Next Article in Journal
2022 WUOF/SIU International Consultation on Urological Diseases: Hereditary Renal Cell Carcinoma Syndromes
Previous Article in Journal
2022 WUOF/SIU International Consultation on Urological Diseases: Kidney Cancer Screening and Epidemiology
 
 
Société Internationale d’Urologie Journal is published by MDPI from Volume 5 Issue 1 (2024). Previous articles were published by another publisher in Open Access under a CC-BY (or CC-BY-NC-ND) licence, and they are hosted by MDPI on mdpi.com as a courtesy and upon agreement with Société Internationale d’Urologie.
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

2022 WUOF/SIU International Consultation on Urological Diseases: Genetics and Tumor Microenvironment of Renal Cell Carcinoma

by
Sari Khaleel
1,
Christopher Ricketts
2,
W. Marston Linehan
2,
Mark Ball
2,
Brandon Manley
3,
Samra Turajilic
4,5,
James Brugarolas
6,7 and
Ari Hakimi
1,*
1
Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, United States
2
Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, United States
3
Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, United States
4
The Francis Crick Institute, London, United Kingdom
5
Renal Unit, The Royal Marsden Hospital, London, United Kingdom
6
Kidney Cancer Program, Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, United States
7
Department of Internal Medicine, UT Southwestern Medical Center, Dallas, United States
*
Author to whom correspondence should be addressed.
Soc. Int. Urol. J. 2022, 3(6), 386-396; https://doi.org/10.48083/BLPV3411
Submission received: 30 August 2022 / Revised: 1 October 2022 / Accepted: 3 October 2022 / Published: 16 November 2022

Abstract

:
Renal cell carcinoma is a diverse group of diseases that can be distinguished by distinct histopathologic and genomic features. In this comprehensive review, we highlight recent advancements in our understanding of the genetic and microenvironmental hallmarks of kidney cancer. We begin with clear cell renal cell carcinoma (ccRCC), the most common subtype of this disease. We review the chromosomal and genetic alterations that drive initiation and progression of ccRCC, which has recently been shown to follow multiple highly conserved evolutionary trajectories that in turn impact disease progression and prognosis. We also review the diverse genetic events that define the many recently recognized rare subtypes within non-clear cell RCC. Finally, we discuss our evolving understanding of the ccRCC microenvironment, which has been revolutionized by recent bulk and single-cell transcriptomic analyses, suggesting potential biomarkers for guiding systemic therapy in the management of advanced ccRCC.

Introduction

Understanding the genomic landscape of clear cell renal cell carcinoma (ccRCC), which accounts for approximately 75% of all renal cell carcinomas, has been critical to the development of targeted systemic therapies to treat this classically chemo- and radiotherapy-resistant disease. However, malignant cells exist in a dynamic and heterogeneous ecosystem of immune cells, stromal cells, cytokines, and extracellular proteins that together constitute the tumor microenvironment (TME) [1], which modulates tumor development and response to systemic therapies in RCC [2]. Better understanding of the TME of ccRCC has helped understand the heterogeneity of response to systemic therapies within ccRCC patients, particularly in the age of immuno-oncologic agent-based therapies. Furthermore, while ccRCC constitutes the majority of RCC tumors, the remaining 25% are represented by an ever-expanding group of tumor subtypes, each with unique histologies and genetic features. This review begins with a summary of recent molecular analyses of clear and non-clear cell RCC subtypes, followed by a discussion of the current understanding of the TME of ccRCC and its role in driving the response to systemic therapies in advanced ccRCC.

Genetics of Clear Cell RCC

The first step toward malignant transformation in ccRCC is the loss of the short arm of chromosome 3 (3p loss), which harbors 4 tumor suppressor genes that constitute the most common sites of mutation in RCC: VHL on 3p.25, and PBRM1, BAP1, and SETD2 on 3p.21 [3,4]. Of these genes, VHL is the most commonly altered in both hereditary and sporadic RCC, through point mutations and methylation in 70%–80% and 5%–10% of patients, respectively [3,5]. Inactivation of the VHL protein results in loss of regulation and thus constitutive activation of its ubiquitin ligase target, the protein HIF. Resulting de-regulation of HIF targets, including vascular endothelial growth factor (VEGF), promotes tumor cell proliferation, neoangiogenesis, and metastases [6,7]. PBRM1 is the second most commonly mutated gene (40% of cases) [3,4], and encodes BAF180 [6,8], a component of the switching defective/sucrose non-fermenting (SWI/ SNF) family of chromatin-remodeling complexes, which determine DNA accessibility to transcription factors and polymerases [8,9,10]. Similarly, BAP1, mutated in 10%–15% of ccRCC patients [11], encodes a nuclear deubiqutinase protein that interacts with host cell factor-1 (HCF-1), which is involved in chromatin remodeling [12,13]. Interestingly, BAP1 and PBRM1 mutations are generally mutually exclusive [3,5,14]. Lastly, while the mechanism by which SETD2, mutated in 10%–15% of ccRCC, affects tumorigenesis remains unclear, it is suspected to involve DNA double-strand break repair, DNA methylation, and RNA splicing [4,15] (Table 1).
The mechanism of 3p loss that results in loss of heterogeneity (LOH) for the above genes frequently involves chromothripsis, a process in which some chromosomes undergo multiple breaks simultaneously, followed by random joining of chromosomal fragments, resulting in hundreds of genomic rearrangements [16]. This initial 3p loss constitutes the “first hit” event and occurs somatically years before the presentation of ccRCC.
A “second hit” resulting in biallelic inactivation of VHL then promotes malignant transformation through upregulation of the hypoxia response in the presence of normoxia. This is usually followed by mutations involving the neighboring PBRM1, SETD2, and BAP1 genes, and less frequently, alterations of TP53, mTOR, TSC1, TSC2, PIK3CA, PTEN, KDM5C and SMARCA4 [17].
Although the repertoire of mutations and somatic copy number alterations (SCNAs) that drive ccRCC is relatively narrow, molecular diversity is achieved through clonal evolution, i.e., selection of cell subpopulations characterized by different driver mutations, resulting in intratumor heterogeneity (ITH) [6]. Consequently, molecular profiling of tumor samples collected from a single spatial location may capture clonal events propagated in all the cancer cells of a given tumor, but can easily miss events in subclones, and under- or over-estimate the frequency of altered genes, an issue that is amplified by the particularly high levels of ITH in ccRCC [18,19]. Therefore, multi-region sampling is critical to capturing the clonal evolution of ccRCC, as demonstrated by the TRACERx Renal program [19]. In the interim analysis TRACERx, molecular profiling of > 1200 primary tumor regions from 100 patients demonstrated clear evidence for highly conserved evolutionary mutational patterns in ccRCC within different clones [20]. Broadly, 2 modes of evolution were observed: linear, in which only a single clonal population is evident, with consequently low ITH; and branched, which involves multiple subclonal populations with high ITH. These populations then evolve either through a linear Darwinian-like process of sequentially selected mutational events, or punctuated evolution, which is noted by short bursts of many genomic alterations occurring in a relatively brief period early in the tumor’s evolution, most likely due to SCNAs and structural chromosomal alterations [21].
ccRCC tumors in the TRACERx cohort that were characterized by linear evolution harbored only 3p loss and VHL mutation/methylation with low ITH, and were thus termed “VHL mono drivers” [20]. These tumors were enriched for small renal masses (SRMs, < 4 cm in maximal dimension), with limited progression and metastatic risk given the limited fitness advantage provided by isolated VHL mutation [22]. In contrast, ccRCC tumors characterized by branched evolution harbored high levels of ITH and parallel evolution [20], i.e., repeat selection of distinct driver mutations in the same gene or pathway, with a highly conserved order of genomic events across clones. Intriguingly, these tumors were larger and more likely to produce metastases than their VHL mono driver counterparts, but with an intermediate metastatic efficiency resulting in solitary metastasis or oligometastases [20]. However, other studies suggest that VHL mutations alone are not sufficient for ccRCC development [23,24]. In contrast, ccRCC tumors characterized by punctuated evolution had low ITH and were dominated by a single clone but exhibited additional molecular alterations in the dominant clone that distinguished them from the similarly monoclonal VHL mono drivers. This tumor evolution group included a VHL-wildtype subtype, a VHL-followed by BAP1 mutation (BAP1-driven) subtype, and tumors with multiple clonal driver mutations (PBRM1, BAP1, STED2 or PTEN). These tumors grew rapidly to a large size and were linked to widespread and rapid metastases [20]. Within this group, BAP1-deficient tumors were associated with higher grade and aggressiveness than PBRM1-deficient tumors [11,24,25], while PBRM1 loss was associated with metastasis tropism to the pancreas [26], and these tumors are characteristically indolent. However, PBRM1-deficient tumors can become more aggressive with further evolution and mutations in the mTOR pathway [24] or SETD2 [27], with which PBRM1 cooperates [25].

Genetics of Non-Clear Cell Carcinoma

Papillary renal cell carcinoma

Papillary renal cell carcinoma has been classically subdivided into 2 subtypes on the basis of histology and genetic features [28]. Genetically, type 1 pRCC is associated with frequent gains of chromosomes 7 and 17, as well as less frequent gains of chromosomes 2, 3, 12, 16, and 20 [29,30,31,32]. The most frequent somatic mutational events in type 1 pRCC are activating mutations of the MET oncogene on chromosome 7, present in 10%–15% of type 1 pRCC cases [33]. Notably, germline activating mutations of the MET oncogene are the pathogenic cause of hereditary papillary renal cell carcinoma (HPRC) syndrome, in which patients present with bilateral, multifocal type 1 pRCCs [31,34] (Table 1).
In contrast, type 2 pRCC tumors are not associated with a specific pattern of copy number alterations, and are now seen to represent a heterogenous group of what are now distinct RCC subtypes, including translocation RCC, FH-deficient RCC, and SDH-deficient RCC. In light of the above heterogeneity and the absence of characteristic genomic features for this group, pRCC type 2 tumors may also be interpreted as aggressive, unclassified RCC that exhibit papillary features but require specific genomic subclassification for clinical outcome prediction [35]. Similarly, while type I pRCC is considered the “classical” morphologic entity, certain neoplasms that exhibit its features may also be considered variants or potential new RCC entities [36], with distinct molecular features, such as papillary renal neoplasm with reversed polarity (PRNRP) [37] and biphasic hyalinizing psammomatous RCC (BHP RCC) [38], which have distinct driver mutations (KRAS and NF2, respectively).

Chromophobe renal cell carcinoma (chRCC)

Like ccRCC and pRCC type 1 tumors, most chRCC are characterized by a distinct pattern of chromosomal alterations, defined by combined loss of chromosomes 1, 2, 6, 10, 13, and 17, seen in approximately 80% of chRCC. Less frequent additional individual losses can occur for chromosomes 3, 5, 8, 9, 11, 18, and 21q in 12%–58% of cases [39,40]. The histology of chRCC can include a rarer eosinophilic variant in which the classic pattern of chromosomal losses is less common. ChRCC have a lower mutation burden than ccRCC or pRCC-1; only TP53 and PTEN are frequently mutated in ~30% and ~8% of cases, respectively [33,41]. Loss of CDKN2A, by either loss of 9p21 or hypermethylation, is the next most common alteration, affecting 19.8% [33] (Table 1). Increased TERT expression has been observed in approximately 17% of ChRCC, resulting from either mutations or genomic rearrangements in the TERT gene promoter, the latter including intra-chromosomal rearrangements and translocations with chromosome 13.11 [42].

Medullary Renal Carcinoma

Renal medullary carcinoma (RMC) is a rare and aggressive subtype of kidney cancer that accounts for less than 1% of all RCC and has a propensity for early metastases, resulting in a median overall survival of little more than a year [43,44,45]. RMC predominantly afflicts individuals with sickle cell trait, creating a preponderance of patients with African or Mediterranean descent, and the young, with a median age from 19 to 22 years [43,44,45,46,47,48,49]. The characteristic genetic and immunohistochemical feature of RMC is the near universal loss of expression of the SWI/SNF-related matrix-associated actin-dependent regulator of chromatin subfamily B member 1 (SMARCB1) protein, also known as integrase interactor 1 (INI1), BRG1-associated factor 47 (BAF47), or sucrose non-fermenting 5 (SNF5). The SMARCB1 protein is encoded by the SMARCB1 gene on chromosome 22q11.23, and in most tumors both copies of this gene are lost through a combination of mutation and chromosomal deletion (Table 1) [50]. SMARCB1 is a core subunit of the SWI/SNF chromatin remodeling complex; its loss results in transcriptional dysregulation of many pathways [9,51].

FH-deficient and SDH-deficient renal cell carcinoma

Hereditary leiomyomatosis and renal cell carcinoma (HLRCC) is a familial cancer syndrome characterized by the development of cutaneous and uterine leiomyomas and a highly aggressive form of kidney cancer [52,53,54,55]. HLRCC is associated with germline mutation of the Krebs cycle enzyme gene fumarate hydratase (FH); because the associated tumors demonstrate loss of FH enzyme activity, they are referred to as FH-deficient RCC [56,57,58]. FH can also be mutated somatically. Similarly, germline mutations of several subunits of the Krebs cycle succinate dehydrogenase enzyme, including SDHB, SDHC, or SDHD, have been associated with increased risk for paraganglioma (PGL), pheochromocytoma, gastrointestinal stromal tumor (GIST), and RCC [59,60,61].
The complete loss of either FH or SDH enzyme activity impairs the normal function of the Krebs cycle, resulting in accumulation of intracellular fumarate and succinate, respectively [62,63]. This accumulation promotes a pseudo-hypoxic state that upregulates several enzymes, particularly enzymes involved in chromatin hypermethylation [32,64,65,66,67]. Furthermore, FH and SDH loss results in aberrant succination of KEAP1 protein, which promotes constitutive upregulation of the NRF2-antioxidant response element (ARE) pathway and inactivation of the core factors responsible for replication and proofreading of mitochondrial DNA (mtDNA), resulting in both a significant decrease in mtDNA content and increased mtDNA mutation [68,69].
While SDH and FH-deficient tumors share similar genetic characteristics, a recent germline analysis comparing these tumors noted that while most of these tumors harbored germline alterations in their respective genes, SDH-deficient RCCs had a lower mutation burden and SCNA burden than FH-deficient RCCs [70]. In addition to patients with germline mutation, a small number of sporadic tumors have also been shown to have complete somatic loss of FH, resulting in a non-hereditary form of FH-deficient RCC [32].

Translocation renal cell carcinoma involving

TFE3, TFEB, or MITF gene fusions

Translocation renal cell carcinomas (T-RCCs) are driven by somatic chromosomal translocations that fuse members of the MiT transcription factor family genes, TFE3, TFEB, or MITF, with various partner genes that result in fusion proteins [31,71,72] that affect many pathways, such as organelle biogenesis, cell proliferation, and cellular fate commitment, all of which may promote tumorigenesis [72,73,74]. T-RCCs represent one of the most common forms of RCC in children and young adults, making up 20%–50% of pediatric RCC patients and 15% of RCC patients under the age of 45 [72,75]. T-RCC in adults can present with a variety of histologies, including both papillary or clear cell [32,72]. To date, fusions involving TFE3 are the most common, followed by TFEB [31,33,71,72,76].

Tumor Microenvironment of RCC

While initial profiling studies of the TME of RCC tumors grouped its tumor phenotypes into either immune-infiltrated or excluded phenotypes [77,78], more recent studies have found infiltrating T cell populations to exist in a continuum from activated antitumor to dysfunctional “exhausted” T cells [79,80]. Similarly, while the function of tumor-associated macrophages (TAMs) has classically been divided into either the proinflammatory/antitumor M1 or the anti-inflammatory/pro-tumor M2 phenotypes (polarizations) [81,82,83,84,85], recent evidence shows that TAM populations are highly plastic, existing in more of a phenotypic spectrum between the M1 and M2 phenotypes in vivo [81,83,84,86].
More recent studies have shifted to transcriptomic analyses that utilize microarray and next generation sequencing (RNA-seq) technologies along with computational techniques to deconvolute the TME to its cellular components and explore their role in tumor response to systemic therapies through an array of gene expression signatures representative of novel cell phenotypes and processes [77,87,88,89]. Such studies noted a generally negative correlation between enrichment of T-helper subtype 2 (Th2) cells and T-reg cells and survival in ccRCC [77], explaining previously reported negative association between T cell infiltration and clinical outcomes in ccRCC [83,84,90,91]. Similarly, worse overall survival and lower likelihood of response to TKI agents were associated with higher levels of M2-type macrophage infiltration in the TME [92]. This understanding of the TME led to investigations of prognostic and theranostic transcriptomic gene signatures that may predict survival and response to systemic therapy in advanced RCC. These include angiogenesis-associated signatures to predict response to tyrosine kinase inhibitors [89,92,93] or immune signatures to predict response to immune checkpoint blockage (ICB)-based combinations [89,93,94]; and transcriptomic classifiers such as the 4 molecular subtypes (ccRCC 1-4) described by Beuselinck et al. for metastatic ccRCC (m-ccRCC) [95], which were shown to predict both survival outcomes as well as therapeutic response to TKI (sunitinib or pazopanib) monotherapy, which were attributed to inherent differences in the their underlying TME [95,96,97]. Prospective patient selection for ICB-based or TKI-monotherapy based on these subtypes was recently evaluated in the phase II BIONIKK trial, which demonstrated the feasibility of biomarker-driven tailored systemic therapy in m-ccRCC [98], potentially maximizing therapeutic benefit while reducing unnecessary toxicity from systemic therapy regimens in m-ccRCC.
Understanding of the TME was further revolutionized by single-cell based analyses such as single-cell RNA sequencing (scRNA-seq) and single-cell mass cytometry (scMC), which allow for massively parallel, high-dimensional analyses of specific cell populations in the TME, enabling prediction of potential interactions between various cell populations based on their expressed surface molecules, promoting a much more granular understanding of the dynamics of the TME of RCC than what was offered by bulk RNA-sequencing approaches, which are bound to oversimplify tumor cell populations and their dynamic interactions [81,83,84,86,99]. In this regard, Chevrier et al. [86] used scMC to profile adaptive and innate (T cell and TAM) populations in the TME of 73 patients with untreated advanced RCC and 5 healthy matched kidney samples. Using computational phenotype clustering, they identified 22 T cell and macrophage phenotypes [86], noting a “terminally exhausted” PD1+ cluster and a corresponding “progenitor exhausted” cluster of potentially ICB-responsive T cells. They also noted 17 different TAM clusters, arguing that the M1/M2 polarization phenotypes are an oversimplification of this plastic and dynamically changing cell population. Finally, they noted immunosuppressed T cell compartments to be associated with high levels of regulatory CD4 cells and a pro-tumor TAM population [86].
Following this study, Braun et al. [83] performed scRNA-seq and T cell receptor (TCR) sequencing of ccRCC tissue from 13 patients with tumors of a range of clinical stages to explore changes in the immune TME with advancing disease. They again noted significant diversity within the TAM and T cell populations, and found T cells to exhibit an overall trend of progressive dysfunction and exhaustion with advancement in disease stage, which was associated with a concurrent shift from M1 to M2-like signatures in the TAM population and increasing T cell and TAM interactions, again confirming that TAMs play a key role in the progression of T cells toward exhaustion in ccRCC [83].
To examine the influence of ICB on the RCC TME, Krishna et al. [81] used scRNA-seq to compare the TME of multi-regional tumor samples from 4 ICB-treated to 2 ICB-naïve advanced ccRCC patients. They noted significant intratumoral and inter-patient heterogeneity, along with differences in the overall TME between ICB-treated versus naïve patients. Focusing on tumor specimens from an ICB-treated patient that exhibited complete response, they noted enrichment of CD8A+ tissue-resident populations and low TAM infiltration in all tumor regions. In contrast, specimens from ICB-resistant patients exhibited high TAM infiltration but low T cell enrichment (i.e., T cell exclusion) [81]. Similarly, Bi et al. compared tumors from 5 ICB-exposed to 3 ICB-naïve patients with advanced ccRCC, and noted that while ICB-exposed tumors were enriched in CD8+ T cells that expressed costimulatory molecules associated with the “progenitor exhausted” phenotype described by Chevrier et al. [86], they also paradoxically expressed inhibitory molecules associated with terminally exhausted T cells, suggesting that these ICB-responsive cells were potentially undergoing a shift toward terminal exhaustion as well. Similarly, antitumor TAM populations in ICB-exposed patients were noted to paradoxically express molecules that correlate with a pro-inflammatory, antitumor phenotype, but again with upregulation of immune checkpoint and anti-inflammatory signaling genes. The authors proposed that these seemingly paradoxical changes in both T cell and TAM populations within the tumors of ICB-exposed patients may explain the initial response and eventual transition to resistance to ICB agents noted in ccRCC [84]. All 3 scRNA-seq studies also identified and externally validated novel gene signatures that may allow for the detection of specific T cell and TAM populations [81,83,84].

Summary

The genetic determinants of RCC have become more clearly defined, which has led to increased understanding of its evolution and metastatic development, particularly in ccRCC. While increasing data support the role of the TME in determining therapeutic response, the molecular links to immune response are only beginning to be characterized. Future studies of both human tissue and murine models will facilitate further progress in the quest to understand and better manage this disease.

Conflicts of Interest

None declared.

Abbreviations

ccRCCclear cell renal cell carcinoma
FHfumarate hydratase
RCCrenal cell carcinoma
SCNAssomatic copy number alterations
SWI/SNF switching defective/sucrose non-fermenting
TAMstumor-associated macrophages
TMEtumor microenvironment

References

  1. Whiteside, T.L. The tumor microenvironment and its role in promoting tumor growth. Oncogene. 2008, 27, 5904–5912. Available at: https:// www.nature.com/articles/onc2008271. Accessed April 13, 2022. [CrossRef] [PubMed]
  2. Rappold, P.M.; Silagy, A.W.; Kotecha, R.R.; Hakimi, A.A. Immune checkpoint blockade in renal cell carcinoma. J Surg Oncol. 2021, 123, 739–750. Available at: https://onlinelibrary.wiley.com/doi/10.1002/jso.26339. Accessed August 12, 2021. [CrossRef]
  3. Sato, Y.; Yoshizato, T.; Shiraishi, Y.; Maekawa, S.; Okuno, Y.; Kamura, T.; et al. Integrated molecular analysis of clear-cell renal cell carcinoma. Nat Genet. 2013, 45, 860–867. Available at: http://www.nature.com/ articles/ng.2699. Accessed October 7, 2021. [CrossRef] [PubMed]
  4. Cancer Genome Atlas Research Network TCGAR. Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature. 2013, 499, 43–49. Available at: http://www.ncbi.nlm.nih.gov/ pubmed/23792563. Accessed December 24, 2021. [CrossRef] [PubMed]
  5. Mitchell, T.J.; Turajlic, S.; Rowan, A.; Nicol, D.; Farmery, J.H.R.; O'Brien, T.; et al. Timing the landmark events in the evolution of clear cell renal cell cancer: TRACERx Renal. Cell. 2018, 173, 611–623.e17. Available at: https://www.sciencedirect.com/science/article/pii/ S0092867418301648?pes=vor. Accessed March 28, 2022. [CrossRef] [PubMed]
  6. Brugarolas, J. Molecular genetics of clear-cell renal cell carcinoma. J Clin Oncol. 2014, 32, 1968–1976. Available at: http://ascopubs.org/doi/10.1200/JCO.2012.45.2003. Accessed December 24, 2021. [CrossRef]
  7. D’Avella, C.; Abbosh, P.; Pal, S.K.; Geynisman, D.M. Mutations in renal cell carcinoma. Urol Oncol Semin Orig Investig. 2020, 38, 763–773. Available at: https://www.sciencedirect.com/science/article/pii/S1078143918304368. Accessed December 24, 2021. [CrossRef]
  8. Varela, I.; Tarpey, P.; Raine, K.; Huang, D.; Ong, C.K.; Stephens, P.; et al. Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma. Nature. 2011, 469, 539–542. Available at: http://www.nature.com/articles/nature09639. Accessed April 7, 2022. [CrossRef] [PubMed]
  9. Masliah-Planchon, J.; Bièche, I.; Guinebretière, J.-M.; Bourdeaut, F.; Delattre, O. SWI/SNF chromatin remodeling and human malignancies. Annu Rev Pathol. 2015, 10, 145–171. Available at: https://www.annualreviews.org/doi/10.1146/annurev-pathol-012414-040445. Accessed April 3, 2022. [CrossRef]
  10. Wilson, B.G.; Roberts, C.W.M. SWI/SNF nucleosome remodellers and cancer. Nat Rev Cancer. 2011, 11, 481–492. Available at: http://www. nature.com/articles/nrc3068. Accessed April 7, 2022. [CrossRef]
  11. Peña-Llopis, S.; Vega-Rubín-de-Celis, S.; Liao, A.; Leng, N.; Pavía-Jiménez, A.; Wang, S.; et al. BAP1 loss defines a new class of renal cell carcinoma. Nat Genet. 2012, 44, 751–759. Available at: http://www.nature.com/articles/ng.2323. Accessed December 24, 2021. [CrossRef] [PubMed]
  12. Yu, H.; Mashtalir, N.; Daou, S.; Hammond-Martel, I.; Ross, J.; Sui, G.; et al. The ubiquitin carboxyl hydrolase BAP1 forms a ternary complex with YY1 and HCF-1 and is a critical regulator of gene expression. Mol Cell Biol. 2010, 30, 5071–5085. Available at: https://journals.asm.org/doi/10.1128/MCB.00396-10. Accessed April 7, 2022. [CrossRef] [PubMed]
  13. Machida, Y.J.; Machida, Y.; Vashisht, A.A.; Wohlschlegel, J.A.; Dutta, A. The deubiquitinating enzyme BAP1 regulates cell growth via interaction with HCF-1. J Biol Chem. 2009, 284, 34179–34188. Available at: https://linkinghub.elsevier.com/retrieve/pii/S0021925820376997. Accessed April 24, 2022. [CrossRef]
  14. Peña-Llopis, S.; Christie, A.; Xie, X.-J.; Brugarolas, J. Cooperation and antagonism among cancer genes: the renal cancer paradigm. Cancer Res. 2013, 73, 4173–4179. Available at: http://cancerres.aacrjournals.org/lookup/doi/10.1158/0008-5472.CAN-13-0360. Accessed April 24, 2022. [CrossRef]
  15. González-Rodríguez, P.; Engskog-Vlachos, P.; Zhang, H.; Murgoci, A.-N.; Zerdes, I.; Joseph, B. SETD2 mutation in renal clear cell carcinoma suppress autophagy via regulation of ATG12. Cell Death Dis. 2020, 11, 1–15. Available at: https://doi.org/10.1038/s41419-020-2266-x. Accessed April 19, 2022. [CrossRef]
  16. Stephens, P.J.; Greenman, C.D.; Fu, B.; Yang, F.; Bignell, G.R.; Mudie, L.J.; et al. Massive genomic rearrangement acquired in a single catastrophic event during cancer development. Cell. 2011, 144, 27–40. Available at: https://www.sciencedirect.com/science/article/pii/S0092867410013772. Accessed April 7, 2022. [CrossRef] [PubMed]
  17. Turajlic, S.; Larkin, J.; Swanton, C. SnapShot: renal cell carcinoma. Cell. 2015, 163, 1556–1556.e1. Available at: https://www.sciencedirect.com/science/article/pii/S0092867415015457?via%3Dihub. Accessed April 7, 2022. [CrossRef] [PubMed]
  18. Gerlinger, M.; Rowan, A.J.; Horswell, S.; Math, M.; Larkin, J.; Endesfelder, D.; et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med. 2012, 366, 883–892. Available at: http://www.nejm.org/doi/abs/10.1056/NEJMoa1113205. Accessed April 7, 2022. [CrossRef]
  19. Turajlic, S.; Xu, H.; Litchfield, K.; Rowan, A.; Chambers, T.; Lopez, J.I.; et al. Tracking cancer evolution reveals constrained routes to metastases: TRACERx Renal. Cell. 2018, 173, 581–594.e12. [Google Scholar] [CrossRef]
  20. Turajlic, S.; Xu, H.; Litchfield, K.; Rowan, A.; Horswell, S.; Chambers, T.; et al. Deterministic evolutionary trajectories influence primary tumor growth: TRACERx Renal. Cell. 2018, 173, 595–610.e11. Available at: https://www.sciencedirect.com/science/article/pii/S0092867418303751?pes=vor. Accessed March 28, 2022. [CrossRef]
  21. Davis, A.; Gao, R.; Navin, N. Tumor evolution: linear, branching, neutral or punctuated? Biochim Biophys Acta Rev Cancer. 2017, 1867, 151–161. [Google Scholar] [CrossRef] [PubMed]
  22. Mandriota, S.J.; Turner, K.J.; Davies, D.R.; Murray, P.G.; Morgan, N.V.; Sowter, H.M.; et al. HIF activation identifies early lesions in VHL kidneys: evidence for site-specific tumor suppressor function in the nephron. Cancer Cell. 2002, 1, 459–468. Available at: https://www.sciencedirect.com/science/article/pii/ S1535610802000715?via%3Dihub. Accessed April 7, 2022. [CrossRef] [PubMed]
  23. Wang, S.-S.; Gu, Y.-F.; Wolff, N.; Stafanius, K.; Christie, A.; Dey, A.; et al. Bap1 is essential for kidney function and cooperates with Vhl in renal tumorigenesis. Proc Natl Acad Sci. 2014, 111, 16538–16543. Available at: https://pnas.org/doi/full/10.1073/pnas.1414789111. Accessed April 13, 2022. [CrossRef] [PubMed]
  24. Gu, Y.-F.; Cohn, S.; Christie, A.; McKenzie, T.; Wolff, N.; Do, Q.N.; et al. Modeling renal cell carcinoma in mice: Bap1 and Pbrm1 inactivation drive tumor grade. Cancer Discov. 2017, 7, 900–917. Available at: http://cancerdiscovery.aacrjournals.org/lookup/doi/10.1158/2159-8290.CD-17-0292. Accessed April 24, 2022. [CrossRef] [PubMed]
  25. Kapur, P.; Peña-Llopis, S.; Christie, A.; Zhrebker, L.; Pavía-Jiménez, A.; Rathmell, W.K.; et al. Effects on survival of BAP1 and PBRM1 mutations in sporadic clear-cell renal-cell carcinoma: a retrospective analysis with independent validation. Lancet Oncol. 2013, 14, 159–167. Available at: https://www.sciencedirect. com/science/article/pii/S1470204512705843?via%3Dihub. Accessed December 23, 2021. [CrossRef] [PubMed]
  26. Singla, N.; Xie, Z.; Zhang, Z.; Gao, M.; Yousuf, Q.; Onabolu, O.; et al. Pancreatic tropism of metastatic renal cell carcinoma. JCI Insight. 2020, 5, e134564. Available at: https://insight.jci.org/articles/view/134564. Accessed April 24, 2022. [CrossRef]
  27. Ho, T.H.; Kapur, P.; Joseph, R.W.; Serie, D.J.; Eckel-Passow, J.E.; Tong, P.; et al. Loss of histone H3 lysine 36 trimethylation is associated with an increased risk of renal cell carcinoma-specific death. Mod Pathol. 2016, 29, 34–42. Available at: http://www.nature.com/articles/modpathol2015123. Accessed April 24, 2022. [CrossRef] [PubMed]
  28. Delahunt, B.; Eble, J.N. Papillary renal cell carcinoma: a clinicopathologic and immunohistochemical study of 105 tumors. Mod Pathol. 1997, 10, 537–544. Available at: http://www.ncbi.nlm.nih.gov/ pubmed/9195569. Accessed October 2, 2022. [PubMed]
  29. Jiang, F.; Richter, J.; Schraml, P.; Bubendorf, L.; Gasser, T.; Sauter, G.; et al. Chromosomal imbalances in papillary renal cell carcinoma: genetic differences between histological subtypes. Am J Pathol. 1998, 153, 1467–1473. [Google Scholar] [CrossRef]
  30. Foster, K.; Crossey, P.; Cairns, P.; Hetherington, J.W.; Richards, F.M.; Jones, M.H.; et al. Molecular genetic investigation of sporadic renal cell carcinoma: analysis of allele loss on chromosomes 3p, 5q, 11p, 17 and 22. Br J Cancer. 1994, 69, 230–234. Available at: http://www.nature.com/articles/bjc199444. Accessed April 3, 2022. [CrossRef]
  31. Durinck, S.; Stawiski, E.W.; Pavía-Jiménez, A.; Modrusan, Z.; Kapur, P.; Jaiswal, B.S.; et al. Spectrum of diverse genomic alterations define non-clear cell renal carcinoma subtypes. Nat Genet. 2015, 47, 13–21. [Google Scholar] [CrossRef] [PubMed]
  32. The Cancer Genome Atlas Research Network. Comprehensive molecular characterization of papillary renal-cell carcinoma. N Engl J Med. 2016, 374, 135–145. Available at: http://www.nejm.org/doi/10.1056/NEJMoa1505917. Accessed April 3, 2022.
  33. Ricketts, C.J.; De Cubas, A.A.; Fan, H.; Smith, C.C.; Lang, M.; Reznik, E.; et al. The cancer genome atlas comprehensive molecular characterization of renal cell carcinoma. Cell Rep. 2018, 23, 313–326.e5. Available at: https://www.sciencedirect.com/ science/article/pii/S2211124718304364?via%3Dihub. Accessed April 3, 2022. [CrossRef] [PubMed]
  34. Schmidt, L.; Duh, F.-M.; Chen, F.; Kishida, T.; Glenn, G.; Choyke, P.; et al. Germline and somatic mutations in the tyrosine kinase domain of the MET proto- oncogene in papillary renal carcinomas. Nat Genet. 1997, 16, 68–73. Available at: http://www.nature.com/articles/ng0597-68. Accessed April 3, 2022. [CrossRef] [PubMed]
  35. Chen, Y.-B.; Xu, J.; Skanderup, A.J.; Dong, Y.; Brannon, A.R.; Wang, L.; et al. Molecular analysis of aggressive renal cell carcinoma with unclassified histology reveals distinct subsets. Nat Commun. 2016, 7, 13131. Available at: http://www.nature.com/articles/ncomms13131. Accessed April 12, 2022. [CrossRef] [PubMed]
  36. Moch, H.; Amin, M.B.; Berney, D.M.; Compérat, E.M.; Gill, A.J.; Hartmann, A.; et al. The 2022 World Health Organization Classification of Tumours of the Urinary System and Male Genital Organs—Part A: Renal, Penile, and Testicular Tumours. Eur Urol. 2022. Available at: https://www.sciencedirect.com/science/article/pii/S0302283822024678#b0220. Accessed October 2, 2022. [CrossRef] [PubMed]
  37. Al-Obaidy, K.I.; Eble, J.N.; Cheng, L.; Williamson, S.R.; Sakr, W.A.; Gupta, N.; et al. Papillary renal neoplasm with reverse polarity. Am J Surg Pathol. 2019, 43, 1099–1111. Available at: https://journals.lww.com/00000478-201908000-00011. Accessed October 2, 2022. [CrossRef] [PubMed]
  38. Argani, P.; Reuter, V.E.; Eble, J.N.; Vlatkovic, L.; Yaskiv, O.; Swanson, D.; et al. Biphasic hyalinizing psammomatous renal cell carcinoma (BHP RCC): a distinctive neoplasm associated with somatic NF2 mutations. Am J Surg Pathol. 2020, 44, 901–916. Available at: http://www.ncbi.nlm.nih.gov/pubmed/32217839. Accessed October 2, 2022. [CrossRef] [PubMed]
  39. Brunelli, M.; Eble, J.N.; Zhang, S.; Martignoni, G.; Delahunt, B.; Cheng, L. Eosinophilic and classic chromophobe renal cell carcinomas have similar frequent losses of multiple chromosomes from among chromosomes 1, 2, 6, 10, and 17, and this pattern of genetic abnormality is not present in renal oncocytoma. Mod Pathol. 2005, 18, 161–169. Available at: http://www.nature.com/articles/3800286. Accessed April 3, 2022. [CrossRef]
  40. Speicher, M.R.; Schoell, B.; du Manoir, S.; Schröck, E.; Ried, T.; Cremer, T.; et al. Specific loss of chromosomes 1, 2, 6, 10, 13, 17, and 21 in chromophobe renal cell carcinomas revealed by comparative genomic hybridization. Am J Pathol. 1994, 145, 356–364. [Google Scholar]
  41. Ball, M.W.; Gorin, M.A.; Drake, C.G.; Hammers, H.J.; Allaf, M.E. The landscape of whole-genome alterations and pathologic features in genitourinary malignancies: an analysis of the cancer genome atlas. Eur Urol Focus. 2017, 3, 584–589. [Google Scholar] [CrossRef] [PubMed]
  42. Casuscelli, J.; Weinhold, N.; Gundem, G.; Wang, L.; Zabor, E.C.; Drill, E.; et al. Genomic landscape and evolution of metastatic chromophobe renal cell carcinoma. JCI Insight. 2017, 2, e92688. Available at: https://insight.jci.org/articles/view/92688. Accessed April 12, 2022. [CrossRef] [PubMed]
  43. Iacovelli, R.; Modica, D.; Palazzo, A.; Trenta, P.; Piesco, G.; Cortesi, E. Clinical outcome and prognostic factors in renal medullary carcinoma: a pooled analysis from 18 years of medical literature. Can Urol Assoc J. 2015, 9, E172–E177. [Google Scholar] [CrossRef] [PubMed]
  44. Hakimi, A.A.; Koi, P.T.; Milhoua, P.M.; Blitman, N.M.; Li, M.; Hugec, V.; et al. Renal medullary carcinoma: the Bronx experience. Urology. 2007, 70, 878–882. Available at: https://www.sciencedirect.com/science/article/pii/S0090429507018092?via%3Dihub. Accessed April 3, 2022. [CrossRef] [PubMed]
  45. Ezekian, B.; Englum, B.; Gilmore, B.F.; Nag, U.P.; Kim, J.; Leraas, H.J.; et al. Renal medullary carcinoma: a national analysis of 159 patients. Pediatr Blood Cancer. 2017, 64, e26609. Available at: https://onlinelibrary.wiley.com/doi/10.1002/pbc.26609. Accessed April 3, 2022. [CrossRef] [PubMed]
  46. Davis, C.J.; Mostofi, F.K.; Sesterhenn, I.A. Renal medullary carcinoma. The seventh sickle cell nephropathy. Am J Surg Pathol. 1995, 19, 1–11. [Google Scholar] [CrossRef] [PubMed]
  47. Maroja Silvino, M.C.; Venchiarutti Moniz, C.M.; Munhoz Piotto, G.H.; Siqueira, S.; Galapo Kann, A.; Dzik, C. Renal medullary carcinoma response to chemotherapy: a referral center experience in Brazil. Rare Tumors. 2013, 5, e44. Available at: http://journals.sagepub.com/doi/10.4081/rt.2013.e44. Accessed April 3, 2022. [CrossRef] [PubMed]
  48. Marsh, A.; Golden, C.; Hoppe, C.; Quirolo, K.; Vichinsky, E. Renal medullary carcinoma in an adolescent with sickle cell anemia. Pediatr Blood Cancer. 2014, 61, 567–567. Available at: https://onlinelibrary.wiley.com/doi/10.1002/pbc.24795. Accessed April 3, 2022. [CrossRef] [PubMed]
  49. Berman, L.B. Sickle cell nephropathy. JAMA. 1974, 228, 1279. Available at: http://jama.jamanetwork.com/article.aspx?doi=10.1001/jama.1974.03230350051035. Accessed April 3, 2022. [CrossRef]
  50. Msaouel, P.; Malouf, G.G.; Su, X.; Yao, H.; Tripathi, D.N.; Soeung, M.; et al. Comprehensive molecular characterization identifies distinct genomic and immune hallmarks of renal medullary carcinoma. Cancer Cell. 2020, 37, 720–734.e13. [Google Scholar] [CrossRef]
  51. Kadoch, C.; Copeland, R.A.; Keilhack, H. PRC2 and SWI/SNF chromatin remodeling complexes in health and disease. Biochemistry. 2016, 55, 1600–1614. Available at: https://pubs.acs.org/doi/10.1021/acs.biochem.5b01191. Accessed April 3, 2022. [CrossRef] [PubMed]
  52. Launonen, V.; Vierimaa, O.; Kiuru, M.; Isola, J.; Roth, S.; Pukkala, E.; Sistonen, P.; et al. Inherited susceptibility to uterine leiomyomas and renal cell cancer. Proc Natl Acad Sci U S A. 2001, 98, 3387–3392. [Google Scholar] [CrossRef] [PubMed]
  53. Merino, M.J.; Torres-Cabala, C.; Pinto, P.; Linehan, W.M. The morphologic spectrum of kidney tumors in hereditary leiomyomatosis and renal cell carcinoma (HLRCC) syndrome. Am J Surg Pathol. 2007, 31, 1578–1585. [Google Scholar] [CrossRef] [PubMed]
  54. Grubb, R.L.; Franks, M.E.; Toro, J.; Middleton, L.; Choyke, L.; Fowler, S.; et al. Hereditary leiomyomatosis and renal cell cancer: a syndrome associated with an aggressive form of inherited renal cancer. J Urol. 2007, 177, 2074–2079; discussion 2079–2080. [Google Scholar] [CrossRef] [PubMed]
  55. Schmidt, L.S.; Linehan, W.M. Hereditary leiomyomatosis and renal cell carcinoma. Int J Nephrol Renovasc Dis. 2014, 7, 253–260. [Google Scholar] [CrossRef] [PubMed]
  56. Toro, J.R.; Nickerson, M.L.; Wei, M.-H.; Warren, M.B.; Glenn, G.M.; Turner, M.L.; et al. Mutations in the fumarate hydratase gene cause hereditary leiomyomatosis and renal cell cancer in families in North America. Am J Hum Genet. 2003, 73, 95–106. [Google Scholar] [CrossRef] [PubMed]
  57. Wei, M.-H.; Toure, O.; Glenn, G.M.; Pithukpakorn, M.; Neckers, L.; Stolle, C.; et al. Novel mutations in FH and expansion of the spectrum of phenotypes expressed in families with hereditary leiomyomatosis and renal cell cancer. J Med Genet. 2006, 43, 18–27. [Google Scholar] [CrossRef] [PubMed]
  58. Multiple-Leiomyoma-Consortium. Germline mutations in FH predispose to dominantly inherited uterine fibroids, skin leiomyomata and papillary renal cell cancer. Nat Genet. 2002, 30, 406–410. Available at: http://www.nature.com/articles/ng849z. Accessed April 3, 2022. [CrossRef]
  59. Baysal, B.E.; Ferrell, R.E.; Willett-Brozick, J.E.; Lawrence, E.C.; Myssiorek, D.; Bosch, A.; et al. Mutations in SDHD, a mitochondrial complex II gene, in hereditary paraganglioma. Science. 2000, 287, 848–851. Available at: https://www.science.org/doi/10.1126/science.287.5454.848. Accessed April 3, 2022. [CrossRef]
  60. Niemann, S.; Müller, U. Mutations in SDHC cause autosomal dominant paraganglioma, type 3. Nat Genet. 2000, 26, 268–270. Available at: http://www.nature.com/articles/ng1100_268. Accessed April 3, 2022. [CrossRef]
  61. Astuti, D.; Latif, F.; Dallol, A.; Dahia, P.L.; Douglas, F.; George, E.; et al. Gene mutations in the succinate dehydrogenase subunit SDHB cause susceptibility to familial pheochromocytoma and to familial paraganglioma. Am J Hum Genet. 2001, 69, 49–54. [Google Scholar] [CrossRef] [PubMed]
  62. Tong, W.-H.; Sourbier, C.; Kovtunovych, G.; Jeong, S.Y.; Vira, M.; Ghosh, M.; et al. The glycolytic shift in fumarate-hydratase-deficient kidney cancer lowers AMPK levels, increases anabolic propensities and lowers cellular iron levels. Cancer Cell. 2011, 20, 315–327. Available at: https://www.sciencedirect.com/science/article/pii/S1535610811002686?via%3Dihub. Accessed April 3, 2022. [CrossRef] [PubMed]
  63. Yang, Y.; Lane, A.N.; Ricketts, C.J.; Sourbier, C.; Wei, M.-H.; Shuch, B.; et al. Metabolic reprogramming for producing energy and reducing power in fumarate hydratase null cells from hereditary leiomyomatosis renal cell carcinoma. PLoS One. 2013, 8, e72179. Available at: https://dx.plos.org/10.1371/journal.pone.0072179. Accessed April 3, 2022. [CrossRef] [PubMed]
  64. Pollard, P.J.; Brière, J.J.; Alam, N.A.; Barwell, J.; Barclay, E.; Wortham, M.C.; et al. Accumulation of Krebs cycle intermediates and over- expression of HIF1α in tumours which result from germline FH and SDH mutations. Hum Mol Genet. 2005, 14, 2231–2239. Available at: http://academic.oup.com/hmg/article/14/15/2231/551734/Accumulation-of-Krebs-cycle- intermediates-and. Accessed April 3, 2022. [CrossRef] [PubMed]
  65. Saxena, N.; Maio, N.; Crooks, D.R.; Ricketts, C.J.; Yang, Y.; Wei, M.-H.; et al. SDHB-deficient cancers: the role of mutations that impair iron sulfur cluster delivery. J Natl Cancer Inst. 2016, 108, djv287. [Google Scholar] [CrossRef] [PubMed]
  66. Xiao, M.; Yang, H.; Xu, W.; Ma, S.; Lin, H.; Zhu, H.; et al. Inhibition of α-KG- dependent histone and DNA demethylases by fumarate and succinate that are accumulated in mutations of FH and SDH tumor suppressors. Genes Dev. 2012, 26, 1326–1338. [Google Scholar] [CrossRef] [PubMed]
  67. Isaacs, J.S.; Jung, Y.J.; Mole, D.R.; Lee, S.; Torres-Cabala, C.; Chung, Y.-L.; et al. HIF overexpression correlates with biallelic loss of fumarate hydratase in renal cancer: novel role of fumarate in regulation of HIF stability. Cancer Cell. 2005, 8, 143–153. [Google Scholar] [CrossRef] [PubMed]
  68. Bardella, C.; El-Bahrawy, M.; Frizzell, N.; Adam, J.; Ternette, N.; Hatipoglu, E.; et al. Aberrant succination of proteins in fumarate hydratase-deficient mice and HLRCC patients is a robust biomarker of mutation status. J Pathol. 2011, 225, 4–11. Available at: https:// onlinelibrary.wiley.com/doi/10.1002/path.2932. Accessed April 3, 2022. [CrossRef] [PubMed]
  69. Adam, J.; Hatipoglu, E.; O’Flaherty, L.; Ternette, N.; Sahgal, N.; Lockstone, H.; et al. Renal cyst formation in Fh1-deficient mice is independent of the Hif/Phd pathway: roles for fumarate in KEAP1 succination and Nrf2 signaling. Cancer Cell. 2011, 20, 524–537. Available at: https://www.sciencedirect.com/science/article/pii/S1535610811003540?via%3Dihub. Accessed April 3, 2022. [CrossRef]
  70. Yoo, A.; Tang, C.; Zucker, M.; Fitzgerald, K.; DiNatale, R.G.; Rappold, P.M.; et al. Genomic and metabolic hallmarks of SDH- and FH-deficient renal cell carcinomas. Eur Urol Focus. 2022. Online ahead of print. Available at: https://www.eu-focus.europeanurology.com/article/S2405-4569(21)00312-6/fulltext. Accessed April 12, 2022. [CrossRef]
  71. Argani, P. MiT family translocation renal cell carcinoma. Semin Diagn Pathol. 2015, 32, 103–113. Available at: https://www.sciencedirect.com/science/article/pii/S0740257015000040?via%3Dihub. Accessed April 3, 2022. [CrossRef]
  72. Kauffman, E.C.; Ricketts, C.J.; Rais-Bahrami, S.; Yang, Y.; Merino, M.J.; Bottaro, D.P.; et al. Molecular genetics and cellular features of TFE3 and TFEB fusion kidney cancers. Nat Rev Urol. 2014, 11, 465–475. Available at: http://www.nature.com/articles/nrurol.2014.162. Accessed April 3, 2022. [CrossRef]
  73. La Spina, M.; Contreras, P.S.; Rissone, A.; Meena, N.K.; Jeong, E.; Martina, J.A. MiT/TFE family of transcription factors: an evolutionary perspective. Front Cell Dev Biol. 2021, 8, 1580. Available at: https://www.frontiersin.org/articles/10.3389/fcell.2020.609683/full. Accessed April 3, 2022. [CrossRef]
  74. Chen, F.; Zhang, Y.; Şenbabaoğlu, Y.; Ciriello, G.; Yang, L.; Reznik, E.; et al. Multilevel genomics-based taxonomy of renal cell carcinoma. Cell Rep. 2016, 14, 2476–2489. [Google Scholar] [CrossRef]
  75. Argani, P.; Reuter, V.E.; Zhang, L.; Sung, Y.S.; Ning, Y.; Epstein, J.I.; et al. TFEB-amplified renal cell carcinomas: an aggressive molecular subset demonstrating variable melanocytic marker expression and morphologic heterogeneity. Am J Surg Pathol. 2016, 40, 1484–1495. [Google Scholar] [CrossRef]
  76. Xia, Q.-Y.; Wang, X.-T.; Ye, S.-B.; Wang, X.; Li, R.; Shi, S.-S.; et al. Novel gene fusion of PRCC-MITF defines a new member of MiT family translocation renal cell carcinoma: clinicopathological analysis and detection of the gene fusion by RNA sequencing and FISH. Histopathology. 2018, 72, 786–794. Available at: https://onlinelibrary.wiley.com/doi/10.1111/his.13439. Accessed April 3, 2022. [CrossRef]
  77. Şenbabaoğlu, Y.; Gejman, R.S.; Winer, A.G.; Liu, M.; Van Allen, E.M.; de Valasco, G.; et al. Tumor immune microenvironment characterization in clear cell renal cell carcinoma identifies prognostic and immunotherapeutically relevant messenger RNA signatures. Genome Biol. 2016, 17, 231. Available at: https://genomebiology.biomedcentral.com/articles/10.1186/s13059-016-1092-z. Accessed August 12, 2021. [CrossRef] [PubMed]
  78. Wang, T.; Lu, R.; Kapur, P.; Jaiswal, B.S.; Hannan, R.; Zhang, Z.; 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. 2018, 8, 1142–1155. Available at: http://cancerdiscovery.aacrjournals.org/lookup/doi/10.1158/2159-8290.CD-17-1246. Accessed April 24, 2022. [CrossRef] [PubMed]
  79. Wherry, E.J. T cell exhaustion. Nat Immunol. 2011, 12, 492–499. Available at: http://www.nature.com/articles/ni.2035. Accessed April 13, 2022. [CrossRef] [PubMed]
  80. Speiser, D.E.; Ho, P.-C.; Verdeil, G. Regulatory circuits of T cell function in cancer. Nat Rev Immunol. 2016, 16, 599–611. Available at: http://www. nature.com/articles/nri.2016.80. Accessed April 13, 2022. [CrossRef]
  81. Krishna, C.; DiNatale, R.G.; Kuo, F.; Srivastava, R.M.; Vuong, L.; Chowell, D.; et al. Single-cell sequencing links multiregional immune landscapes and tissue-resident T cells in ccRCC to tumor topology and therapy efficacy. Cancer Cell. 2021, 39, 662–677. Available at: https://www.sciencedirect.com/science/article/pii/S1535610821001653?via%3Dihub. Accessed August 12, 2021. [CrossRef]
  82. Vuong, L.; Kotecha, R.R.; Voss, M.H.; Hakimi, A.A. Tumor microenvironment dynamics in clear- cell renal cell carcinoma. Cancer Discov. 2019, 9, 1349–1357. Available at: www.aacrjournals.org. Accessed September 14, 2021. [CrossRef]
  83. Braun, D.A.; Street, K.; Burke, K.P.; Cookmeyer, D.L.; eDenize, T.; Pederson, C.B.; et al. Progressive immune dysfunction with advancing disease stage in renal cell carcinoma. Cancer Cell. 2021, 39, 632–648.e8. Available at: https://www.sciencedirect.com/science/article/pii/S153561082100115X. Accessed April 12, 2022. [CrossRef] [PubMed]
  84. Bi, K.; He, M.X.; Bakouny, Z.; Kanodia, A.; Napolitano, S.; Wu, J.; et al. Tumor and immune reprogramming during immunotherapy in advanced renal cell carcinoma. Cancer Cell. 2021, 39, 649–661.e5. Available at: https://www.sciencedirect.com/science/article/pii/ S1535610821001173?via%3Dihub. Accessed April 12, 2022. [CrossRef]
  85. Chakiryan, N.H.; Kimmel, G.J.; Kim, Y.; Hajiran, A.; Aydin, A.M.; Zemp, L.; et al. Spatial clustering of CD68+ tumor associated macrophages with tumor cells is associated with worse overall survival in metastatic clear cell renal cell carcinoma. PLoS One. 2021, 16, e0245415. Available at: https://dx.plos.org/10.1371/journal.pone.0245415. Accessed April 12, 2022. [CrossRef]
  86. Chevrier, S.; Levine, J.H.; Zanotelli, V.R.T.; Silina, K.; Schulz, D.; Bacac, M.; et al. An immune atlas of clear cell renal cell carcinoma. Cell. 2017, 169, 736–749. Available at: https://www.sciencedirect.com/science/article/pii/S0092867417304294. Accessed April 12, 2022. [CrossRef]
  87. Bindea, G.; Mlecnik, B.; Tosolini, M.; Kirilovsky, A.; Wladner, M.; Obenauf, A.C.; et al. Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity. 2013, 39, 782–795. Available at: https://www.sciencedirect.com/science/article/pii/S1074761313004378?via%3Dihub. Accessed August 12, 2021. [CrossRef]
  88. Motzer, R.J.; Robbins, P.B.; Powles, T.; Albiges, L.; Haanen, J.B.; Larkin, J.; et al. Avelumab plus axitinib versus sunitinib in advanced renal cell carcinoma: biomarker analysis of the phase 3 JAVELIN Renal 101 trial. Nat Med. 2020, 26, 1733–1741. [Google Scholar] [CrossRef]
  89. McDermott, D.F.; Huseni, M.A.; Atkins, M.B.; Motzer, R.J.; Rini, B.I.; Escudier, B.; 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. 2018, 24, 749–757. Available at: http://www.nature.com/articles/s41591-018-0053-3. Accessed September 12, 2021. [CrossRef]
  90. Braun, D.A.; Hou, Y.; Bakouny, Z.; Ficial, M.; Sant' Angleo, M.; Forman, J.; et al. Interplay of somatic alterations and immune infiltration modulates response to PD-1 blockade in advanced clear cell renal cell carcinoma. Nat Med. 2020, 26, 909–918. Available at: http://www.nature.com/articles/s41591-020-0839-y. Accessed December 16, 2021. [CrossRef]
  91. Jansen, C.S.; Prokhnevska, N.; Master, V.A.; Sanda, M.G.; Carlisle, J.W.; Bilen, M.A.; et al. An intra-tumoral niche maintains and differentiates stem-like CD8 T cells. Nature. 2019, 576, 465–470. Available at: http://www.nature.com/articles/s41586-019-1836-5. Accessed April 12, 2022. [CrossRef] [PubMed]
  92. Hakimi, A.A.; Voss, M.H.; Kuo, F.; Sanchez, A.; Liu, M.; Nixon, B.G.; et al. Transcriptomic profiling of the tumor microenvironment reveals distinct subgroups of clear cell renal cell cancer: data from a randomized phase III trial. Cancer Discov. 2019, 9, 510–525. Available at: http://cancerdiscovery.aacrjournals.org/lookup/doi/10.1158/2159-8290.CD-18-0957. Accessed August 12, 2021. [CrossRef] [PubMed]
  93. Motzer, R.J.; Choueiri, T.K.; McDermott, D.F.; Powles, T.; Yao, J.; Ammar, R.; et al. Biomarker analyses from the phase III CheckMate 214 trial of nivolumab plus ipilimumab (N+I) or sunitinib (S) in advanced renal cell carcinoma (aRCC). J Clin Oncol. 2020, 38, 5009–5009. Available at: https://ascopubs.org/doi/10.1200/JCO.2020.38.15_suppl.5009. Accessed December 21, 2021. [CrossRef]
  94. Rini, B.I.; Powles, T.; Atkins, M.B.; Escudier, B.; McDermott, D.F.; Suarez, C.; et al. Atezolizumab plus bevacizumab versus sunitinib in patients with previously untreated metastatic renal cell carcinoma (IMmotion151): a multicentre, open-label, phase 3, randomised controlled trial. Lancet. 2019, 393, 2404–2415. Available at: https://www.sciencedirect.com/science/article/pii/ S0140673619307238?via%3Dihub. Accessed December 17, 2021. [CrossRef] [PubMed]
  95. Beuselinck, B.; Job, S.; Becht, E.; Karadimou, A.; Verkarre, V.; Couchy, G.; et al. Molecular subtypes of clear cell renal cell carcinoma are associated with sunitinib response in the metastatic setting. Clin Cancer Res. 2015, 21, 1329–1339. Available at: http://www.ncbi.nlm.nih.gov/pubmed/25583177. Accessed January 17, 2022. [CrossRef] [PubMed]
  96. Verbiest, A.; Couchy, G.; Job, S.; Caruana, L.; Lerut, E.; Oyen, R.; et al. Molecular subtypes of clear-cell renal cell carcinoma are prognostic for outcome after complete metastasectomy. Eur Urol. 2018, 74, 474–480. Available at: https://www.sciencedirect.com/science/article/pii/S0302283818300976. Accessed October 2, 2022. [CrossRef]
  97. Beuselinck, B.; Verbiest, A.; Couchy, G.; Job, S.; de Reynies, A.; Meiller, C.; et al. Pro-angiogenic gene expression is associated with better outcome on sunitinib in metastatic clear-cell renal cell carcinoma. Acta Oncol. 2018, 57, 498–508. Available at: https://www.tandfonline.com/doi/full/10.1080/0284186X.2017.1388927. Accessed January 17, 2022. [CrossRef]
  98. Vano, Y.-A.; Elaidi, R.; Bennamoun, M.; Chevreau, C.; Borchiellini, D.; Pannier, D.; et al. Nivolumab, nivolumab–ipilimumab, and VEGFR- tyrosine kinase inhibitors as first-line treatment for metastatic clear-cell renal cell carcinoma (BIONIKK): a biomarker-driven, open-label, non-comparative, randomised, phase 2 trial. Lancet Oncol. 2022, 23, 612–624. Available at: https://www.sciencedirect.com/science/article/pii/S1470204522001280#bib6. Accessed October 2, 2022. [CrossRef]
  99. Koh, M.Y.; Sayegh, N.; Agarwal, N. Seeing the forest for the trees—single- cell atlases link CD8+ T cells and macrophages to disease progression and treatment response in kidney cancer. Cancer Cell. 2021, 39, 594–596. Available at: https://www.sciencedirect.com/science/article/pii/S1535610821001665?via%3Dihub. Accessed April 12, 2022. [CrossRef]
Table 1. Summary of most common genetic alterations of reviewed RCC subtypes.
Table 1. Summary of most common genetic alterations of reviewed RCC subtypes.
Siuj 03 00386 i001

Share and Cite

MDPI and ACS Style

Khaleel, S.; Ricketts, C.; Linehan, W.M.; Ball, M.; Manley, B.; Turajilic, S.; Brugarolas, J.; Hakimi, A. 2022 WUOF/SIU International Consultation on Urological Diseases: Genetics and Tumor Microenvironment of Renal Cell Carcinoma. Soc. Int. Urol. J. 2022, 3, 386-396. https://doi.org/10.48083/BLPV3411

AMA Style

Khaleel S, Ricketts C, Linehan WM, Ball M, Manley B, Turajilic S, Brugarolas J, Hakimi A. 2022 WUOF/SIU International Consultation on Urological Diseases: Genetics and Tumor Microenvironment of Renal Cell Carcinoma. Société Internationale d’Urologie Journal. 2022; 3(6):386-396. https://doi.org/10.48083/BLPV3411

Chicago/Turabian Style

Khaleel, Sari, Christopher Ricketts, W. Marston Linehan, Mark Ball, Brandon Manley, Samra Turajilic, James Brugarolas, and Ari Hakimi. 2022. "2022 WUOF/SIU International Consultation on Urological Diseases: Genetics and Tumor Microenvironment of Renal Cell Carcinoma" Société Internationale d’Urologie Journal 3, no. 6: 386-396. https://doi.org/10.48083/BLPV3411

Article Metrics

Back to TopTop