Abstract
Purpose of review:
As molecular profiling of renal cell carcinoma (RCC) continues to elucidate novel targets for non-clear cell histologies, understanding the landscape of these targets is of utmost importance. In this review, we highlight the genomic landscape of non-clear cell RCC and its implications for current and future systemic therapies.
Recent findings:
Several genomic studies have described the mutational burden among non-clear cell histologies. These studies have highlighted the importance of MET in papillary RCC and led to several clinical trials evaluating the efficacy of MET inhibitors for papillary RCC. The success of immune checkpoint inhibitors, such as ipilimumab and nivolumab, in clear cell RCC has led to ongoing trials evaluating these novel therapeutics in non-clear cell RCC.
Summary:
Genomic profiling has allowed for the evaluation of novel targets for non-clear cell RCC. This evolving therapeutic landscape is being explored in promising, ongoing trials that have the potential for changing how non-clear cell RCC is managed.
Keywords: Non-clear cell renal cell carcinoma, RCC, genomics, papillary, chromophobe
Introduction
Approximately 76,000 new cases and 14,000 deaths due to kidney cancer are expected in 2021 in the United states alone [1]. Ninety percent of these cases are due to renal cell carcinoma (RCC), which arises from the nephron and contains many histologic subtypes. Most patients with RCC (75–80%) have clear cell histology (ccRCC), while other common histologies are collectively referred to as non-clear cell RCC (nccRCC; Table 1). Treatment of RCC presents a unique challenge as about 30% of patients initially present with metastatic RCC and an additional one-third of RCC patients have recurrence with distant metastases after surgical resection [2,3]. The recent successes of targeted and immunotherapies have shifted the paradigm in the management of metastatic ccRCC [4]. However, therapeutic advances in nccRCC have been limited by the lower incidence and limited clinical trial successes. In this review, we will highlight the mutational landscape of nccRCC based on genomic studies and recent treatment advances for specific subtypes of nccRCC based on clinical trial evidence.
Table 1:
Type | Incidence | Chromosomal mutation | Mutations |
---|---|---|---|
Papillary | 10–15% |
Type I Gain of Ch 7, 16, 17 Loss of 1p36 Type II Loss of Ch 9p21, 3p, 14p, 22q Gain of 5q, 7–16, 17 |
Type I MET Type II CDKN2A, SETD2, BAP1, PBRM1, NRF2-ARE, TFE3 fusions |
Chromophobe | 4–5% | Loss of Ch 1, 2, 6, 10, 13, 17, 21 | TP53, PTEN, mTOR, TERT, FAAH2, PDHB, PDXDC1, ZNF765 |
Collecting duct | <1% | Loss of 8p, 16p, 1p, 9p Gain at 13q |
mtDNA genome mutations |
Translocation | Xp11.23, 6p21 | BIRC7 expression | |
Renal medullary carcinoma | SMARCB1 | ||
Unclassified | NF2, SETD2, BAP1, KMT2C, MTOR, TSC1, TSC2, PTEN | ||
Sarcomatoid† | 5% | TP53, VHL, CDKN2A, NF2 |
can coexist with any RCC histology
Ch: chromosome; mtDNA: mitochondrial DNA
The Genomic Landscape of nccRCC
Over the last three decades the mutational landscape within ccRCC tumors has been well characterized through multiple large-scale genomic studies [5–8]. While ccRCC has a wide variety of mutations, the loss of chromosome 3p is reported in ~90% of sporadic ccRCC, with inactivating mutations and loss of heterozygosity in the VHL gene seen in >90% of ccRCC tumors [5,9,10]. Studying the consequences of VHL alterations has helped uncover aberrant degradation of hypoxia-inducible factor 1 and 2 alpha (HIF1-2α), which in turn promotes pathologic cell proliferation and angiogenesis via upregulation of vascular endothelial growth factor, platelet-derived growth factor, and transforming growth factor-α, which promote tumorigenesis. Discovery of these pathways has helped uncover a myriad of novel therapeutics for treating advanced and/or metastatic ccRCC [4,11].
Papillary RCC (pRCC) type I and II and chromophobe RCC (chRCC) represent 10–15% and 4–5%, respectively, of all RCC. Other nccRCC histologies such as medullary, translocation, collecting duct, and unclassified types, while rare, also constitute nccRCC histologies. Similar to ccRCC, specific chromosomal rearrangements and mutational profiles have been described for various histologies of nccRCC (Table 1) and many are associated with hereditary RCC syndromes (Table 2) [12–17]. While detection of these mutations has been important for classification and diagnosis, it has been difficult to translate these discoveries for therapeutic purposes.
Table 2:
Syndrome | Gene | Inheritance | RCC Histology | Clinical Features |
---|---|---|---|---|
Von Hippel-Lindau | VHL | AD | Clear cell | Retinal angiomas, bilateral multifocal RCC, Brain, spine, or retinal hemangioblastomas |
Hereditary papillary renal carcinoma | MET | AD | Type I papillary | Bilateral multifocal RCC |
Birt-Hogg-Dubé | FLCN | AD | Chromophobe | Cutaneous fibrofolliculoma Pulmonary cysts Spontaneous pneumothorax |
Tuberous sclerosis complex |
TSC1 TSC2 |
AD | Angiomyolipoma Clear cell Cystic oncocytoma |
Angiofibromas Shagreen patches Retinal nodular hamartomas Lymphangioleiomyomatosis Multiple renal cysts |
Hereditary leiomyomatosis and RCC | FH | AD | Type II papillary | Leiomyomas of uterus and skin Adrenal adenoma Aggressive renal tumors |
BAP1 tumor predisposition syndrome | BAP1 | AD | Clear cell Chromophobe |
Melanoma RCC Mesothelioma |
Hereditary paraganglioma-pheochromocytoma syndrome |
SDHA SDHB SDHC SDHD |
AD | Clear cell Chromophobe Type II papillary Oncocytoma |
Head and neck paraganglioma Adrenal or extra-adrenal pheochromocytoma GIST tumors Lung lesions |
Cowden syndrome | PTEN | AD | Papillary Chromophobe Clear cell |
Mucocutaneous lesions Breast cancer Follicular thyroid cancer Endometrial cancer Others |
AD: autosomal dominant; RCC: renal cell carcinoma
Papillary RCC
pRCC is the second most common RCC histology and is subdivided based on morphologic features into types I and II [18]. The initial work on characterizing pRCC identified mutations in the MET proto-oncogene among patients with the hereditary (HPRC) and sporadic type I pRCC and mutations in FH in type II pRCC [19,20]. Following this, evaluation of 220 samples of sporadic pRCC by Albiges et al. found that MET expression was significantly elevated across all types of pRCC and that copy number alterations were present in 81% and 46% of type I and II pRCC, respectively [21]. Additional characterization of nccRCC demonstrated that MET, NF2, SLC5A3, PNKD, and CPQ were mutated in pRCC [13]. Approximately 15% of pRCC tumors in this study contained mutations in MET. The Cancer Genome Atlas subsequently performed the largest multidimensional genomic mapping study with whole exome sequencing, copy-number analysis, mRNA and miRNA sequencing, methylation analysis, and proteomic analysis on 161 pRCCs, which was expanded to include 274 pRCCs from among a larger pool of 843 RCCs representing a comprehensive study of all the major RCC subtypes [9]. This study confirmed differences in types I and II pRCC and identified 4 distinct molecular groupings with progressively worse survival (C1, C2a, C2b, and C2c) [12]. C1 comprised of predominantly type I pRCC tumors, which had a gain of chromosome 7 and 17. Among type I pRCC tumors, somatic mutations in MET were present in 17% of these tumors.
C2a was predominantly type II pRCC and was associated with early-stage tumors and a distinct DNA methylation pattern, whereas C2b consisted exclusively of type II pRCC, presenting with later stage (III and IV) tumors and mutations in SETD2. Tumors in the C2c group were associated with CpG island methylator phenotype (CIMP) and had the worse overall survival among all pRCC tumors. This study also identified other mutations found in type II pRCC, including the deletion of CDKN2a, gene fusions involving the microphthalmia family (MiTF) members TFE3 and TFEB, mutations in chromosomal remodeling genes, activation of the NRF2 oxidative stress pathway, and mutations in FH which are seen in both sporadic Type II pRCC and hereditary leiomyomatosis and renal cell cancer (HLRCC) [22–24].
Chromophobe RCC
Germline mutations have also been described with chRCC. For example, germline mutation in FLCN in Birt-Hogg-Dubé syndrome is associated with increased likelihood of chRCC [25]. Additionally, germline mutations in PTEN causes Cowden syndrome, in which 5–29% patients can develop various histologies of RCC, including the chromophobe subtype. Comprehensive profiling has been used by Davis et al. and Ricketts et al. to characterize somatic mutations among 66 and 81 primary chRCC tumors, respectively [9,14]. Loss of one copy of chromosomes 1, 2, 6, 10, 13, and 17 was seen in 86% of cases. Furthermore, mitochondrial DNA mutations were implicated in upregulation of oxidative phosphorylation among chRCC compared to ccRCC and normal kidney cells. Additionally, whole genome sequencing identified a pattern of localized hypermutation within the TERT promoter region that led to increased TERT expression, suggesting these genetic mutations may lead to immortalization and a mechanism for metabolic upregulation. Other profiling studies by Durinck et al. identified mutations in TP53, PTEN, FAAH2, PDHB, PDXDC1, and ZNF765 among the chRCC tumors [13].
Other rare nccRCC Histologies
Other rare nccRCC histologies include translocation RCC, collecting duct RCC, sarcomatoid RCC, and unclassified RCC. MiTF family translocation RCC is characterized by translocation involving Xp11.23 and 6p21, with tumors predominantly occurring in children and young adults [26]. Gene fusions of MiTF genes TFE3 and TFEB occur with different fusion partners. Collecting duct carcinoma is an extremely rare type of nccRCC with highly aggressive behavior. Frequent alterations of NF2 and SMARCB1 were initially identified and further studies highlighted mutations in CDKN2A and SLC7A11, which was associated with cisplatin-resistance [27,28]. Sarcomatoid RCC is a histologic subtype that can occur with any other type of RCC [29]. This histology is associated with poor clinical outcomes, but treatment with systemic immunotherapy has shown promising response rates [30,31].
Comparative genomic profiling of 26 sarcomatoid RCC against 56 ccRCC tumors and datasets from The Cancer Genome Atlas that included ccRCC, pRCC, and chRCC demonstrated that TP53, VHL, CDKN2A, and NF2 were most commonly mutated among the sarcomatoid RCC [32]. This study highlighted that sarcomatoid RCC could be subdivided based on mutations in TP53 or NF2. Additional profiling efforts by Wang et al. comparing sarcomatoid RCC to other RCC subtypes demonstrated that sarcomatoid RCC is less likely to have two-hit loss of VHL and PBRM1 and is more likely to have mutations in PTEN, TP53, and RELN, which are associated with worse prognosis [33]. Further profiling of metastatic sarcomatoid RCC using RNA-seq by Pal et al. demonstrated elevated aurora kinase A activity, which was associated with increased activity within the mammalian target of rapamycin (mTOR) pathway [34]. Unclassified RCC are comprised of aggressive nccRCC that have no standard therapy. A thorough molecular analysis of 62 unclassified RCC identified somatic mutations in 29 genes, which included NF2, SETD2, BAP1, KMT2C, and MTOR [35]. This study further demonstrated that 26% of unclassified RCC with loss of NF2 and dysregulated Hippo-YAP pathway were associated with worse survival, whereas 21% of unclassified RCC with mutations in MTOR, TSC1, TSC2, or PTEN, and increased mTOR signaling were associated with better clinical outcome.
Comprehensive profiling efforts have helped uncover numerous genetic mutations among the various histologic subtypes of nccRCC, which are often overlapping. These discoveries are still nascent efforts towards better understanding the complex, intersecting pathways that promote tumorigenesis among nccRCC. Understanding the molecular underpinnings of nccRCC is of utmost importance in further categorization of nccRCC and improved targeting with novel therapeutic strategies.
nccRCC trials and tribulations
The therapeutic armamentarium available to treat advanced and metastatic ccRCC expanded rapidly over the last decade [4]. However, the same therapeutic strategies that have been successful in ccRCC have displayed muted responses in patients with nccRCC (Table 3). Therefore, for metastatic nccRCC the National Comprehensive Cancer Network (NCCN) guidelines recommend enrollment in clinical trials or treatment with the vascular endothelial growth factor receptor (VEGFR)-inhibitor sunitinib [36].
Table 3:
Treatment | Clinical trial | Mechanism of action | Control | Cohort (Line of therapy) | Endpoint(s) |
---|---|---|---|---|---|
Sunitinib |
SUPAP [44] NCT00541008 |
VEGFR & PDGFR inhibitor | N/A | pRCC (1st) |
Type I Median PFS: 6.6 mo, 95% CI 2.8–14.8 mo Median OS: 17.8 mo, 95% 5.7–26.1 mo Type II Median PFS: 5.5 mo, 95% CI 3.8–7.1 mo Median OS: 12.4 mo, 95% 8.2–14.3 mo |
Everolimus |
RAPTOR [68] NCT00688753 |
mTOR inhibitor | N/A | pRCC (1st) | Median PFS: 4.1 mo, 95% CI 3.6–5.5 mo Median OS: 21.4 mo, 95% CI 15.4–28.4 mo |
Sunitinib |
ESPN [45] NCT01185366 |
VEGFR & PDGFR inhibitor | Everolimus | nccRCC Sarcomatoid RCC |
Median PFS: 6.1 vs 4.1 mo, p=0.6 Median OS: 16.2 vs 14.9 mo, p=0.18 |
Sunitinib | ASPEN [46] | VEGFR & PDGFR inhibitor | Everolimus | nccRCC | PFS: HR1.41, 80% CI 1.03–1.92; p=0.16 |
Sunitinib (1st) Everolimus (2nd) |
RECORD-3 [69] NCT00903175 |
VEGFR & PDGFR inhibitor mTOR inhibitor |
Everolimus (1st) Sunitinib (2nd) |
ccRCC (1st) nccRCC (1st) |
PFS: HR 1.2, 95% CI 0.9–1.6 Median OS: HR 1.1, 95% 0.9–1.4 |
Sunitinib | NCT01219751 [70] | VEGFR & PDGFR inhibitor | N/A | nccRCC | Median PFS: 6.4 mo, 95% CI 4.2–8.6 mo 1-year PFS rate: 40% |
Foretinib | NCT00726323 [47] | MET & VEGFR2 inhibitor | N/A | pRCC (1st/2nd) | ORR: 13.5% Median PFS: 9.3 mo |
Crizotinib |
CREATE [48] NCT01524926 |
MET, ALK, ROS1 inhibitor | N/A | Type I pRCC (1st) |
MET+ ORR: 50%, 95% CI 6.8–93.2% 1-yr PFS: 75%, 95% CI 12.8–96.1% 1-yr OS: 75%, 95% CI 12.8–96.1% MET− ORR: 6.3%, 95% CI 0.2–30.2% 1-yr PFS: 27.3%, 95% CI 8.5–50.4% 1-yr OS: 71.8%, 95% CI 41.1–88.4% |
Erlotinib |
SWOG S0317 [71] NCT00060307 |
EGFR inhibitor | N/A | pRCC | ORR: 11%, 95% CI 3–24% Median OS: 27 mo, 95% CI 13–36 mo |
Tivantinib Erlotinib |
SWOG S1107 [72] NCT01688973 |
MET inhibitor EGFR inhibitor |
Tivantinib | pRCC (1st/2nd) | RR 0% (Study closed early) Median PFS 2 mo vs 3.9 mo |
Savolitinib |
SAVOIR [49] NCT03091192 |
MET inhibitor | Sunitinib | MET-driven, pRCC (1st/2nd) | PFS: HR 0.71, 95% CI 0.37–1.36, p=0.31 OS: HR 0.51, 95% CI 0.21–1.17, p=0.11 |
Savolitinib Durvalumab |
CALYPSO [52,53] NCT02819596 |
MET inhibitor PD-L1 inhibitor |
N/A | pRCC (1st/2nd) | ORR: 27% Median PFS: 4.9 mo, 95% CI 2.5–12 mo |
Atezolizumab Bevacizumab |
NCT02724878 [73] | Anti-PD-L1 mAb Anti-VEGF mAb |
N/A | nccRCC Sarcomatoid RCC |
ORR: 26% nccRCC and 50% sarcomatoid Median PFS: 8.3 mo, 95% CI 5.7–10.9 mo |
Ipilimumab Nivolumab |
CheckMate 214 [55] Post hoc analysis |
Anti-PD1 mAb Anti-CTLA4 mAb |
Sunitinib | Sarcomatoid RCC | Median OS: HR 0.45, 95% CI 0.3–0.7, p=0.0004 PFS: HR 0.54, 95% CI 0.33–0.86, p=0.0093 ORR: 60.8% vs 23.1% CR: 18.9% vs 3.1% |
Atezolizumab Bevacizumab |
IMmotion151 [54] Subgroup analysis |
Anti-PD-L1 mAb Anti-VEGF mAb |
Sunitinib | Sarcomatoid RCC | PFS: HR 0.52, 95% CI 0.34–0.79 ORR: 49% vs 14% CR: 10% vs 3% |
PD-L1: programmed death-ligand 1; VEGFR2: vascular endothelial growth factor receptor 2; EGFR: epidermal growth factor receptor; PDGFR: platelet-derived growth factor receptor; mTOR: mammalian target of rapamycin; PD-1: programmed cell death protein 1; CTLA4: cytotoxic T-lymphocyte associated protein 4; mAb: monoclonal antibody; pRCC: papillary renal cell carcinoma; nccRCC: non-clear cell renal cell carcinoma; ccRCC: clear cell renal cell carcinoma; PFS: progression-free survival; OS: overall survival; ORR: overall response rate; CR: complete response; HR: hazard ratio
Clinical trials for nccRCC have included a large variety of nccRCC histologies, making interpretation and clinical implementation difficult. Conversely, histology-specific trials have faced a unique challenge of accrual given the lower incidence of certain histologies. As a result, current evidence is mostly comprised of retrospective analyses, subgroup analyses of expanded access program studies, or small phase II studies [37–40]. Furthermore, translating the aforementioned genomic targets into viable therapeutic targets or biomarkers for assessing treatment efficacy have compounded these issues. Nevertheless, efforts are underway to determine whether the success with targeted therapies and immune checkpoint inhibitors in treating advanced ccRCC can be extended to advanced nccRCC (Table 4); the results of such studies may potentially transform the management paradigm of nccRCC.
Table 4:
Clinical trial | Phase | Experimental Arm | Control Arm | Cohort (Line of therapy) |
Primary Endpoint(s) |
---|---|---|---|---|---|
PAPMET, SWOG S1500 NCT02761057 |
II | Cabozantinib Crizotinib Savolitinib Sunitinib |
N/A | pRCC (1st/2nd) | PFS |
NCT02019693 | II | Capmatinib | N/A | pRCC (1st/2nd/3rd) | ORR |
NCT01130519 | II | Bevacizumab+Erlotinib | N/A | pRCC (1st/2nd/3rd) HLRCC (1st/2nd/3rd) |
ORR |
NCT02495103 | I/II | Vandetanib+Metformin | N/A | HLRCC SDH-associated pRCC |
ORR |
CABOSUN 2 NCT03541902 |
II | Cabozantinib | Sunitinib | Non-clear cell (1st/2nd) | PFS |
BONSAI NCT03354884 |
II | Cabozantinib | N/A | Collecting duct | ORR |
NCT02915783 | II | Lenvatinib+Everolimus | N/A | Non-clear cell (1st) | ORR |
CYTOSHRINK NCT04090710 |
II | Ipilimumab+Nivolumab+SBRT | Ipilimumab+Nivolumab | Clear cell (1st) Non-clear cell (1st) |
PFS |
SUNIFORECAST NCT03075423 |
II | Ipilimumab+Nivolumab | Sunitinib | Non-clear cell (1st) | OS at 1 year |
CA209-9KU NCT03635892 |
II | Nivolumab+Cabozantinib | N/A | Non-clear cell (1st/2nd) | ORR |
NCT03170960 | I/II | Atezolizumab+Cabozantinib | N/A | Clear cell (1st) Non-clear cell (2nd) |
MTD ORR |
ANZUP1602, UNISoN NCT03177239 |
II | Nivolumab (1st) followed by Ipilimumab+Nivolumab (2nd) | N/A | Non-clear cell (1st/2nd) | ORR |
CheckMate-920 NCT02982954 |
IIIb/IV | Ipilimumab+Nivolumab | N/A | Clear cell (1st) Non-clear cell (1st) |
IMAE |
KEYNOTE 427 NCT02853344 |
II | Pembrolizumab | N/A | Clear cell (1st) Non-clear cell (1st) |
ORR |
CheckMate 374 NCT02596035 |
IIIb/IV | Nivolumab | N/A | Clear cell (1st/2nd/3rd) Non-clear cell (1st/2nd/3rd) |
IMAE |
OMNIVORE NCT03203473 |
II | Ipilimumab+Nivolumab | N/A | Clear cell Non-clear celll |
PR/CR at 1 year after discontinuing nivolumab PR/CR at 1 year after adding ipilmumab |
AREN1721 NCT03595124 |
II | Axitinib+Nivolumab | Axitinib only Nivolumab only |
Translocation RCC | PFS |
PROSPER RCC NCT03055013 |
III | Perioperative Nivolumab and Nephrectomy | Nephrectomy alone | Biopsy proven RCC ≥T2Nx or TanyN+ or M1* |
EFS |
M1 disease has to be resected/definitively treated at the same time or within 12-weeks of surgery such that patient is considered “No Evidence of Disease”
DLT: dose-limiting toxicity; RCC: renal cell carcinoma; ccRCC: clear cell RCC; pRCC: papillary RCC; HLRCC: hereditary leiomyomatosis and RCC; PFS: progression-free survival; OS: overall survival; ORR: overall response rate; MTD: maximum tolerated dose; IMAE: incidence of high-grade immune-mediated adverse events; EFS: event-free survival; PR: partial response; CR: complete response; N/A: not applicable
Treatment landscape of nccRCC
The therapeutic strategies in nccRCC have predominantly been adapted from the successes of various agents in the management of ccRCC (Table 3 & 4) [4]. With the approval of multiple VEGF and mTOR inhibitors for metastatic ccRCC in the early 2000s [41–43], investigators consequently performed post-hoc analysis of sunitinib, sorafenib, and temsirolimus in nccRCC patients. [37–40]. nccRCC patients had less robust responses to these systemic therapies compared to their ccRCC counterparts. However, the studies collectively demonstrated objective response rates of 5–15% and provided support for targeting the mTOR and VEGF pathways. Subsequently, a prospective phase II trial, SUPAP, demonstrated median progression-free survival (PFS) of 5–7 months in patients who received sunitinib as first-line therapy for metastatic type I and II pRCC [44]. Two phase II trials, ESPN and ASPEN, evaluated sunitinib versus everolimus in metastatic nccRCC [45,46]. The ESPN trial included patients with a mix of nccRCC histology and ccRCC harboring >20% sarcomatoid histology. The trial did not demonstrate a significant difference in median PFS (6.1 vs 4.1 mo, p=0.6) or OS (16.2 vs 14.9 mo, p=0.18) between sunitinib and everolimus [45]. While the ASPEN trial showed a significant increase in median PFS in patients treated with sunitinib compared to everolimus (8.3 vs 5.6 mo, p=0.16), this analysis was done with a prespecified Type I error of 20% compared to the conventional 5% and did not translate to significant increase in OS (HR 1.12, 95% CI 0.7–2.1, p=0.60) [46]. Patients with good risk disease (HR 2.9, 80% CI 1.5–5.7) and papillary histology (HR 1.6, 80% CI 1.1–2.3) had a longer PFS with sunitinib compared to everolimus.
Following the availability of additional genomic data, therapeutic development for nccRCC focused on targets with high mutation rates that could be easily targeted. For example, the MET gene is mutated among 17% of pRCC [12,13]. A phase II trial looking at the efficacy of foretinib, a dual MET/VEGFR inhibitor, among patients with pRCC demonstrated median PFS of 9.3 months, and with 50%, 20%, and 9% partial response (PR) among patients with germline MET mutation, somatic MET mutation, and no MET mutation, respectively [47]. Despite these promising results, further development of this inhibitor was discontinued. Similar results were obtained when crizotinib, a MET/ALK/ROS1 inhibitor, was evaluated for type I pRCC in the CREATE trial [48]. In MET+ patients, 50% had PR with 1-year PFS rate of 75% compared to 6.3% PR and 1-year PFS rate of 27.3% in MET- patients. A potent MET inhibitor, savolitinib was recently evaluated in the phase III SAVOIR trial [49], building on a previous study that demonstrated significant improvement in PFS among patients with MET-driven pRCC [50]. Patients underwent baseline genomic profiling and those with MET-driven disease were randomized to sunitinib or savolitinib. Unfortunately, this study was discontinued due to poor accrual and did not demonstrate a statistical difference in PFS or OS between the two treatment arms.
Immune checkpoint inhibitors are effective in ccRCC, but data on their activity in nccRCC has been lacking until recently. A retrospective analysis of patients who had received nivolumab, an anti-PD1 antibody, for various nccRCC histologies demonstrated median PFS of 3.5 months and median OS that was not reached [51]. However, data from this study is difficult to interpret given several confounders such as different histologies, selection bias inherent to retrospective studies, and short follow up time.
More recently, preliminary results from a phase II study evaluating savolitinib in combination with durvalumab, an anti-PD-L1 antibody, for metastatic pRCC demonstrated ORR of 27% with median PFS of 4.9 months [52,53]. Notably patients receiving first-line therapy had a response rate of 33% and median PFS of 12.3 months. Interestingly, PD-L1 and MET expression were not associated with higher response rates or longer OS [53]. Further supporting the strong activity of the concept of dual inhibition, McGregor et al. performed a multicenter phase II trial to evaluate atezolizumab combined with bevacizumab in patients with nccRCC or sarcomatoid RCC; they demonstrated a remarkable ORR among nccRCC (26%) and sarcomatoid RCC (50%), with median PFS of 8.3 months.
This data further corroborates a subgroup analysis from the IMmotion 151 clinical trial, which evaluated these agents against sunitinib in metastatic ccRCC; this trial demonstrated higher median PFS (8.3 vs 5.3 mo) and ORR (49% vs 14%) among patients with sarcomatoid RCC [54]. Post-hoc analysis of patients with sarcomatoid histology from a similar trial, CheckMate 214, that evaluated the combination of nivolumab and ipilimumab against sunitinib for metastatic ccRCC demonstrated a higher OS (HR 0.45, 95% CI 0.3–0.7, p=0.0004) and improved PFS (HR 0.54, 95% CI 0.33–0.86, p=0.0093), with ORR of 60.8% in favor of nivolumab and ipilimumab [55]. Taken together, these results suggest that combination strategies may yield promising results in both nccRCC and sarcomatoid RCC. Further efforts to perform post-hoc analyses from pivotal phase III trials will help better tailor the use of targeted therapies and immunotherapies.
Ongoing clinical trials
The results of the aforementioned trials and other ongoing trials are leading to significant progress towards developing therapeutic regimens for metastatic nccRCC and sarcomatoid RCC. (Table 4). Notably, the SWOG S1500 (PAPMET) trial is prospectively assessing the role of targeting MET in patients with pRCC by randomizing an estimated 180 patients to either sunitinib, crizotinib, cabozantinib, or savolitinib [56]. The crizotinib and savolitinib arms were prematurely closed based on futility analysis and the final analysis, which is expected in 2021, will evaluate the efficacy of sunitinib and cabozantinib. Together with the CABOSUN II trial, this trial will help establish the role of cabozantinib, a MET/VEGFR/AXL inhibitor, in treating metastatic nccRCC, especially pRCC.
The paradigm-shifting phase III clinical trials, including KEYNOTE 426, JAVELIN 101, and CheckMate 214, that have transformed the management of metastatic ccRCC have suggested a potential role for combination therapy with checkpoint inhibitors in nccRCC [57–59]. CheckMate 920 and CheckMate 374 are phase IIIb/IV trials to confirm the safety and efficacy of ipilimumab+nivolumab and nivolumab alone, respectively, and thereby may help expand the indications for these regimens to include nccRCC [60,61]. In addition, the SUNIFORECAST trial will evaluate the efficacy of ipilimumab+nivolumab as first-line therapy against sunitinib in nccRCC in a European multicenter phase II trial [62]. The combination of a checkpoint inhibitor and a tyrosine kinase inhibitor, nivolumab and cabozantinib, is also being assessed in the CA209–9KU trial.
Preliminary results from NCT01130519, a trial of bevacizumab and erlotinib in patients with advanced HLRCC or sporadic pRCC, demonstrated a median PFS of 14.3 months and overall response rates of 54.2%, 72%, and 35% in the entire, HLRCC, and sporadic pRCC cohorts, respectively [63]. This is the first prospective study in HLRCC and suggests encouraging activity of bevacizumab and erlotinib particularly in this cohort. Final results from the overall study are eagerly awaited.
One notable addition to the list of ongoing clinical trials is the PROSPER RCC trial; in this study patients with biopsy-proven RCC of any histology who have locally advanced or oligometastatic disease that can be completely resected via nephrectomy receive perioperative nivolumab versus nephrectomy alone [64,65]. This study is the first-of-its-kind by including any RCC histology and incorporating a checkpoint inhibitor earlier in the treatment course.
The multitude of ongoing trials for nccRCC is an important step towards developing a variety of biology-driven therapies for nccRCC. Genomic and metabolic studies have clearly demonstrated key differences between ccRCC and nccRCC and among the various subtypes of nccRCC [9]. Leveraging this vast space of genomic information to target nccRCC is seeing new light in the era of better targeted and checkpoint inhibitor therapies.
Conclusions
Comprehensive profiling techniques have revolutionized our understanding of the mutational burden that underlies various nccRCC. While these efforts have helped in understanding the basis of mutations underlying both RCC-associated syndromes and sporadic RCC (Table 2) [66], the studies have revealed an extraordinary number of mutations associated with nccRCC, underscoring the great diversity of nccRCC [12–14,67]. Validation of these targets has yet to be thoroughly performed, leaving a significant gap between target identification, role in pathogenesis and drug development. Using previous and prospective genomic profiling may help identify biomarkers that better point to which therapies are best suited for which histologic subtypes. Signaling pathways and complex tumor microenvironment interactions could undermine drug efficacy, especially in histologies with significant heterogeneity. Furthermore, selection pressures from multiple lines of therapy could also result in metabolic alterations and responses that might lead to decreased efficacy.
While our understanding of nccRCC continues to evolve, a robust platform for understanding the complexities of tumor biology is needed. The results of the SWOG S1500 trial will be met with enthusiasm as it will provide clarity on the clinical significance of MET inhibition in pRCC and the efficacy of cabozantinib in nccRCC. The expansion of clinical trials into the nccRCC space provides optimism as newer, proven ccRCC treatment strategies, such as checkpoint inhibition and combinatorial therapy, are being applied to nccRCC and sarcomatoid RCC. A concerted effort needs to be made to include multiple RCC histologies in future clinical trials with planned subgroup analyses in order to further advance the treatment of both clear cell and non-clear cell RCC.
Key Points.
Non-clear cell RCC is comprised of various histologies with significantly different genetic mutations, posing a challenge for translating genomic mutations into clinically significant targets.
Successes of therapeutics in clear cell RCC has not yielded similar results for non-clear cell RCC; thus, clinical trial participation should be considered for all advanced or metastatic non-clear cell RCC cases.
Ongoing trials with novel combination therapies provide a promising future for treating patients with non-clear cell RCC.
Financial support and sponsorship
The research was funded by the Intramural Research Program of the National Institutes of Health, National Cancer Institute, Center for Cancer Research, Bethesda, Maryland, USA, and by a grant from the National Cancer Institute (P30CA072720).
Footnotes
Conflicts of interest
None
References
- 1.Siegel RL, Miller KD, Fuchs HE, Jemal A. Cancer statistics, 2021. CA Cancer J Clin. 2021;71(1):7–33. [doi: 10.3322/caac.21654] [DOI] [PubMed] [Google Scholar]
- 2.Zisman A, Pantuck AJ, Wieder J, et al. Risk group assessment and clinical outcome algorithm to predict the natural history of patients with surgically resected renal cell carcinoma. J Clin Oncol. 2002;20(23):4559–4566. [doi: 10.1200/JCO.2002.05.111] [DOI] [PubMed] [Google Scholar]
- 3.Nerich V, Hugues M, Paillard MJ, et al. Clinical impact of targeted therapies in patients with metastatic clear-cell renal cell carcinoma. Onco Targets Ther. 2014;7:365–374. [doi: 10.2147/OTT.S56370] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Patel HV, Shinder B, Srinivasan R, Singer EA. Challenges and opportunities in the management of metastatic renal cell carcinoma: Combination therapy and the role of cytoreductive surgery. Curr Opin Oncol. 2020;32(3):240–249. [doi: 10.1097/CCO.0000000000000621] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Cancer Genome Atlas Research N. Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature. 2013;499(7456):43–49. [doi: 10.1038/nature12222] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Sato Y, Yoshizato T, Shiraishi Y, et al. Integrated molecular analysis of clear-cell renal cell carcinoma. Nat Genet. 2013;45(8):860–867. [doi: 10.1038/ng.2699] [DOI] [PubMed] [Google Scholar]
- 7.Scelo G, Riazalhosseini Y, Greger L, et al. Variation in genomic landscape of clear cell renal cell carcinoma across europe. Nat Commun. 2014;5:5135. [doi: 10.1038/ncomms6135] [DOI] [PubMed] [Google Scholar]
- 8.Turajlic S, Xu H, Litchfield K, et al. Deterministic evolutionary trajectories influence primary tumor growth: Tracerx renal. Cell. 2018;173(3):595–610 e511. [doi: 10.1016/j.cell.2018.03.043] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Ricketts CJ, De Cubas AA, Fan H, et al. The cancer genome atlas comprehensive molecular characterization of renal cell carcinoma. Cell Rep. 2018;23(12):3698. [doi: 10.1016/j.celrep.2018.06.032] [DOI] [PubMed] [Google Scholar]
- 10.Gnarra JR, Tory K, Weng Y, et al. Mutations of the vhl tumour suppressor gene in renal carcinoma. Nat Genet. 1994;7(1):85–90. [doi: 10.1038/ng0594-85] [DOI] [PubMed] [Google Scholar]
- 11.The nobel prize in physiology or medicine 2019. [press release]. Nobel Media AB 20202020. [Google Scholar]
- 12.Cancer Genome Atlas Research N, Linehan WM, Spellman PT, et al. Comprehensive molecular characterization of papillary renal-cell carcinoma. N Engl J Med. 2016;374(2):135–145. [doi: 10.1056/NEJMoa1505917] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Durinck S, Stawiski EW, Pavia-Jimenez A, et al. Spectrum of diverse genomic alterations define non-clear cell renal carcinoma subtypes. Nat Genet. 2015;47(1):13–21. [doi: 10.1038/ng.3146] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Davis Caleb F, Ricketts CJ, Wang M, et al. The somatic genomic landscape of chromophobe renal cell carcinoma. Cancer Cell. 2014;26(3):319–330. [doi: 10.1016/j.ccr.2014.07.014] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Carlo MI, Hakimi AA, Stewart GD, et al. Familial kidney cancer: Implications of new syndromes and molecular insights. Eur Urol. 2019;76(6):754–764. [doi: 10.1016/j.eururo.2019.06.015] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Linehan WM, Ricketts CJ. The cancer genome atlas of renal cell carcinoma: Findings and clinical implications. Nat Rev Urol. 2019;16(9):539–552. [doi: 10.1038/s41585-019-0211-5] [DOI] [PubMed] [Google Scholar]
- 17.Linehan WM, Schmidt LS, Crooks DR, et al. The metabolic basis of kidney cancer. Cancer Discov. 2019;9(8):1006–1021. [doi: 10.1158/2159-8290.CD-18-1354] [DOI] [PubMed] [Google Scholar]; *A comprehensive summary of findings from The Cancer Genome Atlas profiling efforts.
- 18.Delahunt B, Eble JN. Papillary renal cell carcinoma: A clinicopathologic and immunohistochemical study of 105 tumors. Mod Pathol. 1997;10(6):537–544. [doi: [PubMed] [Google Scholar]
- 19.Schmidt L, Duh FM, Chen F, et al. Germline and somatic mutations in the tyrosine kinase domain of the met proto-oncogene in papillary renal carcinomas. Nat Genet. 1997;16(1):68–73. [doi: 10.1038/ng0597-68] [DOI] [PubMed] [Google Scholar]
- 20.Hagenkord JM, Gatalica Z, Jonasch E, Monzon FA. Clinical genomics of renal epithelial tumors. Cancer Genet. 2011;204(6):285–297. [doi: 10.1016/j.cancergen.2011.06.001] [DOI] [PubMed] [Google Scholar]
- 21.Albiges L, Guegan J, Le Formal A, et al. Met is a potential target across all papillary renal cell carcinomas: Result from a large molecular study of prcc with cgh array and matching gene expression array. Clin Cancer Res. 2014;20(13):3411–3421. [doi: 10.1158/1078-0432.CCR-13-2173] [DOI] [PubMed] [Google Scholar]
- 22.Ooi A, Wong JC, Petillo D, et al. An antioxidant response phenotype shared between hereditary and sporadic type 2 papillary renal cell carcinoma. Cancer Cell. 2011;20(4):511–523. [doi: 10.1016/j.ccr.2011.08.024] [DOI] [PubMed] [Google Scholar]
- 23.Tomlinson IP, Alam NA, Rowan AJ, et al. Germline mutations in fh predispose to dominantly inherited uterine fibroids, skin leiomyomata and papillary renal cell cancer. Nat Genet. 2002;30(4):406–410. [doi: 10.1038/ng849] [DOI] [PubMed] [Google Scholar]
- 24.Crooks DR, Maio N, Lang M, et al. Mitochondrial DNA alterations underlie an irreversible shift to aerobic glycolysis in fumarate hydratase-deficient renal cancer. Sci Signal. 2021;14(664). [doi: 10.1126/scisignal.abc4436] [DOI] [PMC free article] [PubMed] [Google Scholar]; **This study highlights the mechanism for the Warburg effect in HLRCC.
- 25.Nickerson ML, Warren MB, Toro JR, et al. Mutations in a novel gene lead to kidney tumors, lung wall defects, and benign tumors of the hair follicle in patients with the birt-hogg-dube syndrome. Cancer Cell. 2002;2(2):157–164. [doi: 10.1016/s1535-6108(02)00104-6] [DOI] [PubMed] [Google Scholar]
- 26.Argani P, Olgac S, Tickoo SK, et al. Xp11 translocation renal cell carcinoma in adults: Expanded clinical, pathologic, and genetic spectrum. Am J Surg Pathol. 2007;31(8):1149–1160. [doi: 10.1097/PAS.0b013e318031ffff] [DOI] [PubMed] [Google Scholar]
- 27.Pal SK, Choueiri TK, Wang K, et al. Characterization of clinical cases of collecting duct carcinoma of the kidney assessed by comprehensive genomic profiling. Eur Urol. 2016;70(3):516–521. [doi: 10.1016/j.eururo.2015.06.019] [DOI] [PubMed] [Google Scholar]
- 28.Wang J, Papanicolau-Sengos A, Chintala S, et al. Collecting duct carcinoma of the kidney is associated with cdkn2a deletion and slc family gene up-regulation. Oncotarget. 2016;7(21):29901–29915. [doi: 10.18632/oncotarget.9093] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Ged Y, Chen YB, Knezevic A, et al. Metastatic chromophobe renal cell carcinoma: Presence or absence of sarcomatoid differentiation determines clinical course and treatment outcomes. Clin Genitourin Cancer. 2019;17(3):e678–e688. [doi: 10.1016/j.clgc.2019.03.018] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Iacovelli R, Ciccarese C, Bria E, et al. Patients with sarcomatoid renal cell carcinoma - re-defining the first-line of treatment: A meta-analysis of randomised clinical trials with immune checkpoint inhibitors. Eur J Cancer. 2020;136:195–203. [doi: 10.1016/j.ejca.2020.06.008] [DOI] [PubMed] [Google Scholar]
- 31.Aly R, Aujla AS, Gupta S, et al. Evolving paradigms in the management and outcomes of sarcomatoid renal cell carcinoma in the era of immune checkpoint inhibitors. World J Oncol. 2020;11(5):183–187. [doi: 10.14740/wjon1325] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Malouf GG, Ali SM, Wang K, et al. Genomic characterization of renal cell carcinoma with sarcomatoid dedifferentiation pinpoints recurrent genomic alterations. Eur Urol. 2016;70(2):348–357. [doi: 10.1016/j.eururo.2016.01.051] [DOI] [PubMed] [Google Scholar]
- 33.Wang Z, Kim TB, Peng B, et al. Sarcomatoid renal cell carcinoma has a distinct molecular pathogenesis, driver mutation profile, and transcriptional landscape. Clin Cancer Res. 2017;23(21):6686–6696. [doi: 10.1158/1078-0432.CCR-17-1057] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Pal SK, He M, Tong T, et al. Rna-seq reveals aurora kinase-driven mtor pathway activation in patients with sarcomatoid metastatic renal cell carcinoma. Mol Cancer Res. 2015;13(1):130–137. [doi: 10.1158/1541-7786.MCR-14-0352] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Chen YB, Xu J, Skanderup AJ, et al. Molecular analysis of aggressive renal cell carcinoma with unclassified histology reveals distinct subsets. Nat Commun. 2016;7:13131. [doi: 10.1038/ncomms13131] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Network NCC. Nccn clinical practice guidelines in oncology kidney cancer 2020. Available from: https://www.nccn.org/professionals/physician_gls/pdf/kidney.pdf. [DOI] [PubMed]
- 37.Choueiri TK, Plantade A, Elson P, et al. Efficacy of sunitinib and sorafenib in metastatic papillary and chromophobe renal cell carcinoma. J Clin Oncol. 2008;26(1):127–131. [doi: 10.1200/JCO.2007.13.3223] [DOI] [PubMed] [Google Scholar]
- 38.Dutcher JP, de Souza P, McDermott D, et al. Effect of temsirolimus versus interferon-alpha on outcome of patients with advanced renal cell carcinoma of different tumor histologies. Med Oncol. 2009;26(2):202–209. [doi: 10.1007/s12032-009-9177-0] [DOI] [PubMed] [Google Scholar]
- 39.Gore ME, Szczylik C, Porta C, et al. Safety and efficacy of sunitinib for metastatic renal-cell carcinoma: An expanded-access trial. Lancet Oncol. 2009;10(8):757–763. [doi: 10.1016/S1470-2045(09)70162-7] [DOI] [PubMed] [Google Scholar]
- 40.Stadler WM, Figlin RA, McDermott DF, et al. Safety and efficacy results of the advanced renal cell carcinoma sorafenib expanded access program in north america. Cancer. 2010;116(5):1272–1280. [doi: 10.1002/cncr.24864] [DOI] [PubMed] [Google Scholar]
- 41.Motzer RJ, Hutson TE, Tomczak P, et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med. 2007;356(2):115–124. [doi: 10.1056/NEJMoa065044] [DOI] [PubMed] [Google Scholar]
- 42.Hudes G, Carducci M, Tomczak P, et al. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med. 2007;356(22):2271–2281. [doi: 10.1056/NEJMoa066838] [DOI] [PubMed] [Google Scholar]
- 43.Escudier B, Eisen T, Stadler WM, et al. Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med. 2007;356(2):125–134. [doi: 10.1056/NEJMoa060655] [DOI] [PubMed] [Google Scholar]
- 44.Ravaud A, Oudard S, De Fromont M, et al. First-line treatment with sunitinib for type 1 and type 2 locally advanced or metastatic papillary renal cell carcinoma: A phase ii study (supap) by the french genitourinary group (getug)dagger. Ann Oncol. 2015;26(6):1123–1128. [doi: 10.1093/annonc/mdv149] [DOI] [PubMed] [Google Scholar]
- 45.Tannir NM, Jonasch E, Albiges L, et al. Everolimus versus sunitinib prospective evaluation in metastatic non-clear cell renal cell carcinoma (espn): A randomized multicenter phase 2 trial. Eur Urol. 2016;69(5):866–874. [doi: 10.1016/j.eururo.2015.10.049] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Armstrong AJ, Halabi S, Eisen T, et al. Everolimus versus sunitinib for patients with metastatic non-clear cell renal cell carcinoma (aspen): A multicentre, open-label, randomised phase 2 trial. Lancet Oncol. 2016;17(3):378–388. [doi: 10.1016/S1470-2045(15)00515-X] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Choueiri TK, Vaishampayan U, Rosenberg JE, et al. Phase ii and biomarker study of the dual met/vegfr2 inhibitor foretinib in patients with papillary renal cell carcinoma. J Clin Oncol. 2013;31(2):181–186. [doi: 10.1200/JCO.2012.43.3383] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Schoffski P, Wozniak A, Escudier B, et al. Crizotinib achieves long-lasting disease control in advanced papillary renal-cell carcinoma type 1 patients with met mutations or amplification. Eortc 90101 create trial. Eur J Cancer. 2017;87:147–163. [doi: 10.1016/j.ejca.2017.10.014] [DOI] [PubMed] [Google Scholar]
- 49.Choueiri TK, Heng DYC, Lee JL, et al. Efficacy of savolitinib vs sunitinib in patients with met-driven papillary renal cell carcinoma: The savoir phase 3 randomized clinical trial. JAMA Oncol. 2020;6(8):1247–1255. [doi: 10.1001/jamaoncol.2020.2218] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Choueiri TK, Plimack E, Arkenau HT, et al. Biomarker-based phase ii trial of savolitinib in patients with advanced papillary renal cell cancer. J Clin Oncol. 2017;35(26):2993–3001. [doi: 10.1200/JCO.2017.72.2967] [DOI] [PubMed] [Google Scholar]
- 51.Koshkin VS, Barata PC, Zhang T, et al. Clinical activity of nivolumab in patients with non-clear cell renal cell carcinoma. J Immunother Cancer. 2018;6(1):9. [doi: 10.1186/s40425-018-0319-9] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Powles T, Larkin JMG, Patel P, et al. A phase ii study investigating the safety and efficacy of savolitinib and durvalumab in metastatic papillary renal cancer (calypso). Journal of Clinical Oncology. 2019;37(7_suppl):545–545. [doi: 10.1200/JCO.2019.37.7_suppl.545] [DOI] [PubMed] [Google Scholar]
- 53.Rodriguez CS, Larkin JMG, Patel P, et al. Overall survival results for durvalumab and savolitinib in metastatic papillary renal cancer. Journal of Clinical Oncology. 2020;38(6_suppl):619–619. [doi: 10.1200/JCO.2020.38.6_suppl.619] [DOI] [Google Scholar]
- 54.Rini BI, Motzer RJ, Powles T, et al. Atezolizumab plus bevacizumab versus sunitinib for patients with untreated metastatic renal cell carcinoma and sarcomatoid features: A prespecified subgroup analysis of the immotion151 clinical trial. Eur Urol. 2020. [doi: 10.1016/j.eururo.2020.06.021] [DOI] [PMC free article] [PubMed] [Google Scholar]; **This subgroup analysis demonstrates the efficacy of anti-PD-L1 and anti-VEGF combination therapy for sarcomatoid RCC.
- 55.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. 2020. [doi: 10.1158/1078-0432.CCR-20-2063] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Pal SK, Tangen CM, Thompson IM, et al. A randomized, phase ii efficacy assessment of multiple met kinase inhibitors in metastatic papillary renal carcinoma (prcc): Swog s1500. Journal of Clinical Oncology. 2017;35(15_suppl):TPS4599–TPS4599. [doi: 10.1200/JCO.2017.35.15_suppl.TPS4599] [DOI] [Google Scholar]; *Ongoing PAPMET trial is currently evaluating the efficacy of cabozantinib, a MET/VEGFR/AXL inhibitor, in treating metastatic nccRCC.
- 57.Motzer RJ, Penkov K, Haanen J, et al. Avelumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. N Engl J Med. 2019;380(12):1103–1115. [doi: 10.1056/NEJMoa1816047] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Motzer RJ, Tannir NM, McDermott DF, et al. Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma. N Engl J Med. 2018;378(14):1277–1290. [doi: 10.1056/NEJMoa1712126] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Rini BI, Plimack ER, Stus V, et al. Pembrolizumab plus axitinib versus sunitinib for advanced renal-cell carcinoma. New England Journal of Medicine. 2019. [doi: 10.1056/NEJMoa1816714] [DOI] [PubMed] [Google Scholar]
- 60.Vogelzang NJ, Olsen MR, McFarlane JJ, et al. Safety and efficacy of nivolumab in patients with advanced non-clear cell renal cell carcinoma: Results from the phase iiib/iv checkmate 374 study. Clin Genitourin Cancer. 2020. [doi: 10.1016/j.clgc.2020.05.006] [DOI] [PubMed] [Google Scholar]; **This study demonstrate the efficacy of nivolumab only for treatment of advanced nccRCC.
- 61.Emamekhoo H, Olsen M, Carthon BC, et al. Safety and efficacy of nivolumab plus ipilimumab (nivo+ipi) in patients with advanced renal cell carcinoma (arcc) with brain metastases: Interim analysis of checkmate 920. Journal of Clinical Oncology. 2019;37(15_suppl):4517–4517. [doi: 10.1200/JCO.2019.37.15_suppl.4517] [DOI] [Google Scholar]
- 62.Ahrens M, Escudier B, Boleti E, et al. A randomized phase ii study of nivolumab plus ipilimumab versus standard of care in previously untreated and advanced non-clear cell renal cell carcinoma (suniforecast). Journal of Clinical Oncology. 2020;38(15_suppl):TPS5103–TPS5103. [doi: 10.1200/JCO.2020.38.15_suppl.TPS5103] [DOI] [Google Scholar]
- 63.Gurram S, Al Harthy M, Singer EA, et al. Phase ii study of bevacizumab and erlotinib in subjects with advanced hereditary leiomyomatosis and renal cell cancer (hlrcc) or sporadic papillary renal cell cancer. Kidney Cancer. 2020;18(4 Suppl):28–29. [doi: [Google Scholar]
- 64.Patel HD, Karam JA, Allaf ME. Surgical management of advanced kidney cancer: The role of cytoreductive nephrectomy and lymphadenectomy. J Clin Oncol. 2018:JCO2018790246. [doi: 10.1200/JCO.2018.79.0246] [DOI] [PubMed] [Google Scholar]
- 65.Patel HV, Srivastava A, Singer EA. To be or “node” to be: Nodal disease and the role of lymphadenectomy in the treatment of renal cell carcinoma. Med Res Arch. 2020;8(5). [doi: 10.18103/mra.v8i5.2091] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Lang M, Vocke CD, Ricketts CJ, et al. Clinical and molecular characterization of microphthalmia-associated transcription factor (mitf)-related renal cell carcinoma. Urology. 2020. [doi: 10.1016/j.urology.2020.11.025] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Dudani S, de Velasco G, Wells JC, et al. Evaluation of clear cell, papillary, and chromophobe renal cell carcinoma metastasis sites and association with survival. JAMA Netw Open. 2021;4(1):e2021869. [doi: 10.1001/jamanetworkopen.2020.21869] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Escudier B, Molinie V, Bracarda S, et al. Open-label phase 2 trial of first-line everolimus monotherapy in patients with papillary metastatic renal cell carcinoma: Raptor final analysis. Eur J Cancer. 2016;69:226–235. [doi: 10.1016/j.ejca.2016.08.004] [DOI] [PubMed] [Google Scholar]
- 69.Knox JJ, Barrios CH, Kim TM, et al. Final overall survival analysis for the phase ii record-3 study of first-line everolimus followed by sunitinib versus first-line sunitinib followed by everolimus in metastatic rcc. Ann Oncol. 2017;28(6):1339–1345. [doi: 10.1093/annonc/mdx075] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Lee JL, Ahn JH, Lim HY, et al. Multicenter phase ii study of sunitinib in patients with non-clear cell renal cell carcinoma. Ann Oncol. 2012;23(8):2108–2114. [doi: 10.1093/annonc/mdr586] [DOI] [PubMed] [Google Scholar]
- 71.Gordon MS, Hussey M, Nagle RB, et al. Phase ii study of erlotinib in patients with locally advanced or metastatic papillary histology renal cell cancer: Swog s0317. J Clin Oncol. 2009;27(34):5788–5793. [doi: 10.1200/JCO.2008.18.8821] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Twardowski PW, Tangen CM, Wu X, et al. Parallel (randomized) phase ii evaluation of tivantinib (arq197) and tivantinib in combination with erlotinib in papillary renal cell carcinoma: Swog s1107. Kidney Cancer. 2017;1(2):123–132. [doi: 10.3233/KCA-170018] [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.McGregor BA, McKay RR, Braun DA, et al. Results of a multicenter phase ii study of atezolizumab and bevacizumab for patients with metastatic renal cell carcinoma with variant histology and/or sarcomatoid features. J Clin Oncol. 2020;38(1):63–70. [doi: 10.1200/JCO.19.01882] [DOI] [PMC free article] [PubMed] [Google Scholar]