Next Article in Journal
Patient-Reported Outcomes Before and After Radiotherapy for Brain Metastases—A Prospective Cohort Study of 239 Non-Small-Cell Lung Cancer Patients
Previous Article in Journal
Correction: Guarneri et al. Diagnostic Performance of PIVKA-II in Italian Patients with Hepatocellular Carcinoma. Cancers 2025, 17, 167
Previous Article in Special Issue
Characteristics and Outcomes of T1a Renal Cell Carcinoma Presenting with Metastasis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Systemic Treatment of Locally Advanced or Metastatic Non-Clear Cell Renal Cell Carcinoma

by
Joseph Vento
1,*,
Tian Zhang
1,
Payal Kapur
2,
Hans Hammers
1,
James Brugarolas
1 and
Qian Qin
1
1
Division of Hematology and Oncology, Department of Internal Medicine, University of Texas Southwestern, Dallas, TX 75235, USA
2
Department of Pathology, University of Texas Southwestern, Dallas, TX 75235, USA
*
Author to whom correspondence should be addressed.
Cancers 2025, 17(9), 1527; https://doi.org/10.3390/cancers17091527
Submission received: 1 April 2025 / Revised: 24 April 2025 / Accepted: 28 April 2025 / Published: 30 April 2025
(This article belongs to the Special Issue Advances in Locally Advanced and Metastatic Kidney Cancer)

Simple Summary

Non-clear cell renal cell carcinoma represents a diverse group of rare kidney cancers. As an increased number of treatments emerge, it is essential to understand systemic options for these cancers. Here, we review systemic therapies in non-clear cell renal cell carcinoma, with special attention to recent prospective clinical trials and treatment options for specific disease subtypes.

Abstract

Non-clear cell renal cell carcinoma (nccRCC) represents a heterogenous group of malignancies with varying degrees of clinical aggressiveness and response to different systemic therapies. As the characterization of subtypes of nccRCC continues to evolve, it is important to understand the evidence around systemic treatments used in advanced or metastatic stages of specific subtypes. Here, we review the literature on systemic therapies in nccRCC, with a focus on prospective trials that included patients with papillary renal cell carcinoma (RCC), chromophobe RCC, RCC not further classified/unclassified RCC, translocation RCC, collecting duct RCC, and renal medullary carcinoma. We also review emerging treatments for other molecularly defined subtypes of this disease.

1. Introduction

The most common subtype of renal cell carcinoma (RCC) in adults is clear cell renal cell carcinoma (ccRCC), which makes up 75–80% of cases [1]. All other RCCs are grouped under the umbrella term of non-clear cell renal cell carcinoma (nccRCC), which encompasses a heterogenous group of malignancies that vary in characteristics such as cell of origin, morphology, and molecular features. Common subtypes included in nccRCC are papillary RCC (10–15% of all RCCs), chromophobe RCC (5–10%), and RCC not further classified (NOS) (previously referred to as unclassified RCC) (5%), while less common subtypes include collecting duct carcinoma (2%) and SMARCB1-deficient renal medullary carcinoma (<1%). The most recent 2022 World Health Organization (WHO) classification of kidney cancers included for the first time a category of molecularly defined subtypes of RCCs, which includes TFE3-rearranged RCC and TFEB-altered RCC (translocation RCC), fumarate hydratase (FH)-deficient RCC, succinate dehydrogenase (SDH)-deficient RCC, ELOC-mutated RCC, and anaplastic lymphoma kinase (ALK)-rearranged RCC [2].
Though a large number of all RCC diagnoses occur at early stages and may be curable with localized treatments (e.g., surgery), cancer-specific mortality drops significantly for patients who present with advanced stages that warrant systemic therapies (27–28% at 5 years) [1]. The rarity and diversity of nccRCC subtypes have made these cancers challenging to study prospectively; however, an increasing number of published and ongoing clinical trials have focused specifically on systemic therapies in these diseases. Of note, aggressive focal therapy (e.g., surgery and radiation) can play important roles in the management of nccRCC patients, and, though not discussed explicitly in this review, understanding the likelihood of response to systemic therapy can aid in personalizing treatment approaches to patients with specific disease subtypes.
The primary types of systemic therapies with prospective data in nccRCC subtypes are immune checkpoint inhibitors (ICIs), vascular endothelial growth factor receptor (VEGFR)-targeting tyrosine kinase inhibitors (TKIs), mammalian target of rapamycin (mTOR) inhibitors, vascular endothelial growth factor (VEGF)-targeting monoclonal antibodies, and chemotherapies. Mirroring the success of ICI-based doublets in ccRCC, several prospective clinical trials have tested ICI-based doublets in single-arm phase 2 trials for nccRCC patients [3]. We discuss these trials and changes to the systemic treatment landscape for nccRCC subtypes.

2. Results

Updates from the 2022 WHO classification of nccRCC are provided to contextualize the nccRCC systemic therapy literature published prior to these recent changes. A summary of prospective trial efficacy results for papillary, chromophobe, unclassified, and translocation RCC for TKIs and mTOR inhibitors is provided in Table 1 and for ICI monotherapies and combination strategies in Table 2. Collecting duct and renal medullary RCC data are reported separately given their unique biology and chemotherapy-based treatment strategies. Finally, other molecularly defined subtypes are discussed, with a focus on FH-deficient RCC.
The primary endpoint for most prospective trials in nccRCC subtypes is overall response rate (ORR), which reports the number of complete responses (CRs, no evidence of viable tumor) or partial responses (PRs, 30% shrinkage or more of tumor index lesions) out of the total number of patients in a trial with measurable disease. While useful as a surrogate marker of efficaciousness in early-phase trials, this outcome falls short in explaining duration of response or survival time. These metrics are better captured through reporting median progression-free survival (mPFS) or median overall survival (mOS) times, which require longer follow-up times for patients who receive a specific therapy. These metrics are provided when available for the trials discussed, and ongoing trials should continue to emphasize these meaningful outcomes in patients with nccRCC.
Additionally, patient comorbidities can play a significant role in systemic therapy selection and their sequencing for patients across subtypes of nccRCC. VEGFR-targeted TKIs and VEFR antibodies should be avoided in patients with poorly controlled hypertension, recent major cardiovascular events, or known gastrointestinal fistulas or perforations [23]. ICIs should be avoided in patients with pre-existing autoimmune conditions requiring high levels of immunosuppression, and close co-management with autoimmune disease experts is frequently required when treatment is attempted [24]. mTOR inhibitors must be used with caution in patients with poorly controlled diabetes, hyperlipidemia, or severe pulmonary disease due to risks of metabolic abnormalities and pneumonitis [25]. Local therapies such as stereotactic body radiation therapy (SBRT) can play important roles for palliation or disease control in patients with limited performance status or oligoprogressive disease [26].

2.1. Updates in nccRCC Classification

It is important to review recent updates in nccRCC subtype classification to contextualize the nccRCC literature that utilized prior classification schemes. The 2022 WHO RCC Classification made significant changes to how nccRCC subtypes are classified, in particular for papillary RCC [2]. A review of morphologic features of representative nccRCC subtypes is shown in Figure 1.
Prior to the 2022 WHO classification, papillary RCC was divided into type 1 and type 2 papillary RCC [27]. “Type 1” papillary RCC was defined by thin fibrovascular cores lined by a single layer of cells typically with pale cytoplasm and low-grade nuclei and high frequency of MET gene alterations [28]. “Type 2” papillary RCC characterized by pseudostratification, eosinophilic cytoplasm, and prominent nucleoli as well as gain in chromosomes 12, 16, and 20. Importantly, many type 2 papillary RCC show significant variability in both their morphology and clinical behavior and, with growing use of molecular studies, are now recognized as distinct molecular or histologic types, such as FH-deficient RCC, MiT family translocation RCC, ALK-rearranged RCC, and acquired cystic-disease-associated RCC (ACD-RCC) [29]. Further, recent analyses have shown that papillary RCC are better prognostically classified by the WHO grade rather than the type 1/type 2 subtyping and, therefore, in the 2022 WHO, the former “type 1” papillary RCC is regarded as the classic papillary RCC and subtyping into type 1 or 2 is no longer recommended.
Ongoing innovations in artificial intelligence may drive more accurate nccRCC classification into already existing disease subtypes as well as lead to additional methods of classification [30]. These methods will become increasingly important as subtype classification relies more on complex multimodal data ranging from histopathology to multi-omic molecular analyses.

2.2. Papillary RCC

Papillary RCC is believed to originate from the renal proximal tubule similar to ccRCC, though single-cell studies have highlighted that some of these tumors may originate from the collecting duct [31]. Due to the previously discussed updates in the 2022 WHO classification of papillary RCC, trial results that included this subtype must be interpreted with the context that the modern definition of papillary RCC consists largely of what was previously defined as type 1 papillary RCC, with much of type 2 papillary RCC now reclassified into new independent subtypes.
Analogous to the history of systemic therapies in advanced ccRCC, the prospective trials in papillary RCC initially focused on VEGFR TKI monotherapies, with cabozantinib (inhibitor of MET, AXL, and VEGFR2) outperforming sunitinib (inhibitor of PDGFR, VEGFR1-3, KIT, and RET) in the only direct comparison of these agents [5]. Next came ICI monotherapies and then, more recently, an increasing number of studies have evaluated ICI/ICI and VEGFR-TKI/ICI combination strategies, with single-arm prospective studies of cabozantinib with nivolumab (PD-1 antibody) and lenvatinib (inhibitor of VEGFR1-3, FGFR1-4, PDGFRA, KIT, and RET) with pembrolizumab (PD-1 antibody) showing compelling efficacies. Additional, randomized trials are ongoing and further discussed below (NCT05411081) [32].

2.2.1. VEGFR TKI Monotherapy

Single-arm studies of VEGFR TKIs enrolling patients with papillary RCC are summarized in Table 1 and include the papillary-exclusive AXIPAP trial for axitinib (inhibitor of VEGFR1-3, ORR 28.6%) [4], NCT01538238 for pazopanib (inhibitor of PDGFR, VEGFR1-3, FGFR1/3, and KIT, ORR 39% for papillary subgroup) [10], NCT02915783 for lenvatinib with everolimus (mTOR inhibitor, ORR 15% for papillary subgroup) [8], and an NCT00502307 subgroup analysis for tivozanib (inhibitor of VEGFR1-3, ORR 87.5% for papillary subgroup) [11]. Though the subgroup had a small number of patients, the tivozanib study in particular showed a high response rate for papillary RCC, with seven out of the eight patients responding to treatment [11]. Additional single-arm studies include the SUPAP study for sunitinib, which divided outcomes by the historical type 1 (ORR 13%, mOS 18.7 months (mo), 95% confidence interval [CI] 5.7–26.1 mo) and type 2 (ORR 11%, mOS 12.4 mo, 95% CI 8.2–14.3 mo) papillary RCC [33].
Randomized trials of VEGFR TKI monotherapy include the ESPN and ASPEN trials comparing sunitinib to everolimus for nccRCC [6,7]. The ESPN trial enrolled 31% papillary RCC patients with an ORR of 9% for sunitinib and 2.8% for everolimus. The trial did not find a significant mPFS difference, with 6.1 mo for sunitinib and 4.1 mo for everolimus (p = 0.6). In the ASPEN trial, sunitinib showed a prolonged mPFS over everolimus (8.3 versus (vs.) 5.6 mo, hazard ratio (HR) 1.41, 80% confidence interval (CI) 1.03–1.92, p = 0.16) with an ORR of 18% vs. 9% [7]. Of the 70 patients (65%) with papillary RCC, ORR was higher at 24% for sunitinib vs. 5% with everolimus. The difference in findings between these trials may be driven by the nonpapillary subgroups, which encompassed 60% of the total patients in ESPN but only 35% in ASPEN.
More recently, the PAPMET trial randomized patients with papillary RCC to sunitinib, cabozantinib, savolitinib (MET inhibitor), and crizotinib (inhibitor of ALK, HGFR, and ROS1). Cabozantinib demonstrated an improved ORR and mPFS over sunitinib (ORR: 23% vs. 4%, two-sided p = 0.010; mPFS: 9.0 vs. 5.6 mo, HR 0.60, 95% CI 0.37–0.97, p = 0.019) [5]. The savolitinib and crizotinib arms were halted at the prespecified futility analysis. Important toxicities in the cabozantinib arm included fatigue (70%, 15% grade 3+), diarrhea (56%, 7% grade 3+), and hand–foot syndrome (49%, 21% grade 3+), with dose reductions in 34% of patients and a discontinuation rate of 23%. For sunitinib, toxicities included fatigue (62%, 9% grade 3+), diarrhea (49%, 7% grade 3+), and nausea (44%, 9%, 21% grade 3+), with dose reductions in 38% of patients and a discontinuation rate of 24%
Finally, while most studies evaluated VEGFR TKI monotherapy in the first line, the UNICAB trial was designed to address the role of cabozantinib in the post-ICI or ICI-ineligible setting. However, the trial had low recruitment in the setting of the COVID-19 pandemic. Of the total 31 patients enrolled with nccRCC (including 10 papillary type 1 and 4 papillary type 2), the ORR was 22.6% (95% CI 9.6–41.1 mo) [34].

2.2.2. mTOR Inhibitors

Everolimus monotherapy has been utilized as a control arm in multiple randomized trials with relatively low efficacy in papillary RCC (Table 1). The prior single-arm phase II RAPTOR study of everolimus in papillary RCC reported similar low efficacy with an ORR of 1/88 (1%) and an mOS of 21.4 mo (95% CI 15.4–28.4 mo). In combination, the NCT02915783 phase II trial of everolimus plus lenvatinib showed a 15% ORR, while the NCT01399918 phase II trial of everolimus plus bevacizumab (VEGF antibody) showed an ORR rate of 35% in patients with treatment-naïve, papillary RCC [9].

2.2.3. ICI Monotherapy

Prior to the exploration of ICI combinations, several trials investigated ICI monotherapy strategies. In the phase II KEYNOTE-427, pembrolizumab achieved an ORR of 26.7% in 165 treatment-naïve, nccRCC patients, of which 71.5% with papillary RCC achieved an ORR of 28.8% [12]. Of the responders, 59.7% had durable response lasting longer than 12 mo. The phase IIIb/IV CheckMate 374 trial evaluated nivolumab monotherapy in patients with up to three prior lines of therapy and found an ORR of 13.6% (including two partial responses (PRs) in patients with papillary RCC) and an mPFS of 2.2 mo.
The UNISoN trial was a single-arm trial evaluating nivolumab monotherapy followed by nivolumab plus ipilimumab at progression [14]. The ORR for nivolumab alone was 16.9% with 14.7% ORR across papillary RCC patients. The ORR in patients who received doublet ICI therapy after progression on nivolumab was 10%.

2.2.4. ICI Combination Therapies

KEYNOTE-B61 was a multicenter single-arm phase 2 trial examining the combination of pembrolizumab and lenvatinib in advanced nccRCC [15]. In total, 93 (59%) of the 158 enrolled patients had papillary RCC, and the primary outcome of ORR for patients with papillary RCC was 54% (8 CRs and 42 PRs). mPFS was 18 months (95% CI 14 mo to Not Reached). Toxicity profiles were similar to the CLEAR trial of this ICI–TKI combination therapy in ccRCC [35], with 51% of patients having grade 3 or higher toxicities and the most common adverse events including hypertension (34% overall, 23% grade 3+), diarrhea (41% overall, 3% grade 3+), and hypothyroidism (36% overall, 1% grade 3+). Thirty-four percent of patients reduced their dose, while 14% of patients discontinued lenvatinib due to toxicity. Fifteen percent of patients discontinued pembrolizumab and seven percent discontinued both drugs due to toxicity.
CA209-9KU was a single-center phase II trial evaluating the combination of nivolumab and cabozantinib in advanced nccRCC [16]. A total of 32 of the 47 enrolled patients had papillary RCC with an ORR of 47% (95% CI 30–64%, 1 CR, and 14 PR) and an mPFS of 13 mo (95% CI 7–16 mo) [36]. Across all enrolled patients, toxicity profiles were similar to the CheckMate 9ER trial of this IO–TKI combination in ccRCC [37], with 55% grade 3 or above toxicities. The most common adverse events were fatigue (70%, 0% grade 3+), palmar–plantar erythrodysesthesia (68%, 5% grade 3+), and diarrhea (63%, 7.5% grade 3+). Of the patients, 88% required a dose reduction, 40% discontinued cabozantinib, 40% discontinued nivolumab, and 33% discontinued both drugs due to side effects.
Prospective trials of ICI–ICI combination therapy in nccRCC include the multicohort CheckMate 920 phase 3b/4 study of nivolumab plus ipilimumab (anti-CTLA4 antibody). This trial enrolled patients excluded from registrational Checkmate studies and included an nccRCC cohort [17]. Detailed subtype-level outcomes are not available, though, of the 52 patients enrolled, 18 had papillary RCC. In the overall, response-evaluable cohort of 46 patients, ORR was 19.6% (95% CI 9.4–33.9), mPFS was 3.7 mo (95% CI 2.7–4.6 mo), and mOS was 21.2 mo (95% CI 16.6 mo—not evaluable). Within the papillary RCC cohort, one patient achieved a CR and four patients achieved PRs. Overall toxicities were consistent with the CheckMate 214 trial of this ICI–ICI combination in ccRCC [38], with 36.5% of patients having grade 3 or higher toxicities. The most common toxicities were fatigue (48.1%, 3.8% grade 3+), diarrhea (30.8%, 3.8% grade 3+), and nausea (26.9%, 0% grade 3+).
The SUNNIFORECAST trial offered additional prospective data for the nivolumab/ipilimumab combination [18]. This multicenter phase II trial randomized 309 patients with nccRCC, including 178 patients (57.6%) with papillary RCC, to either nivolumab plus ipilimumab or standard of care (SOC). In the SOC arm of 152 patients, 124 received TKI monotherapy and 17 received ICI–TKI combination therapy. The primary outcome was 12-month overall survival (OS) rate, for which the nivolumab plus ipilimumab arm outperformed the SOC arm (86.9% vs. 76.8%, p = 0.014). However, the OS rates at 6 mo (94.7% vs. 90.0%, p = 0.067), the OS rates at 18 mo (76.6% vs. 69.1%, p = 0.084), and the mOS of 42.4 vs. 33.9 mo did not reach statistical significance (p = 0.292). The ORR was 32.8% in the nivolumab/ipilimumab group vs. 19.6% in the SOC group and 29.2% vs. 21.0% in the papillary RCC subgroup. In the subgroup analysis, for the 26 patients whose tumor had a PD-L1 expression of greater than or equal to 1%, there was a trend towards improved OS, HR 0.56 (95% CI 0.33–0.95).
Outside of ICI–ICI and ICI–TKI combinations, additional ICI combinations have been explored in papillary RCC. These include savolitinib with durvalumab (PD-L1 antibody) in the CALYPSO trial [20], cabozantinib with atezolizumab (PD-L1 antibody) in COSMIC-021 [21], and bevacizumab with atezolizumab in NCT02724878 [22]. Notably, the CALYPSO trial of savolitinib with durvalumab had an overall ORR of 29% with a higher ORR of 53% in the MET-driven subgroup, defined as chromosome 7 gain, MET amplification, MET kinase domain variations, or hepatocyte growth factor amplification. The ongoing phase III SAMETA trial compares savolitinib plus durvalumab with control arms of sunitinib or durvalumab monotherapy in MET-driven papillary RCC [39].
The CAN-I trial (NCT04413123) investigating the triplet cabozantinib, nivolumab, and ipilimumab combination in nccRCC reported data from the first 39 treated patients, with an ORR of 21% and an mPFS of 9.7 mo (95% CI 6.1–12.7) [40]. The Alliance ICONIC trial investigates the same triplet therapy in rare genitourinary cancers, including nccRCC subsets of collecting duct RCC, sarcomatoid RCC, papillary RCC, chromophobe RCC, and renal medullary cancer [41]. The SWOG S2200/PAPMET2 (NCT05411081), randomized trial of cabozantinib with or without atezolizumab for patients with papillary RCC may provide additional insights [42].

2.2.5. Papillary RCC Summary

Though existing trials in papillary RCC are largely single-arm and rely on outdated classification criteria to define papillary RCC, they demonstrate compelling efficacy for the TKI–ICI combinations, including cabozantinib/nivolumab and lenvatinib/pembrolizumab. Additionally, randomized data from the PAPMET trial support the use of cabozantinib monotherapy for this subtype, and the ongoing, randomized, PAPMET2 investigates cabozantinib with or without atezolizumab. Increasing evidence, including the recent SUNNIFORECAST study, highlights ICI–ICI combination therapy as another option in this disease subtype. Efficacy interpretation of ICI-based approaches, especially ICI monotherapy or ICI–ICI combination therapy, is limited by reliance on metrics such as ORR, when it may be better assessed by outcomes such as response duration and overall survival. Future directions for this nccRCC subtype include understanding how to stratify treatment by biomarkers such as MET-driven disease, as well as sequence lines of therapies, given the majority of these studies occurred in the treatment-naïve setting. Ongoing trials in papillary RCC and other nccRCC subtypes are summarized in Table 3.

2.3. Chromophobe RCC

Chromophobe RCC makes up 5% of RCC cases and arises from the collecting duct intercalated cells [47]. It is associated with chromosomal aneuploidy, as well as TP53 and PTEN mutations. In addition, the folliculin gene (FLCN) is mutated in the context of Birt–Hogg–Dube syndrome and the TSC1 or TSC2 genes are mutated in the context of tuberous sclerosis complex [48]. While the majority of chromophobe RCC tumors do not metastasize, they can dedifferentiate into more aggressive subtypes; this process is typically associated with TP53 mutation and whole-genome duplication [49].
No prospective clinical trials have focused solely on chromophobe RCC and, given the relative rarity of this disease, collective nccRCC trials do not selectively test treatments for chromophobe RCC. Of the nccRCC trials with a chromophobe cohort (Table 1), efficacy varies widely, with some demonstrating promising efficacy, while others show no response. Compared to papillary and other nccRCC subtypes, chromophobe RCC patients seem to have higher responses to mTOR inhibition. For example, everolimus monotherapy showed a 33% ORR (2/6 patients) in the ASPEN trial, while lenvatinib and everolimus showed a 44.4% ORR (4/9 patients) in NCT02915783 [7,8].
Response to ICI monotherapy and ICI combination therapy varies widely. With ICI monotherapy, a 9.5% ORR (2/21 patients) to pembrolizumab in KEYNOTE-427, a 28.6% ORR (2/7 patients) to nivolumab in Checkmate 374, and a 7.7% ORR (1/13 patients) to nivolumab in UNISoN was observed [12,50,51]. With ICI–TKI and ICI–ICI combination therapies, no response was seen in CA209-9KU with cabozantinib–nivolumab and only one response (ORR 9.1%) was seen in NCT04413123 with cabozantinib–nivolumab–ipilimumab [16,40]. Conversely, a 28% ORR (8/29 patients) was seen in KEYNOTE-B61 with lenvatinib and pembrolizumab and a 25.9% ORR (7/27 patients) was seen in SUNNIFORECAST with nivolumab–ipilimumab [15,18]. Additional meaningful ICI efficacy endpoints such as duration of response and OS are not yet available.
Several retrospective reviews supplement the paucity of data in prospective trials for this disease and report similar ORR rates to subgroups of prospective trials (see Table 4). A retrospective review by Moussa et al. showed activity for nivolumab–ipilimumab in chromophobe RCC (ORR 25% in 3/12 patients), and a review specific to chromophobe RCC demonstrated responses to this disease with TKI monotherapy (ORR 15%), ICI–TKI combination therapy (30%), and ICI–ICI combination therapy (ORR 12.5%) [52,53]. Two retrospective reviews on cabozantinib use in nccRCC demonstrated ORR rates for chromophobe RCC patients of 30% (3/10 patients) and 17% (1/6 patients) [54,55].
The biology of chromophobe RCC is vastly different from papillary RCC and other nccRCC subtypes and, although some responses have been shown with existing treatment strategies, generally, prognosis is poor for metastatic disease. Recent work on the drivers of tumorigenesis in chromophobe RCC has highlighted the role of both IL-15 and ferroptosis in the disease, and future treatments targeting these pathways could be considered [48].

2.4. Not Otherwise Specified (Also Referred to in the Literature as Unclassified) RCC

RCC-NOS is a diverse group of RCC that do not fit into currently recognized diagnostic categories of RCC. These tumors generally have a heterogeneous morphology/immunohistochemistry (IHC) profile and typically include high-grade RCCs. Pure sarcomatoid morphology, where a differentiated component has been completely effaced, may also be included [58]. These tumors often need liberal sampling and IHC workup to rule out extrarenal and urothelial-origin tumors. With increasing use of molecular studies, this category has become infrequent. In the 2022 WHO classification, unclassified RCC has been renamed to RCC-NOS to emphasize that it is not a distinct subtype [2].
Similar to chromophobe RCC, no prospective clinical trials have included solely RCC-NOS, and retrospective studies mirror the ORRs reported in subgroups of prospective trials [52,54]. Treatment recommendations for this subtype largely mirror that of papillary RCC, with options including ICI–TKI combination strategies or cabozantinib monotherapy. Additionally, the heterogeneity of this subtype may mask ccRCC biology, with one cohort reporting 11.9% of unclassified RCC patients harboring a VHL mutation [59]. Thus, systemic treatment approaches used in ccRCC may be considered, including ICI–ICI combination therapy.

2.5. Translocation RCC

Translocation RCC, or microphthalmia-associated transcription factor family translocation RCC (MiTF-tRCC), encompasses a rare group of diseases involving translocations affecting the short arm of the X chromosomes and fusions of the TFE3, TFEB, or MITF genes [60,61]. In the 2022 WHO classification, this disease subtype has been broken up further into molecularly defined entities including TFE3-rearranged RCC (Xp11 translocation RCC) and TFEB-altered RCC, given the different behaviors of these specific subtypes [2]. TFEB-altered RCC includes TFEB-rearranged RCC and TFEB-amplified RCC. TFEB-amplification RCC was recently described harboring amplification of the 6p21 region with resultant TFEB overexpression [62]. TFEB-amplified RCC occurs in older patients and has a worse outcome. Morphologically, translocation RCC can often mimic other common RCCs such as clear cell and papillary RCC and are, therefore, often misdiagnosed. For example, in IMmotion151, more than 15 tumors classified as ccRCC were found to harbor TFE fusions [63]. Molecular subset clustering from this trial suggests these tumors may have a permissive immune microenvironment; however, significant benefit from immune checkpoint inhibitor-based regimens has not been consistently observed.
As with chromophobe and unclassified RCCs, assessment of response in translocation RCCs is limited to subgroup analysis of nccRCC trials. ICI–TKI combination approaches appear promising, with 4/6 translocation RCC patients responding to lenvatinib plus pembrolizumab in KEYNOTE-B61 and 1/2 translocation RCC patients responding to cabozantinib plus nivolumab in CA209-9KU [15,36], but the small numbers of patients with this subtype limit the generalizability of these findings. Two retrospective reviews of 29 and 22 translocation RCC patients demonstrated ORRs of 5.5% and 14% for ICI–ICI combination therapy and ORRs of 36% and 54% for ICI–TKI combination therapies [56,57]. The phase II NCT03595124 trial is prospectively investigating the activity of nivolumab with or without axitinib specifically in patients with TFE/translocation RCC and is estimated to complete accrual in 2026 [43]. Ongoing investigations into how the fusions that define this nccRCC subtype drive tumorigenesis have highlighted MET and RET kinases as potential pathways for therapeutic targets [64].

2.6. Collecting Duct Carcinoma

Collecting duct carcinoma arises from the distal convoluted tubules and generally presents at advanced or metastatic stages. These tumors can involve mutations in NF2, SETD2, SMARCB1, and CDKN2A [65,66]. Histologically, collecting duct carcinoma is a diagnosis that requires diligent exclusion from another epithelial tumor, urothelial carcinoma, SMARCB1-deficient renal medullary carcinoma (RMC), FH-deficient RCC, and high-grade RCC and is vanishingly rare. Treatments for collecting duct carcinoma traditionally involve chemotherapy, based on biologic similarity to urothelial cancers, with systemic therapy trials summarized in Table 5.
Prospective data for modern chemotherapies include the phase II GETUG study evaluating gemcitabine plus a platinum chemotherapy agent, which found an ORR of 6/23 (26%, 95% CI 8–44%) [67]. The addition of sorafenib (inhibitor of VEGFR1-3, PDGFR, and RAF kinases) to chemotherapy was studied in a single-arm phase 2 study that showed an mPFS of 8.8 mo (95% CI 6.7–10.9 mo) and ORR of 30.8% in 26 patients with collecting duct carcinoma. The phase II BEVABEL-GETUG/AFU24 trial examined the addition of bevacizumab to gemcitabine and platinum chemotherapy and found an ORR of 41.2% (95% CI 25–57%), with an mPFS of 5.9 mo (5.3–9.3 mo) and mOS of 11.1 mo (7.6–24.2 mo) [68].
More recently, the BONSAI trial showed the efficacy of first-line cabozantinib monotherapy, which achieved an ORR of 35% (95% CI 16–57%) and an mPFS of 4 months (95% CI 3–13 mo) in 23 patients with collecting duct carcinoma [69]. Collecting duct carcinoma was excluded from all prospective nccRCC IO–TKI combination trials, and only two patients in the IO–IO CheckMate 920 trial had collecting duct RCC, with one patient having a PR [17]. Nine patients in the SUNNIFORECAST trial evaluating ICI–ICI combination therapy had collecting duct carcinoma, although a subgroup analysis is not yet available [18].
Table 5. Prospective and retrospective data in collecting duct and renal medullary carcinoma.
Table 5. Prospective and retrospective data in collecting duct and renal medullary carcinoma.
StudyStudy Drug(s)nnccRCC Subtype (s)ORR (95% CI)mPFS (95% CI)
Prospective Clinical Trials
BEVABEL-GETUG/
AFU24 [68]
Gemcitabine + platinum chemo + bevacizumab 34Collecting Duct
Renal Medullary
41.2% (25–57%)5.9 mo (5.3–9.3 mo)
GETUG 2007 [67]Gemcitabine + platinum chemo23Collecting Duct26% (8–44%)7.1 mo (3–11.3 mo)
BONSAI [69]Cabozantinib22Collecting Duct35% (16–57%)4 mo (3–31 mo)
BONSAI-2 [70]Nivolumab (Post-cabo)425%PFS 2.8–19.9 mo
NCT02721732 [71]Pembrolizumab (1 front-line)5Renal Medullary0/5 (0%) 8.7 weeks
Retrospective Reviews
Shah et al. [72]
(at any line of therapy)
Carboplatin/
paclitaxel (±bev)
21Renal Medullary7/21
33%
N/A
Gemcitabine/
cisplatin (±bev)
124/12
33%
N/A
Gemcitabine/
doxorubicin (±paclitaxel/bev)
113/11
27.3%
N/A
Gemcitabine/
carboplatin/
paclitaxel
21/2
50%
N/A
Gemcitabine/
cisplatin/
paclitaxel
11/1
100%
N/A
Wilson et al. [73]Gemcitabine + doxorubicin (second-line)16Renal Medullary18.8%2.8 mo (0–6 mo)
Bev—bevacizumab, CI—confidence interval, ICI—immune checkpoint inhibitor, mo—month, TKI—tyrosine kinase inhibitor, N/A—not available, NR—not reached, ORR—overall response rate.

2.7. Renal Medullary Carcinoma

RMC is an aggressive malignancy arising from the collecting duct and is associated with sickle hemoglobinopathies. In the latest 2022 WHO classification system, RMC is now a molecularly defined nccRCC subtype characterized by loss of SMARCB1 expression [74]. Patients with RMC have been largely excluded from nccRCC trials, and RMCs generally have poor response to treatments effective for other RCCs such as VEGFR TKIs [72,75]. Trial and review data for RMC are summarized in Table 5. Based on available data, platinum-based chemotherapy with or without bevacizumab is the mainstay first-line systemic therapy, though responses are generally not durable and outcomes are poor across all stages of disease [68,72].
A phase II basket trial evaluating pembrolizumab in rare cancers included five patients with RMC, none of whom had a significant response [71]. Ongoing trials are investigating ICI-based combination therapies, including NCT05347212 with nivolumab plus relatlimab (LAG3 antibody) in RMC and NCT05286801 with tiragolumab (TIGIT antibody) plus atezolizumab in SMARCB1- or SMARCA4-deficient RCC [44,45].

2.8. Fumarate-Hydratase-Deficient RCC and Other Molecularly Defined nccRCC Subtypes

A molecularly defined subtype of nccRCC is FH-deficient RCC, which is driven by a sporadic or hereditary FH mutation [76]. A hereditary form of the disease can be associated with hereditary leiomyomatosis and renal cell cancer (HLRCC) and includes a subgroup of what was previously defined as type 2 papillary RCCs.
A phase II study of bevacizumab and erlotinib (epidermal growth factor receptor (EGFR) inhibitor) that enriched for patients with FH-deficient RCCs (either HLRCC or papillary RCC) reported an overall ORR of 51% (42/83 patients, CI 40–61%) and an HLRCC cohort ORR of 64% (27/42 patients, 95% CI 49–77%) [77]. Biochemically, FH-deficient tumors have accumulation of hypoxia-inducible factor (HIF) that can upregulate VEGF and be stimulated by EGFR pathways, leading to trials studying both EGFR and VEGF blockade for patients with these tumors [76]. Ongoing trials exploring expansion upon this dual blockade strategy include NCT04981509 examining bevacizumab, erlotinib, and atezolizumab for patients with HLRCC or papillary RCC [46].
Regarding ICI combination therapies, a phase II trial of the poly ADP ribose polymerase inhibitor (PARPi) talazoparib combined with avelumab (PD-L1 antibody) in genomically defined metastatic kidney cancer reported an ORR of 0% in a cohort of eight patients that included four FH-deficient RCC and one SDH-deficient RCC [78]. Of note, six had previously received ICI in prior lines of therapy. Recently, encouraging phase II data were presented on the ICI–TKI combination of lenvatinib plus tislelizumab (PD-1 antibody) in FH-deficient RCC tumors, which showed an ORR of 93.3% (14/15 patients), including CRs in 3 of 15 patients (20%) [79]. These approaches highlight the importance of ongoing studies for ICI combination strategies in this molecular subtype.
Emphasizing the value of thorough molecular characterization and consideration of additional molecular-targeting approaches for rare subtypes of nccRCC, Pal et al. highlight three case reports of ALK-rearranged RCC having excellent responses to the ALK kinase inhibitor alectinib, even after multiple other lines of other systemic therapies [80]. Thorough molecular characterization of tissue samples and/or tumor cell free DNA (cfDNA) provides additional opportunities to identify targeted approaches to treating patients with these rare cancers, especially with an increasing number of tumor-agnostic predictive molecular biomarkers for specific systemic therapies [81].

2.9. Sarcomatoid Differentiation and PD-L1 Status

The presence of sarcomatoid differentiation may be a useful predictive biomarker of ICI response in patients with ccRCC [82]. Several ICI monotherapy and ICI combination studies in nccRCC also provide subgroup outcomes for patients with sarcomatoid differentiation and PD-L1 expression ≥ 1% (Table 6). Just as with ccRCC, sarcomatoid differentiation appears to be a useful biomarker for increased responsiveness to ICI-based therapies. However, no generalizable statements can be taken from these single-arm studies with multiple layers of heterogeneity (e.g., varying histologic subtypes and simultaneous TKI treatments). PD-L1 positivity (PD-L1 ≥ 1%) may also predict improved immunotherapy responses in nccRCC subtypes and, of particular interest, was associated with improved OS for patients treated with nivolumab–ipilimumab in the SUNNIFORECAST trial (HR 0.56, 95% CI 0.33–0.95). Future studies in nccRCC subtypes should continue to investigate these biomarkers when including ICI-based interventions.

3. Discussion

Non-clear cell RCC encompasses a diverse set of malignancies with varying response to different classes and combinations of systemic therapies. The diversity of these diseases may not be adequately reflected in clinical guidelines. For example, the current NCCN guidelines do not differentiate among nccRCC subtypes for preferred systemic therapy regimens, which include clinical trial, cabozantinib, cabozantinib with nivolumab, or lenvatinib with pembrolizumab [83]. Evidence for these recommendations mainly comes from single-arm studies where the largest enrolling subtype is papillary RCC. The rarity of most nccRCC subtypes make clinical trial design in these diseases challenging, particularly in the locally advanced or metastatic setting, where systemic therapies are frequently warranted. However, more precise nccRCC classification, especially molecularly defined disease subtypes, opens the doors to an increasing number of collaborative multicenter and innovative trial designs that have shown benefit across a variety of rare cancers [84]. The nuances of nccRCC subtypes discussed across this review, in particular the unique biology of subtypes such as collecting duct carcinoma and renal medullary carcinoma, argue against systemic treatment guidelines aimed to encompass all nccRCC subtypes.
Acknowledging the limitations of available evidence, TKI–ICI combinations (pembrolizumab with lenvatinib or nivolumab with cabozantinib) or cabozantinib monotherapy present the most compelling evidence for first-line treatment in papillary RCC. The randomized PAPMET trial of papillary RCC patients demonstrated a benefit of cabozantinib over sunitinib, and the ongoing randomized PAPMET2 trial is exploring the benefit of cabozantinib/atezolizumab when compared to cabozantinib monotherapy. Systemic treatment options for chromophobe RCC, unclassified RCC, and translocation RCC are largely similar to papillary RCC, though evidence is limited to subgroup analyses of nccRCC trials and retrospective reviews. While everolimus-based treatments have shown encouraging activity in patients with chromophobe RCC, additional insights into the unique biology of this disease may present opportunities to expand treatment options. Similarly, for translocation RCC, ongoing investigations into the molecular fusions that drive this disease may open up novel treatment targets. The ICI–ICI combination of nivolumab with ipilimumab has growing evidence for use across specific nccRCC subtypes (papillary, chromophobe, unclassified, and translocation RCC), and the potential for durable responses may not be adequately reflected in commonly reported efficacy outcomes such as ORR. Long-term follow-up for meaningful ICI efficacy endpoints, such as duration of response and overall survival, may shed additional light.
Available evidence for managing collecting duct RCC and RMC is less robust and comes from small prospective trials and retrospective reports. Based on these data, an effective treatment in both subtypes includes platinum-based chemotherapy combinations with consideration for the addition of bevacizumab. In collecting duct RCC, there are prospective data to consider cabozantinib monotherapy as an alternative treatment option. Additional subgroup analyses of ongoing and future studies will demonstrate whether ICI combination strategies are effective in these diseases. For RMC patients, classes of systemic treatments outside of chemotherapy such as ICIs and TKIs have not yet demonstrated effectiveness, though ongoing trials are investigating novel combinations of these therapies.
Finally, the 2022 WHO classification of RCC added additional molecular-based subtypes that may expand precision oncology approaches for nccRCC systemic therapies. Joint EGFR and VEFR inhibition with bevacizumab and erlotinib in FH-deficient RCC has proven a promising approach, and ICI combinations appear effective in this disease subtype as well. Tumor-agnostic predictive biomarkers may provide additional treatment options for nccRCC patients, highlighting the importance of thorough molecular testing for these tumors. Ongoing studies in nccRCC have begun to align with molecular characteristics of specific nccRCC subtypes, such as MET-driven papillary RCC.

4. Conclusions

Currently available clinical trial data highlight ICI–TKI combination treatment (lenvatinib plus pembrolizumab or cabozantinib plus nivolumab) or cabozantinib monotherapy as the systemic therapy options with the strongest evidence for papillary RCC, unclassified RCC, chromophobe, and translocation RCC. A growing body of literature highlights that ICI–ICI combination therapies may be considered in these nccRCC subtypes. Additionally, chromophobe RCC may benefit from everolimus-based approaches. For both chromophobe RCC and translocation RCC, translational discoveries into their unique biologies are opening up new targets for future treatments. Platinum-based chemotherapy combinations remain the best available systemic therapies for collecting duct RCC and RMC; cabozantinib monotherapy may be an additional option for collecting duct RCC. Thorough molecular characterization of nccRCC tumors is essential for evaluating potential precision oncology treatment approaches.

Author Contributions

Conceptualization, J.V. and Q.Q.; data curation, J.V.; writing—original draft preparation, J.V.; writing—review and editing, J.V., T.Z., P.K., H.H., J.B., and Q.Q. All authors have read and agreed to the published version of the manuscript.

Funding

National Cancer Institute (NCI) Cancer Center Support Grant (P30CA142543).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

Joseph Vento: none; Tian Zhang: PI/research funding to institution—Merck, Janssen, Astra Zeneca, Pfizer, Astellas, Eli Lilly, Tempus, ALX Oncology, Janux Therapeutics, OncoC4, Exelixis, Bayer; Advisory Board—Merck, Exelixis, Sanofi-Aventis, Janssen, Astra Zeneca, Pfizer, Amgen, BMS, Eisai, Aveo, Eli Lilly, Bayer, Gilead, Novartis, EMD Serono, Dendreon, Xencor; Consultant—Pfizer, MJH Associates, Vaniam, Aptitude Health, PeerView, Aravive, Dava Oncology; Payal Kapur: none; Hans Hammers: Research Funding—Bristol-Myers Squibb, Merck, Surface Oncology, NGM Biopharmaceuticals, Aveo, Hibercell, NiKang Therapeutics, Exelixis, Werewolf Therapeutics; Honoraria—Bristol-Myers Squibb; Consulting—Bristol-Myers Squibb, Pfizer, Exelixis, Bayer Novartis, Merck, ARMO BioSciences, Corvus Pharmaceuticals, Surface Oncology, Eli Lilly; Travel—Bristol-Myers Squibb, Merck, Pfizer, Eli Lilly, Novartis; James Brugarolas: Research Funding—SPORE award (NIH/NCI P50CA196516); Consultant—Regeneron Pharmaceuticals; Qian (Janie) Qin: Consulting—Exelixis, Eisai, Janssen; Honorarium—JH Life Sciences, DAVA Oncology, Mashup Media.

Abbreviations

The following abbreviations are used in this manuscript:
ALKAnaplastic lymphoma kinase
ccRCCClear cell renal cell carcinoma
CIConfidence interval
EGFREpidermal growth factor receptor
FHFumarate hydratase
HRHazard ratio
ICIImmune checkpoint inhibitor
IHCImmunohistochemistry
mOSMedian overall survival
mPFSMedian progression-free survival
mTORMammalian target of rapamycin
nccRCCNon-clear cell renal cell carcinoma
ORROverall response rate
OSOverall survival
PARPiPoly ADP ribose polymerase inhibitor
RCCRenal cell carcinoma
SDHSuccinate Dehydrogenase
SOCStandard of Care
TKITyrosine kinase inhibitor
VEGFRVascular endothelial growth factor receptor
WHOWorld Health Organization

References

  1. Rose, T.L.; Kim, W.Y. Renal Cell Carcinoma: A Review. J. Am. Med. Assoc. 2024, 332, 1001–1010. [Google Scholar] [CrossRef] [PubMed]
  2. Goswami, P.R.; Singh, G.; Patel, T.; Dave, R. The WHO 2022 Classification of Renal Neoplasms (5th Edition): Salient Updates. Cureus 2022, 16, e58470. [Google Scholar] [CrossRef]
  3. Chen, Y.W.; Wang, L.; Panian, J.; Dhanji, S.; Derweesh, I.; Rose, B.; Bagrodia, A.; McKay, R.R. Treatment Landscape of Renal Cell Carcinoma. Curr. Treat. Options Oncol. 2023, 24, 1889–1916. [Google Scholar] [CrossRef] [PubMed]
  4. Negrier, S.; Rioux-Leclercq, N.; Ferlay, C.; Gross-Goupil, M.; Gravis, G.; Geoffrois, L.; Chevreau, C.; Boyle, H.; Rolland, F.; Blanc, E.; et al. Axitinib in first-line for patients with metastatic papillary renal cell carcinoma: Results of the multicentre, open-label, single-arm, phase II AXIPAP trial. Eur. J. Cancer 2020, 129, 107–116. [Google Scholar] [CrossRef]
  5. Pal, S.K.; Tangen, C.; Thompson, I.M.; Balzer-Haas, N.; George, D.J.; Heng, D.Y.C.; Shuch, B.; Stein, M.; Tretiakova, M.; Humphrey, P.; et al. A comparison of sunitinib with cabozantinib, crizotinib, and savolitinib for treatment of advanced papillary renal cell carcinoma: A randomised, open-label, phase 2 trial. Lancet 2021, 397, 695–703. [Google Scholar] [CrossRef]
  6. Tannir, N.M.; Jonasch, E.; Albiges, L.; Altinmakas, E.; Ng, C.S.; Matin, S.F.; Wang, X.; Qiao, W.; Dubauskas Lim, Z.; Tamboli, P.; 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, 866–874. [Google Scholar] [CrossRef]
  7. Armstrong, A.J.; Halabi, S.; Eisen, T.; Broderick, S.; Stadler, W.M.; Jones, R.J.; Garcia, J.A.; Vaishampayan, U.N.; Picus, J.; Hawkins, R.E.; 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, 378–388. [Google Scholar] [CrossRef]
  8. Hutson, T.E.; Michaelson, M.D.; Kuzel, T.M.; Agarwal, N.; Molina, A.M.; Hsieh, J.J.; Vaishampayan, U.N.; Xie, R.; Bapat, U.; Ye, W.; et al. A Single-arm, Multicenter, Phase 2 Study of Lenvatinib Plus Everolimus in Patients with Advanced Non-Clear Cell Renal Cell Carcinoma. Eur. Urol. 2021, 80, 162–170. [Google Scholar] [CrossRef]
  9. Voss, M.H.; Molina, A.M.; Chen, Y.B.; Woo, K.M.; Chaim, J.L.; Coskey, D.T.; Redzematovic, A.; Wang, P.; Lee, W.; Selcuklu, S.D.; et al. Phase II Trial and Correlative Genomic Analysis of Everolimus Plus Bevacizumab in Advanced Non–Clear Cell Renal Cell Carcinoma. J. Clin. Oncol. 2016, 34, 3846–3853. [Google Scholar] [CrossRef]
  10. Pazopanib for the Treatment of Non-clear Cell Renal Cell Carcinoma: A Single-Arm, Open-Label, Multicenter, Phase II Study. Available online: https://www.e-crt.org/journal/view.php?doi=10.4143/crt.2016.584 (accessed on 3 March 2025).
  11. Barata, P.C.; Chehrazi-Raffle, A.; Allman, K.D.; Asnis-Alibozek, A.; Kasturi, V.; Pal, S.K. Activity of Tivozanib in Non-clear Cell Renal Cell Carcinoma: Subgroup Analysis from a Phase II Randomized Discontinuation Trial. Oncologist 2023, 28, 894–900. [Google Scholar] [CrossRef]
  12. McDermott, D.F.; Lee, J.L.; Ziobro, M.; Suarez, C.; Langiewicz, P.; Matveev, V.B.; Wiechno, P.; Gafanov, R.A.; Tomczak, P.; Pouliot, F.; et al. Open-Label, Single-Arm, Phase II Study of Pembrolizumab Monotherapy as First-Line Therapy in Patients with Advanced Non-Clear Cell Renal Cell Carcinoma. J. Clin. Oncol. 2021, 39, 1029–1039. [Google Scholar] [CrossRef] [PubMed]
  13. Vogelzang, N.J.; Olsen, M.R.; McFarlane, J.J.; Arrowsmith, E.; Bauer, T.M.; Jain, R.K.; Somer, B.; Lam, E.T.; Kochenderfer, M.D.; Molina, A.; 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, 18, 461–468.e3. [Google Scholar] [CrossRef] [PubMed]
  14. Conduit, C.; Davis, I.D.; Goh, J.C.; Kichenadasse, G.; Gurney, H.; Harris, C.A.; Pook, D.; Krieger, L.; Parnis, F.; Underhill, C.; et al. A phase II trial of nivolumab followed by ipilimumab and nivolumab in advanced non-clear-cell renal cell carcinoma. BJU Int. 2024, 133 (Suppl. S3), 57–67. [Google Scholar] [CrossRef] [PubMed]
  15. Albiges, L.; Gurney, H.; Atduev, V.; Suarez, C.; Climent, M.A.; Pook, D.; Tomczak, P.; Barthelemy, P.; Lee, J.L.; Stus, V.; et al. Pembrolizumab plus lenvatinib as first-line therapy for advanced non-clear-cell renal cell carcinoma (KEYNOTE-B61): A single-arm, multicentre, phase 2 trial. Lancet Oncol. 2023, 24, 881–891. [Google Scholar] [CrossRef]
  16. Lee, C.H.; Voss, M.H.; Carlo, M.I.; Chen, Y.B.; Zucker, M.; Knezevic, A.; Lefkowitz, R.A.; Shapnik, N.; Dadoun, C.; Reznik, E.; et al. Phase II Trial of Cabozantinib Plus Nivolumab in Patients with Non-Clear-Cell Renal Cell Carcinoma and Genomic Correlates. J. Clin. Oncol. 2022, 40, 2333–2341. [Google Scholar] [CrossRef]
  17. Tykodi, S.S.; Gordan, L.N.; Alter, R.S.; Arrowsmith, E.; Harrison, M.R.; Percent, I.; Singal, R.; Veldhuizen, P.V.; George, D.J.; Hutson, T.; et al. Safety and efficacy of nivolumab plus ipilimumab in patients with advanced non-clear cell renal cell carcinoma: Results from the phase 3b/4 CheckMate 920 trial. J. Immunother. Cancer 2022, 10, e003844. [Google Scholar] [CrossRef]
  18. Bergmann, L.; Ahrens, M.; Albiges, L.; Goupil, M.G.; Boleti, E.; Gravis, G.; Flechon, A.; Grimm, M.O.J.; Rausch, S.; Barthelemy, P.; et al. LBA75 Prospective randomised phase-II trial of ipilimumab/nivolumab versus standard of care in non-clear cell renal cell cancer: Results of the SUNNIFORECAST trial. Ann. Oncol. 2024, 35, S1263. [Google Scholar] [CrossRef]
  19. McGregor, B.A.; Huang, J.; Xie, W.; Xu, W.; Bilen, M.A.; Braun, D.A.; Zhang, T.; McKay, R.R.; McDermott, D.F.; Hammers, H.J.; et al. Phase II study of cabozantinib (Cabo) with nivolumab (Nivo) and ipilimumab (Ipi) in advanced renal cell carcinoma with variant histologies (RCCvh). J. Clin. Oncol. 2023, 41 (Suppl. S16), 4520. [Google Scholar] [CrossRef]
  20. Suárez, C.; Larkin, J.M.G.; Patel, P.; Valderrama, B.P.; Rodriguez-Vida, A.; Glen, H.; Thistlethwaite, F.; Ralph, C.; Srinivasan, G.; Mendez-Vidal, M.J.; et al. Phase II Study Investigating the Safety and Efficacy of Savolitinib and Durvalumab in Metastatic Papillary Renal Cancer (CALYPSO). J. Clin. Oncol. 2023, 41, 2493–2502. [Google Scholar] [CrossRef]
  21. Pal, S.K.; McGregor, B.; Suárez, C.; Tsao, C.K.; Kelly, W.; Vaishampayan, U.; Pagliaro, L.; Maughan, B.L.; Loriot, Y.; Castellano, D.; et al. Cabozantinib in Combination with Atezolizumab for Advanced Renal Cell Carcinoma: Results From the COSMIC-021 Study. J. Clin. Oncol. 2021, 39, 3725–3736. [Google Scholar] [CrossRef]
  22. McGregor, B.A.; McKay, R.R.; Braun, D.A.; Werner, L.; Gray, K.; Flaifel, A.; Signoretti, S.; Hirsch, M.S.; Steinharter, J.A.; Bakouny, Z.; 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, 63–70. [Google Scholar] [CrossRef] [PubMed]
  23. Franczyk, B.; Rysz, J.; Ławiński, J.; Ciałkowska-Rysz, A.; Gluba-Brzózka, A. Cardiotoxicity of Selected Vascular Endothelial Growth Factor Receptor Tyrosine Kinase Inhibitors in Patients with Renal Cell Carcinoma. Biomedicines 2023, 11, 181. [Google Scholar] [CrossRef] [PubMed]
  24. Li, H.; Huntington, S.; Gross, C.; Wang, S.Y. Immunotherapy utilization patterns in patients with advanced cancer and autoimmune disease. PLoS ONE 2024, 19, e0300789. [Google Scholar] [CrossRef]
  25. Motzer, R.; Porta, C.; Alekseev, B.; Rha, S.Y.; Choueiri, T.K.; Mendez-Vidal, M.J.; Hong, S.H.; Kapoor, A.; Goh, J.C.; Eto, M.; et al. Health-related quality-of-life outcomes in patients with advanced renal cell carcinoma treated with lenvatinib plus pembrolizumab or everolimus versus sunitinib (CLEAR): A randomised, phase 3 study. Lancet Oncol. 2022, 23, 768–780. [Google Scholar] [CrossRef] [PubMed]
  26. Raj, R.K.; Upadhyay, R.; Wang, S.J.; Singer, E.A.; Dason, S. Incorporating Stereotactic Ablative Radiotherapy into the Multidisciplinary Management of Renal Cell Carcinoma. Curr. Oncol. 2023, 30, 10283–10298. [Google Scholar] [CrossRef]
  27. Lobo, J.; Ohashi, R.; Amin, M.B.; Berney, D.M.; Compérat, E.M.; Cree, I.A.; Gill, A.J.; Hartmann, A.; Menon, S.; Netto, G.J.; et al. WHO 2022 landscape of papillary and chromophobe renal cell carcinoma. Histopathology 2022, 81, 426–438. [Google Scholar] [CrossRef]
  28. The Cancer Genome Atlas Research Network. Comprehensive Molecular Characterization of Papillary Renal-Cell Carcinoma. N. Engl. J. Med. 2016, 374, 135–145. [Google Scholar] [CrossRef]
  29. Stewart, G.D.; Uzzo, R.G.; Choueiri, T.K. 2nd Wuof/Siu Icud on Kidney Cancer. In Wiuf/Siu/Icud; World Urologic Oncology Federation: Seville, Spain, 2022. [Google Scholar]
  30. Khurshid, H. Role of artificial intelligence in cancer diagnosis and treatment: Current trends and future directions. Glob. J. Basic Sci. 2024, 1, 1991. [Google Scholar]
  31. Wang, Q.; Zhang, Y.; Zhang, B.; Fu, Y.; Zhao, X.; Zhang, J.; Zuo, K.; Xing, Y.; Jiang, S.; Qin, Z.; et al. Single-cell chromatin accessibility landscape in kidney identifies additional cell-of-origin in heterogenous papillary renal cell carcinoma. Nat. Commun. 2022, 13, 31. [Google Scholar] [CrossRef]
  32. National Cancer Institute (NCI). A Phase II Randomized Trial of Cabozantinib (NSC #761968) With or Without Atezolizumab (NSC #783608) in Patients with Advanced Papillary Renal Cell Carcinoma (PAPMET2). 2025. Available online: https://clinicaltrials.gov/study/NCT05411081 (accessed on 17 March 2025).
  33. Ravaud, A.; Oudard, S.; De Fromont, M.; Chevreau, C.; Gravis, G.; Zanetta, S.; Theodore, C.; Jimenez, M.; Sevin, E.; Laguerre, B.; 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). Ann. Oncol. 2015, 26, 1123–1128. [Google Scholar] [CrossRef]
  34. Harris, C.A.; Link, E.; Goh, J.C.; Parnis, F.; Gurney, H.; Kichenadasse, G.; Underhill, C.; Torres, J.; Roncolato, F.; Inderjeeth, A.J.; et al. UNICAB: Cabozantinib in locally advanced or metastatic non-clear cell renal cell carcinoma post immunotherapy or in those unsuitable for immunotherapy (ANZUP 1802). J. Clin. Oncol. 2024, 42, 428. [Google Scholar] [CrossRef]
  35. Motzer, R.; Alekseev, B.; Rha, S.Y.; Porta, C.; Eto, M.; Powles, T.; Grünwald, V.; Hutson, T.E.; Kopyltsov, E.; Méndez-Vidal, M.J.; et al. Lenvatinib plus Pembrolizumab or Everolimus for Advanced Renal Cell Carcinoma. N. Engl. J. Med. 2021, 384, 1289–1300. [Google Scholar] [CrossRef] [PubMed]
  36. Fitzgerald, K.N.; Lee, C.H.; Voss, M.H.; Carlo, M.I.; Knezevic, A.; Peralta, L.; Chen, Y.; Lefkowitz, R.A.; Shah, N.J.; Owens, C.N.; et al. Cabozantinib Plus Nivolumab in Patients with Non–Clear Cell Renal Cell Carcinoma: Updated Results from a Phase 2 Trial. Eur. Urol. 2024, 86, 90–94. [Google Scholar] [CrossRef] [PubMed]
  37. Choueiri, T.K.; Powles, T.; Burotto, M.; Escudier, B.; Bourlon, M.T.; Zurawski, B.; Juárez, V.M.O.; Hsieh, J.J.; Basso, U.; Shah, A.Y.; et al. Nivolumab plus Cabozantinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2021, 384, 829–841. [Google Scholar] [CrossRef]
  38. Motzer, R.J.; Tannir, N.M.; McDermott, D.F.; Frontera, O.A.; Melichar, B.; Choueiri, T.K.; Plimack, E.R.; Barthélémy, P.; Porta, C.; George, S.; et al. Nivolumab plus Ipilimumab versus Sunitinib in Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2018, 378, 1277–1290. [Google Scholar] [CrossRef]
  39. Choueiri, T.K.; Xu, W.; Poole, L.; Telaranta-Keerie, A. SAMETA: An open-label, three-arm, multicenter, phase III study of savolitinib + durvalumab versus sunitinib and durvalumab monotherapy in patients with MET-driven, unresectable, locally advanced/metastatic papillary renal cell carcinoma (PRCC). J. Clin. Oncol. 2022, 40, 4601. [Google Scholar] [CrossRef]
  40. McGregor, B.A.; Paul, M.; Xie, W.; Xu, W.; Bilen, M.A.; Braun, D.A.; Berg, S.; Zhang, T.; McKay, R.R.; McDermott, D.F.; et al. 1702P Updated Results of Phase II Study of Cabozantinib (Cabo) with Nivolumab (Nivo) and Ipilimumab (Ipi) in Advanced Renal Cell Carcinoma with Divergent Histologies (RCCdh)–Annals of Oncology. Available online: https://www.annalsofoncology.org/article/S0923-7534(24)03314-3/fulltext (accessed on 18 March 2025).
  41. National Cancer Institute (NCI). A Phase II Study of Ipilimumab, Cabozantinib, and Nivolumab in Rare Genitourinary Cancers (ICONIC). 2025. Available online: https://clinicaltrials.gov/study/NCT03866382 (accessed on 3 March 2025).
  42. Maughan, B.L.; Plets, M.; Pal, S.K.; Ged, Y.; Tangen, C.; Vaishampayan, U.N.; Lerner, S.P.; Thompson, I.M. SWOG S2200 (PAPMET2): A phase II randomized trial of cabozantinib with or without atezolizumab in patients with advanced papillary renal cell carcinoma (PRCC). J. Clin. Oncol. 2024, 42 (Suppl. S4), TPS493. [Google Scholar] [CrossRef]
  43. National Cancer Institute (NCI). A Randomized Phase 2 Trial of Axitinib/Nivolumab Combination Therapy vs. Single Agent Nivolumab for the Treatment of TFE/Translocation Renal Cell Carcinoma (tRCC) Across All Age Groups. 2025. Available online: https://clinicaltrials.gov/study/NCT03595124 (accessed on 4 March 2025).
  44. 44. M.D. Anderson Cancer Center. Phase II Trial of Immunotherapy in Patients with Carcinomas Arising from the Renal Medulla. 2024. Available online: https://clinicaltrials.gov/study/NCT05347212 (accessed on 22 April 2025).
  45. National Cancer Institute (NCI). A Phase 1/2 Study of Tiragolumab (NSC# 827799) and Atezolizumab (NSC# 783608) in Patients with Relapsed or Refractory SMARCB1 or SMARCA4 Deficient Tumors. 2025. Available online: https://clinicaltrials.gov/study/NCT05286801 (accessed on 22 April 2025).
  46. National Cancer Institute (NCI). A Phase 2 Study of Bevacizumab, Erlotinib and Atezolizumab in Subjects with Advanced Hereditary Leiomyomatosis and Renal Cell Cancer (HLRCC) Associated or Sporadic Papillary Renal Cell Cancer. 2025. Available online: https://clinicaltrials.gov/study/NCT04981509 (accessed on 22 April 2025).
  47. Garje, R.; Elhag, D.; Yasin, H.A.; Acharya, L.; Vaena, D.; Dahmoush, L. Comprehensive review of chromophobe renal cell carcinoma. Crit. Rev. Oncol. Hematol. 2021, 160, 103287. [Google Scholar] [CrossRef]
  48. Henske, E.P.; Cheng, L.; Hakimi, A.A.; Choueiri, T.K.; Braun, D.A. Chromophobe renal cell carcinoma. Cancer Cell 2023, 41, 1383–1388. [Google Scholar] [CrossRef]
  49. Kapur, P.; Zhong, H.; Le, D.; Mukhopadhyay, R.; Miyata, J.; Carrillo, D.; Rakheja, D.; Rajaram, S.; Durinck, S.; Modrusan, Z.; et al. Molecular underpinnings of dedifferentiation and aggressiveness in chromophobe renal cell carcinoma. JCI Insight 2024, 9, e176743. [Google Scholar] [CrossRef]
  50. McFarlane, J.J.; Kochenderfer, M.D.; Olsen, M.R.; Bauer, T.M.; Molina, A.; Hauke, R.J.; Reeves, J.A.; Babu, S.; Veldhuizen, P.V.; Somer, B.; et al. Safety and Efficacy of Nivolumab in Patients with Advanced Clear Cell Renal Cell Carcinoma: Results from the Phase IIIb/IV CheckMate 374 Study. Clin. Genitourin. Cancer 2020, 18, 469–476.e4. [Google Scholar] [CrossRef] [PubMed]
  51. Conduit, C.; Kichenadasse, G.; Harris, C.A.; Gurney, H.; Ferguson, T.; Parnis, F.; Goh, J.C.; Morris, M.F.; Underhill, C.; Pook, D.W.; et al. Sequential immunotherapy in rare variant renal cell carcinomafinal report of UNISoN (ANZUP 1602): Nivolumab then ipilimumab + nivolumab in advanced nonclear cell renal cell carcinoma. J. Clin. Oncol. 2022, 40 (Suppl. S16), 4537. [Google Scholar] [CrossRef]
  52. Moussa, M.J.; Khandelwal, J.; Wilson, N.R.; Malikayil, K.L.; Surasi, D.S.; Bathala, T.K.; Lin, Y.; Rao, P.; Tamboli, P.; Sircar, K.; et al. Efficacy and safety of nivolumab plus ipilimumab in patients with metastatic variant histology (non-clear cell) renal cell carcinoma. J. Immunother. Cancer 2025, 13, e010958. [Google Scholar] [CrossRef] [PubMed]
  53. Paffenholz, P.; Casuscelli, J.; Zschaebitz, S.; Rinderknecht, E.; Mattigk, A.; Volk, A.L.; Ivanyi, P.; Hilser, T.; Darr, C.; Flegar, L.; et al. Real world evidence from a retrospective multi-center analysis on first-line therapy for metastatic renal cell carcinoma with chromophobe histology. J. Clin. Oncol. 2025, 43 (Suppl. S5), 488. [Google Scholar] [CrossRef]
  54. Martínez Chanzá, N.; Xie, W.; Asim Bilen, M.; Dzimitrowicz, H.; Burkart, J.; Geynisman, D.M.; Balakrishnan, A.; Bowman, I.A.; Jain, R.; Stadler, W.; et al. Cabozantinib in advanced non-clear-cell renal cell carcinoma: A multicentre, retrospective, cohort study. Lancet Oncol. 2019, 20, 581–590. [Google Scholar] [CrossRef]
  55. Campbell, M.T.; Bilen, M.A.; Shah, A.Y.; Lemke, E.; Jonasch, E.; Venkatesan, A.M.; Altinmakas, E.; Duran, C.; Msaouel, P.; Tannir, N.M. Cabozantinib for the treatment of patients with metastatic non-clear cell renal cell carcinoma: A retrospective analysis. Eur. J. Cancer 2018, 104, 188–194. [Google Scholar] [CrossRef]
  56. Alhalabi, O.; Thouvenin, J.; Négrier, S.; Vano, Y.A.; Campedel, L.; Hasanov, E.; Bakouny, Z.; Hahn, A.W.; Bilen, M.A.; Msaouel, P.; et al. Immune Checkpoint Therapy Combinations in Adult Advanced MiT Family Translocation Renal Cell Carcinomas. Oncologist 2023, 28, 433–439. [Google Scholar] [CrossRef]
  57. Ged, Y.; Touma, A.; Meza Contreras, L.; Elias, R.; Van Galen, J.; Cupo, O.; Baraban, E.; Singla, N.; Lee, C.H.; Pal, S.; et al. Multi-institutional Analysis of Immune-Oncology Combination Therapy for Metastatic MiT Family Translocation Renal Cell Carcinoma. J. Immunother. 2025, 48, 113. [Google Scholar] [CrossRef]
  58. Akgul, M.; Cheng, L. Immunophenotypic and pathologic heterogeneity of unclassified renal cell carcinoma: A study of 300 cases. Hum. Pathol. 2020, 102, 70–78. [Google Scholar] [CrossRef]
  59. Yekeduz, E.; Braun, D.A.; El Hajj Chehade, R.; Eid, M.; Labaki, C.; Machaalani, M.; Nassar, A.; Nawfal, R.; Saad, E.; Saliby, R.M.; et al. Applying genomic analysis to refine unclassified renal cell carcinoma. J. Clin. Oncol. 2024, 42 (Suppl. S16), 4551. [Google Scholar] [CrossRef]
  60. Ged, Y.; Feinaj, A.; Baraban, E.; Singla, N. Management of translocation carcinomas of the kidney. Trans. Cancer Res. 2024, 13, 11. [Google Scholar] [CrossRef] [PubMed]
  61. Durinck, S.; Stawiski, E.W.; Pavía-Jiménez, A.; Modrusan, Z.; Kapur, P.; Jaiswal, B.S.; Zhang, N.; Toffessi-Tcheuyap, V.; Nguyen, T.T.; Pahuja, K.B.; et al. Spectrum of diverse genomic alterations define non–clear cell renal carcinoma subtypes. Nat. Genet. 2015, 47, 13–21. [Google Scholar] [CrossRef] [PubMed]
  62. Gupta, S.; Johnson, S.H.; Vasmatzis, G.; Porath, B.; Rustin, J.G.; Rao, P.; Costello, B.A.; Leibovich, B.C.; Thompson, R.H.; Cheville, J.C.; et al. TFEB-VEGFA (6p21.1) co-amplified renal cell carcinoma: A distinct entity with potential implications for clinical management. Mod. Pathol. 2017, 30, 998–1012. [Google Scholar] [CrossRef] [PubMed]
  63. Motzer, R.J.; Banchereau, R.; Hamidi, H.; Powles, T.; McDermott, D.; Atkins, M.B.; Escudier, B.; Liu, L.F.; Leng, N.; Abbas, A.R.; et al. Molecular Subsets in Renal Cancer Determine Outcome to Checkpoint and Angiogenesis Blockade. Cancer Cell 2020, 38, 803–817.e4. [Google Scholar] [CrossRef]
  64. Prakasam, G.; Mishra, A.; Christie, A.; Miyata, J.; Carrillo, D.; Tcheuyap, V.T.; Ye, H.; Do, Q.N.; Wang, Y.; Torras, O.R.; et al. Comparative genomics incorporating translocation renal cell carcinoma mouse model reveals molecular mechanisms of tumorigenesis. J. Clin. Investig. 2024, 134, e170559. [Google Scholar] [CrossRef]
  65. Cabanillas, G.; Montoya-Cerrillo, D.; Kryvenko, O.N.; Pal, S.K.; Arias-Stella, J.A. Collecting duct carcinoma of the kidney: Diagnosis and implications for management. Urol. Oncol. Semin. Orig. Investig. 2022, 40, 525–536. [Google Scholar] [CrossRef]
  66. Pal, S.K.; Choueiri, T.K.; Wang, K.; Khaira, D.; Karam, J.A.; Van Allen, E.; Palma, N.A.; Stein, M.N.; Johnson, A.; Squillace, R.; et al. Characterization of Clinical Cases of Collecting Duct Carcinoma of the Kidney Assessed by Comprehensive Genomic Profiling. Eur. Urol. 2016, 70, 516–521. [Google Scholar] [CrossRef]
  67. Oudard, S.; Banu, E.; Vieillefond, A.; Fournier, L.; Priou, F.; Medioni, J.; Banu, A.; Duclos, B.; Rolland, F.; Escudier, B.; et al. Prospective Multicenter Phase II Study of Gemcitabine Plus Platinum Salt for Metastatic Collecting Duct Carcinoma: Results of a GETUG (Groupe d’Etudes des Tumeurs Uro-Génitales) Study. J. Urol. 2007, 177, 1698–1702. [Google Scholar] [CrossRef]
  68. Thibault, C.; Fléchon, A.; Albiges, L.; Joly, C.; Barthelemy, P.; Gross-Goupil, M.; Chevreau, C.; Coquan, E.; Rolland, F.; Laguerre, B.; et al. Gemcitabine plus platinum-based chemotherapy in combination with bevacizumab for kidney metastatic collecting duct and medullary carcinomas: Results of a prospective phase II trial (BEVABEL-GETUG/AFU24). Eur. J. Cancer 2023, 186, 83–90. [Google Scholar] [CrossRef]
  69. Procopio, G.; Sepe, P.; Claps, M.; Buti, S.; Colecchia, M.; Giannatempo, P.; Guadalupi, V.; Mariani, L.; Lalli, L.; Fucà, G.; et al. Cabozantinib as First-line Treatment in Patients with Metastatic Collecting Duct Renal Cell Carcinoma: Results of the BONSAI Trial for the Italian Network for Research in Urologic-Oncology (Meet-URO 2 Study). JAMA Oncol. 2022, 8, 910–913. [Google Scholar] [CrossRef]
  70. Buti, S.; Trentini, F.; Sepe, P.; Claps, M.; Isella, L.; Verzoni, E.; Procopio, G. BONSAI-2 study: Nivolumab as therapeutic option after cabozantinib failure in metastatic collecting duct carcinoma patients. Tumori J. 2023, 109, 418–423. [Google Scholar] [CrossRef] [PubMed]
  71. Nze, C.; Msaouel, P.; Derbala, M.H.; Stephen, B.; Abonofal, A.; Meric-Bernstam, F.; Tannir, N.M.; Naing, A. A Phase II Clinical Trial of Pembrolizumab Efficacy and Safety in Advanced Renal Medullary Carcinoma. Cancers 2023, 15, 3806. [Google Scholar] [CrossRef] [PubMed]
  72. Shah, A.Y.; Karam, J.A.; Malouf, G.G.; Rao, P.; Lim, Z.D.; Jonasch, E.; Xiao, L.; Gao, J.; Vaishampayan, U.N.; Heng, D.Y.; et al. Management and outcomes of patients with renal medullary carcinoma: A multicentre collaborative study. BJU Int. 2017, 120, 782–792. [Google Scholar] [CrossRef] [PubMed]
  73. Wilson, N.R.; Wiele, A.J.; Surasi, D.S.; Rao, P.; Sircar, K.; Tamboli, P.; Shah, A.Y.; Genovese, G.; Karam, J.A.; Wood, C.G.; et al. Efficacy and safety of gemcitabine plus doxorubicin in patients with renal medullary carcinoma. Clin. Genitourin. Cancer 2021, 19, e401–e408. [Google Scholar] [CrossRef]
  74. Lebenthal, J.M.; Kontoyiannis, P.D.; Hahn, A.W.; Lim, Z.D.; Rao, P.; Cheng, J.P.; Chan, B.; Daw, N.C.; Sheth, R.A.; Karam, J.A.; et al. Clinical Characteristics, Management, and Outcomes of Patients with Renal Medullary Carcinoma: A Single-center Retrospective Analysis of 135 Patients. Eur. Urol. Oncol. 2024, 8, 315–323. [Google Scholar] [CrossRef]
  75. Molina, A.M.; Feldman, D.R.; Ginsberg, M.S.; Kroog, G.; Tickoo, S.K.; Jia, X.; Georges, M.; Patil, S.; Baum, M.S.; Reuter, V.E.; et al. Phase II trial of sunitinib in patients with metastatic non-clear cell renal cell carcinoma. Investig. New Drugs 2012, 30, 335–340. [Google Scholar] [CrossRef]
  76. Lindner, A.K.; Tulchiner, G.; Seeber, A.; Siska, P.J.; Thurnher, M.; Pichler, R. Targeting strategies in the treatment of fumarate hydratase deficient renal cell carcinoma. Front. Oncol. 2022, 12, 6014. [Google Scholar] [CrossRef]
  77. Srinivasan, R.; Gurram, S.; Al Harthy, M.; Singer, E.A.; Sidana, A.; Shuch, B.M.; Ball, M.W.; Friend, J.C.; Mac, L.; Purcell, E.; et al. Results from a phase II study of bevacizumab and erlotinib in subjects with advanced hereditary leiomyomatosis and renal cell cancer (HLRCC) or sporadic papillary renal cell cancer. J. Clin. Oncol. 2020, 38 (Suppl. S15), 5004. [Google Scholar] [CrossRef]
  78. Kotecha, R.R.; Doshi, S.D.; Knezevic, A.; Chaim, J.; Chen, Y.; Jacobi, R.; Zucker, M.; Reznik, E.; McHugh, D.; Shah, N.J.; et al. A Phase 2 Trial of Talazoparib and Avelumab in Genomically Defined Metastatic Kidney Cancer. Eur. Urol. Oncol. 2024, 7, 804–811. [Google Scholar] [CrossRef]
  79. Kong, W.; Wu, G.; Xu, Y.; Wang, Z.; Zhang, J. Lenvatinib plus tislelizumab as first-line therapy for advanced fumarate hydratase-deficient renal cell carcinoma: A single-center, single-arm, phase II study. J. Clin. Oncol. 2025, 43 (Suppl. S5), 443. [Google Scholar] [CrossRef]
  80. Pal, S.; Bergerot, P.; Dizman, N.; Bergerot, C.; Adashek, J.; Madison, R.; Chung, J.; Ali, S.; Jones, J.; Salgia, R. Responses to Alectinib in ALK-rearranged Papillary Renal Cell Carcinoma. Eur. Urol. 2018, 74, 124–128. [Google Scholar] [CrossRef] [PubMed]
  81. Subbiah, V.; Gouda, M.A.; Ryll, B.; Burris, H.A., III; Kurzrock, R. The evolving landscape of tissue-agnostic therapies in precision oncology. CA Cancer J. Clin. 2024, 74, 433–452. [Google Scholar] [CrossRef] [PubMed]
  82. Rathmell, W.K.; Rumble, R.B.; Van Veldhuizen, P.J.; Al-Ahmadie, H.; Emamekhoo, H.; Hauke, R.J.; Louie, A.V.; Milowsky, M.I.; Molina, A.M.; Rose, T.L.; et al. Management of Metastatic Clear Cell Renal Cell Carcinoma: ASCO Guideline. J. Clin. Oncol. 2022, 40, 2957–2995. [Google Scholar] [PubMed]
  83. Guidelines Detail. NCCN. Available online: https://www.nccn.org/guidelines/guidelines-detail?category=1&id=1440 (accessed on 26 March 2025).
  84. Subbiah, V.; Othus, M.; Palma, J.; Cuglievan, B.; Kurzrock, R. Designing Clinical Trials for Patients with Rare Cancers: Connecting the Zebras. Am. Soc. Clin. Oncol. Educ. Book 2025, 45, e100051. [Google Scholar] [CrossRef]
Figure 1. Microscopic features of morphologically defined RCCs. (A). Papillary RCC showing papillary architecture with fibrovascular lined by columnar cells with low-grade nuclei (typical of type 1). (B). Chromophobe RCC with sheets of neoplastic cells with abundant admixed clear and eosinophilic cytoplasm. The cells have prominent cell borders and wrinkled nuclei. (C). TFE3-rearranged RCC showing papillary architecture. Cells have abundant clear to eosinophilic cytoplasm and grade 3 nuclei. (D). Fumarate-hydratase-deficient RCC also demonstrating a papillary architecture. The cells have abundant eosinophilic cytoplasm and large nuclei with macronucleoli with focally perinucleolar clearing (all 100× magnification).
Figure 1. Microscopic features of morphologically defined RCCs. (A). Papillary RCC showing papillary architecture with fibrovascular lined by columnar cells with low-grade nuclei (typical of type 1). (B). Chromophobe RCC with sheets of neoplastic cells with abundant admixed clear and eosinophilic cytoplasm. The cells have prominent cell borders and wrinkled nuclei. (C). TFE3-rearranged RCC showing papillary architecture. Cells have abundant clear to eosinophilic cytoplasm and grade 3 nuclei. (D). Fumarate-hydratase-deficient RCC also demonstrating a papillary architecture. The cells have abundant eosinophilic cytoplasm and large nuclei with macronucleoli with focally perinucleolar clearing (all 100× magnification).
Cancers 17 01527 g001
Table 1. Prospective clinical trials for TKIs and mTOR inhibitors in papillary, chromophobe, unclassified, and translocation RCCs.
Table 1. Prospective clinical trials for TKIs and mTOR inhibitors in papillary, chromophobe, unclassified, and translocation RCCs.
Trial NameStudy Drug(s)nEfficacy (ORR *, mPFS, mOS with 95% CI)
Papillary RCC-Only Trials
AXIPAP [4]Axitinib44ORR 12/42, 28.6% (15.7–44.6)
mPFS 6.6 mo (5.5–9.2)
PAPMET [5]Cabozantinib44ORR 23%
mPFS 9.0 mo (6–12)
mOS 21.5 mo (12.0–28.1)
Sunitinib46ORR 4%
mPFS 5.6 mo (3)
mOS 17.3 mo (12.8–21.8)
Savolitinib29ORR 3%
Crizotinib28ORR 0%
Trials With Multiple nccRCC Subtypes
Papillary RCCChromophobe RCCUnclassified RCCTranslocation RCC
ESPN [6]Sunitinib34ORR 3/33 (9%)
mPFS 5.7 mo
(1.4–19.8)
mPFS 8.9 mo
(2.9–20.1)
mPFS 9.4 mo (3.3–15.4)mPFS 6.1 mo
(6.0–8.8)
Everolimus38ORR 1/35 (3%)
mPFS 4.1 mo
(1.5–7.48)
NAmPFS 4.7 mo (2.6–NA)mPFS 3.0
(1.3–NA)
ASPEN [7]Sunitinib51ORR 8/33
24%
ORR 1/10
10%
ORR 0/8
0%
-
Everolimus57ORR 2/37
5%
ORR 2/6
33%
ORR 1/14
7%
-
NCT02915783 [8]Lenvatinib +
everolimus
31ORR 3/20
15%
ORR 4/9
44.4%
ORR 1/2
50%
-
NCT01399918 [9]Everolimus +
Bevacizumab
35ORR 7/18 **
38.9%
ORR 2/5
40%
ORR 1/9
11.1%
-
NCT01538238 [10]Pazopanib28ORR 7/18
39%
ORR 1/3
33%
ORR 0/2
0%
NA
NCT00502307 [11]Tivozanib46ORR 7/8
87.5%
ORR 2/2
100%
ORR 18/27
66.7%
-
* ORR can only be estimated for a subsection of the total n with measurable disease. ** Includes unspecified RCC with papillary RCC features. CI—confidence intervals, Mo—month, mOS—median overall survival, mPFS—median progression-free survival, NA—not available, ORR—overall response rate, RCC—renal cell carcinoma.
Table 2. Prospective clinical trials for immune checkpoint inhibitor monotherapies and combination therapies in papillary, chromophobe, unclassified, and translocation RCCs.
Table 2. Prospective clinical trials for immune checkpoint inhibitor monotherapies and combination therapies in papillary, chromophobe, unclassified, and translocation RCCs.
Trial NameStudy Drug(s)nEfficacy (ORR *, mPFS with 95% CI)
Papillary RCCChromophobe RCCUnclassified RCCTranslocation RCC
Immune Checkpoint Inhibitor Monotherapy
KEYNOTE-427 [12]Pembrolizumab165ORR 34/118
28.8% (20.8–37.9)
ORR 2/21
9.5% (1.2–30.4)
ORR 8/26
30.8% (14.3–51.8)
NA
CheckMate 374 [13]Nivolumab44ORR 2/24
8.3%
ORR 2/7
28.6%
ORR 1/8
12.5%
NA
UNISoN [14]Nivolumab80ORR 5/34
14.7%
ORR 1/13
8%
ORR 1/8
13%
ORR 2/5
40%
Nivolumab +
Ipilimumab (post-nivo)
41ORR 10%
(95% CI 3–23)
Immune Checkpoint Inhibitor Combination Therapies
KEYNOTE-B61 [15]Lenvatinib +
Pembrolizumab
158ORR 50/93
54% (43–64)
mPFS 17.5 mo
(15-NR)
ORR 8/29
28% (13–64)
mPFS 12.5 mo
(3.9-NR)
ORR 11/21
52% (30–74)
ORR 4/6
67% (22–96)
CA209–9KU [16]Cabozantinib +
Nivolumab
47ORR 15/32
47% (30–64)
mPFS 13 mo
(7–16)
ORR 0/7
0% (0–41.0)
ORR 3/6
50% (12–88)
mPFS 8 mo
(1-NE)
ORR 1/2
50% (1–99)
mPFS 14 mo
(5–23)
CheckMate 920 [17]Nivolumab + Ipilimumab46Overall ORR 9/46
19.6% (9.4–33.9%)
1 CR, 4 PR
18 patients **
0 CR, 0 PR
7 patients **
1 CR, 3 PR
22 patients **
0 CR, 0 PR
2 patients **
SUNNIFORECAST [18]
(intervention arm)
Nivolumab +
Ipilimumab
125ORR 21/72
29.2%
ORR 7/27
25.9%
NANA
NCT04413123 [19]Cabozantinib + nivolumab + ipilimumab40ORR 6/19
31.6%
ORR 1/11
9.1%
ORR 1/1
100%
ORR 0/5
0%
CALYPSO [20]
MET-driven subgroup
Savolitinib +
Durvalumab
41ORR 12/41
29% (16–46)
---
17ORR 9/17
53% (28–77)
---
COSMIC-021 [21]Cabozantinib + Atezolizumab32ORR 7/15
47%
ORR 1/9
11%
--
NCT02724878 [22]Bevacizumab +
Atezolizumab
42ORR 3/12
25%
ORR 1/10
10%
ORR 3/9
3.3%
ORR 1/5
20%
* ORR can only be estimated for a subsection of the total n with measurable disease. ** Breakdown of patients with measurable disease by subtype not provided; unable to calculate ORR. CI—confidence interval, CR—complete response, PR—partial response, mo—month, mPFS—median progression-free survival, NA—not available, NE—not estimable, nivo—nivolumab, NR—not reached, ORR—overall response rate, RCC—renal cell carcinoma.
Table 3. Select ongoing clinical trials for subtypes of nccRCC.
Table 3. Select ongoing clinical trials for subtypes of nccRCC.
StudyStudy Drug(s)nccRCC Subtype(s)Primary
Outcome
Goal
Recruitment (n)
Estimated
Completion
ICONIC [41] (NCT03866382)Nivolumab,
Ipilimumab, and cabozantinib
Papillary
Chromophobe
Collecting Duct
Sarcomatoid (~50%)
Renal Medullary
ORR
Secondary:
PFS, OS
314February 2026
PAPMET2 [42] (NCT05411081)Cabozantinib +/- AtezolizumabPapillaryPFS
Secondary: OS, ORR
200July 2027
SAMETA [39] (NCT05043090)Durvalumab +/- savolitinib vs. sunitinibMET-driven PapillarymPFS
Secondary: OS, ORR
147June 2026
NCT03595124 [43]Axitinib +/- NivolumabTranslocationmPFS
Secondary: toxicities
15January 2026
NCT05347212 [44] Nivolumab + RelatlimabRenal MedullaryORR
Secondary: OS, PFS
30July 2027
NCT05286801 [45]Tiragolumab + atezolizumabRenal MedullaryORR
Secondary: PFS, OS, PK
86June 2026
NCT04981509 [46]Bevacizumab + erlotinib +
atezolizumab
Papillary
HLRCC
CR
Secondary: ORR,
65December 2027
CR—complete response, HLRCC—hereditary leiomyomatosis and renal cell carcinoma, ORR—overall response rate, OS—overall survival, PK—pharmacokinetics, PFS—progression-free survival.
Table 4. Select retrospective reviews for subtypes of nccRCC.
Table 4. Select retrospective reviews for subtypes of nccRCC.
StudyStudy Drug(s)nnccRCC Subtype(s)ORRmPFS (95% CI)
Moussa et al. [52]Nivolumab +
Ipilimumab
55Papillary
Chromophobe
Unclassified
12/25 (48%)
3/12 (25%)
5/18 (27.8%)
10.6 mo (2.8–22.8)
3.6 mo (0.9–NE)
3 mo (2.1–7)
Alhalabi et al. [56]ICI + TKI18Translocation1/18 (5.5%)5.4 mo
ICI + ICI114/11(36%)2.8 mo
Ged et al. [57]ICI + TKI
ICI + ICI
14
8
Translocation1/7 (14%)
6/11 (54%)
TTF 1.2 mo
TTF 6.2 mo
Paffenholz et al. [53]ICI + TKI10Chromophobe3/10 (30%)8 mo (0–19.2)
ICI + ICI81/8 (12.5%)3 mo (2.4–3.6)
TKI132/13 (15%)4 mo (0–10.2)
Chanza et al. [54]Cabozantinib112Papillary
Chromophobe
Unclassified
Translocation
18/66 (27%)
3/10 (30%)
2/15 (13%)
5/17 (29%)
6.9 mo (4.6–10.1)
5.7 mo (1.1–7.8)
6.0 mo (1.4–9.9)
8.3 mo (4.6–NR)
Campbell et al. [55]Cabozantinib30Papillary
Chromophobe
Unclassified
Translocation
1/15 (7%)
1/6 (17%)
1/3 (33%)
0/2 (0%)
8.6 mo (6.1–14.7)
CI—confidence interval, ICI—immune checkpoint inhibitor, mo—month, NE—not estimable, NR—not reached, ORR—overall response rate, TKI—tyrosine kinase inhibitor, TTF—time to treatment failure.
Table 6. ORR by sarcomatoid differentiation and PD-L1 status in nccRCC ICI trials.
Table 6. ORR by sarcomatoid differentiation and PD-L1 status in nccRCC ICI trials.
Trial NameStudy Drug(s)nObjective Response Rates (ORR) *
OverallSarcomatoid DifferentiationPD-L1 ≥ 1%
Immune Checkpoint Inhibitor Monotherapy
KEYNOTE-427 [12]Pembrolizumab16526.7%16/38
42.1% (95% CI, 26.3–59.2%)
36/102
35.3% (26.1–45.4)
CheckMate 374 [13]Nivolumab4413.6%2/4
50%
NA
mOS 16.3 mo
UNISoN [14]Nivolumab8016.9%NANA
Immune Checkpoint Inhibitor Combination Therapies
KEYNOTE-B61 [15]Lenvatinib +
Pembrolizumab
15849%9/18
47% (95% CI 24–71)
54/93
58% (95% CI 47–68)
CA209-9KU [16]Cabozantinib +
Nivolumab
4747.5%NANA
CheckMate 920 [17]Nivolumab +
Ipilimumab
4619.6%5/14
35.7% (95% CI 12.8–64.9)
4/13
30.8%
SUNNIFORECAST [18]Nivolumab +
Ipilimumab
12532.8%NANA
HR for OS 0.56 (95% CI 0.33–0.95)
NCT04413123 [19]Cabozantinib + nivolumab + ipilimumab4021%3/9
33.3%
NA
CALYPSO [20]Savolitinib +
Durvalumab
4129%NA9/27
33% (95% CI 17–54)
COSMIC-021 [21]Cabozantinib + Atezolizumab3223%NA5/23
22%
NCT02724878 [22]Bevacizumab +
Atezolizumab
4226%3/8
38%
9/23
36%
* ORR can only be estimated for a subsection of the total n with measurable disease. Mo—month, NA—not available, nivo—nivolumab, ORR—overall response rate, OS—overall survival, RCC—renal cell carcinoma.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Vento, J.; Zhang, T.; Kapur, P.; Hammers, H.; Brugarolas, J.; Qin, Q. Systemic Treatment of Locally Advanced or Metastatic Non-Clear Cell Renal Cell Carcinoma. Cancers 2025, 17, 1527. https://doi.org/10.3390/cancers17091527

AMA Style

Vento J, Zhang T, Kapur P, Hammers H, Brugarolas J, Qin Q. Systemic Treatment of Locally Advanced or Metastatic Non-Clear Cell Renal Cell Carcinoma. Cancers. 2025; 17(9):1527. https://doi.org/10.3390/cancers17091527

Chicago/Turabian Style

Vento, Joseph, Tian Zhang, Payal Kapur, Hans Hammers, James Brugarolas, and Qian Qin. 2025. "Systemic Treatment of Locally Advanced or Metastatic Non-Clear Cell Renal Cell Carcinoma" Cancers 17, no. 9: 1527. https://doi.org/10.3390/cancers17091527

APA Style

Vento, J., Zhang, T., Kapur, P., Hammers, H., Brugarolas, J., & Qin, Q. (2025). Systemic Treatment of Locally Advanced or Metastatic Non-Clear Cell Renal Cell Carcinoma. Cancers, 17(9), 1527. https://doi.org/10.3390/cancers17091527

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop