor. Somatic SSNVs in these three genes and LOH of 3p are hallmarks of ccRCC (28), and the evidence that these mutations predate the split of carcinomatous and sarcomatoid elements provides strong evidence that these tumors initially arise as ccRCC.

Table S3.

Gene burden analysis of somatic mutations in 19 nonhypermutated tumors, by tumor component

Gene Known ccRCC driver Pan-Cancer gene Coding size, bp No. of LOF or damaging missense mutations P value No. of other nonsynonymous mutations No. of silent mutations
Shared mutations
 VHL Yes Yes 645 11 2.96e-32 0 0
 SETD2 Yes Yes 7,716 4 2.60e-6 0 0
 PBRM1 Yes Yes 4,837 3 1.98e-5 1 0
 PTEN Yes Yes 1,221 2 1.00e-4 0 0
 UBXN7 No No 1,481 2 1.51e-04 0 0
 KLHL7 No No 1,772 2 1.65e-04 0 0
 GBA No No 1,622 2 1.75e-04 0 0
 PIK3CA Yes Yes 3,227 2 5.39e-4 0 0
 USP53 No No 3,237 2 5.42e-04 0 0
 NEDD4 No No 3,766 2 9.48e-04 0 0
 TLR4 No Yes 2,523 1 0.029 0 0
 TSHZ2 No Yes 3,107 1 0.035 0 0
 KDM6A Yes Yes 4,235 1 0.042 0 0
Sarcomatoid-specific mutations
 TP53 Yes Yes 1,192 6 2.28e-14 0 0
 FAT2 No No 13,047 4 4.84e-05 0 0
 ARID1A Yes Yes 6,878 3 5.04e-05 0 0
 TSG101 No No 1,183 2 6.94e-05 0 0
 PTEN Yes Yes 1,221 2 9.80e-05 0 0
 BAP1 Yes Yes 2,207 2 2.41e-04 0 1
 HIF1A No No 2,496 2 3.05e-04 0 0
 LRIF1 No No 2,314 2 5.23e-04 0 0
 PBRM1 Yes Yes 4,837 2 2.44e-03 0 0
 NRAS No Yes 1,797 1 8.55e-03 0 0
 PTPN11 No Yes 1,559 1 0.020 0 0
 TBL1XR1 No Yes 2,969 1 0.023 0 0
 EPHA3 No Yes 3,955 1 0.043 0 0
 ERBB4 No Yes 574 1 0.044 0 0
Carcinomatous-specific mutations
 RLIM No No 1,878 2 5.20e-05 0 0
 PRPF8 No No 7,050 2 7.08e-04 0 0
 KDM5C Yes Yes 4,709 1 0.025 0 0
 MTOR Yes Yes 7,707 1 0.035 0 0
 PBRM1 Yes Yes 4,837 1 0.041 0 0

Significance threshold was 0.05 for known Pan-Cancer genes and 2.5 x 10−6 for all other genes. P values in bold indicate genes surpassing significance threshold. LOF, loss of function mutations—nonsense, splice site, or small insertion/deletions; damaging missense mutations—mutations at highly conserved positions or predicted as damaging by PolyPhen-2.

Increased Burden of Cancer Driver Mutations in Sarcomatoid Elements.

Among somatic mutations that were specific to either sarcomatoid or carcinomatous elements, sarcomatoid components had a significantly higher burden of unique SSNVs (mean 45 vs. 18 SSNVs per tumor, P = 6.2 × 10−4; Fig. 2A). Similarly, sarcomatoid components had nearly twice the length of element-specific somatic LOH (median 913 Mb vs. 460 Mb, P < 0.05; Fig. 1D).

Fig. 2.

Fig. 2.

Comparison of somatic mutations in carcinomatous and sarcomatoid elements. (A) Mean number of somatic mutations by tumor component for the 19 nonhypermutated tumors. Among all mutations, 41.7% were shared between tumor components. Sarcomatoid regions had a significantly higher number of component-specific mutations (mean 45 vs. 18, P = 6.2 × 10−4 by Wilcoxon signed-rank text). (B) Mean number of nonsynonymous somatic mutations in known Pan-Cancer genes by tumor component. Sarcomatoid regions had a significantly higher number of component-specific mutations (1.42 vs. 0.26, P = 0.002 by Wilcoxon signed-rank test). (C) Ratio of nonsynonymous to synonymous mutations in known Pan-Cancer genes by tumor component.

The minor allele frequencies (MAFs) of component-specific SSNVs were significantly lower than those of shared SSNVs. Among carcinomatous components, the median MAFs were 15.3% vs. 21.4% for component-specific and shared SSNVs, respectively (P < 2.2 × 10−16 by Mann–Whitney u test). Similarly, in sarcomatoid elements the median MAFs were 19.4% vs. 27.0% for component-specific and shared SSNVs, respectively (P < 2.2 × 10−16 by Mann–Whitney u test). These findings are consistent with many component-specific mutations arising after clonal lineage separation.

Component-specific, nonsynonymous SSNVs in known cancer genes were significantly more frequent in sarcomatoid than carcinomatous elements (respectively 27 total, mean 1.4 SSNVs per tumor, vs. 5 total, mean 0.26; P = 0.002 by Wilcoxon signed rank test; Fig. 2B). Nonsynonymous somatic mutations in known cancer genes also occurred more often than expected by chance in sarcomatoid elements (P = 1.7 × 10−6) but not carcinomatous elements (P = 0.08). Consistent with this finding, the ratio of nonsynonymous/synonymous (NS/S) SSNVs was 5.4 for cancer genes vs. 3.7 for other genes in sarcomatoid elements, whereas the NS/S ratio was not elevated among Pan-Cancer genes compared with other genes in carcinomatous elements (2.5 vs. 2.9) (Fig. 2C). These findings lend further support to the evolution of sarcomatoid elements from carcinomatous elements by acquisition of somatic mutations in cancer drivers.

Sarcomatoid-Specific Mutations in Tumor Protein p53, AT-Rich Interaction Domain 1A, and BRCA1 Associated Protein 1.

Among sarcomatoid-specific SSNVs in known cancer genes, the frequency of mutation in tumor protein p53 (TP53) was remarkable (Table S3). There were no SSNVs or segments of LOH involving TP53 among carcinomatous regions in these tumors. In contrast, six sarcomatoid elements acquired biallelic TP53 mutations (six NS SSNVs that were all homozygous/hemizygous via LOH), an event highly unlikely to occur by chance (P = 5.47 × 10−17; Fig. 1C). Phylogenetic trees of the carcinomatous and sarcomatoid components of these six tumors are shown in Fig. 3. Sarcomatoid-specific mutations also occurred in other cancer driver genes, including two mutations in BRCA1 associated protein 1 (BAP1) and three in AT-rich interaction domain 1A (ARID1A). With the exception of one ARID1A mutation, all were accompanied by LOH (P = 3.24 × 10−5 and P = 1.54 × 10−5 for presence of SSNV and LOH in ARID1A and BAP1, respectively). Interestingly, all biallelic TP53, ARID1A, and BAP1 mutations were mutually exclusive (P = 0.08, Monte Carlo simulation), suggesting that these SSNVs may represent alternative pathways toward sarcomatoid transformation (Fig. 1C). Consistent with this interpretation, mutual exclusivity of ARID1A and TP53 is commonly observed in ovarian and endometrial malignancies (33, 34).

Fig. 3.

Fig. 3.

Phylogenetic trees of six TP53-mutant, nonhypermutated ccRCC tumors. Branch and trunk lengths correspond to the number of somatic mutations in each tumor component, including shared, carcinomatous-specific, and sarcomatoid-specific mutations. Mutations in previously described ccRCC genes, Pan-Cancer genes, and other recurrently mutated genes (TSG101, RQCD1, LRIF1, PTK7, and FAT family) are shown. Sample IDs are labeled at top left of each phylogenetic tree.

Among tumors with genome-wide LOH data in both components, several chromosomes showed recurrent segments of sarcomatoid-specific LOH that were unlikely to have occurred by chance. These segments included chromosome 1p (57%, all including ARID1A, q = 0.030); chromosome 9 (86%, all including CDKN2A, q = 0.007), chromosome 10 [36%, all including phosphatase and tensin homolog (PTEN), q = 0.108] chromosome 14 (64%, q = 0.108), chromosome 17p (43%, all including TP53, q = 0.030), chromosome 18 (50%, q = 0.188), and chromosome 22 (29% tumors, q = 0.210) (Fig. 1 D and E).

We also sought other genes with SSNVs that occurred more often than expected by chance on either lineage (Table S3). FAT atypical cadherin 2 (FAT2) was the second most frequently mutated gene in the sarcomatoid-specific gene burden analysis (four SSNVs) and the top mutated gene not previously implicated in ccRCC (P = 4.84 × 10−5). Six other genes not previously implicated in ccRCC harbored sarcomatoid-specific SSNVs in two tumors each, some in segments of LOH. These genes include two additional FAT genes, FAT1 and FAT3, as well as tumor susceptibility 101 (TSG101), ligand-dependent nuclear receptor interacting factor 1 (LRIF1), required for cell differentiation1 homolog (RQCD1), and protein tyrosine kinase 7 (PTK7). Details of these and other SSNVs in driver genes are shown in Table S4. Phylogenetic trees of all tumors, by tumor component and including mutated cancer genes, are shown in Fig. S3.

Table S4.

Nonsynonymous somatic mutations in select genes

Sample Samples with mutation* Gene Chr Pos Nucleotide change No. of ref sarc No. of nonref sarc No. of ref carc No. of nonref carc AA change No. of nonconserved species PolyPhen-2 score LOH§
CCC-FP-10 CS TP53 17 7578406 C > T 39 5 70 10 R175H 2 0.999(D) CS
CCC-FP-12 S TP53 17 7579355 A > G 61 32 130 0 L111P 5 1(D) S
CCC-FP-13 S TP53 17 7577081 T > C 121 32 248 0 E286G 1 0.995(D) S
CCC-FP-19 S TP53 17 7578413 C > T 8 7 61 0 V173M 3 1(D) S
CCC-FP-21 S TP53 17 7578211 C > T 138 19 271 1 R213Q 1 1(D) S
CCC-FP-26 S TP53 17 7577551 TCATGCCGCCCATGCAGGAACTGTTACACATGTAGTT > T 55 8 112 0 M246in_frame_del NA NA S
CCC-FP-28 S TP53 17 7578538 T > A 94 99 185 0 N131I 5 1(D) S
CCC-FP-1 CS VHL 3 10188253 A > T 118 42 128 46 Q132 NA NA CS
CCC-FP-10 CS VHL 3 10191512 CTAGTCA > C 123 15 96 47 L169 NA NA CS
CCC-FP-12 CS VHL 3 10183725 C > A 13 8 18 3 S65X NA NA CS
CCC-FP-15 CS VHL 3 10188251 CA > C 139 180 191 62 Q132fs NA NA CS
CCC-FP-16 CS VHL 3 10183703 CG > C 23 9 14 1 R58fs NA NA CS
CCC-FP-2 CS VHL 3 10191476 ACT > A 29 62 98 35 T157fs NA NA CS
CCC-FP-20 CS VHL 3 10183800 ACTT > A 9 6 20 5 N90in_frame_del NA NA CS
CCC-FP-26 CS VHL 3 10188245 G > C 99 90 194 14 V130L 0 0.979(D) CS
CCC-FP-28 CS VHL 3 10183762 C > A 9 8 15 10 C77X NA NA CS
CCC-FP-4 CS VHL 3 10183861 CA > C 6 6 1 5 H110fs NA NA CS
CCC-FP-5 CS VHL 3 10188295 TA > T 152 87 167 81 P146fs NA NA CS
CCC-FP-7 CS VHL 3 10183797 T > A 5 3 11 7 L89H 4 1(D) CS
CCC-FP-8 CS VHL 3 10188210 T > G 331 39 214 80 L118R 1 1(D) CS
CCC-FP-10 S ARID1A 1 27101611 GC > G 20 9 34 3 Q1414fs NA NA CS
CCC-FP-15 S ARID1A 1 27099030 C > G 80 88 228 0 S1149X NA NA S
CCC-FP-20 S ARID1A 1 27105553 C > T 99 10 170 3 R1505X NA NA
CCC-FP-6 S ARID1A 1 27099947 C > T 39 88 128 12 R1276X NA NA S
CCC-FP-14 S BAP1 3 52436820 CTTCTCTACCTCCTCCTTGAGGCACGCCTCATAGTTTGCAATCTC > c 80 13 133 1 E653fs NA NA CS
CCC-FP-7 S BAP1 3 52443860 G > A 63 14 42 0 P12L 1 0.997(D) CS
CCC-FP-1 CS PBRM1 3 52682418 A > G 194 122 171 79 I252T 2 0.901(P) CS
CCC-FP-12 CS PBRM1 3 52613113 CACAG > C 76 56 125 14 V1139fs NA NA CS
CCC-FP-15 CS PBRM1 3 52643672 CC > AA 49 45 48 13 E742X, P741P NA NA CS
CCC-FP-19 C PBRM1 3 52582176 GC > C 146 0 230 29 A1551fs NA NA CS
CCC-FP-20 S PBRM1 3 52668672 AATCAGGGTATT > A 276 54 230 2 K416fs NA NA CS
CCC-FP-21 CS PBRM1 3 52713619 T > TC 55 32 121 31 R37fs NA NA CS
CCC-FP-29 S PBRM1 3 52643477 T > G 214 27 239 0 I807fs NA NA CS
CCC-FP-21 S PTEN 10 89711913 T > A 145 62 417 0 Y177X NA NA CS
CCC-FP-26 S PTEN 10 89624292 CT > C 161 117 384 1 D22fs NA NA CS
CCC-FP-7 CS PTEN 10 89720677 T > A 120 43 180 68 N276K 0 1(D) CS
CCC-FP-7 CS PTEN 10 89717724 G > CCAC 239 33 338 48 C250in_frame_ins NA NA CS
CCC-FP-16 CS SETD2 3 47164856 G > A 51 23 60 16 R424X NA NA CS
CCC-FP-21 CS SETD2 3 47147612 T > G 27 17 48 16 2 bp upstream of exon 6 NA NA CS
CCC-FP-3 CS SETD2 3 47129632 G > A 299 36 311 63 Q1750X NA NA CS
CCC-FP-7 CS SETD2 3 47162421 CA > C 21 11 39 29 L1235fs NA NA CS
CCC-FP-15 CS FAT1 4 187628245 C > T 17 11 30 8 V913M 35 0.143(B)
CCC-FP-26 CS FAT1 4 187539105 T > C 193 99 364 45 K2879E 13 0.005(B) S
CCC-FP-16 CS FAT2 5 150945470 AG > A 12 2 15 11 L1008fs NA NA
CCC-FP-19 S FAT2 5 150914030 AGAC > A 18 9 51 0 V3122in_frame_del NA NA
CCC-FP-20 S FAT2 5 150947852 C > T 22 5 24 1 R214Q 10 0.958(D)
CCC-FP-26 S FAT2 5 150911450 C > T 30 6 24 0 R3170H 6 0.996(D)
CCC-FP-3 S FAT2 5 150923703 C > T 168 32 158 0 G2329R 0 1(D)
CCC-FP-4 S FAT3 11 92085964 T > C 73 9 100 0 L229P 2 1(D) S
CCC-FP-13 S FAT3 11 92620268 G > C 218 11 203 0 G4347A 7 0.792(D)
CCC-FP-12 S PTK7 6 43098336 C > A 99 40 154 1 A250D 1 1(D)
CCC-FP-17 CS PTK7 6 43111336 G > T 133 18 25 4 E613D 0 0.999(D) S
CCC-FP-19 S RQCD1 2 219457100 G > A 36 14 80 0 R205H 0 1(D)
CCC-FP-20 CS RQCD1 2 219457091 C > T 311 5 158 1 T202M 0 1(D) S
CCC-FP-19 S LRIF1 1 111494368 CA > C 20 12 66 1 D380fs NA NA
CCC-FP-29 S LRIF1 1 111492586 G > C 272 52 285 0 R586G 1 0.992(D)
CCC-FP-3 S TSG101 11 18503287 AT > TG 385 10 343 0 L324F, Y325N 0 0.999(D)
CCC-FP-29 S TSG101 11 18531165 TCCAAA > CCCAA 211 52 299 0 F135fs NA NA

AA, amino acid; carc, carcinomatous components; Chr, chromosome; nonref, number of nonreference reads; Pos, chromosomal position; ref, number of reference reads; sarc, sarcomatoid components.

*Tumor components with mutation [classification (C), exclusive to carcinomatous region; (S), exclusive to sarcomatoid region; (CS), shared by sarcomatoid and carcinomatous regions].

Number of vertebrate species with substitutions at this position.

PolyPhen-2 score [classification (D), probably damaging; (P), possibly damaging; (B), benign].

§Tumor components with LOH [classification (C), exclusive to carcinomatous region; (S), exclusive to sarcomatoid region; (CS), shared by sarcomatoid and carcinomatous regions].

Hypermutated tumors.

Discussion

The presence of sarcomatoid features has long been recognized as an extremely poor prognostic factor in kidney cancer. However, until now, a genetic basis of sarcomatoid transformation has remained largely unknown. Our finding that 43% of somatic mutations are shared between carcinomatous and sarcomatous elements provides conclusive evidence that these elements arise from a common clonal ancestor. Despite the suggestion of a shared clonal origin, there has previously been little evidence that the sarcomatoid component arose in a process of dedifferentiation from a preexisting carcinomatous component (16). The data herein provides strong evidence of a carcinomatous origin. The most frequently mutated cancer drivers that are shared by carcinomatous and sarcomatous elements are the genes that are characteristically mutated in ccRCC. Second, the burden of component-specific SSNVs in known cancer drivers is more than fivefold higher on the sarcomatoid than carcinomatous regions. Third, there is a highly significant burden of sarcomatoid-specific mutation of TP53, implicating a specific gene in development of sarcomatoid elements, along with recurrent mutations and/or segments of LOH affecting other known cancer genes. These findings support a pathogenic sequence in which somatic mutations occurring in a ccRCC drives dedifferention to a sarcomatoid state. Importantly, the finding of highly significant sarcomatoid-specific mutation signals is inconsistent with the observed differences being the result of simple heterogeneity within tumors, in which case differences between carcinomatous and sarcomatoid elements would be expected to be stochastic.

The high frequency of biallelic TP53 mutations in sarcomatoid elements was striking. These biallelic mutations were found in 6 of 19 (31.5%) nonhypermutated tumors. TP53 mutations are otherwise rare in ccRCC (28, 35). For example, among 395 ccRCCs reported by TCGA that do not have sarcomatoid elements, only 6 of 395 (1.5%) have TP53 mutation (P = 3 × 10−6, Fisher exact test, odds ratio 29), and only two of these mutations are in segments of LOH (Fig. S4). These findings are consistent with prior work from Oda et al. who performed a candidate gene study of 14 tumors with sarcomatoid transformation and noted a higher incidence of TP53 mutations in the sarcomatoid region by using immunohistochemistry and Sanger sequencing (36). TP53 alterations may link the EMT pathway to sarcomatoid transformation, because p53 loss can reduce expression of miR-200c, which contributes to EMT (37). Additionally, one hypermutated tumor contained an R175H alteration in TP53, a known gain-of-function mutation that results in up-regulation of TWIST1, an important EMT transcriptional regulator (38). Interestingly, in Wilms’ tumor, another form of renal cancer, loss of TP53 also leads to histologic dedifferentiation (anaplasia) and a poor prognosis (39, 40).

Fig. S4.

Fig. S4.

Comparison of frequency of somatic mutation of most frequently mutated in sarcomatoid regions (red outline) (n = 19) and that observed in nonsarcomatoid clear cell tumors in the TCGA (blue outline) (n = 395). Heterozygous alterations are shaded gray, whereas hemizygous alterations are in white. Fisher’s exact test was used for comparisons. *P < 0.05. TP53, ARID1A, RQCD1, LRIF1, TSG101, PTK7, and FAT1/2/3 were all significantly increased in the sarcomatoid components.

We identified somatic mutations in genes that are characteristic for ccRCC, including VHL, PBRM1, SETD2, PTEN, ARID1A, and BAP1. Notably, all ARID1A and BAP1 SSNVs were exclusive to sarcomatoid regions, all but one were in segments of LOH, and were mutually exclusive with each other and with TP53 mutations. Deficiency of ARID1A and BAP1 has been associated with worse prognosis, higher tumor grade, and a higher incidence of sarcomatoid histology (41, 42). However, many other tumors show only LOH at these loci, suggesting these events may be permissive of, but insufficient for, sarcomatoid transformation. Although mutual exclusivity of mutation in TP53 and ARID1A has been described in ovarian and endometrial cancer, the explanation has been unclear. Recently, their gene products have been shown to form a complex that regulates transcription of CDKN1A and SMAD4 (33), suggesting that mutation of these genes may be equivalent and sufficient to promote tumorigenesis via a common pathway.

The incidence of VHL SSNVs was 57.9% (11/19 tumors). Additionally, all tumors had LOH of chromosome 3p. Consistent with its role as an early event in tumorigenesis (43, 44), all VHL mutations were shared in carcinomatous and sarcomatoid elements. VHL alterations (mutation and hypermethylation) have been considered the hallmark of ccRCC (45, 46). For centrally reviewed ccRCC, the incidence of VHL mutation is as high as 81.3% (47), which suggests that our cohort had a lower incidence of VHL mutation (35, 47). Similarly, it has been shown that wild-type VHL ccRCC has a more aggressive phenotype (48, 49), perhaps related to an increased propensity for sarcomatoid transformation.

Mutations in genes not implicated in ccRCC may be relevant to sarcomatoid transformation. Sarcomatoid-specific mutations in FAT2 were found in five tumors. Mutations in other members of the FAT family, including FAT1 and FAT3, were in two tumors each. FAT family mutations were rarely found in ccRCC in TCGA (Fig. S4). FAT proteins play multiple roles in cell adhesion, motility, polarity, signaling, and proliferation, and mutations are implicated in a variety of cancers (5052). Loss of FAT1 has been shown to promote WNT signaling, a critical mediator of EMT (53). Further exploration of these genes in sarcomatoid elements will be required to assess the significance of the role of FAT genes. Mutations in several other genes were of interest, but will also require larger numbers of samples to assess significance. These mutated genes include TSG101, a member of the ESCRT-I complex involved in ubiquitinated protein trafficking (54); PTK7, a tyrosine kinase regulator of cell motility, adhesion, polarity, and WNT signaling (55); and RQCD1 and LRIF1, both retinoic-acid receptor transcriptional cofactors (56, 57). Several of these genes were enriched in specimens listed as having sarcomatoid transformation in the TCGA dataset (Fig. S4).

Effective systemic therapy for individuals with sarcomatoid renal tumors is an unmet need in oncology. TP53 and ARID1A are among potential sarcomatoid-specific targets for which therapeutics are in development (58, 59). As drugs that mitigate effects of mutation in these genes enter clinical trials, their study in sarcomatoid renal tumors may be warranted. Similarly, hypermutability in sarcomatoid tumors also has implications for treatment of both tumor components. Loss of key mismatch repair genes can sensitize tumors to radiation and some types of chemotherapy (60). These tumors have also shown sensitivity to immunotherapy for several cancer types, perhaps due to the increased burden of somatic mutation-derived epitopes (61, 62). Lastly, PD-1 and PDL-1 expression has also recently been found to be greater in tumors with sarcomatoid features (63), raising the possibility that these tumors may be responsive to immune checkpoint inhibitor immunotherapy.

Acknowledgments

We thank the staff of the Yale Center for Genome analysis for technical excellence in sample preparation, capture, and DNA sequencing, and Marta Boeke, PhD, and Brandon Castor assisted with specimen acquisition. This publication was made possible by Clinical and Translational Science Awards (CTSA) Grant KL2 TR000140 (to B.S.) from the National Center for Research Resources and the National Center for Advancing Translational Science. J.C. was supported through National Cancer Institute Grant K08-CA191019. This work was supported in part by Gilead Sciences. R.P.L. is an Investigator of the Howard Hughes Medical Institute.

Footnotes

The authors declare no conflict of interest.

Data deposition: The data reported in this paper have been deposited in the NCBI dbGap, www.ncbi.nlm.nih.gov (accession no. phs000744).

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1525735113/-/DCSupplemental.

References

  • 1.Shuch B, et al. Cytoreductive nephrectomy for kidney cancer with sarcomatoid histology--is up-front resection indicated and, if not, is it avoidable? J Urol. 2009;182(5):2164–2171. doi: 10.1016/j.juro.2009.07.049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.de Peralta-Venturina M, et al. Sarcomatoid differentiation in renal cell carcinoma: A study of 101 cases. Am J Surg Pathol. 2001;25(3):275–284. doi: 10.1097/00000478-200103000-00001. [DOI] [PubMed] [Google Scholar]
  • 3.Shuch B, et al. Understanding pathologic variants of renal cell carcinoma: Distilling therapeutic opportunities from biologic complexity. Eur Urol. 2015;67(1):85–97. doi: 10.1016/j.eururo.2014.04.029. [DOI] [PubMed] [Google Scholar]
  • 4.Shuch B, et al. Impact of pathological tumour characteristics in patients with sarcomatoid renal cell carcinoma. BJU Int. 2012;109(11):1600–1606. doi: 10.1111/j.1464-410X.2011.10785.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Zhang BY, et al. A novel prognostic model for patients with sarcomatoid renal cell carcinoma. BJU Int. 2015;115(3):405–411. doi: 10.1111/bju.12781. [DOI] [PubMed] [Google Scholar]
  • 6.Delahunt B, et al. Members of the ISUP Renal Tumor Panel The International Society of Urological Pathology (ISUP) grading system for renal cell carcinoma and other prognostic parameters. Am J Surg Pathol. 2013;37(10):1490–1504. doi: 10.1097/PAS.0b013e318299f0fb. [DOI] [PubMed] [Google Scholar]
  • 7.Cheville JC, et al. Sarcomatoid renal cell carcinoma: An examination of underlying histologic subtype and an analysis of associations with patient outcome. Am J Surg Pathol. 2004;28(4):435–441. doi: 10.1097/00000478-200404000-00002. [DOI] [PubMed] [Google Scholar]
  • 8.Merrill MM, et al. Clinically nonmetastatic renal cell carcinoma with sarcomatoid dedifferentiation: Natural history and outcomes after surgical resection with curative intent. Urol Oncol. 2015;33(4):166.e21–166.e29. doi: 10.1016/j.urolonc.2014.11.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Shuch B, Bratslavsky G, Linehan WM, Srinivasan R. Sarcomatoid renal cell carcinoma: A comprehensive review of the biology and current treatment strategies. Oncologist. 2012;17(1):46–54. doi: 10.1634/theoncologist.2011-0227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Golshayan AR, et al. Metastatic sarcomatoid renal cell carcinoma treated with vascular endothelial growth factor-targeted therapy. J Clin Oncol. 2009;27(2):235–241. doi: 10.1200/JCO.2008.18.0000. [DOI] [PubMed] [Google Scholar]
  • 11.Voss MH, et al. 2014. Treatment outcome with mTOR inhibitors for metastatic renal cell carcinoma with nonclear and sarcomatoid histologies. Ann Ocol 25(3):663–668.
  • 12.Nanus DM, Garino A, Milowsky MI, Larkin M, Dutcher JP. Active chemotherapy for sarcomatoid and rapidly progressing renal cell carcinoma. Cancer. 2004;101(7):1545–1551. doi: 10.1002/cncr.20541. [DOI] [PubMed] [Google Scholar]
  • 13.Farrow GM, Harrison EG, Jr, Utz DC. 1968. Sarcomas and sarcomatoid and mixed malignant tumors of the kidney in adults. Cancer 22(1–3):545–563.
  • 14.Anani W, Amin M, Pantanowitz L, Parwani AV. A series of collision tumors in the genitourinary tract with a review of the literature. Pathol Res Pract. 2014;210(4):217–223. doi: 10.1016/j.prp.2013.12.005. [DOI] [PubMed] [Google Scholar]
  • 15.Darai-Ramqvist E, Nilsonne G, Flores-Staino C, Hjerpe A, Dobra K. Microenvironment-dependent phenotypic changes in a SCID mouse model for malignant mesothelioma. Front Oncol. 2013;3:203. doi: 10.3389/fonc.2013.00203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Delahunt B. Sarcomatoid renal carcinoma: The final common dedifferentiation pathway of renal epithelial malignancies. Pathology. 1999;31(3):185–190. doi: 10.1080/003130299104945. [DOI] [PubMed] [Google Scholar]
  • 17.Jones TD, et al. Clonal divergence and genetic heterogeneity in clear cell renal cell carcinomas with sarcomatoid transformation. Cancer. 2005;104(6):1195–1203. doi: 10.1002/cncr.21288. [DOI] [PubMed] [Google Scholar]
  • 18.Oda H, Machinami R. Sarcomatoid renal cell carcinoma. A study of its proliferative activity. Cancer. 1993;71(7):2292–2298. doi: 10.1002/1097-0142(19930401)71:7<2292::aid-cncr2820710720>3.0.co;2-3. [DOI] [PubMed] [Google Scholar]
  • 19.Kuroiwa K, Konomoto T, Kumazawa J, Naito S, Tsuneyoshi M. Cell proliferative activity and expression of cell-cell adhesion factors (E-cadherin, alpha-, beta-, and gamma-catenin, and p120) in sarcomatoid renal cell carcinoma. J Surg Oncol. 2001;77(2):123–131. doi: 10.1002/jso.1082. [DOI] [PubMed] [Google Scholar]
  • 20.Shuch B, et al. Histologic evaluation of metastases in renal cell carcinoma with sarcomatoid transformation and its implications for systemic therapy. Cancer. 2010;116(3):616–624. doi: 10.1002/cncr.24768. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Conant JL, Peng Z, Evans MF, Naud S, Cooper K. Sarcomatoid renal cell carcinoma is an example of epithelial--mesenchymal transition. J Clin Pathol. 2011;64(12):1088–1092. doi: 10.1136/jclinpath-2011-200216. [DOI] [PubMed] [Google Scholar]
  • 22.Goh G, et al. Recurrent activating mutation in PRKACA in cortisol-producing adrenal tumors. Nat Genet. 2014;46(6):613–617. doi: 10.1038/ng.2956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Li H, Durbin R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics. 2009;25(14):1754–1760. doi: 10.1093/bioinformatics/btp324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Cibulskis K, et al. Sensitive detection of somatic point mutations in impure and heterogeneous cancer samples. Nat Biotechnol. 2013;31(3):213–219. doi: 10.1038/nbt.2514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Zhao S, et al. Landscape of somatic single-nucleotide and copy-number mutations in uterine serous carcinoma. Proc Natl Acad Sci USA. 2013;110(8):2916–2921. doi: 10.1073/pnas.1222577110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Abecasis GR, et al. 1000 Genomes Project Consortium An integrated map of genetic variation from 1,092 human genomes. Nature. 2012;491(7422):56–65. doi: 10.1038/nature11632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Kandoth C, et al. Mutational landscape and significance across 12 major cancer types. Nature. 2013;502(7471):333–339. doi: 10.1038/nature12634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Cancer Genome Atlas Research Network Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature. 2013;499(7456):43–49. doi: 10.1038/nature12222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Cerami E, et al. The cBio cancer genomics portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov. 2012;2(5):401–404. doi: 10.1158/2159-8290.CD-12-0095. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Alexandrov LB, et al. Australian Pancreatic Cancer Genome Initiative ICGC Breast Cancer Consortium ICGC MMML-Seq Consortium ICGC PedBrain Signatures of mutational processes in human cancer. Nature. 2013;500(7463):415–421. doi: 10.1038/nature12477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Pritchard CC, et al. Complex MSH2 and MSH6 mutations in hypermutated microsatellite unstable advanced prostate cancer. Nat Commun. 2014;5:4988. doi: 10.1038/ncomms5988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Davis CF, et al. Cancer Genome Atlas Research Network The somatic genomic landscape of chromophobe renal cell carcinoma. Cancer Cell. 2014;26(3):319–330. doi: 10.1016/j.ccr.2014.07.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Guan B, Wang TL, Shih IeM. ARID1A, a factor that promotes formation of SWI/SNF-mediated chromatin remodeling, is a tumor suppressor in gynecologic cancers. Cancer Res. 2011;71(21):6718–6727. doi: 10.1158/0008-5472.CAN-11-1562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Bosse T, et al. Loss of ARID1A expression and its relationship with PI3K-Akt pathway alterations, TP53 and microsatellite instability in endometrial cancer. Mod Pathol. 2013;26(11):1525–1535. doi: 10.1038/modpathol.2013.96. [DOI] [PubMed] [Google Scholar]
  • 35.Sato Y, et al. Integrated molecular analysis of clear-cell renal cell carcinoma. Nat Genet. 2013;45(8):860–867. doi: 10.1038/ng.2699. [DOI] [PubMed] [Google Scholar]
  • 36.Oda H, Nakatsuru Y, Ishikawa T. Mutations of the p53 gene and p53 protein overexpression are associated with sarcomatoid transformation in renal cell carcinomas. Cancer Res. 1995;55(3):658–662. [PubMed] [Google Scholar]
  • 37.Chang CJ, et al. p53 regulates epithelial-mesenchymal transition and stem cell properties through modulating miRNAs. Nat Cell Biol. 2011;13(3):317–323. doi: 10.1038/ncb2173. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Kogan-Sakin I, et al. Mutant p53(R175H) upregulates Twist1 expression and promotes epithelial-mesenchymal transition in immortalized prostate cells. Cell Death Differ. 2011;18(2):271–281. doi: 10.1038/cdd.2010.94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Bardeesy N, Beckwith JB, Pelletier J. Clonal expansion and attenuated apoptosis in Wilms’ tumors are associated with p53 gene mutations. Cancer Res. 1995;55(2):215–219. [PubMed] [Google Scholar]
  • 40.Maschietto M, et al. TP53 mutational status is a potential marker for risk stratification in Wilms tumour with diffuse anaplasia. PLoS One. 2014;9(10):e109924. doi: 10.1371/journal.pone.0109924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Peña-Llopis S, et al. BAP1 loss defines a new class of renal cell carcinoma. Nat Genet. 2012;44(7):751–759. doi: 10.1038/ng.2323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Kapur P, 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(2):159–167. doi: 10.1016/S1470-2045(12)70584-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Gerlinger M, et al. Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat Genet. 2014;46(3):225–233. doi: 10.1038/ng.2891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Gerlinger M, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med. 2012;366(10):883–892. doi: 10.1056/NEJMoa1113205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Gnarra JR, et al. Mutations of the VHL tumour suppressor gene in renal carcinoma. Nat Genet. 1994;7(1):85–90. doi: 10.1038/ng0594-85. [DOI] [PubMed] [Google Scholar]
  • 46.Herman JG, et al. Silencing of the VHL tumor-suppressor gene by DNA methylation in renal carcinoma. Proc Natl Acad Sci USA. 1994;91(21):9700–9704. doi: 10.1073/pnas.91.21.9700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Nickerson ML, et al. 2008. Improved identification of von Hippel-Lindau gene alterations in clear cell renal tumors. Clin Cancer Res 14(15):4726–4734.
  • 48.Patard JJ, et al. Absence of VHL gene alteration and high VEGF expression are associated with tumour aggressiveness and poor survival of renal-cell carcinoma. Br J Cancer. 2009;101(8):1417–1424. doi: 10.1038/sj.bjc.6605298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Gordan JD, et al. HIF-alpha effects on c-Myc distinguish two subtypes of sporadic VHL-deficient clear cell renal carcinoma. Cancer Cell. 2008;14(6):435–446. doi: 10.1016/j.ccr.2008.10.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Furukawa T, et al. Whole exome sequencing reveals recurrent mutations in BRCA2 and FAT genes in acinar cell carcinomas of the pancreas. Sci Rep. 2015;5:8829. doi: 10.1038/srep08829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Morris LG, Ramaswami D, Chan TA. The FAT epidemic: A gene family frequently mutated across multiple human cancer types. Cell Cycle. 2013;12(7):1011–1012. doi: 10.4161/cc.24305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Katoh M. Function and cancer genomics of FAT family genes (review) Int J Oncol. 2012;41(6):1913–1918. doi: 10.3892/ijo.2012.1669. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Morris LG, et al. Recurrent somatic mutation of FAT1 in multiple human cancers leads to aberrant Wnt activation. Nat Genet. 2013;45(3):253–261. doi: 10.1038/ng.2538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Feng GH, Lih CJ, Cohen SN. TSG101 protein steady-state level is regulated posttranslationally by an evolutionarily conserved COOH-terminal sequence. Cancer Res. 2000;60(6):1736–1741. [PubMed] [Google Scholar]
  • 55.Peradziryi H, Tolwinski NS, Borchers A. The many roles of PTK7: A versatile regulator of cell-cell communication. Arch Biochem Biophys. 2012;524(1):71–76. doi: 10.1016/j.abb.2011.12.019. [DOI] [PubMed] [Google Scholar]
  • 56.Hiroi N, et al. Mammalian Rcd1 is a novel transcriptional cofactor that mediates retinoic acid-induced cell differentiation. EMBO J. 2002;21(19):5235–5244. doi: 10.1093/emboj/cdf521. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Li HJ, Haque ZK, Chen A, Mendelsohn M. RIF-1, a novel nuclear receptor corepressor that associates with the nuclear matrix. J Cell Biochem. 2007;102(4):1021–1035. doi: 10.1002/jcb.21340. [DOI] [PubMed] [Google Scholar]
  • 58.Hong B, van den Heuvel AP, Prabhu VV, Zhang S, El-Deiry WS. Targeting tumor suppressor p53 for cancer therapy: Strategies, challenges and opportunities. Curr Drug Targets. 2014;15(1):80–89. doi: 10.2174/1389450114666140106101412. [DOI] [PubMed] [Google Scholar]
  • 59.Bitler BG, et al. Synthetic lethality by targeting EZH2 methyltransferase activity in ARID1A-mutated cancers. Nat Med. 2015;21(3):231–238. doi: 10.1038/nm.3799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Martin SA, Lord CJ, Ashworth A. Therapeutic targeting of the DNA mismatch repair pathway. Clin Cancer Res. 2010;16(21):5107–5113. doi: 10.1158/1078-0432.CCR-10-0821. [DOI] [PubMed] [Google Scholar]
  • 61.Rizvi NA, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348(6230):124–128. doi: 10.1126/science.aaa1348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Le DT, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015;372(26):2509–2520. doi: 10.1056/NEJMoa1500596. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Joseph RW, et al. PD-1 and PD-L1 expression in renal cell carcinoma with sarcomatoid differentiation. Cancer Immunol Res. 2015;3(12):1303–1307. doi: 10.1158/2326-6066.CIR-15-0150. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Proceedings of the National Academy of Sciences of the United States of America are provided here courtesy of National Academy of Sciences

RESOURCES