Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Mar 1.
Published in final edited form as: Hum Pathol. 2022 Aug 4;133:22–31. doi: 10.1016/j.humpath.2022.07.022

The expanding role of BAP1 in clear cell renal cell carcinoma

Payal Kapur 1,2,3,*, Satwik Rajaram 3,4, James Brugarolas 3,5
PMCID: PMC9898467  NIHMSID: NIHMS1852153  PMID: 35932824

Abstract

Mutations drive renal cell carcinoma (RCC) biology and tumor growth. The BRCA1-associated protein-1 (BAP1) gene is frequently mutated in clear cell renal cell carcinoma (ccRCC) and has emerged as a prognostic and putative predictive biomarker. In this review, we discuss the role of BAP1 as a signature event of a subtype of ccRCC marked by aggressiveness, inflammation, and possibly a heightened response to immunotherapy.

Introduction

Clear cell renal cell carcinoma (ccRCC), the most common renal cell carcinoma (RCC) subtype, is a riveting tumor. Genes frequently mutated in other tumors such as lung, breast, colon or prostate cancer to name a few, are infrequently mutated in ccRCC. Instead, ccRCC is characterized by the inactivation of the gene von Hippel-Lindau (VHL) which is rarely mutated in other cancers. VHL is a two-hit tumor suppressor gene, located on chromosome 3p25. Deletions of this region, observed in approximately 90% of ccRCC1-3, generally involve a large segment of chromosome 3p resulting in loss of one allele of VHL. 3p loss typically involves a chromothripsis event that is accompanied by 5q amplification. The second allele of VHL is inactivated either by mutation or methylation4. VHL inactivation results in accumulation of the alpha subunit of HIF (hypoxia-inducible factor) and constitutive expression of target genes5 promoting angiogenesis through upregulation of the vascular endothelial growth factor (VEGF)6,7. VHL inactivation has been found in preneoplastic renal cysts and has been speculated to occur several decades before patients are diagnosed with ccRCC. In genetically engineered mice, VHL inactivation alone does not result in ccRCC8-10. Thus, while VHL inactivation is the critical founding step, additional cooperating driver mutations are needed for ccRCC genesis.

Interestingly, chromosome 3p contains 3 other tumor suppressor genes frequently mutated in ccRCC (PBRM1, SETD2 and BAP1). PBRM1 (Polybromo-1), is mutated in ~40% of ccRCCs11-14. The SETD2 (Set domain-containing protein 2) gene encodes a nonredundant histone 3 lysine 36 trimethyltransferase and is mutated in ~15% of ccRCC15. BAP1 (BRCA1 associated protein 1) is mutated in ~12% of ccRCC12,13,16. Notably, all three genes are involved in epigenetic regulation. Other genes have been implicated in ccRCC pathogenesis including other chromatin-modifying genes such as KDM5C, KDM6A, and MLL2; mTOR pathway genes; and cell cycle regulators such as CDKN2A13,17,18.

The BAP1 tumor suppressor protein

BAP1 has emerged as a critical tumor suppressor gene across multiple cancer types including melanoma, mesothelioma and RCC. BAP1 is mutated both somatically, as well as in the germline. The BAP1 gene is located on chromosome 3p21, has 17 exons, and encodes a 729 amino acid protein of 90 kDa. The BAP1 protein is a deubiquitinating enzyme with an N-terminal ubiquitin carboxy-terminal hydrolase (UCH) domain, a host cell factor 1 binding motif (HBM), and a C-terminal domain containing a coiled-coil motif that interacts with ASXL1/2. BAP1 localizes to the nucleus and contains a nuclear localization signal (NLS) in the C-terminus19,20. BAP1 binds to chromatin and deubiquitinates histone H2A thereby opposing the silencing function of polycomb repressive complexes which ubiquitinate H2A21.

Mutations in BAP1 are distributed throughout the open reading frame. Independently of their destabilizing effects, truncating mutations often interfere with the C-terminal NLS, which is believed to be necessary for BAP1 function22. Missense mutations cluster in the catalytic domain, which is required for tumor suppressor function12,14. Furthermore, BAP1 undergoes self-deubiquitylation which influences subcellular localization23.

Discovered in 1998, BAP1 was found to bind the RING finger domain of the BRCA1 protein24. However, this may not be true in all mammalian cells under normal conditions25. BAP1 regulates processes including DNA damage repair, cellular differentiation, and cell cycle control26.

The familial BAP1 syndrome

Inherited in an autosomal dominant pattern, BAP1 mutations confer a high risk of malignancy. The BAP1 syndrome is characterized by uveal and cutaneous melanoma, mesothelioma, and ccRCC27. It is also associated with an increased risk of benign and malignant tumors including melanocytic tumors/atypical spitz tumors, basal cell carcinoma, hepatocellular carcinoma, cholangiocarcinoma, meningioma and others28-32. RCCs have been reported in 10% of probands, occur at a younger age, and are more aggressive than their sporadic counterparts33. As germline BAP1 mutation predisposes to ccRCCs that are indistinguishable from sporadic ccRCC, BAP1 loss may initiate ccRCC development in some settings.

Advancing our understanding of BAP1 with an immunohistochemistry assay

Using a combination of whole genome and exome sequencing, followed by targeted sequencing, we identified BAP1 mutations in 14% of primary ccRCCs12. Most mutations were predicted to abrogate protein expression and a surrogate immunohistochemistry (IHC) test was developed12. We developed an IHC assay with an antibody raised against the C-terminus of BAP1 and utilized genetically characterized tumor samples as controls12. We applied this assay to a cohort of 175 tumors with BAP1 sequencing data. Positive nuclear staining was observed in 150 cases of which 148 were wild-type. The 2 discordant cases had missense mutations, which likely abrogated protein function, but did not affect the generation of a protein product12. Twenty-five cases were negative by IHC including 22 cases with BAP1 mutations. Three samples lacking a mutation did not express BAP1 protein on western blot and therefore were likely to be BAP1-deficient. This suggested that there may be other mechanism of inactivation besides intragenic mutations. Overall, compared to gene sequencing, the IHC test had a greater sensitivity for BAP1 loss although slightly lower specificity. The positive and negative predictive values of the IHC test were 92% (for detecting BAP1 loss) and 98%, respectively. Moreover, IHC allowed the study of protein expression at the single cell level. This enabled, for example, resolving samples that were seemingly double mutant for BAP1 and PBRM1 based on mutation data, which turned out to be samples with different regions mutated for BAP1 and PBRM1. Another advantage of IHC is its routine, straightforward application and the utilization of formalin fixed paraffin embedded (FFPE) specimens, which are readily available.

Wild-type BAP1 results in nuclear immunostaining in ccRCC tumor cells. BAP1 IHC is not a quantitative assay but nearly all mutations are biallelic (intragenic accompanied by 3p loss) and abrogate nuclear staining. Mutations interfering with expression of the NLS may result in cytoplasmic staining and tumor cells where only cytoplasmic staining is noted are likely BAP1-deficient. In the absence of tumor nuclear staining, it is also important to assess the positive staining in the stromal and inflammatory cells, which offers a positive internal control. The BAP1 IHC assay has been utilized extensively (in more than 5,000 ccRCCs) at the Clinical Laboratory Improvement Amendments (CLIA)–certified University of Texas Southwestern Medical Center (UTSW) laboratory and has enabled significant discoveries12,34-39.

BAP1 anticorrelation with PBRM1 sets foundation for ccRCC molecular classification

Interestingly, we discovered that simultaneous BAP1 and PBRM1 mutations co-occurred far less frequently than one might expect based on their individual frequencies alone12. This mutual anticorrelation was confirmed in meta-analyses as well as in multiple studies including sequencing studies utilizing multiregional sampling17,39. In a meta-analysis, the odds of having mutations in BAP1 were reduced in PBRM1-mutated tumors40. In a cohort involving ~1,300 patients the odds ratio for simultaneous loss of BAP1 and PBRM1 was 0.18 (95% CI, 0.11-0.28; p<0.00001)39. This is in contrast to SETD2 mutations where the odds are increased by two fold in PBRM1-mutated tumors, which is suggestive of cooperativity40. These data show a strong negative selection for simultaneous BAP1 and PBRM1 mutations. Though there is precedent for tumors with loss of BAP1 and PBRM1 in different cell populations of the same tumor41, tumors with simultaneous loss of BAP1 and PBRM1 in the same cell populations are infrequent.

Mutation exclusivity is often observed amongst genes in the same pathway. However, as discussed below, BAP1- and PBRM1-mutant tumors exhibit different histological features, biology, and outcomes12,34,36,39,40. When compared to ccRCC with PBRM1 mutations, BAP1-mutant tumors have characteristic, but independent, gene expression signatures36. We analyzed 308 ccRCC from the TCGA that had RNA-Seq data available and found that when compared to the rest, the BAP1- and PBRM1-mutant signatures did not overlap beyond what is expected by chance alone, indicating that they regulate different processes12. PBRM1 encodes BAF180 (herein referred to as PBRM1), a defining component of a switch/sucrose nonfermentable (SWI/SNF) nucleosome remodeling complex known as PBAF. PBRM1 recruits this complex to nucleosomes with a specific acetylation pattern and regulates DNA accessibility42,43.

Gene mutations drive RCC growth and, similar to other cancers, define different subtypes. Therefore, given the mutual exclusivity, divergent gene expression and biologic behavior (discussed below), these mutations lay the foundation for a molecular genetic classification of sporadic ccRCC that has clinical implications.

BAP1 loss drives tumor aggressiveness

PBRM1-mutant tumors (especially in the absence of additional mutations, i.e. SETD2 mutations) tend to be of low grade. In contrast, BAP1-mutated tumors are of high grade (Figure 2) and are associated with aggressive features such as tumor necrosis, sarcomatoid differentiation and mTORC1 activation12,36,44. An association of aggressive histology with advanced stage was observed in multiple cohorts including the TCGA cohort13,14,36,44,45. Nucleolar size is the main determinant of grade and is controlled by mTORC1, and we found an association between BAP1 loss with mTORC1 activation12.

Figure 2.

Figure 2.

Representative images of clear cell renal cell carcinoma with loss of (A) PBRM1; (B) BAP1; (C) Both BAP1 and PBRM1. Tumors with BAP1 loss are frequently high grade in contrast to those with PBRM1 loss. Loss of BAP1 and PBRM1 anticorrelate and in the rare event when they co occur the tumors have most aggressive features (400x magnification).

The strongest evidence implicating BAP1 (and PBRM1) in promoting ccRCC tumorigenesis and regulating aggressiveness is provided by genetically engineered mouse models (GEMM). Using a Cre line driven by promoter sequences of Pax8 (and Six2) we showed that Vhl targeting alone fails to generate ccRCC. Instead, homozygous deletion of Vhl together with either Bap1 or Pbrm1 successfully caused neoplasms in the mouse kidney9,10. Bap1- and Pbrm1-deficient tumors mirrored the morphology, grade and biologic behavior of human ccRCC10. Bap1-deficient tumors were of high grade, had mTORC1 activation, and increased proliferation rates (by ki-67 staining). In contrast, Pbrm1-deficient tumor exhibited small nest architecture, delicate interconnected vascular networks, low-grade nuclei, and longer latency than Bap1-deficient tumors. Interestingly, in Pbrm1-deficient kidneys, disrupting Tsc1 accelerated tumorigenesis, resulting in higher grade tumors with mTORC1 activation41. These data suggest that mTORC1 activation and BAP1 loss drive tumor grade and aggressiveness in ccRCC. Studies in mice also suggested that the cell of origin for ccRCC may be in the Bowman’s capsule. Overall, these data unequivocally implicate BAP1 and PBRM1 not only as drivers of ccRCC, but also of tumor grade.

Given the association between BAP1 and PBRM1 with nuclear grade12,36, it is not surprising that BAP1- and PBRM1-deficient tumors are associated with different outcomes in patients. In patients with localized ccRCC, median overall survival (OS) for patients with BAP1-mutant tumors was 4.6 years (95% CI, 2.1-7.2), compared to 10.6 years (95% CI, 9.8-11.5) for patients with PBRM1-mutant tumors, corresponding to ~3x higher risk for BAP1 patients of dying from their disease (hazard ratio [HR], 2.7; 95% CI, 0.99-7.6; p=0.044) in an institutional cohort36. These results were validated in a second cohort from TCGA13,36. In addition, similar results were observed using the Mayo registry (a cohort of ~1,400 patients with non-metastatic ccRCC), where loss of BAP1 by IHC was associated with a markedly reduced rate of RCC-specific survival (HR, 3.06; 95% CI, 2.28 - 4.10; p = 6·10−14)34,39 and from multi-institutional tissue microarray that included a cohort of 559 non-metastatic ccRCCs in which BAP1 negative tumors were associated with significantly worse disease-free survival (HR, 2.9; 95% CI, 1.8-4.7; p<0.0001) and OS (HR, 2.0; 95% CI, 1.3-3.1; p=0.001) than patients with BAP1 positive tumors35. These results have been reproduced by multiple groups13,14,17,44,45. One study suggests that the particular type of mutation may affect outcomes. Specifically, investigators found that truncating mutations may be associated with worse cancer-specific survival44, but results have not been reproducible. Although other genes other genes have been associated with an unfavorable prognosis in ccRCC44,46,47, BAP1 is the only one that has been consistently reported to correlate with poor prognostic factors including tumor grade, stage, size, necrosis, metastasis at presentation44,46, and sarcomatoid histology18,48.

Furthermore, we found that patients with loss of both BAP1 and PBRM1, though rare, were associated with even worse outcomes (median RCC-specific survival at 10 years of 42% vs. 87% for patients with wild type tumors for both BAP1 and PBRM1)36,39,49.

BAP1 and PBRM1 (as well as SETD2) loss has also been observed in other RCCs, particularly papillary RCC, though at much lower frequency. The association of PBRM1 loss with survival has been variable, though BAP1 loss has consistently been associated with poor prognosis across RCC subtypes13,38. In papillary RCC, inactivation of PBRM1, BAP1 and SETD2 occurs primarily through mutation and loss of chromosome 3p is rarely observed50. Whether these mutations are driver mutations or whether PBRM1, BAP1, and SETD2 are haploinsufficient in papillary RCC remains to be determined.

The molecular evolution of BAP1-deficient tumors

Sato et al. determined that the mutant allele ratio of BAP1 is similar to VHL in 70-80% of ccRCCs suggesting that BAP1 mutations are subclonal in 20-30%14. Using BAP1 IHC on a single representative section per tumor we identified focal loss of BAP1 in ~3% of ccRCCs34,37. More recently, to ascertain tumor phylogeny and intratumoral heterogeneity, the TRACERx program sequenced ~1200 samples from 105 ccRCC tumors of 100 patients (rare tumors with TCEB1 mutation as well as clear and papillary histology were included). TRACERx confirmed many of the prior studies. They showed mutual exclusivity of BAP1 with PBRM1/SETD2 mutations at a clonal level. BAP1 mutations were associated with low intratumoral heterogeneity (ITH), higher tumor grade and proliferation, suggesting that BAP1 loss confers sufficient fitness advantage to outcompete other mechanisms. In addition, they found that BAP1 had a propensity for being a lone driver as majority of these tumors had no additional driver mutations. Based on the conserved patterns of sequential mutations, they constructed an evolutionary classification that could be applied to ~60% of the tumors (63 of the 105 RCC). Amongst the 20 ccRCCs with BAP1 mutations, 17 could be classified: 12 were categorized as BAP1-driven and 5 as multiple clonal drivers (3 with an additional PBRM1 mutation and one with a SETD2 mutation). The mean time to progression after nephrectomy for these two evolutionary subtypes with BAP1 mutations was half that for subtypes driven by PBRM1 mutations (5.9, 4.7 respectively vs. 11.7 months).

BAP1 loss predicts worse outcome in small renal masses

Loss of BAP1 generally correlates with poor risk features, even in low stage tumors. We found that in small renal masses (SRMs) BAP1 predicted outcomes independently of known prognostic factors (including UCLA integrated staging system [UISS] nomogram) and in patients with low SSIGN (stage, size, grade, and necrosis) score34. This is particularly important as most patients diagnosed with renal cancer today present with SRMs (≤ 4 cm). Given the low risk of metastasis in these patients, many can be followed with active surveillance protocols. However, a small subset behaves aggressively. In a recent study of ~2000 small SRM, BAP1 loss was significantly associated with metastases in multivariable analyses51. The finding that BAP1 is independently prognostic in SRM has practical applications since BAP1 status on biopsy is particularly accurate52. Recent work that included 178 patients with paired core biopsies and subsequent resection of the renal mass found that histology accuracy on the biopsy was excellent, biopsies underestimated grade, necrosis, and sarcomatoid/ rhabdoid features52. Amongst all prognostic variables evaluated in the ccRCC subset, BAP1 status had the highest agreement between biopsies and resection (93.5%; kappa [w] 0.78; 95% CI 0.59-0.96)52. Thus, BAP1 status on needle biopsy may better stratify patients with SRMs.

Deep learning algorithms exploit morphological characteristics of BAP1 deficient tumors

BAP1 has been reported to have a role in cytoskeleton remodeling26. BAP1 loss in cutaneous tumors is associated with characteristic histologic features including epithelioid morphology53. Similarly, ccRCC with BAP1 loss exhibit a characteristic architecture and cytologic features54. These tumors have a tubulopapillary or expanded nested/trabecular architecture with cells characterized by moderate to abundant clear or granular eosinophilic cytoplasm, cytoplasmic globules and well-defined cell borders35,54. Most distinctive are the nuclear features consisting of central round uniform nuclei, smooth open chromatin, large and prominent eosinophilic nucleoli, and subtle perinucleolar clearing (unpublished data and reference54). These tumors frequently have tumor necrosis and a prominent intratumoral lymphocytic infiltrate. These findings set the foundation for evaluating deep learning algorithms to predict BAP1 status from hematoxylin and eosin stained ccRCC images. We have shown that deep learning algorithms can reliability predict BAP1 status using H&E slides alone (AUC=0.87 [range in different hold out cohorts=0.82-0.91]). The discriminative power went beyond grade and enabled the detection of subclonal loss55. The algorithms were also accurate when applied to PDX models55, where the human stroma is replaced by mouse stroma, showing that the morphology-BAP1 relationship is intrinsic to tumor cell characteristics.

BAP1 and inflammation - implications for immunotherapy

The frequency of BAP1 loss is higher in patients with metastasis (17-25%)18,56-58. Similar to localized disease, BAP1 loss has been shown to have poor prognosis in advanced disease.

BAP1 studies have recently shifted focus towards immunological phenotypes59,60. Tumors that respond to immune checkpoint inhibitors (ICI) are commonly associated with increased tumor-infiltrating immune cells, PDL-1 expression, and an inflammatory TME61. Furthermore, there is an association between BAP1 loss and PD-L1 expression; BAP1 negative ccRCCs are more likely to be PD-L1 positive than PBRM1 negative tumors (17.2% versus 8.3%, p=0.01)39.

Interestingly, in the GEMM models, the Bap1-deficient tumors were observed to be associated with lymphocytes (CD4 and CD8 T cell) while Pbrm1-deficient tumors were not, suggesting a causal relationship. To investigate this further we undertook a novel empirical approach. We dissected the gene expression of RCC tumor cells by leveraging PDXs (where only the tumor cells expand)62,63 and generated, by subtraction from the corresponding patient tumor, an empirically derived gene expression signature of the tumor microenvironment (eTME)60. Based on the eTME, two major RCC tumor subtypes were identified: an inflamed and an angiogenesis-enriched/non-inflamed subtype. Inflamed tumors had abundant infiltration of Tregs, NK cells, Th1 cells, neutrophils, macrophages, B cells, CD8+ T cells, and were enriched for BAP1 mutations (p=7.7x10−5). Additionally, an IFN-γ gene signature and PD1 expression strongly correlated with the inflamed subtype. In contrast, non-inflamed tumors were enriched for PBRM1 mutations, an angiogenesis signature, dendritic cells, and mast cells. Provocatively, patients with the inflamed subtype had indicators of systemic inflammation, such as thrombocytosis, and anemia, which are well validated prognostic factors in the IMDC (International Metastatic RCC Database Consortium) risk model64,65. Overall, these analyses show that the molecular makeup of tumor cells could modulate the immune response in the TME.

Thus, mirroring the antithetical relationship observed between PBRM1 and BAP1, are signatures of angiogenesis and inflammation that also tend not to overlap in ccRCC (Figure 3) and have therapeutic implications60,66,67. PBRM1-mutated tumors have been reported to have inferior outcomes to ICI-only regimens compared with those with an angiogenesis-targeting agent66. Others have demonstrated that expression of endogenous retroviruses is enriched in BAP1 mutated tumors and correlates with IO-response68,69. Furthermore, BAP1 mutated tumors demonstrate a more inflamed immune microenvironment suggesting that some form of immune-targeting strategy may benefit these patients59,71. In patients enrolled in two phase III trials (COMPARZ [first line sunitinib vs pazopanib] and RECORD-3 [first line sunitinib vs everolimus] presence of a BAP1 mutation was associated with worse outcomes57,70. Mutations in PBRM1 correlated with statistically longer time to treatment failure with VEGF-targeted therapy. However, a correlation between BAP1 loss and sensitivity to everolimus was not found. These data resulted in an improved genomically-annotated MSKCC risk model that incorporated presence of any mutation in BAP1 or TP53 or both and absence of any mutations in PBRM1 (C-index: original MSKCC model, 0.595 [95% CI 0.557-0.634] vs new model, 0.637 [0.595-0.679])70.

Figure 3.

Figure 3.

Representative images of clear cell renal cell carcinoma with loss of BAP1. Tumors with BAP1 loss are frequently high grade inflamed tumors that express PDL1 (200x magnification).

RNA-seq approaches have been used to predict response by identification of inflamed or angiogenic subtypes. One of the largest gene expression profiling studies to predict treatment response was undertaken for the ~800 metastatic ccRCC patients enrolled in the randomized phase 3 IMmotion151 trial evaluating the combination of atezolizumab (anti-PD-L1) and bevacizumab (anti-VEGF) vs. sunitinib (VEGFR2 inhibitor)71,72. Unsupervised analyses of the most variably expressed genes identified 7 clusters that broadly included an angiogenic and a proliferative group. One of the proliferative clusters (Cluster 4) with the highest BAP1 mutation rate (40%) had the highest PD-L1 protein expression rate (80%). This cluster was characterized by a high immune signature, and showed preferential responsiveness to ICI-containing regimen.

Current treatments for metastatic ccRCC involve ICI, angiogenesis inhibitors and their combinations. Representing two extremes of the angiogenic to proliferative/inflamed tumor spectrum are ccRCC with pancreatic metastases versus those with sarcomatoid differentiation respectively. ccRCC with metastasis to the pancreas (independently of other organs) is associated with remarkably indolent clinical course that is independent of both metastatic tumor burden and IMDC risk scores. These tumors are characterized by low grade, PBRM1 loss, BAP1 retention, and angiogenesis73-75. Consistent with these findings, they exhibit preferential responsiveness to antiangiogenic drugs and are resistant to nivolumab73-75. In contrast, sarcomatoid ccRCCs are inflamed and devoid of vasculature, have high frequency of BAP1 loss (40%) and are resistant to antiangiogenic drugs, but responsive to ICI18,71,76. As immune and vascular traits are readily accessible through analyses of histological slides,73 histology is well poised to advance the biomarker field.

Conclusions

In summary, the BAP1 two-hit tumor suppressor gene cooperates with VHL loss and drives ccRCC with characteristic morphological features including hallmarks of aggressive tumors such as high grade. BAP1 loss is associated with mTORC1 activation and an inflamed, poorly vascularized, tumor microenvironment. Its mutual exclusivity with PBRM1 (which drives low grade, angiogenic tumors) set the foundation for a molecular classification with prognostic and potentially therapeutic implications (Figure 4). BAP1 prognostic role is particularly significant in SRMs, where it functions as an independent prognostic factor. Future studies will dissect the mechanism of tumor suppression and inflammation in the hopes of identifying therapeutic targets that may boost, in particular, immunotherapy.

Figure 4.

Figure 4.

Diagram illustrating that BAP1 and PBRM1 are mutually exclusive, cooperate with VHL loss and drives ccRCC with characteristic morphological features. BAP1 loss is associated with an inflamed, poorly vascularized, high grade ccRCC unlike PBRM1 that drives low grade and angiogenic ccRCC.

Figure 1.

Figure 1.

Representative section of a clear cell renal cell carcinoma with truncating mutation in BAP1 gene. (A)Hematoxylin and eosin-stained section reveals a high-grade tumor with spaced out vasculature and tumor cells with abundant cytoplasm, and high-grade nuclei. (B) Immunohistochemical stain for BAP1 (arrow) shows lack of nuclear immunoreactivity in the tumor nuclei. Background endothelial and inflammatory cells retain BAP1 and serve as internal control (100x magnification).

Acknowledgements

PK is supported by grants from NIH (P50CA196516, R01CA244579, R01CA15447, and R01DK115986), DOD (W81XWH-16-1-0474, W81XWH1910710 and KC200285), and CPRIT (RP200233 and RP220294). SR is supported by grants from CPRIT (RP220294) and DOD (KC200285) and startup funds provided through the Lyda Hill Department of Bioinformatics at UTSW. JB is supported by a grant from NIH (P50 CA196516) and from the Cancer Research & Prevention Institute of Texas (RP180192, and RP180191).

Footnotes

Declaration of Interests

None

References:

  • 1.Zbar B, Brauch H, Talmadge C, Linehan M. Loss of alleles of loci on the short arm of chromosome 3 in renal cell carcinoma. Nature. Jun 25-Jul 1 1987;327(6124):721–4. doi: 10.1038/327721a0 [DOI] [PubMed] [Google Scholar]
  • 2.Beroukhim R, Brunet JP, Di Napoli A, et al. Patterns of gene expression and copy-number alterations in von-hippel lindau disease-associated and sporadic clear cell carcinoma of the kidney. Cancer Res. Jun 1 2009;69(11):4674–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Chen M, Ye Y, Yang H, et al. Genome-wide profiling of chromosomal alterations in renal cell carcinoma using high-density single nucleotide polymorphism arrays. Int J Cancer. Jun 11 2009;125(10):2342–2348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Nickerson ML, Jaeger E, Shi Y, et al. Improved identification of von Hippel-Lindau gene alterations in clear cell renal tumors. Clin Cancer Res. Aug 1 2008;14(15):4726–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Chen W, Hill H, Christie A, et al. Targeting renal cell carcinoma with a HIF-2 antagonist. Nature. Nov 3 2016;539(7627):112–117. doi: 10.1038/nature19796 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Giaccia AJ, Simon MC, Johnson R. The biology of hypoxia: the role of oxygen sensing in development, normal function, and disease. Genes Dev. Sep 15 2004;18(18):2183–94. doi: 10.1101/gad.1243304 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Bruick RK, McKnight SL. Building better vasculature. Genes Dev. Oct 1 2001;15(19):2497–502. doi: 10.1101/gad.931601 [DOI] [PubMed] [Google Scholar]
  • 8.Mandriota SJ, Turner KJ, Davies DR, et al. HIF activation identifies early lesions in VHL kidneys: evidence for site-specific tumor suppressor function in the nephron. Cancer Cell. Jun 2002;1(5):459–68. doi: 10.1016/s1535-6108(02)00071-5 [DOI] [PubMed] [Google Scholar]
  • 9.Wang SS, Gu YF, Wolff N, et al. Bap1 is essential for kidney function and cooperates with Vhl in renal tumorigenesis. Research Support, N.I.H., Extramural Research Support, Non-U.S. Gov't. Proceedings of the National Academy of Sciences of the United States of America. Nov 18 2014;111(46):16538–43. doi: 10.1073/pnas.1414789111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Gu YF, Cohn S, Christie A, et al. Modeling Renal Cell Carcinoma in Mice: Bap1 and Pbrm1 Inactivation Drive Tumor Grade. Cancer Discov. Aug 2017;7(8):900–917. doi: 10.1158/2159-8290.CD-17-0292 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Varela I, Tarpey P, Raine K, et al. Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma. Nature. Jan 27 2011;469(7331):539–42. doi: 10.1038/nature09639 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Pena-Llopis S, Vega-Rubin-de-Celis S, Liao A, et al. BAP1 loss defines a new class of renal cell carcinoma. Research Support, N.I.H., Extramural Research Support, Non-U.S. Gov't. Nature genetics. Jun 10 2012;44(7):751–9. doi: 10.1038/ng.2323 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Creighton CJ, Morgan M, Gunaratne PH, et al. Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature. Jul 4 2013;499(7456):43–9. doi: 10.1038/nature12222 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Sato Y, Yoshizato T, Shiraishi Y, et al. Integrated molecular analysis of clear-cell renal cell carcinoma. Nature genetics. Aug 2013;45(8):860–7. doi: 10.1038/ng.2699 [DOI] [PubMed] [Google Scholar]
  • 15.Dalgliesh GL, Furge K, Greenman C, et al. Systematic sequencing of renal carcinoma reveals inactivation of histone modifying genes. Nature. Jan 21 2010;463(7279):360–3. doi: 10.1038/nature08672 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Guo G, Gui Y, Gao S, et al. Frequent mutations of genes encoding ubiquitin-mediated proteolysis pathway components in clear cell renal cell carcinoma. Nature genetics. Dec 4 2011;44(1):17–9. doi: 10.1038/ng.1014 [DOI] [PubMed] [Google Scholar]
  • 17.Turajlic S, Xu H, Litchfield K, et al. Deterministic Evolutionary Trajectories Influence Primary Tumor Growth: TRACERx Renal. Cell. Apr 19 2018;173(3):595–610 e11. doi: 10.1016/j.cell.2018.03.043 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Motzer RJ, Banchereau R, Hamidi H, et al. Molecular Subsets in Renal Cancer Determine Outcome to Checkpoint and Angiogenesis Blockade. Cancer Cell. Dec 14 2020;38(6):803–817 e4. doi: 10.1016/j.ccell.2020.10.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Okino Y, Machida Y, Frankland-Searby S, Machida YJ. BRCA1-associated protein 1 (BAP1) deubiquitinase antagonizes the ubiquitin-mediated activation of FoxK2 target genes. J Biol Chem. Jan 16 2015;290(3):1580–91. doi: 10.1074/jbc.M114.609834 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Daou S, Hammond-Martel I, Mashtalir N, et al. The BAP1/ASXL2 Histone H2A Deubiquitinase Complex Regulates Cell Proliferation and Is Disrupted in Cancer. J Biol Chem. Nov 27 2015;290(48):28643–63. doi: 10.1074/jbc.M115.661553 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Scheuermann JC, de Ayala Alonso AG, Oktaba K, et al. Histone H2A deubiquitinase activity of the Polycomb repressive complex PR-DUB. Nature. May 13 2010;465(7295):243–7. doi: 10.1038/nature08966 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Ventii KH, Devi NS, Friedrich KL, et al. BRCA1-associated protein-1 is a tumor suppressor that requires deubiquitinating activity and nuclear localization. Cancer Res. Sep 1 2008;68(17):6953–62. doi: 10.1158/0008-5472.CAN-08-0365 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Mashtalir N, Daou S, Barbour H, et al. Autodeubiquitination protects the tumor suppressor BAP1 from cytoplasmic sequestration mediated by the atypical ubiquitin ligase UBE2O. Mol Cell. May 8 2014;54(3):392–406. doi: 10.1016/j.molcel.2014.03.002 [DOI] [PubMed] [Google Scholar]
  • 24.Jensen DE, Proctor M, Marquis ST, et al. BAP1: a novel ubiquitin hydrolase which binds to the BRCA1 RING finger and enhances BRCA1-mediated cell growth suppression. Oncogene. Mar 5 1998;16(9):1097–112. [DOI] [PubMed] [Google Scholar]
  • 25.Hauri S, Comoglio F, Seimiya M, et al. A High-Density Map for Navigating the Human Polycomb Complexome. Cell Rep. Oct 4 2016;17(2):583–595. doi: 10.1016/j.celrep.2016.08.096 [DOI] [PubMed] [Google Scholar]
  • 26.White AE, Harper JW. Cancer. Emerging anatomy of the BAP1 tumor suppressor system. Science. Sep 21 2012;337(6101):1463–4. doi: 10.1126/science.1228463 [DOI] [PubMed] [Google Scholar]
  • 27.Carbone M, Yang H, Pass HI, Krausz T, Testa JR, Gaudino G. BAP1 and cancer. Nat Rev Cancer. Mar 2013;13(3):153–9. doi: 10.1038/nrc3459 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Testa JR, Cheung M, Pei J, et al. Germline BAP1 mutations predispose to malignant mesothelioma. Nature genetics. Aug 28 2011;43(10):1022–5. doi: 10.1038/ng.912 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Abdel-Rahman MH, Pilarski R, Cebulla CM, et al. Germline BAP1 mutation predisposes to uveal melanoma, lung adenocarcinoma, meningioma, and other cancers. J Med Genet. Dec 2011;48(12):856–9. doi: 10.1136/jmedgenet-2011-100156 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Farley MN, Schmidt LS, Mester JL, et al. A novel germline mutation in BAP1 predisposes to familial clear-cell renal cell carcinoma. Molecular cancer research : MCR. Sep 2013;11(9):1061–1071. doi: 10.1158/1541-7786.MCR-13-0111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Popova T, Hebert L, Jacquemin V, et al. Germline BAP1 mutations predispose to renal cell carcinomas. Am J Hum Genet. Jun 6 2013;92(6):974–80. doi: 10.1016/j.ajhg.2013.04.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Walpole S, Pritchard AL, Cebulla CM, et al. Comprehensive Study of the Clinical Phenotype of Germline BAP1 Variant-Carrying Families Worldwide. J Natl Cancer Inst. Dec 1 2018;110(12):1328–1341. doi: 10.1093/jnci/djy171 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Rai K, Pilarski R, Cebulla CM, Abdel-Rahman MH. Comprehensive review of BAP1 tumor predisposition syndrome with report of two new cases. Clin Genet. Mar 2016;89(3):285–94. doi: 10.1111/cge.12630 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.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. Apr 1 2014;120(7):1059–67. doi: 10.1002/cncr.28521 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Kapur P, Christie A, Raman JD, et al. BAP1 immunohistochemistry predicts outcomes in a multi-institutional cohort with clear cell renal cell carcinoma. J Urol. Mar 2014;191(3):603–10. doi: 10.1016/j.juro.2013.09.041 [DOI] [PubMed] [Google Scholar]
  • 36.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. The Lancet Oncology. Feb 2013;14(2):159–167. doi: 10.1016/S1470-2045(12)70584-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Eckel-Passow JE, Serie DJ, Cheville JC, et al. BAP1 and PBRM1 in metastatic clear cell renal cell carcinoma: tumor heterogeneity and concordance with paired primary tumor. BMC Urol. Mar 21 2017;17(1):19. doi: 10.1186/s12894-017-0209-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.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. Jan 2015;33(1):23 e9–23 e14. doi: 10.1016/j.urolonc.2014.10.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Joseph RW, Kapur P, Serie DJ, et al. Clear Cell Renal Cell Carcinoma Subtypes Identified by BAP1 and PBRM1 Expression. J Urol. Jan 2016;195(1):180–7. doi: 10.1016/j.juro.2015.07.113 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Pena-Llopis S, Christie A, Xie XJ, Brugarolas J. Cooperation and antagonism among cancer genes: the renal cancer paradigm. Cancer Res. Jul 15 2013;73(14):4173–9. doi: 10.1158/0008-5472.CAN-13-0360 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Gerlinger M, Horswell S, Larkin J, et al. Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nature genetics. Mar 2014;46(3):225–33. doi: 10.1038/ng.2891 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Brugarolas J PBRM1 and BAP1 as novel targets for renal cell carcinoma. Cancer J. Jul-Aug 2013;19(4):324–32. doi: 10.1097/PPO.0b013e3182a102d1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Yuan J, Chen K, Zhang W, Chen Z. Structure of human chromatin-remodelling PBAF complex bound to a nucleosome. Nature. May 2022;605(7908):166–171. doi: 10.1038/s41586-022-04658-5 [DOI] [PubMed] [Google Scholar]
  • 44.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 KIRC TCGA Research Network. Clin Cancer Res. Jun 15 2013;19(12):3259–67. doi: 10.1158/1078-0432.CCR-12-3886 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Gossage L, Murtaza M, Slatter AF, et al. Clinical and pathological impact of VHL, PBRM1, BAP1, SETD2, KDM6A, and JARID1c in clear cell renal cell carcinoma. Research Support, Non-U.S. Gov't. Genes, chromosomes & cancer. Jan 2014;53(1):38–51. doi: 10.1002/gcc.22116 [DOI] [PubMed] [Google Scholar]
  • 46.Manley BJ, Zabor EC, Casuscelli J, et al. Integration of Recurrent Somatic Mutations with Clinical Outcomes: A Pooled Analysis of 1049 Patients with Clear Cell Renal Cell Carcinoma. Eur Urol Focus. Oct 2017;3(4-5):421–427. doi: 10.1016/j.euf.2016.06.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Hakimi AA, Mano R, Ciriello G, et al. Impact of recurrent copy number alterations and cancer gene mutations on the predictive accuracy of prognostic models in clear cell renal cell carcinoma. J Urol. Jul 2014;192(1):24–9. doi: 10.1016/j.juro.2014.01.088 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Bi M, Zhao S, Said JW, et al. Genomic characterization of sarcomatoid transformation in clear cell renal cell carcinoma. Proceedings of the National Academy of Sciences of the United States of America. Feb 23 2016;113(8):2170–5. doi: 10.1073/pnas.1525735113 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Piva F, Santoni M, Matrana MR, et al. BAP1, PBRM1 and SETD2 in clear-cell renal cell carcinoma: molecular diagnostics and possible targets for personalized therapies. Expert Rev Mol Diagn. 2015;15(9):1201–10. doi: 10.1586/14737159.2015.1068122 [DOI] [PubMed] [Google Scholar]
  • 50.Kovac M, Navas C, Horswell S, et al. Recurrent chromosomal gains and heterogeneous driver mutations characterise papillary renal cancer evolution. Nat Commun. Mar 19 2015;6:6336. doi: 10.1038/ncomms7336 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Kapur P, Zhong H, Araj E, et al. Predicting Oncologic Outcomes in Small Renal Tumors European Urology Oncology. 2022;(under revision) [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Kapur P, Setoodeh S, Araj E, et al. Improving Renal Tumor Biopsy Prognostication With BAP1 Analyses. Arch Pathol Lab Med. May 21 2021;doi: 10.5858/arpa.2020-0413-OA [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Wiesner T, Obenauf AC, Murali R, et al. Germline mutations in BAP1 predispose to melanocytic tumors. Nature genetics. Aug 28 2011;43(10):1018–21. doi: 10.1038/ng.910 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Gallan AJ, Parilla M, Segal J, Ritterhouse L, Antic T. BAP1-Mutated Clear Cell Renal Cell Carcinoma. Am J Clin Pathol. Apr 26 2021;155(5):718–728. doi: 10.1093/ajcp/aqaa176 [DOI] [PubMed] [Google Scholar]
  • 55.Acosta P, Panwar V, Jarmale V, et al. Intratumoral resolution of driver gene mutation heterogeneity in renal cancer using deep learning. Cancer Res. Jun 3 2022;doi: 10.1158/0008-5472.CAN-21-2318 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Carlo MI, Manley B, Patil S, et al. Genomic Alterations and Outcomes with VEGF-Targeted Therapy in Patients with Clear Cell Renal Cell Carcinoma. Kidney Cancer. Jul 26 2017;1(1):49–56. doi: 10.3233/KCA-160003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Hsieh JJ, Chen D, Wang PI, et al. Genomic Biomarkers of a Randomized Trial Comparing First-line Everolimus and Sunitinib in Patients with Metastatic Renal Cell Carcinoma. Eur Urol. Mar 2017;71(3):405–414. doi: 10.1016/j.eururo.2016.10.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Turajlic S, Xu H, Litchfield K, et al. Tracking Cancer Evolution Reveals Constrained Routes to Metastases: TRACERx Renal. Cell. Apr 19 2018;173(3):581–594 e12. doi: 10.1016/j.cell.2018.03.057 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Clark DJ, Dhanasekaran SM, Petralia F, et al. Integrated Proteogenomic Characterization of Clear Cell Renal Cell Carcinoma. Cell. Oct 31 2019;179(4):964–983 e31. doi: 10.1016/j.cell.2019.10.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Wang T, Lu R, Kapur P, et al. An Empirical Approach Leveraging Tumorgrafts to Dissect the Tumor Microenvironment in Renal Cell Carcinoma Identifies Missing Link to Prognostic Inflammatory Factors. Cancer Discov. Sep 2018;8(9):1142–1155. doi: 10.1158/2159-8290.CD-17-1246 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Vuong L, Kotecha RR, Voss MH, Hakimi AA. Tumor Microenvironment Dynamics in Clear-Cell Renal Cell Carcinoma. Cancer Discov. Oct 2019;9(10):1349–1357. doi: 10.1158/2159-8290.CD-19-0499 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Sivanand S, Pena-Llopis S, Zhao H, et al. A validated tumorgraft model reveals activity of dovitinib against renal cell carcinoma. Science translational medicine. Jun 6 2012;4(137):137ra75. doi: 10.1126/scitranslmed.3003643 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Elias R, Tcheuyap VT, Kaushik AK, et al. A renal cell carcinoma tumorgraft platform to advance precision medicine. Cell Rep. Nov 23 2021;37(8):110055. doi: 10.1016/j.celrep.2021.110055 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Motzer RJ, Bacik J, Murphy BA, Russo P, Mazumdar M. Interferon-alfa as a comparative treatment for clinical trials of new therapies against advanced renal cell carcinoma. J Clin Oncol. Jan 1 2002;20(1):289–96. doi: 10.1200/JCO.2002.20.1.289 [DOI] [PubMed] [Google Scholar]
  • 65.Heng DY, Xie W, Regan MM, et al. Prognostic factors for overall survival in patients with metastatic renal cell carcinoma treated with vascular endothelial growth factor-targeted agents: results from a large, multicenter study. J Clin Oncol. Dec 1 2009;27(34):5794–9. doi: 10.1200/jco.2008.21.4809 [DOI] [PubMed] [Google Scholar]
  • 66.McDermott DF, Huseni MA, Atkins MB, et al. Clinical activity and molecular correlates of response to atezolizumab alone or in combination with bevacizumab versus sunitinib in renal cell carcinoma. Nat Med. Jun 2018;24(6):749–757. doi: 10.1038/s41591-018-0053-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Brannon AR, Reddy A, Seiler M, et al. Molecular Stratification of Clear Cell Renal Cell Carcinoma by Consensus Clustering Reveals Distinct Subtypes and Survival Patterns. Genes Cancer. Feb 1 2010;1(2):152–163. doi: 10.1177/1947601909359929 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Smith CC, Beckermann KE, Bortone DS, et al. Endogenous retroviral signatures predict immunotherapy response in clear cell renal cell carcinoma. J Clin Invest. Nov 1 2018;128(11):4804–4820. doi: 10.1172/JCI121476 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Panda A, de Cubas AA, Stein M, et al. Endogenous retrovirus expression is associated with response to immune checkpoint blockade in clear cell renal cell carcinoma. JCI Insight. Aug 23 2018;3(16)doi: 10.1172/jci.insight.121522 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Voss MH, Reising A, Cheng Y, et al. Genomically annotated risk model for advanced renal-cell carcinoma: a retrospective cohort study. The Lancet Oncology. Dec 2018;19(12):1688–1698. doi: 10.1016/S1470-2045(18)30648-X [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Motzer RJ, Banchereau R, Hamidi H, et al. Molecular subsets in renal cancer determine outcome to checkpoint and angiogenesis blockade. Cancer Cell. 2020; [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Brugarolas J, Rajaram S, Christie A, Kapur P. The Evolution of Angiogenic and Inflamed Tumors: The Renal Cancer Paradigm. Cancer Cell. Dec 14 2020;38(6):771–773. doi: 10.1016/j.ccell.2020.10.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Kapur P, Christie A, Rajaram S, Brugarolas J. What morphology can teach us about renal cell carcinoma clonal evolution Review. Kidney Cancer Journal. 2020;18(3):68–75. [PMC free article] [PubMed] [Google Scholar]
  • 74.Singla N, Xie Z, Zhang Z, et al. Pancreatic tropism of metastatic renal cell carcinoma. JCI Insight. 2020; [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Cai Q, Christie A, Rajaram S, et al. Ontological analyses reveal clinically-significant clear cell renal cell carcinoma subtypes with convergent evolutionary trajectories into an aggressive type. EBioMedicine. May 2020;51:102526. doi: 10.1016/j.ebiom.2020.102707 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Tannir NM, Signoretti S, Choueiri TK, et al. Efficacy and Safety of Nivolumab Plus Ipilimumab versus Sunitinib in First-line Treatment of Patients with Advanced Sarcomatoid Renal Cell Carcinoma. Clin Cancer Res. Jan 1 2021;27(1):78–86. doi: 10.1158/1078-0432.CCR-20-2063 [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES