Next Article in Journal
Secular Trends of Liver Cancer Mortality and Years of Life Lost in Wuhan, China 2010–2019
Previous Article in Journal
Real-Life Effectivity of Dose Intensity Reduction of First-Line mFOLFIRI-Based Treatment of Metastatic Colorectal Cancers: Sometimes Less Is More
Previous Article in Special Issue
DPYD Exon 4 Deletion Associated with Fluoropyrimidine Toxicity and Importance of Copy Number Variation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Navigating the Current Landscape of Non-Clear Cell Renal Cell Carcinoma: A Review of the Literature

1
Department of Medical Oncology, Eastern Health, Melbourne, VIC 3128, Australia
2
Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
3
Eastern Health Clinical School, Monash University, Melbourne, VIC 3128, Australia
4
Department of Surgery, University of Melbourne, Melbourne, VIC 3010, Australia
*
Author to whom correspondence should be addressed.
Curr. Oncol. 2023, 30(1), 923-937; https://doi.org/10.3390/curroncol30010070
Submission received: 8 December 2022 / Revised: 24 December 2022 / Accepted: 6 January 2023 / Published: 9 January 2023
(This article belongs to the Special Issue The Evolving Landscape of Systemic Therapies and Oncology Outcomes)

Abstract

:
Non-clear cell renal cell carcinoma (nccRCC) is an entity comprised of a heterogeneous constellation of RCC subtypes. Genomic profiling has broadened our understanding of molecular pathogenic mechanisms unique to individual nccRCC subtypes. To date, clinical trials evaluating the use of immunotherapies and targeted therapies have predominantly been conducted in patients with clear cell histology. A comprehensive review of the literature has been undertaken in order to describe molecular pathogenic mechanisms pertaining to each nccRCC subtype, and concisely summarise findings from therapeutic trials conducted in the nccRCC space.

1. Introduction

Renal cell carcinoma (RCC) is the seventh-most common malignancy globally [1], representing 5% of adult malignancies in males and 3% in females [2]. Recognised risk factors include cigarette smoking, obesity and hypertension. Underlying disorders such as von Hippel-Lindau (VHL) syndrome, end stage renal failure, acquired renal cystic disease, dialysis dependence and prior renal transplant predispose to development of RCC [3]. Clear cell RCC (ccRCC) constitutes approximately 80% of all RCC, whilst the less common non-clear cell RCC (nccRCC) can be further stratified by subtype [4]. The World Health Organisation (WHO) has updated the classification system of RCC in 2022, distinguishing subtypes on the basis of both morphological appearance as well as underlying molecular aberrations (e.g., TFE3 and ALK-rearranged RCC, SMARCB1-deficient medullary-like RCC), thus offering an integrated approach to characterizing tumour subtypes [5].
The majority of advanced RCC clinical trials are targeted to patients with ccRCC. Management paradigms within this space have historically consisted of cytokine therapy (such as interferon-alpha and IL-2) and oral targeted therapies (tyrosine kinase inhibitors, such as sunitinib); however, immunotherapy (IO) has recently emerged as a mainstay of initial treatment of metastatic ccRCC [6]. This shift has occurred due to improvements in survival with immunotherapy in large randomized clinical trials (CheckMate-214, KEYNOTE-426, CheckMate-9ER, CLEAR and JAVELIN-101) [7,8,9,10,11]. Notably, only patients with a clear cell component of RCC were included in these practice-informing trials. Comparatively, nccRCC- a heterogeneous group of malignancies with diverse clinical and molecular features- has been less well studied. Much of the treatment evidence for advanced nccRCC arises from smaller, non-randomised or retrospective studies and expert-level evidence only. Moreover, there remains a dearth of established treatments and clinical trials specifically designed for this patient population. Herein, we provide an overview of nccRCC with a focus on evidence for systemic therapy of advanced disease.

2. Pathological, Molecular and Clinical Features of nccRCC Subtypes

Certain subtypes of metastatic nccRCC are associated with more aggressive disease behaviour and poorer survival outcomes compared to ccRCC. Within nccRCC, subtypes exhibit different clinical courses. The incidence of morphological subtypes of nccRCC are listed in Table 1. Recent genomic sequencing studies of nccRCC have revealed molecular aberrations amongst different nccRCC subtypes that parallel different patterns of clinical behavior, therapy response and prognosis. Herein, we discuss nccRCC subtypes with unique histological and molecular features in further detail.

2.1. Papillary

Papillary RCC is denoted histologically by the presence of basophilic cells arranged in a papillary fashion [13]. Papillary RCC can be further divided into two types, each demonstrating a unique biological basis and clinical phenotype. Type 1 papillary RCC is typically characterized by papillae with shallower layers of columnar epithelial cells that possess minimal cytoplasm and small ovoid nuclei. Additional features include presence of foamy macrophages and psammoma bodies [13]. In contrast, type 2 papillary RCC exhibits more aggressive histological features such as pleomorphic irregular nuclei with nucleoli and eosinophilic cytoplasm-containing large cells [14]. Tumour stage and grade at time of diagnosis are consequently higher with type 2 papillary RCC [13]. This translates into the clinical setting, with type 1 papillary RCC conferring a better prognosis than ccRCC and type 2 papillary RCC. It is known to have a more indolent clinical course and is less likely to metastasise than type 2 papillary RCC [14].
MET-activating mutations were initially found in the hereditary papillary RCC setting [15]. The pathogenicity of these mutations was subsequently demonstrated in both hereditary and sporadic forms of the disease in further studies reported in the Cancer Genome Atlas (TCGA) profiling [16]. A total of 81% of type 1 papillary RCC demonstrates alteration of MET or increased copy number of chromosome 7, the long arm of which contains the MET proto-oncogene [16]. In contrast, type 2 papillary RCC has a more heterogeneous molecular profile. Recurrent genomic alterations described in this subtype include CDKN2A mutation/promoter hypermethylation, NRF2-ARE overexpression, TFE3 fusions and SETD2 mutations [16].

2.2. Hereditary Leiomyomatosis and Renal Cell Cancer (HLRCC)

HLRCC is a rare subset of nccRCC with an autosomal dominant pattern of inheritance that predisposes to the development of cutaneous and uterine leiomyomas in addition to RCC [17]. Historically, HLRCC was classified as type 2 papillary RCC. It is now recognised as a distinct entity [5], as it is pathogenically driven by germline mutations in the fumarate hydratase (FH) gene. FH is responsible for the formation of L-malate from fumarate as part of the tricarboxylic acid (TCA) cycle. Mutations in FH lead to enhancement of hypoxia-inducible factor 1-alpha (HIF-1α) and downstream factors such as erythropoietin (EPO) and vascular endothelial growth factor (VEGF), in turn leading to tumorigenesis [18].

2.3. Chromophobe

Chromophobe RCC originates from the intercalated cells of the renal collecting duct [19]. Loss of whole chromosomes is a hallmark of chromophobe RCC. A higher frequency of mutations in the tumour suppressor genes TP53 and PTEN have been shown in this subset of nccRCC. TERT rearrangements may also play a role in tumorigenesis of chromophobe RCC [20]. Genetic syndromes associated with chromophobe RCC include the following: tuberous sclerosis, Cowden syndrome, Birt-Hogg-Dube syndrome, hereditary paraganglioma/phaeochromocytoma and BAP1 tumour predisposition syndromes [21]. Clinically, chromophobe RCC is more likely to arise in young female patients and has an indolent clinical course. Fewer than 5% of patients present with advanced disease, and prognosis is more favourable compared with ccRCC [22].

2.4. Medullary

Medullary RCC is a rare form of RCC that is associated with poorer survival. It occurs more frequently in young African males with sickle cell trait [23]. Molecular pathogenesis is similar to urothelial carcinoma, with loss of SMARCB1, NOTCH2 amplifications and deletions of NOTCH1/NOTCH3 commonly seen [24]. Medullary RCC tends to be resistant to both cytokine and chemotherapy [23].

2.5. Collecting Duct

Collecting duct/Bellini duct RCC arises from the principal cells of renal medullary collecting ducts [25]. Histologically it is characterized by desmoplastic stroma, tubular or tubule-papillary infiltration of the renal parenchyma and high-grade nuclear features [26]. Collecting duct tumours are associated with a poor prognosis, and therefore early histological identification is imperative [27]. Next generation sequencing in a small group of patients with collecting duct RCC has revealed molecular abnormalities in SETD2, CDKN2A, SMARCB1 and HF2 [28].

2.6. Sarcomatoid and Rhabdoid Dedifferentiation

Though not categorized as subtypes of nccRCC, note should be made of the phenomena of sarcomatoid and rhabdoid dedifferentiation, which can be seen in approximately 10–15% of RCC, often co-existing with other RCC histologies (e.g., clear cell, papillary, etc.) [29]. Morphologically, sarcomatoid dedifferentiation arises from the process of epithelial-mesenchymal transition. Sarcomatoid RCC therefore contains both carcinoma and sarcomatoid-type histological features [30]. Rhabdoid dedifferentiation is characterized by clusters of large epithelioid cells with vesicular nuclei, prominent nucleoli and paranuclear intracytoplasmic hyaline inclusions [31]. Sarcomatoid and rhabdoid dedifferentiation can occur either concomitantly or independently of each other [32]. These tumours are known to be biologically aggressive and therefore confer a poor prognosis [33,34].
Transcriptomic analyses have demonstrated the role of MYC overexpression, BAP1 mutations and CDKN2A deletions in the pathogenesis and aggressive course of sarcomatoid and rhabdoid dedifferentiated RCC [29]. These tumours are known to have an ‘immune-inflamed phenotype’, typified by immune activation, higher PD-L1 expression, upregulation of antigen presenting machinery genes and enhanced infiltration of cytotoxic immune cells [29]. This translates into the clinical setting, as demonstrated through the responsiveness of these tumours to IO, with response rates of 47–61% and overall survival benefit over sunitinib seen in subgroup analyses of Phase III ccRCC trials [35,36,37].

3. Systemic Therapy

A greater understanding of the molecular biology and pathogenesis of nccRCC has led to a growing number of trials exploring therapeutic agents in this context. Mirroring the trials being conducted in the ccRCC space, both immune checkpoint inhibition and targeted therapy are being investigated. Of note, local therapies such as surgery and ablative techniques (e.g., radiofrequency, microwave and cryoablation) can be utilized in the management of early stage nccRCC [38]. However, it is unclear if differences exist between outcomes in ccRCC and nccRCC managed with local therapies, as comparative studies have not been conducted. As such, systemic therapy will be the focus of this discussion.
Retrospective studies have demonstrated efficacy of IO (ORR 10–35%), and targeted therapies (ORR 15–30%) in nccRCC, although it must be recognised that sample sizes are small and study populations are heterogeneous in these analyses (Table 2).
These data do not demonstrate major differences between survival outcomes with use of targeted therapies vs. IO. Of note, poor outcomes were seen with mTOR inhibitors. Additionally, chromophobe tumours responded poorly to IO, although higher response rates were observed with combination ipilimumab/nivolumab in this subgroup. Results must be interpreted with caution, however, due to the heterogeneity of trial populations, small sample sizes and the retrospective nature of these studies.

3.1. Immunotherapy

To date, other than sarcomatoid and rhabdoid dedifferentiation, no biomarkers have been able to reliably predict response to IO in renal cell carcinoma. PD-L1 expression was shown to vary between nccRCC subtype in a series that demonstrated PD-L1 positivity of 5.6% in chromophobe RCC, 10% in papillary RCC, 20% in Xp11.2 translocation RCC and 20% in collecting duct carcinoma [52]. Broadly speaking, RCC is a tumour with low tumour mutational burden (TMB), with a median of 1.1 mutations/Mb. Of RCC subtypes, lowest TMB is seen with chromophobe RCC (<1 mutation/Mb) [53]. As such, chromophobe RCC is thought to exhibit an immunologically ‘cold’ phenotype. Interestingly, however, both chromophobe and papillary RCC were shown to have the highest insertion deletion (indel) rates proportional to TMB in a pan-cancer whole-exome sequencing analysis of nineteen solid tumour types [54]. Indels are known to generate neoantigenic peptides, and therefore hypothesised to correlate with antitumour response to IO [54].
Notably, IO response rates were poorer in chromophobe RCC in comparison with other nccRCC subtypes in the majority of retrospective analyses listed in Table 2. This may add weight to the hypothesis that chromophobe tumours are indeed immunologically ‘cold’.
Building upon retrospective data, evidence to support use of either single agent or doublet IO in this space was further substantiated in prospective single arm studies (Table 3). Parallel ccRCC cohorts in KEYNOTE-427, Checkmate-374 and HCRN GU16-260 demonstrated superior response rates of immunotherapy in ccRCC when compared with nccRCC [55,56,57].

3.2. Targeted Therapy

Prospective studies have demonstrated activity of mTOR inhibitors and anti-angiogenic VEGF tyrosine kinase inhibitors (TKIs) inhibitors against nccRCC (Table 4). Angiogenesis has been identified as a hallmark of RCC biology [60]. Activation of mTOR is known to lead to angiogenesis through HIF-1α dependent and independent factors, including modulation of nitric oxide and angiopoietins [61]. Head-to-head trials have demonstrated superior outcomes with sunitinib compared with everolimus [62,63,64], although these findings were not statistically significant and conclusions must be cautiously interpreted owing to heterogeneous trial populations, small cohorts and crossover to the sunitinib arm in the ESPN trial [63].
The MET proto-oncogene is implicated in the tumorigenesis of papillary RCC, particularly type 1 papillary RCC. Mechanisms of MET overexpression include gene amplification and fusions, receptor mutations, increased copy number of chromosome 7, and activating point mutations [16]. Though activity of MET inhibitors has been explored in this space, results have been disappointing in the unselected population (Table 5). The SAVOIR study is the only study to date with central confirmation of MET as a driver, demonstrating superior outcomes with the MET inhibitor savolitinib in comparison with sunitinib in an exclusively MET-driven population. Central confirmation of MET alteration is also required in the ongoing SAMETA trial (NCT05043090) [68] which aims to evaluate the efficacy of immune checkpoint and MET inhibitor as monotherapies or combination therapy in an exclusively MET-driven population.
Knowledge of signalling pathways implicated in HLRCC such as HIF upregulation and NRF2 activation [18] has led to targeted therapy trials in this space. Response rates of approximately 50% were demonstrated with the combination of the epithelial growth factor receptor (EGFR) inhibitor erlotinib and bevacizumab, a VEGF inhibitor, in both the phase II AVATAR trial [74] and a Korean retrospective series [75]. Trials evaluating use of immune checkpoint inhibitors in this space are ongoing [18], and preclinical studies of polyadenosine diphosphate-ribose polymerase (PARP) inhibitors hold promise as a therapeutic strategy [76], although more clinical data are necessary.

3.3. Combination Therapies: IO/Targeted Therapy

The interplay of tumour vessels and T-cells creates a vicious cycle within the tumour microenvironment. Tumour vessels have been shown to hinder CD8+ T cell trafficking into the tumour microenvironment, leading to impaired T-cell effector functions and T-cell killing. Additionally, VEGF has the potential to interfere with dendritic cell maturation and priming of T cells. As such, combination IO and VEGF inhibition is hypothesised to have synergistic anti-tumour activity [77].
Single arm prospective studies have presented efficacy data with combination IO and targeted therapy (Table 6), although more prospective, randomised trials are needed to assess superiority over other combinations and individual classes of treatment. A recent phase II single arm trial of nivolumab in combination with cabozantinib demonstrated the striking difference in response rates between patients with chromophobe (0%) vs. papillary/unclassified/translocation associated RCC (47.5%) [78]. Combination therapy with IO and MET inhibitor was explored in the CALYPSO trial in both papillary and clear cell RCC cohorts [79,80] (Table 6). In the ccRCC cohort, the primary objective of ≥50% response rate was not observed either with savolitinib monotherapy or with combination savolitinib/durvalumab.

4. Future Directions

Non-clear cell RCC is a subset of genitourinary cancer that is being increasingly examined. In spite of this, however, there is still no agent or combination of agents that has parallelled response rates seen in ccRCC. As understanding of the underlying molecular pathophysiology of nccRCC grows, so does the potential for development of predictive biomarkers and future therapeutic targets. A tissue-based analysis conducted as part of the ASPEN trial (everolimus vs. sunitinib) demonstrated the relevance of particular prognostic biomarkers in nccRCC. Multivariate analysis revealed the negative and positive predictive values of p-Akt and c-kit expression respectively with regards to overall survival. Moreover, c-kit expression was predictive of benefit from everolimus, and c-MET expression correlated with a poorer response to everolimus or sunitinib [83].
The prognostic and predictive value of PD-L1 level and TMB in nccRCC is unclear, although higher PD-L1 level has been shown to correlate with poorer survival in several studies in the metastatic RCC space (inclusive of both clear cell and non-clear cell RCC) [84].Within the clear cell RCC context, recent data have highlighted the utility of metabolic profiling in predicting clinical outcomes including prognosis and response to therapies through a combination of metabolomics, lipidomics and transcriptomics. There is therefore scope for similar investigation to be prospectively conducted in the non-clear cell RCC space [85].
Other molecular targets are currently under investigation. Preclinical studies have demonstrated the oncogenicity of Hypoxia-inducible factor 1-alpha (HIF-1α) in ccRCC and FH deficient nccRCC [86]. Increased HIF-1α activity results from loss of function of the VHL tumour suppressor gene [86]. Further exploration of this target has led to development of HIF inhibitors. The role of Bezultifan, an oral HIF-2α inhibitor, in VHL associated RCC has been studied in a recent open label phase I/II study in patients, demonstrating an ORR of 49.2% [87]. Fumarate hydratase mutations—which are commonly seen in hereditary type II papillary RCC and are characterised by an aggressive clinical course—have also been hypothesised to lead to HIF-1α overexpression through intracellular accumulation of the oncometabolite fumarate, and consequent destabilisation of HIF-1α [88]. As such, the role of HIF-1α in FH-deficient RCC is ongoingly being examined.

5. Discussion

Though historically poorly characterised, our understanding of nccRCC is rapidly expanding. It is imperative to recognise that nccRCC subtypes exhibit heterogeneity. Recent advances in genomic profiling have enabled identification of underlying molecular abnormalities unique to the tumorigenesis of each subtype of this disease. Prototypically, discovery of the role of MET alterations in hereditary papillary RCC has subsequently led to investigation of its role in the sporadic form of the disease, in turn resulting in therapeutic strategies with several MET inhibitor trials. In a similar vein, the pathogenic role of FH gene mutations in HLRCC has also led to exploration of HIF inhibitors as a therapeutic class. Synthesis of the aforementioned trials raises the question of the optimal first line management strategy of nccRCC subtypes. It is important to acknowledge that to date, only single arm immunotherapy trials have been conducted in this space, and no head-to-head studies of checkpoint inhibitors vs. targeted therapies exist. Indirect comparisons would suggest that response rates to checkpoint inhibitors are superior to targeted therapies with some exceptions (e.g., chromophobe RCC); however, there is no clear overall survival benefit. Additionally, trial populations are heterogeneous in subtype frequency, and inter-trial population differences exist. Given that nccRCC subtypes are biologically and phenotypically distinct, it is also important to recognise variability in efficacy of therapeutic classes between subtypes. In the majority of the listed trials, sample sizes of distinct nccRCC subtypes are small and difficult to draw firm conclusions from.
KEYNOTE-427, the largest prospective immunotherapy trial in the nccRCC space, has demonstrated a discrepancy in ORR with pembrolizumab between papillary and chromophobe RCC (28.8 vs. 9.5%, n = 21 in chromophobe subgroup) [55]. This could reflect the immunologically ‘cold’ phenotype of chromophobe RCC described in the literature [51,52]. Poor response of chromophobe RCC to IO has also been demonstrated in several retrospective analyses. In contrast, an ORR of 29% was observed with nivolumab in the chromophobe cohort of the CheckMate-374 study, albeit with a smaller sample size (n = 7) [55]. As such, it is unclear if poorer response to IO is unequivocally seen with chromophobe RCC. In light of this data, it may seem reasonable to pursue targeted therapy as a first line treatment strategy in patients with chromophobe RCC, whilst further data are awaited. The findings of the phase II single arm nivolumab/cabozantinib study demonstrating no responses in the chromophobe cohort [78] suggest that response rates are not superior with the addition of IO to targeted therapy in this subgroup.
In contrast, checkpoint inhibitors would seem to be preferable to TKI therapy in papillary and other subtypes of RCC, given suggestion of improved response rates.
MET inhibition may prove to be a viable strategy in patients with papillary RCC and confirmed MET alterations, though minimal data exist in MET-exclusive populations to date. Response rates of up to 50% have been noted in MET-driven papillary RCC [69,70], albeit in small sample sizes, and these agents certainly hold promise for the future.
Cabozantinib has demonstrated encouraging efficacy in the treatment of collecting duct RCC, a poor prognosis RCC subtype, in the phase II BONSAI trial [67]. Given that no standard treatments exist for this disease, the observed ORR of 35% suggests that this TKI could be the agent of choice for this subpopulation.
Building upon our understanding of its molecular pathophysiology, in the HLRCC population, the combination of EGFR and VEGF inhibition (erlotinib/bevacizumab) has been shown to be effective, with response rates of up to 50% seen in the AVATAR study [74] and a separate retrospective analysis [75], highlighting this as a valid therapeutic option in this subgroup.
Moving forward, early results of HIF inhibitor trials have been promising, with the oral HIF-2 inhibitor Belzutifan inducing response rates of 49% in patients with VHL-associated RCC [87]. Further studies investigating role of HIF inhibitors in FH deficient RCC such as HLRCC are required to evaluate clinical applicability of preclinical studies using these agents.
Single arm combination IO and targeted therapy trials have been promising so far, although all four studies have demonstrated superior response rates with non-chromophobe (25–60%) than chromophobe (0–15%) cohorts [78,79,81,82]. Importantly, further head-to-head trials comparing combination therapy with both IO and targeted therapies are required to clearly answer the question of whether additional benefit is gained from a combination strategy. Comparison of doublet IO to combination IO/targeted therapy would also be clinically applicable. Moreover, breakdown of efficacy by nccRCC subtype in such studies would also guide the optimal choice of first line management.
The challenges associated with conducting further trials in a heterogeneous population with small numbers of patients with each subtype of the disease must be recognised, highlighting the importance of collaborative efforts between global institutions to establish further evidence-based guidance in the understanding and management of nccRCC.
As our understanding of nccRCC subtypes broadens, it is also important that clinically relevant prognostic and predictive biomarkers are investigated, in order to better shape the therapeutic landscape and personalise treatment for the individual patient. In particular, the value of biomarkers such as PD-L1 expression, TMB and indel rates have not been convincingly predictive of response to immunotherapy, and more work is certainly required in this sphere.

6. Conclusions

Non-clear cell RCC is comprised of a heterogeneous spectrum of biologically distinct subtypes. Understanding of the molecular pathogenesis of subtypes has significantly improved in recent times. This will hopefully pave the way for future trials investigating the role of biomarkers, and selection of the optimal therapeutic strategy in individual nccRCC subtypes.

Author Contributions

Conceptualization, A.J. and A.A.H.; methodology, A.J.; writing—original and draft preparation, A.J.; writing- review and editing, A.J., A.A.H., L.S.; supervision, A.A.H., L.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Padala, S.A.; Barsouk, A.; Thandra, K.C.; Saginala, K.; Mohammed, A.; Vakiti, A.; Rawla, P.; Barsouk, A. Epidemiology of Renal Cell Carcinoma. World J. Oncol. 2020, 11, 79–87. [Google Scholar] [CrossRef] [PubMed]
  2. Capitanio, U.; Bensalah, K.; Bex, A.; Boorjian, S.A.; Bray, F.; Coleman, J.; Gore, J.L.; Sun, M.; Wood, C.; Russo, P. Epidemiology of Renal Cell Carcinoma. Eur. Urol. 2019, 75, 74–84. [Google Scholar] [CrossRef] [PubMed]
  3. Chow, W.-H.; Dong, L.M.; Devesa, S.S. Epidemiology and risk factors for kidney cancer. Nat. Rev. Urol. 2010, 7, 245–257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Escudier, B.; Porta, C.; Schmidinger, M.; Rioux-Leclercq, N.; Bex, A.; Khoo, V.; Grünwald, V.; Gillessen, S.; Horwich, A.; ESMO Guidelines Committee. Renal cell carcinoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2019, 30, 706–720. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Moch, H.; Amin, M.B.; Berney, D.M.; Compérat, E.M.; Gill, A.J.; Hartmann, A.; Menon, S.; Raspollini, M.R.; Rubin, M.A.; Srigley, J.R.; et al. The 2022 World Health Organization Classification of Tumours of the Urinary System and Male Genital Organs—Part A: Renal, Penile, and Testicular Tumours. Eur. Urol. 2022, 82, 458–468. [Google Scholar] [CrossRef]
  6. Ahrens, M.; Scheich, S.; Hartmann, A.; Bergmann, L. IAG-N Interdisciplinary Working Group Kidney Cancer of the German Cancer Society Non-Clear Cell Renal Cell Carcinoma—Pathology and Treatment Options. Oncol. Res. Treat. 2019, 42, 128–135. [Google Scholar] [CrossRef]
  7. Albiges, L.; Tannir, N.M.; Burotto, M.; McDermott, D.; Plimack, E.R.; Barthélémy, P.; Porta, C.; Powles, T.; Donskov, F.; George, S.; et al. Nivolumab plus ipilimumab versus sunitinib for first-line treatment of advanced renal cell carcinoma: Extended 4-year follow-up of the phase III CheckMate 214 trial. ESMO Open 2020, 5, e001079. [Google Scholar] [CrossRef]
  8. Powles, T.; Plimack, E.R.; Soulières, D.; Waddell, T.; Stus, V.; Gafanov, R.; Nosov, D.; Pouliot, F.; Melichar, B.; Vynnychenko, I.; et al. Pembrolizumab plus axitinib versus sunitinib monotherapy as first-line treatment of advanced renal cell carcinoma (KEYNOTE-426): Extended follow-up from a randomised, open-label, phase 3 trial. Lancet Oncol. 2020, 21, 1563–1573. [Google Scholar] [CrossRef]
  9. Choueiri, T.K.; Powles, T.; Burotto, M.; Escudier, B.; Bourlon, M.T.; Zurawski, B.; Oyervides Juárez, V.M.; 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]
  10. 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]
  11. Motzer, R.J.; Penkov, K.; Haanen, J.; Rini, B.; Albiges, L.; Campbell, M.T.; Venugopal, B.; Kollmannsberger, C.; Negrier, S.; Uemura, M.; et al. Avelumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2019, 380, 1103–1115. [Google Scholar] [CrossRef] [PubMed]
  12. Zarrabi, K.; Walzer, E.; Zibelman, M. Immune Checkpoint Inhibition in Advanced Non-Clear Cell Renal Cell Carcinoma: Leveraging Success from Clear Cell Histology into New Opportunities. Cancers 2021, 13, 3652. [Google Scholar] [CrossRef]
  13. Mendhiratta, N.; Muraki, P.; Sisk, A.E.; Shuch, B. Papillary renal cell carcinoma: Review. Urol. Oncol. Semin. Orig. Investig. 2021, 39, 327–337. [Google Scholar] [CrossRef]
  14. Leroy, X.; Zini, L.; Leteurtre, E.; Zerimech, F.; Porchet, N.; Aubert, J.-P.; Gosselin, B.; Copin, M.-C. Morphologic Subtyping of Papillary Renal Cell Carcinoma: Correlation with Prognosis and Differential Expression of MUC1 between the Two Subtypes. Mod. Pathol. 2002, 15, 1126–1130. [Google Scholar] [CrossRef] [Green Version]
  15. Smith, K.E.R.; Bilen, M.A. A Review of Papillary Renal Cell Carcinoma and MET Inhibitors. Kidney Cancer 2019, 3, 151–161. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Cancer Genome Atlas Research Network; Linehan, W.M.; Spellman, P.T.; Ricketts, C.J.; Creighton, C.J.; Fei, S.S.; Davis, C.; Wheeler, D.A.; Murray, B.A.; Schmidt, L.; et al. Comprehensive Molecular Characterization of Papillary Renal-Cell Carcinoma. N. Engl. J. Med. 2016, 374, 135–145. [Google Scholar] [CrossRef]
  17. Chayed, Z.; Kristensen, L.K.; Ousager, L.B.; Rønlund, K.; Bygum, A. Hereditary leiomyomatosis and renal cell carcinoma: A case series and literature review. Orphanet J. Rare Dis. 2021, 16, 34. [Google Scholar] [CrossRef]
  18. Zhang, C.; Li, L.; Zhang, Y.; Zeng, C. Hereditary Leiomyomatosis and Renal Cell Cancer: Recent Insights Into Mechanisms and Systemic Treatment. Front. Oncol. 2021, 11, 686556. [Google Scholar] [CrossRef] [PubMed]
  19. Gerharz, C.D.; Moll, R.; Störkel, S.; Ramp, U.; Thoenes, W.; Gabbert, H.E. Ultrastructural appearance and cytoskeletal architecture of the clear, chromophilic, and chromophobe types of human renal cell carcinoma in vitro. Am. J. Pathol. 1993, 142, 851–859. [Google Scholar]
  20. Davis, C.F.; Ricketts, C.J.; Wang, M.; Yang, L.; Cherniack, A.D.; Shen, H.; Buhay, C.; Kang, H.; Kim, S.C.; Fahey, C.C.; et al. The Somatic Genomic Landscape of Chromophobe Renal Cell Carcinoma. Cancer Cell 2014, 26, 319–330. [Google Scholar] [CrossRef] [Green Version]
  21. Garje, R.; Elhag, D.; Yasin, H.A.; Acharya, L.; Vaena, D.; Dahmoush, L. Comprehensive review of chromophobe renal cell carcinoma. Crit. Rev. Oncol. 2021, 160, 103287. [Google Scholar] [CrossRef] [PubMed]
  22. Casuscelli, J.; Becerra, M.F.; Seier, K.; Manley, B.J.; Benfante, N.; Redzematovic, A.; Stief, C.G.; Hsieh, J.J.; Tickoo, S.K.; Reuter, V.E.; et al. Chromophobe Renal Cell Carcinoma: Results From a Large Single-Institution Series. Clin. Genitourin. Cancer 2019, 17, 373–379.e4. [Google Scholar] [CrossRef] [PubMed]
  23. Shetty, A.; Matrana, M.R. Renal medullary carcinoma: A case report and brief review of the literature. Ochsner J. 2014, 14, 270–275. [Google Scholar] [PubMed]
  24. Msaouel, P.; Malouf, G.G.; Su, X.; Yao, H.; Tripathi, D.N.; Soeung, M.; Gao, J.; Rao, P.; Coarfa, C.; Creighton, C.J.; et al. Comprehensive Molecular Characterization Identifies Distinct Genomic and Immune Hallmarks of Renal Medullary Carcinoma. Cancer Cell 2020, 37, 720–734.e13. [Google Scholar] [CrossRef]
  25. Harbin, A.C.; Styskel, B.A.; Patel, V.; Wang, H.; Eun, D.D. Collecting Duct Renal Cell Carcinoma Found to Involve the Collecting System During Partial Nephrectomy: A Case Report. J. Kidney Cancer VHL 2015, 2, 134–139. [Google Scholar] [CrossRef]
  26. Srigley, J.R.; Eble, J.N. Collecting duct carcinoma of kidney. Semin. Diagn. Pathol. 1998, 15, 54–67. [Google Scholar]
  27. Sui, W.; Matulay, J.T.; Robins, D.J.; James, M.B.; Onyeji, I.C.; Roychoudhury, A.; Wenske, S.; DeCastro, G.J. Collecting duct carcinoma of the kidney: Disease characteristics and treatment outcomes from the National Cancer Database. Urol. Oncol. Semin. Orig. Investig. 2017, 35, 540.e13–540.e18. [Google Scholar] [CrossRef]
  28. Zhang, H.; Lu, X.; Huang, G.; Hua, M.; Zhang, W.; Wang, T.; Huang, L.; Wang, Z.; Chen, Q.; Li, J.; et al. A genomic mutation spectrum of collecting duct carcinoma in the Chinese population. BMC Med. Genom. 2022, 15, 1. [Google Scholar] [CrossRef]
  29. Bakouny, Z.; Braun, D.A.; Shukla, S.A.; Pan, W.; Gao, X.; Hou, Y.; Flaifel, A.; Tang, S.; Bosma-Moody, A.; He, M.X.; et al. Integrative molecular characterization of sarcomatoid and rhabdoid renal cell carcinoma. Nat. Commun. 2021, 12, 808. [Google Scholar] [CrossRef]
  30. Pichler, R.; Compérat, E.; Klatte, T.; Pichler, M.; Loidl, W.; Lusuardi, L.; Schmidinger, M. Renal Cell Carcinoma with Sarcomatoid Features: Finally New Therapeutic Hope? Cancers 2019, 11, 422. [Google Scholar] [CrossRef] [Green Version]
  31. Gökden, N.; Nappi, O.; Swanson, P.E.; Pfeifer, J.D.; Vollmer, R.T.; Wick, M.R.; Humphrey, P.A. Renal Cell Carcinoma With Rhabdoid Features. Am. J. Surg. Pathol. 2000, 24, 1329–1338. [Google Scholar] [CrossRef] [PubMed]
  32. Delahunt, B.; Cheville, J.C.; Martignoni, G.; Humphrey, P.A.; Magi-Galluzzi, C.; McKenney, J.; Egevad, L.; Algaba, F.; Moch, H.; Grignon, D.J.; et al. The International Society of Urological Pathology (ISUP) Grading System for Renal Cell Carcinoma and Other Prognostic Parameters. Am. J. Surg. Pathol. 2013, 37, 1490–1504. [Google Scholar] [CrossRef] [PubMed]
  33. Przybycin, C.G.; McKenney, J.K.; Reynolds, J.P.; Campbell, S.; Zhou, M.; Karafa, M.T.; Magi-Galluzzi, C. Rhabdoid Differentiation Is Associated With Aggressive Behavior in Renal Cell Carcinoma. Am. J. Surg. Pathol. 2014, 38, 1260–1265. [Google Scholar] [CrossRef] [PubMed]
  34. Kyriakopoulos, C.E.; Chittoria, N.; Choueiri, T.K.; Kroeger, N.; Lee, J.-L.; Srinivas, S.; Knox, J.J.; Bjarnason, G.A.; Ernst, S.D.; Wood, L.A.; et al. Outcome of Patients With Metastatic Sarcomatoid Renal Cell Carcinoma: Results From the International Metastatic Renal Cell Carcinoma Database Consortium. Clin. Genitourin. Cancer 2014, 13, e79–e85. [Google Scholar] [CrossRef]
  35. Tannir, N.M.; Signoretti, S.; Choueiri, T.K.; McDermott, D.F.; Motzer, R.J.; Flaifel, A.; Pignon, J.-C.; Ficial, M.; Frontera, O.A.; George, S.; 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. 2021, 27, 78–86. [Google Scholar] [CrossRef]
  36. Rini, B.I.; Plimack, E.R.; Stus, V.; Gafanov, R.; Hawkins, R.; Nosov, D.; Pouliot, F.; Soulieres, D.; Melichar, B.; Vynnychenko, I.; et al. Pembrolizumab (pembro) plus axitinib (axi) versus sunitinib as first-line therapy for metastatic renal cell carcinoma (mRCC): Outcomes in the combined IMDC intermediate/poor risk and sarcomatoid subgroups of the phase 3 KEYNOTE-426 study. J. Clin. Oncol. 2019, 37, 4500. [Google Scholar] [CrossRef]
  37. Choueiri, T.; Larkin, J.; Pal, S.; Motzer, R.; Rini, B.; Venugopal, B.; Alekseev, B.; Miyake, H.; Gravis, G.; Bilen, M.; et al. Efficacy and correlative analyses of avelumab plus axitinib versus sunitinib in sarcomatoid renal cell carcinoma: Post hoc analysis of a randomized clinical trial. ESMO Open 2021, 6, 100101. [Google Scholar] [CrossRef]
  38. Pandolfo, S.D.; Carbonara, U.; Beksac, A.T.; Derweesh, I.; Celia, A.; Schiavina, R.; Elbich, J.; Basile, G.; Hampton, L.J.; Cerrato, C.; et al. Microwave versus cryoablation and radiofrequency ablation for small renal mass: A multicenter comparative analysis. Minerva Urol. Nephrol. 2022. ahead of print. [Google Scholar] [CrossRef]
  39. Colomba, E.; Le Teuff, G.; Eisen, T.; Stewart, G.D.; Fife, K.; Larkin, J.; Biondo, A.; Pickering, L.; Srinivasan, A.; Boyle, H.; et al. Metastatic chromophobe renal cell carcinoma treated with targeted therapies: A Renal Cross Channel Group study. Eur. J. Cancer 2017, 80, 55–62. [Google Scholar] [CrossRef]
  40. Buti, S.; Bersanelli, M.; Maines, F.; Facchini, G.; Gelsomino, F.; Zustovich, F.; Santoni, M.; Verri, E.; De Giorgi, U.; Masini, C.; et al. First-Line PAzopanib in NOn–clear-cell Renal cArcinoMA: The Italian Retrospective Multicenter PANORAMA Study. Clin. Genitourin. Cancer 2017, 15, e609–e614. [Google Scholar] [CrossRef]
  41. Campbell, M.T.; Bilen, M.A.; Shah, A.Y.; Lemke, E.; Jonasch, E.; Venkatesan, A.; Altinmakas, E.; Duran, C.; Msaouel, P.; Tannir, N. 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]
  42. Agarwala, V.; Ramaswamy, A.; Joshi, A.; Patil, V.M.; Noronha, V.; Menon, S.; Popat, B.P.; Nilesh, S.; Prabhash, K. Treatment outcomes of metastatic nonclear cell renal cell carcinoma: A single institution retrospective analysis. South Asian J. Cancer 2018, 07, 226–230. [Google Scholar] [CrossRef] [PubMed]
  43. Chanzá, N.M.; Xie, W.; Bilen, M.A.; 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] [PubMed]
  44. Kim, J.K.; Kim, S.H.; Song, M.K.; Joo, J.; Seo, S.I.; Kwak, C.; Jeong, C.W.; Song, C.; Hwang, E.C.; Seo, I.Y.; et al. Survival and clinical prognostic factors in metastatic non-clear cell renal cell carcinoma treated with targeted therapy: A multi-institutional, retrospective study using the Korean metastatic renal cell carcinoma registry. Cancer Med. 2019, 8, 3401–3410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Choudhary, A.; Joshi, A.; Noronha, V.; Menon, N.; Kapoor, A.; Menon, S.; Prabhash, K. 202MO Real-world outcomes of non-clear cell renal cell carcinoma: Retrospective study from tertiary cancer center in India. Ann. Oncol. 2020, 31, S1320. [Google Scholar] [CrossRef]
  46. McKay, R.R.; Bossé, D.; Xie, W.; Wankowicz, S.A.; Flaifel, A.; Brandao, R.; Lalani, A.-K.A.; Martini, D.J.; Wei, X.X.; Braun, D.A.; et al. The Clinical Activity of PD-1/PD-L1 Inhibitors in Metastatic Non–Clear Cell Renal Cell Carcinoma. Cancer Immunol. Res. 2018, 6, 758–765. [Google Scholar] [CrossRef] [Green Version]
  47. Gupta, R.; Ornstein, M.C.; Li, H.; Allman, K.D.; Wood, L.S.; Gilligan, T.; Garcia, J.A.; Von Merveldt, D.; Hammers, H.J.; Rini, B.I. Clinical Activity of Ipilimumab Plus Nivolumab in Patients With Metastatic Non–Clear Cell Renal Cell Carcinoma. Clin. Genitourin. Cancer 2019, 18, 429–435. [Google Scholar] [CrossRef]
  48. Chahoud, J.; Msaouel, P.; Campbell, M.T.; Bathala, T.; Xiao, L.; Gao, J.; Zurita, A.J.; Shah, A.Y.; Jonasch, E.; Sharma, P.; et al. Nivolumab for the Treatment of Patients with Metastatic Non-Clear Cell Renal Cell Carcinoma (nccRCC): A Single-Institutional Experience and Literature Meta-Analysis. Oncologist 2019, 25, 252–258. [Google Scholar] [CrossRef] [Green Version]
  49. Koshkin, V.S.; Barata, P.C.; Zhang, T.; George, D.J.; Atkins, M.B.; Kelly, W.J.; Vogelzang, N.J.; Pal, S.K.; Hsu, J.; Appleman, L.J.; et al. Clinical activity of nivolumab in patients with non-clear cell renal cell carcinoma. J. Immunother. Cancer 2018, 6, 9. [Google Scholar] [CrossRef] [Green Version]
  50. Schwartzman, W.; Elias, R.; Patel, V.M.; Bowman, A.I.; Chintalapati, S.; Kapur, P.; Hammers, H.J.; Brugarolas, J. Safety and efficacy of immune checkpoint inhibitors (ICI) in metastatic non-clear cell renal cell carcinoma (nccRCC): An institutional experience. J. Clin. Oncol. 2020, 38, 640. [Google Scholar] [CrossRef]
  51. Kilari, D.; Szabo, A.; Ghatalia, P.; Rose, T.L.; Dong, H.; Weise, N.; Zhuang, T.Z.; Alloghbi, A.; Jain, R.K.; Alva, A.S.; et al. Outcomes with novel combinations in nonclear cell renal cell carcinoma (nccRCC): ORACLE study. J. Clin. Oncol. 2022, 40, 4545. [Google Scholar] [CrossRef]
  52. Choueiri, T.; Fay, A.; Gray, K.; Callea, M.; Ho, T.; Albiges, L.; Bellmunt, J.; Song, J.; Carvo, I.; Lampron, M.; et al. PD-L1 expression in nonclear-cell renal cell carcinoma. Ann. Oncol. 2014, 25, 2178–2184. [Google Scholar] [CrossRef] [PubMed]
  53. Yakirevich, E.; Patel, N.R. Tumor mutational burden and immune signatures interplay in renal cell carcinoma. Ann. Transl. Med. 2020, 8, 269. [Google Scholar] [CrossRef]
  54. Turajlic, S.; Litchfield, K.; Xu, H.; Rosenthal, R.; McGranahan, N.; Reading, J.L.; Wong, Y.N.S.; Rowan, A.; Kanu, N.; Al Bakir, M.; et al. Insertion-and-deletion-derived tumour-specific neoantigens and the immunogenic phenotype: A pan-cancer analysis. Lancet Oncol. 2017, 18, 1009–1021. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. McDermott, D.F.; Lee, J.-L.; Bjarnason, G.A.; Larkin, J.M.G.; Gafanov, R.A.; Kochenderfer, M.D.; Jensen, N.V.; Donskov, F.; Malik, J.; Poprach, A.; et al. Open-Label, Single-Arm Phase II Study of Pembrolizumab Monotherapy as First-Line Therapy in Patients With Advanced Clear Cell Renal Cell Carcinoma. J. Clin. Oncol. 2021, 39, 1020–1028. [Google Scholar] [CrossRef] [PubMed]
  56. 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]
  57. Atkins, M.B.; Jegede, O.A.; Haas, N.B.; McDermott, D.F.; Bilen, M.A.; Stein, M.; Sosman, J.A.; Alter, R.; Plimack, E.R.; Ornstein, M.; et al. Phase II Study of Nivolumab and Salvage Nivolumab/Ipilimumab in Treatment-Naive Patients With Advanced Clear Cell Renal Cell Carcinoma (HCRN GU16-260-Cohort A). J. Clin. Oncol. 2022, 40, 2913–2923. [Google Scholar] [CrossRef] [PubMed]
  58. Tykodi, S.S.; Gordan, L.N.; Alter, R.S.; Arrowsmith, E.; Harrison, M.R.; Percent, I.; Singal, R.; Van Veldhuizen, P.; 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]
  59. 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, 4537. [Google Scholar] [CrossRef]
  60. Zhang, X.; Yamashita, M.; Uetsuki, H.; Kakehi, Y. Angiogenesis in renal cell carcinoma: Evaluation of microvessel density, vascular endothelial growth factor and matrix metalloproteinases. Int. J. Urol. 2002, 9, 509–514. [Google Scholar] [CrossRef]
  61. Karar, J.; Maity, A. PI3K/AKT/mTOR Pathway in Angiogenesis. Front. Mol. Neurosci. 2011, 4, 51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. 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] [PubMed]
  63. Tannir, N.M.; Jonasch, E.; Albiges, L.; Altinmakas, E.; Ng, C.S.; Matin, S.F.; Wang, X.; Qiao, W.; Lim, Z.D.; 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. 2015, 69, 866–874. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Knox, J.J.; Barrios, C.H.; Kim, T.M.; Cosgriff, T.; Srimuninnimit, V.; Pittman, K.; Sabbatini, R.; Rha, S.Y.; Flaig, T.W.; Page, R.D.; 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, 1339–1345. [Google Scholar] [CrossRef]
  65. Escudier, B.; Molinie, V.; Bracarda, S.; Maroto, P.; Szczylik, C.; Nathan, P.; Negrier, S.; Weiss, C.; Porta, C.; Grünwald, V.; 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. [Google Scholar] [CrossRef]
  66. Hudes, G.; Carducci, M.; Tomczak, P.; Dutcher, J.; Figlin, R.; Kapoor, A.; Staroslawska, E.; Sosman, J.; McDermott, D.; Bodrogi, I.; et al. Temsirolimus, Interferon Alfa, or Both for Advanced Renal-Cell Carcinoma. N. Engl. J. Med. 2007, 356, 2271–2281. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. 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. JAMA Oncol. 2022, 8, 910. [Google Scholar] [CrossRef]
  68. Choueiri, T.K.; Xu, W.; Poole, L.; Telaranta-Keerie, A.; Hartmaier, R.; Powles, T. 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, TPS4601. [Google Scholar] [CrossRef]
  69. Schöffski, P.; Wozniak, A.; Escudier, B.; Rutkowski, P.; Anthoney, A.; Bauer, S.; Sufliarsky, J.; van Herpen, C.; Lindner, L.H.; Grünwald, V.; 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. [Google Scholar] [CrossRef]
  70. Choueiri, T.K.; Vaishampayan, U.; Rosenberg, J.E.; Logan, T.F.; Harzstark, A.L.; Bukowski, R.M.; Rini, B.I.; Srinivas, S.; Stein, M.N.; Adams, L.M.; 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, 181–186. [Google Scholar] [CrossRef]
  71. Twardowski, P.W.; Tangen, C.M.; Wu, X.; Plets, M.R.; Plimack, E.R.; Agarwal, N.; Vogelzang, N.J.; Wang, J.; Tao, S.; Thompson, I.M.; 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, 123–132. [Google Scholar] [CrossRef] [Green Version]
  72. Choueiri, T.K.; Heng, D.Y.C.; Lee, J.L.; Cancel, M.; Verheijen, R.B.; Mellemgaard, A.; Ottesen, L.H.; Frigault, M.M.; L’Hernault, A.; Szijgyarto, Z.; et al. Efficacy of Savolitinib vs. Sunitinib in Patients WithMET-Driven Papillary Renal Cell Carcinoma. JAMA Oncol. 2020, 6, 1247–1255. [Google Scholar] [CrossRef] [PubMed]
  73. 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] [PubMed]
  74. 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, 5004. [Google Scholar] [CrossRef]
  75. Choi, Y.; Keam, B.; Kim, M.; Yoon, S.; Kim, D.; Choi, J.G.; Seo, J.Y.; Park, I.; Lee, J.L. Bevacizumab Plus Erlotinib Combination Therapy for Advanced Hereditary Leiomyomatosis and Renal Cell Carcinoma-Associated Renal Cell Carcinoma: A Multicenter Retrospective Analysis in Korean Patients. Cancer Res. Treat. 2019, 51, 1549–1556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Sulkowski, P.L.; Sundaram, R.K.; Oeck, S.; Corso, C.D.; Liu, Y.; Noorbakhsh, S.; Niger, M.; Boeke, M.; Ueno, D.; Kalathil, A.N.; et al. Krebs-cycle-deficient hereditary cancer syndromes are defined by defects in homologous-recombination DNA repair. Nat. Genet. 2018, 50, 1086–1092. [Google Scholar] [CrossRef] [PubMed]
  77. Lee, W.S.; Yang, H.; Chon, H.J.; Kim, C. Combination of anti-angiogenic therapy and immune checkpoint blockade normalizes vascular-immune crosstalk to potentiate cancer immunity. Exp. Mol. Med. 2020, 52, 1475–1485. [Google Scholar] [CrossRef]
  78. 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]
  79. Powles, T.; Larkin, J.; Patel, P.; Pérez-Valderrama, B.; Rodriguez-Vida, A.; Glen, H.; Thistlethwaite, F.; Ralph, C.; Srinivasan, G.; Mendez-Vidal, M.J.; et al. A phase II study investigating the safety and efficacy of savolitinib and durvalumab in metastatic papillary renal cancer (CALYPSO). J. Clin. Oncol. 2019, 37, 545. [Google Scholar] [CrossRef]
  80. Powles, T.; Mendez-Vidal, M.J.; Rodriguez-Vida, A.; Pérez-Valderrama, B.; Esteban, E.; Thistlethwaite, F.; Patel, P.M.; Herranz, U.A.; Srinivasan, G.; Hamid, A.; et al. CALYPSO: A three-arm randomized phase II study of durvalumab alone or with savolitinib or tremelimumab in previously treated advanced clear cell renal cancer. J. Clin. Oncol. 2022, 40, LBA4503. [Google Scholar] [CrossRef]
  81. 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]
  82. 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] [PubMed]
  83. Halabi, S.; Yang, Q.; Carmack, A.; Zhang, S.; Foo, W.-C.; Eisen, T.; Stadler, W.; Jones, R.; Garcia, J.; Picus, J.; et al. Tissue based biomarkers in non-clear cell RCC: Correlative analysis from the ASPEN clinical trial. Kidney Cancer J. 2021, 19, 64–72. [Google Scholar] [CrossRef] [PubMed]
  84. Shen, M.; Chen, G.; Xie, Q.; Li, X.; Xu, H.; Wang, H.; Zhao, S. Association between PD-L1 Expression and the Prognosis and Clinicopathologic Features of Renal Cell Carcinoma: A Systematic Review and Meta-Analysis. Urol. Int. 2020, 104, 533–541. [Google Scholar] [CrossRef] [PubMed]
  85. Di Meo, N.A.; Lasorsa, F.; Rutigliano, M.; Loizzo, D.; Ferro, M.; Stella, A.; Bizzoca, C.; Vincenti, L.; Pandolfo, S.D.; Autorino, R.; et al. Renal Cell Carcinoma as a Metabolic Disease: An Update on Main Pathways, Potential Biomarkers, and Therapeutic Targets. Int. J. Mol. Sci. 2022, 23, 14360. [Google Scholar] [CrossRef]
  86. Gudas, L.J.; Fu, L.; Minton, D.R.; Mongan, N.P.; Nanus, D.M. The role of HIF1α in renal cell carcinoma tumorigenesis. J. Mol. Med. 2014, 92, 825–836. [Google Scholar] [CrossRef]
  87. Jonasch, E.; Donskov, F.; Iliopoulos, O.; Rathmell, W.K.; Narayan, V.K.; Maughan, B.L.; Oudard, S.; Else, T.; Maranchie, J.K.; Welsh, S.J.; et al. Belzutifan for Renal Cell Carcinoma in von Hippel–Lindau Disease. N. Engl. J. Med. 2021, 385, 2036–2046. [Google Scholar] [CrossRef]
  88. Ooi, A.; Furge, K.A. Fumarate hydratase inactivation in renal tumors: HIF1 alpha, NRF2, and “cryptic targets” of transcription factors. Chin. J. Cancer 2012, 31, 413–420. [Google Scholar] [CrossRef]
Table 1. Incidence of RCC subtypes [12].
Table 1. Incidence of RCC subtypes [12].
Subtype% of all RCC
Papillary RCC (Types 1 and 2)10–15
Chromophobe RCC5
Collecting duct RCC1
MiT family translocation RCC1
Multilocular cystic renal neoplasm of low
malignant potential
1
Medullary RCC<1%
Tubulocystic RCC<1%
Acquired cystic kidney disease-associated RCC<1%
Hereditary leiomyomatosis with RCC<1%
Succinate dehydrogenase deficient RCC<1%
Unclassified <1%
Table 2. Selected retrospective studies of targeted and immunotherapy in nccRCC [39,40,41,42,43,44,45,46,47,48,49,50,51].
Table 2. Selected retrospective studies of targeted and immunotherapy in nccRCC [39,40,41,42,43,44,45,46,47,48,49,50,51].
TrialYearTreatmentSample SizeResults
mOS (mths)mPFS (mths)ORROther
Endpoints
Colomba et al. [39]2017VEGF/mTORi
VEGFi: 82%
mTORi: 18%
n = 61VEGFi: 22.9
(95% CI 17.8–49.2)
mTORi: 3.2
(95% CI 2.3–NE)
N/AVEGFi: 28.9%
mTORi: 0%
mTTF
VEGFi: 8.0mths
(95% CI 4.1–13.6)
mTORi: 2.3mths
(95% CI 0.7–8.0)
Buti et al. [40]2017Pazopanibn = 37
Papillary: 51%
Chromophobe: 24%
Unclassified: 21%
17.3
(95% CI 11.5–23.0)
15.9
(95% CI 5.9–25.8)
Overall: 27%
Papillary: 21%
Chromophobe: 44%
DCR: 81%
Campbell et al. [41]2018Cabozantinibn = 3025.4
(95% CI 15.5–35.4)
8.6
(95% CI 6.1–14.7)
14.3%DCR: 78.6%
Agarwala et al. [42]2018VEGF/mTORi
Sorafenib: 35%
Sunitinib: 22.5%
Pazopanib: 20%
Everolimus: 17.5%
n = 40
Papillary: 62.5%
Sarcomatoid: 15%
Chromophobe: 12.5%
11.7N/AN/AmEFS: 6.1mths
Chanza et al. [43]2019Cabozantinibn = 112
Papillary: 59%
Translocation: 15%
Unclassified: 13%
Chromophobe: 9%
12.0
(95% CI 9.2–17.0)
7.0
(95% CI 5.7–9.0)
Overall: 27%
Papillary: 27%
Chromophobe: 30%
Collecting duct: 50%
Unclassified: 13%
mTTF: 6.7mths
Kim et al. [44]2019VEGFi vs. mTORin = 156
Papillary: 59.6%
Chromophobe: 12.8%
Collecting duct: 11.5%
Unclassified: 10.3%
N/A10.0 vs. 5.0 (p = 0.0275)N/AmCSS:
27.0 vs. 16.0 (p = 0.1706)
Choudhary et al. [45]2020VEGFi
Sorafenib: 39.2%
Sunitinib: 27.4%
Pazopanib: 21.6%
n = 139
Papillary: 76.2%
Chromophobe: 7.9%
Sarcomatoid: 6.5%
Unclassified: 1.4%
11.9
(95% CI 5.4–18.4)
6.0
(95% CI 2.4-9.6)
16.6%DCR: 54.1%
McKay et al. [46]2018PD-1/PD-L1
inhibitors
n = 43
Papillary: 33%
Chromophobe: 23%
Unclassified: 21%
Sarcomatoid: 16%
Translocation: 7%
12.9
(95% CI 7.4–NR)
N/AOverall: 19%
Papillary: 29%
Chromophobe: 0%
Translocation: 33%
Unclassified: 0%
mTTF: 4.0mths (95% CI 2.8–5.5)
Gupta et al. [47]2020Ipilimumab/
Nivolumab
n = 18N/A7.1Overall: 33.3%
Chromophobe: 20%
Medullary: 0%
Papillary T2: 50%
Translocation: 0%
Unclassified: 33.3%
N/A
Chahoud et al. [48]2020Nivolumabn = 40
Papillary: 30%
Unclassified: 27.5%
Chromophobe: 12.5%
Translocation: 7.5%
21.7
(95% CI 7.83–NR)
4.9
(95% CI 3.53–10.27)
Overall: 21.6%
Papillary T1: 25%
Papillary T2: 0%
Chromophobe: 0%
Translocation: 0%
Unclassified: 44.4%
DCR: 53.4%
Koshkin et al. [49]2018Nivolumabn = 41
Papillary: 39%
Unclassified: 34%
Chromophobe: 12%
Collecting duct: 10%
NR3.5
(95% CI 1.9–5.0)
Overall: 20%
Papillary: 14%
Chromophobe: 0%
Collecting duct: 25%
Translocation: 0%
Unclassified: 36%
10mth OS: 68%
Schwartzmann et al. [50]2020Nivolumab: 46%
Ipilimumab/
nivolumab: 54%
n = 28
Unclassified: 42.9%
Papillary: 28.6%
Chromophobe: 10.7%
HLRCC: 10.7%
15.9
(95% CI 5.9–25.9)
N/A10.7%mTNT: 4.9
(95% CI 1.7–8.1)
ORACLE [51]2021IO/VEGF
(Pembrolizumab/
axitinib,
atezolizumab/
bevacizumab, avelumab/
axitinib)
n = 1924.716.821%mDOR: 23.6mths
DCR: 69%
IO/IO
(ipilimumab/
nivolumab)
n = 4019.213.619%mDOR: 13.6mths
DCR: 46%
VEGF/mTORi
(lenvatinib/
everolimus)
n = 723.1NR0%mDOR: NR
DCR: 72%
mOS: median overall survival, mPFS: median progression free survival, ORR: objective response rate, mEFS: median event free survival, DCR: disease control rate, mTTF: median time to treatment failure, mCSS: median cancer specific survival, mTNT: median time to next treatment, mDOR: median duration of response.
Table 3. Prospective single arm immunotherapy trials in nccRCC [55,56,57,58,59].
Table 3. Prospective single arm immunotherapy trials in nccRCC [55,56,57,58,59].
TrialPhaseCohortTreatmentSample SizeEfficacy
mOS (mths)mPFS (mths)ORR
KEYNOTE-427
[55]
IIA
(Clear Cell)
Pembrolizumab n = 110NR7.1
(95% CI 5.6–11.0)
36.4%
B
(Non-clear cell)
n = 165
Papillary: 71.5%
Unclassified: 15.8%
Chromophobe: 12.7%
28.9
(95% CI 24.3-NR)
4.2
(95% CI 2.9–5.6)
26.7%
Papillary: 28.8%
Chromophobe: 9.5%
Unclassified: 30.8%
CheckMate-374
[56]
III/IVA
(Clear Cell)
Nivolumab n = 9721.8
(95% CI 17.4-NE)
3.6
(95% CI 2.0–5.5)
22.7%
B
(Non-clear cell)
n = 44
Papillary: 54.5%
Chromophobe: 15.9%
Unclassified: 18.2%
Translocation: 5%
Other: 6%
16.3
(95% CI 9.2-NE)
2.2
(95% CI 1.8–5.4)
13.6%
CheckMate-920
[58]
III/IVCohort 2 (nccRCC with KPS ≥70%) ^Ipilimumab/
nivolumab > nivolumab
n = 52
Unclassified: 42.3%
Papillary: 34.6%
Chromophobe: 13.5%
Translocation: 3.8%
Collecting duct: 3.8%
Medullary: 1.9%
21.2
(95% CI 16.6-NE)
3.7 (95% CI 2.7–4.6)19.6%
HCRN
GU16-260
[57]
IIA
(Clear Cell)
Nivolumab
(Part A) +/−
ipilimumab * (Part B)
n = 123N/A7.4 (95% CI 5.5–10.9)Part A: 29.3%
Part B: 11%
B
(Non-clear cell)
n = 35
Papillary: 54%
Chromophobe: 17%
Unclassified: 29%
Part B: n = 16
N/A4.0 (95% CI 2.7–4.3)Part A: 14.3%
Part B: 6%
UNISoN
(ANZUP 1602)
[59]
IIN/A (nccRCC cohort only)Nivolumab +/−
ipilimumab **
n = 85Part 1+2:
24
(95% CI 16–28)
Part 2 (nivo+ipi): 10 (95% CI 6–17)
Part 2: 2.6
(95% CI 2.2–3.8)
Nivo: 17%
Nivo + ipi: 10%
KPS: Karnosfky Performance Score. ^CheckMate-920 included 4 cohorts: predominantly ccRCC with KPS ≥ 70% (cohort 1), nccRCC with KPS≥70% (cohort 2), cc/nccRCC with nonactive brain metastases and KPS≥70% (cohort 3), cc/nccRCC with KPS 50–60% (cohort 4). * Salvage ipilimumab (1mg/kg) and nivolumab (3mg/kg) was administered if PD or SD was found 48 weeks after initiation. ** Salvage ipilimumab (1 mg/kg) was administered in patients refractory to single agent nivolumab at 3 months.
Table 4. Prospective targeted therapy trials in nccRCC [62,63,64,65,66,67].
Table 4. Prospective targeted therapy trials in nccRCC [62,63,64,65,66,67].
TrialPhaseTreatmentComparatorSample SizeEfficacy
mOS (mths)mPFS (mths)ORR
ASPEN [62]IISunitinibEverolimusn = 10831.5 vs. 13.2
HR 1.12
(95% CI 0.7–2.1, p = 0.60)
8.3 vs. 5.6
HR 1.41
(80% CI 1.03–1.92, p = 0.16)
18% vs. 9%
ESPN [63]IISunitinibEverolimusn = 68
(Crossover allowed at PD)
16.2 vs. 14.9 (p = 0.18)1L: 6.1 vs. 4.1 (p = 0.6)
2L: 1.8 vs. 2.8 (p = 0.6)
1L: 9% vs. 3%
2L: 9.5% vs. 8.6%
RECORD-3 [64]II1L everolimus/
2L sunitinib
1L sunitinib/
2L everolimus
n = 47122.4
(95% CI 18.6-33.3)
vs
29.5
(95% CI 22.8–33.1)
21.7
(95% CI 15.1–26.7)
vs
22.2
(95% CI 16.0–29.8)
N/A
RAPTOR [65]IIEverolimusN/An = 92
Papillary: 78%
21.4 (95% CI 15.4–28.4)T1 Papillary: 7.9 (95% CI 2.1–11.0)
T2 Papillary: 5.1 (95% CI 3.3–5.5)
1%
ARCC [66]IIIInterferon alfaTemsirolimus/Combination therapyn = 6267.3 vs. 10.9 vs. 8.43.1 vs. 5.5 vs. 4.74.8% vs. 8.6% vs. 8.1%
BONSAI [67]IICabozantinibN/An = 23
(Collecting duct RCC only)
7.0 (95% CI 3–31)4.0 (95% CI 3–13)35%
(95% CI 15–57)
Table 5. MET inhibitor trials in papillary RCC [69,70,71,72,73].
Table 5. MET inhibitor trials in papillary RCC [69,70,71,72,73].
TrialPhaseTreatmentSample SizeEfficacy
mOS (mths)mPFS (mths)ORR
CREATE
(T1 Papillary RCC) [69]
IICrizotinibn = 23
(MET-altered: n = 4)
N/AN/AMET positive cohort: 50%
(2/4 patients)
MET negative cohort: 6.3% (1/16 patients)
NCT00726323 [70]IIForetinibn = 74
(MET-altered: n = 36)
NR9.3Overall
population: 13.5%
Germline MET mutation
positive: 50% (5/10 patients)
Germline MET mutation
negative: 9% (5/57 patients)
SWOG S1107 [71]IITivantinib/erlotinib
vs
Tivantinib
n = 50
Tivantinib/
erlotinib: n = 25
Tivantinib: n = 25
(MET-altered: n = 1)
10.3 vs. 11.35.4 vs. 2.00% in both arms
SAVOIR [72]IIISavolitinib vs. Sunitinibn = 60 (all MET-driven)
Savolitinib: n = 33
Sunitinib: n = 27
NR vs. 13.2
(HR 0.51, 95% CI 0.2–1.2; p = 0.11)
7.0 vs. 5.6
(HR 0.71; 95% CI 0.4–1.4)
27% vs. 7%
PAPMET [73]IISavolitinib/
Crizotinib/
Sunitinib/
Cabozantinib
n = 152
(MET testing not performed)
N/ANo improvement in PFS with savolitinib/
crizotinib vs. VEGFi
(halted early
after
prespecified
futility
analysis)
N/A
Cabozantinib vs. SunitinibN/A9.0 vs. 5.6
HR 0.60
Table 6. Completed single arm studies of IO plus targeted therapy in nccRCC [78,79,81,82].
Table 6. Completed single arm studies of IO plus targeted therapy in nccRCC [78,79,81,82].
TrialPhaseTreatmentSample SizeEfficacy
NCT02724878 [81]IIAtezolizumab/
Bevacizumab
n = 60
Papillary: n = 12
Chromophobe: n = 10
Unclassified: n = 9
Collecting duct: n = 5
Medullary: n = 1
ORR: 33%
Papillary: 25%
Chromophobe: 10%
Unclassified: 33%
Collecting duct: 40%
Medullary: 100%
COSMIC-021 [82]Ib/IIAtezolizumab/
Cabozantinib
n = 32 (nccRCC cohort)
Papillary: n = 15
Chromophobe: n = 9
Other: n = 7
mPFS: 9.5mths
ORR 31% (80% CI: 20-44)
Papillary: 40%
Chromophobe: 14%
Other: 60%
CALYPSO [79]I/IIDurvalumab/
Savolitinib
n = 41
Papillary: n = 40
ORR 27%
mOS: 12.3mths
mPFS: 4.9mths
NCT03635892 [78]IINivolumab/
Cabozantinib
n = 47
Cohort 1 (papillary,
unclassified,
translocation associated RCC): n = 40
Cohort 2 (chromophobe): n = 7
Cohort 1: ORR 47.5%, mPFS 12.5mths,
mOS: 28mths
Cohort 2: ORR 0%
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

John, A.; Spain, L.; Hamid, A.A. Navigating the Current Landscape of Non-Clear Cell Renal Cell Carcinoma: A Review of the Literature. Curr. Oncol. 2023, 30, 923-937. https://doi.org/10.3390/curroncol30010070

AMA Style

John A, Spain L, Hamid AA. Navigating the Current Landscape of Non-Clear Cell Renal Cell Carcinoma: A Review of the Literature. Current Oncology. 2023; 30(1):923-937. https://doi.org/10.3390/curroncol30010070

Chicago/Turabian Style

John, Alexius, Lavinia Spain, and Anis A. Hamid. 2023. "Navigating the Current Landscape of Non-Clear Cell Renal Cell Carcinoma: A Review of the Literature" Current Oncology 30, no. 1: 923-937. https://doi.org/10.3390/curroncol30010070

Article Metrics

Back to TopTop