Non–Clear Cell Renal Cell Carcinoma, Part 2: Therapy

Volume 13, Issue 6 June 2015

Loana B. Valenca, MD, Michelle S. Hirsch, MD, PhD, Toni K. Choueiri, MD, and Lauren C. Harshman, MD

The authors are affiliated with the Dana-Farber Cancer Institute, Brigham and Women’s Hospital, and Harvard Medical School in Boston, Massachusetts. Dr Valenca is a visiting physician, Dr Hirsch is an associate professor and chief of the genitourinary pathology service in the Department of Pathology at Brigham and Women’s Hospital, Dr Choueiri is an associate professor and the clinical director of the Lank Center for Genitourinary Oncology at the Dana-Farber Cancer Institute, and Dr Harshman is an assistant professor at the Lank Center for
Genitourinary Oncology at the Dana-Farber Cancer Institute.

Lauren C. Harshman, MD

Lank Center for Genitourinary Oncology

Dana-Farber Cancer Institute

Boston, MA 02215

Tel: 617-632-4524

Fax: 617-632-2165

E-mail: laurenc_harshman@dfci.harvard.edu

Abstract: Non–clear cell renal cell carcinomas (RCCs) represent a heterogeneous group of diseases with distinct molecular drivers, histologies, and clinical outcomes. Their low incidence and heterogeneity have resulted in a lack of studies that address the optimal strategies for each subtype. This article (the second in a 2-part series) reviews the current targeted therapies approved for RCC, such as the vascular endothelial growth factor receptor tyrosine kinase inhibitors and the mammalian target of rapamycin inhibitors. Ongoing studies will provide more information regarding the role of these agents in non–clear cell RCC. Ultimately, enhanced understanding of genetic triggers and the development of more tailored treatments remain imperative to improve outcomes in non–clear cell RCC.

Introduction

This article addresses the treatment of non–clear cell renal cell carcinoma (RCC). Non–clear cell RCC includes but is not limited to papillary RCC, chromophobe RCC, collecting duct carcinoma (CDC), renal medullary carcinoma, and renal carcinoma associated with an Xp11.2 translocation. In addition, RCC can be characterized by the presence or absence of a sarcomatoid component (these subtypes are discussed in part 1 of this 2-part series).

Although most clear cell RCCs are sporadic and not attributable to von Hippel-Lindau disease, somatic inactivation of the VHL gene is present in the majority of cases1 and results in altered VHL protein function. The VHL protein negatively regulates hypoxia-inducible factor, which activates genes involved in cell proliferation, neovascularization, and extracellular matrix formation.2 Consequently, RCC represents one of the best clinical models for therapies designed to address alterations in this hypoxia-inducible pathway. Since 2005, multiple agents that target components of this pathway have been approved for the treatment of clear cell RCC. These agents are broadly known as the targeted molecular therapies and have demonstrated a wider range of efficacy and enhanced tolerability compared with traditional cytokine-based immunotherapies, such as interferon-α and high-dose interleukin-2. Seven targeted agents are now available: sunitinib (Sutent, Pfizer), sorafenib (Nexavar, Bayer/Onyx), pazopanib (Votrient, GlaxoSmithKline), axitinib (Inlyta, Pfizer), everolimus (Afinitor, Novartis), temsirolimus (Torisel, Wyeth), and bevacizumab (Avastin, Genentech).3-9 However, data are limited regarding the activity of these drugs in non–clear cell RCC histologies, because most of the clinical trials included only clear cell disease. Previous studies evaluating cytokines and cytotoxic chemotherapy in non–clear cell RCC have shown minimal activity, perhaps with the exception of chemotherapy with CDC.10

Treatment of Metastatic Disease

Once metastatic, non–clear cell RCC histologies are generally characterized by resistance to traditional systemic therapies, and survival rates are low.10-12 Small studies, meta-analyses, expanded-access trials (Table 1), and ongoing prospective trials (Table 2) have tried to address the utility of various agents in non–clear cell RCC.

VEGF Pathway Targeted Agents in Non–Clear Cell RCC

Although loss of function of the VHL gene is not detected in non–clear cell RCC, vascular endothelial growth factor (VEGF) receptors and their ligands can be overexpressed in some subtypes, such as papillary RCC and chromophobe RCC.13,14 However, in the reported retrospective series and prospective trials, the response rates with the tyrosine kinase inhibitors generally have been disappointing compared with those observed in clear cell RCC (Table 2).11,12,15-28 One retrospective review of 21 patients with non–clear cell RCC treated with sunitinib reported a modest overall response rate (ORR) of 14.3%, and overall clinical benefit (objective response or disease stabilization) in 52.4%.12 Choueiri and colleagues retrospectively compared the effects of sorafenib and sunitinib in 53 patients with papillary (77%) and chromophobe (23%) histologies.15 Three (25%) of 12 chromophobe RCC patients achieved a response with sorafenib (n=2) or sunitinib (n=1). Median progression-free survival (PFS) was 10.6 months. In the papillary RCC subgroup, sunitinib achieved a better ORR and longer median PFS at 4.8% and 11.9 months, respectively, compared with sorafenib, which induced no objective responses and produced a median PFS of 5.1 months. Although the number of patients was too small to draw definitive conclusions, the prolonged PFS from sunitinib in papillary RCC was comparable to the data from the large phase 3 trial in treatment-naive, metastatic clear cell RCC patients.3 Ethnicity and a patient’s underlying genetics may also play a role in response. In a small retrospective analysis of 31 Korean patients, most of whom had type 2 papillary RCC, 11 patients (35%) achieved a partial response and 17 (55%) experienced disease stabilization.16 Median PFS was 6.4 months, and median overall survival (OS) was 25.6 months.

The sorafenib expanded access trial provided further insight into the efficacy of sorafenib in 202 patients with non–clear cell RCC histologies.18 In the 107 patients with papillary RCC, partial response and stable disease were observed in 3% and 81%, respectively. In the 20 chromophobe RCC patients, partial response and stable disease were observed in 5% and 85%, respectively. Overall, median PFS was 24 weeks in the non–clear cell RCC subset. In the sunitinib expanded access trial, which enrolled 4371 patients with RCC, 588 patients (13%) had non–clear cell RCC histologies, of whom 11% achieved an objective response.19 Median PFS was 7.8 months, and median OS was 13.4 months. Two other prospective studies encompassing various subtypes of non–clear cell RCC (mostly papillary) have observed response rates of approximately 5%, and stable disease in 53% to 68% of patients.11,20 Overall, chromophobe RCC appears to have better outcomes, consistent with its known better prognosis irrespective of treatment.29

In a prospective phase 2 study, the French Genito-Urinary Group (GETUG) specifically assessed response to VEGF inhibition using sunitinib in 28 treatment-naive patients with papillary RCC.21 Response rate was evaluable in 22 of the 28 patients. Only 1 patient experienced a partial response, but the majority achieved disease stabilization (72.7%; n=16/22).

Despite the poor prognosis of Xp11.2 translocation carcinoma in adults, response to VEGF-targeted agents has been observed, as described in retrospective series and case reports.22,23 Objective responses and PFS seem to be similar to or perhaps slightly lower than those reported for clear cell RCC, at 20% and 7.1 months, respectively.23

With respect to CDC, a few small series and case reports support the use of VEGF inhibitors.24,25 In 1 report, partial response or stable disease was observed in 23% of the patients, and median OS was 4 months.24 Given the aggressive, highly proliferative nature of CDC, platinum- or taxane-based chemotherapy is generally tried first; however, VEGF-targeted agents can be considered for patients unfit for chemotherapy or as second-line options.

VEGF inhibitors can also be effective in RCC with a sarcomatoid component.26-28 One study with 23 patients receiving sunitinib observed objective responses in 30% and stable disease in 22%.26 Another retrospective study analyzed responses to sunitinib, sorafenib, or bevacizumab alone or in combination.27 Overall, 19% of the 43 patients experienced a partial response and nearly half had disease stabilization (49%). Patients with a sarcomatoid component of less than 20% achieved better outcomes.

mTOR Inhibitors

Several downstream effectors of the mammalian target of rapamycin (mTOR) pathway are overexpressed and possibly activated in clear cell RCC as well as the other -histologic subtypes.30 The use of mTOR inhibitors in non–clear cell RCC was supported by a phase 3 registration trial that compared the efficacy and safety of temsirolimus alone vs temsirolimus in combination with interferon-α (IFN-α) or IFN-α alone for the first-line treatment of poor-prognosis RCC.5 In all histologies, response rates were similarly low in all 3 arms, and ranged from 7% to 11%. However, median OS was significantly longer in the temsirolimus monotherapy arm compared with the other 2 arms (10.9 months for temsirolimus, 7.3 months for IFN-α, and 8.4 months for the combination; P=.0069). This study was notable for being the first phase 3 trial of the targeted therapies in RCC that permitted patients with non–clear cell RCC to enroll. Exploratory analyses of non–clear cell RCC and indeterminate histologies (~20% of the patients) showed comparable median OS between clear cell RCC and non–clear cell RCC at 10.7 months and 11.6 months, respectively.31

Everolimus has been prospectively tested in a small phase 2 study of 43 Asian patients and a large, expanded-access program (REACT; RAD001 Expanded Access Clinical Trial in RCC).32,33 In the REACT study, 5.5% (n=75/1367) of patients had non–clear cell RCC. Everolimus elicited similar results in non–clear cell RCC as compared with clear cell RCC.33,34 The median treatment duration was approximately 3 months, and objective responses were similarly dismal in both the non–clear cell and clear cell histologies (1.3% vs 1.7%, respectively). Disease stabilization appears to be the hallmark of clinical benefit with the mTOR inhibitors, and similar rates were observed in the non–clear cell and clear cell cohorts (49.3% vs 51.6%, respectively).

Further indication of the general efficacy of first line mTOR inhibitors in non–clear cell RCC comes from an established international metastatic RCC (mRCC) database, in which retrospective analysis characterized their use and efficacy.35 Of the 127 patients identified, 51 patients (40.2%) had non–clear cell RCC histology and 24 (18.9%) had sarcomatoid features. The majority received temsirolimus (73%). The principal reasons for first-line therapy with an mTOR inhibitor over VEGF blockade were poor-risk status, non–clear cell RCC histologies, and clinical trials. In non–clear cell disease, median PFS was 4.8 months for temsirolimus and 3.3 months for everolimus. Median OS was 14.3 months for temsirolimus and 20.6 months for everolimus in non–clear cell RCC.

A large randomized phase 2 study, RECORD-3 (Renal Cell Cancer Treatment With Oral RAD001 Given Daily), compared first-line everolimus followed by sunitinib at progression with the standard sequence of first-line sunitinib followed by everolimus.36 Of the 471 patients with metastatic RCC, 66 patients (14%) had non–clear cell RCC histology. Everolimus did not achieve noninferiority compared with sunitinib as a first-line therapy. Median PFS was shorter for first-line everolimus (7.9 months) than for first-line sunitinib (10.7 months). The non–clear cell RCC subgroup had a similarly inferior PFS in the everolimus arm (5.1 vs 7.2 months). Overall, the trial results supported the standard sequence of sunitinib followed by everolimus at progression.36

The ESPN trial (Everolimus Versus Sunitinib Prospective Evaluation in Metastatic Non-Clear Cell Renal Cell Carcinoma) was the first direct comparison of VEGF blockade and mTOR inhibition in treatment-naive, non–clear cell RCC.37 All subtypes of non–clear cell RCC and clear cell RCC patients with at least a 20% sarcomatoid component were permitted. The primary objective was to assess whether everolimus would elicit an increase in PFS to 20 weeks, vs a baseline estimate of 12 weeks with sunitinib. Patients could cross over to the other arm at progression. Everolimus and sunitinib both yielded modest efficacy. Patients in the first-line sunitinib arm had trends for longer median PFS (6.1 vs 4.1 months) and OS (16.2 vs 14.9 months), but these were not statistically significant. The ORR for first-line therapy was observed only in patients with chromophobe histology and was 2.8% for everolimus and 6% for sunitinib. Compared with all -histologies, patients with chromophobe RCC achieved a longer median OS (31.6 months in the sunitinib arm and 25.1 months in the everolimus arm).37 Ultimately, this trial was discontinued early for futility at the interim analysis of OS.

The ongoing phase 2 ASPEN (Phase II Study of Afinitor vs. Sutent in Patients With Metastatic Non-Clear Cell Renal Cell Carcinoma) trial continues to assess the antitumor activity of sunitinib and everolimus in non–clear cell disease. In this study, special emphasis is placed on papillary and chromophobe subtypes. Clear cell RCC with sarcomatoid differentiation, medullary carcinoma, and CDC are excluded (NCT01108445).

Pal and colleagues observed overexpression of Aurora A kinase and increased activity of the mTOR pathway in sarcomatoid areas of RCC samples.38 For this reason, mTOR and Aurora kinase inhibitors may be reasonable therapeutic options for metastatic RCC with a sarcomatoid component. In 1 retrospective study including patients with clear cell RCC with sarcomatoid features, of the 23 patients with a sarcomatoid component, median PFS was 3.5 months and median OS was 8.2 months.39

MET Inhibitors

MET inhibition is a rational strategy in RCC.40 Activating mutations of the MET gene are associated with the majority of hereditary papillary type 1 RCC and a small percentage of sporadic papillary RCC.41,42 However, MET activation can occur in all types of papillary RCC owing to increased gene copy number or upregulation of coactivators.42 Increased c-MET expression has been observed in clear cell RCC and may be explained by the fact that, at least in vitro, inactivating VHL evokes constitutive phosphorylation of MET.43 It has been speculated that phosphorylation of the MET protein modifies the intercellular adherence structure, which may induce tumor cell proliferation and resultant oncogenesis.43

Several c-MET targeted agents have been tested in RCC, including foretinib, rilotumumab, tivantinib, and cabozantinib (Cometriq, Exelixis). Foretinib is a tyrosine kinase inhibitor targeting MET, VEGF, and multiple other receptors. In a phase 2 study of 74 papillary RCC patients, Choueiri and colleagues observed evidence of clinical efficacy.44 Although the ORR of 13.5% did not meet the 25% predefined response rate, the PFS of 9.3 months compared favorably with the VEGF receptor inhibitor experience in clear cell disease. Moreover, the activity in patients with a MET germline mutation was especially notable, with 50% of patients achieving a partial response in contrast to 9% whose tumors did not express the mutation.44

Rilotumumab (AMG 102) is a fully human monoclonal antibody that specifically targets hepatocyte growth factor/scattered factor (HGF/SF). A phase 2 study was performed in 3 patients with metastatic RCC, -including all histologies, most of whom had disease refractory to prior systemic therapy.45 Of 7 patients with papillary RCC, four had stable disease. No objective responses were elicited, and median PFS was 3.4 months.

Tivantinib is a selective, noncompetitive inhibitor of c-MET.46 One of the 4 phase 1 solid tumor trials included 5 patients with non–clear cell RCC.47 Stabilization of disease was the best response in 3 patients with non–clear cell RCC. Tivantinib was also evaluated in a phase 2 trial, in which 6 patients with Xp11.2 translocation RCC were enrolled.48 Interestingly, the MET receptor gene is upregulated by microphthalmia transcription factor (MITF), making MET inhibitors logical for the treatment for MITF-associated tumors.49 However, stable disease was achieved in only 3 patients with a disappointing PFS of 2 months, implying negligible efficacy in this aggressive disease.48

Cabozantinib, a multityrosine kinase inhibitor against multiple receptors, including c-MET and the VEGF receptor, has shown promise in clear cell RCC, with an ORR of 28% and median PFS of 12.9 months in a treatment-refractory population.50 Ongoing phase 2 and 3 studies are evaluating this agent further in RCC (NCT01865747; NCT01835158).

Cytotoxic Chemotherapy 

In general, RCC is thought to be resistant to cytotoxic chemotherapy, with objective responses generally elicited in less than 5% of patients.51 Modest efficacy has been observed in small series and in more aggressive subtypes (Table 2). For example, in a phase 2 study of 51 patients with metastatic non–clear cell RCC, capecitabine showed activity with an objective response rate of 26% and disease stabilization in 47% of patients.52 Median PFS was 10.1 months, and OS was 18.3 months. Responses to combination chemotherapy also have been reported in patients with CDC and sarcomatoid variants of RCC. A phase 2 trial reported that the combination of gemcitabine and platinum agents (cisplatin or carboplatin) was active in CDC.53 In 23 patients, objective responses were achieved in 26%, including 1 complete response, and stable disease was achieved in 44%, for an overall clinical benefit rate of 70%. Median PFS was 7.1 months, and median OS was 10.5 months. One small series and 1 case report have demonstrated that the addition of bevacizumab in this combination was also effective and may enhance activity.54,55 Of the 6 patients who were treated with cisplatin/gemcitabine/bevacizumab, there were 2 complete responses, 3 partial responses, and 1 case of stable disease. Median PFS was promising at 15.1 months, with a median OS of 27.8 months.55

Given the more aggressive nature of tumors with sarcomatoid differentiation, chemotherapeutic approaches with agents such as gemcitabine and doxorubicin have been attempted. Various studies have demonstrated objective response rates ranging from 16% to 39%, including some complete responses.56-58 Conversely, a smaller study testing gemcitabine with doxorubicin59 and another study with doxorubicin and ifosfamide did not produce any objective responses in these variants.60 Chemotherapy often is used initially for patients with RCC with sarcomatoid differentiation. However, given the similar responses observed with the VEGF inhibitors at least retrospectively, first-line VEGF inhibition is also reasonable. Ongoing and maturing clinical trials are testing combinations of chemotherapy and VEGF blockade in RCC with sarcomatoid differentiation. The final results of a phase 2 trial testing sunitinib plus gemcitabine in sarcomatoid RCC or high-risk RCC were recently presented. In the group with the sarcomatoid component, the ORR was 26% and the SD was 38%.61 An ongoing phase 2 cooperative group trial is evaluating sunitinib vs sunitinib/gemcitabine (NCT00556049). Finally, another phase 2 study is assessing the safety and efficacy of capecitabine and gemcitabine plus the VEGF antibody bevacizumab in patients with RCC with sarcomatoid differentiation (NCT00496587).

Cytoreductive Nephrectomy and Metastasectomy for Non–Clear Cell RCC

In the immunotherapy era, 2 randomized clinical trials demonstrated an OS benefit to adding cytoreductive nephrectomy prior to systemic therapy with IFN-α in patients with metastatic RCC.62,63 Recently, 2 retrospective studies compared survival in patients with metastatic disease who had cytoreductive nephrectomy in addition to treatment with a targeted therapy.64,65 Most patients appeared to benefit from primary tumor removal, except for those with poor prognostic features according to International mRCC Database Consortium (IMDC) criteria.66 Two ongoing phase 3 trials are prospectively evaluating the importance of nephrectomy in metastatic clear cell RCC treated with sunitinib (NCT00930033; NCT01099423).

There are few data on the role of cytoreductive nephrectomy in metastatic non–clear cell RCC. In a retrospective population-based study of 591 non–clear cell RCC patients treated between 2000 and 2009, patients who underwent cytoreductive nephrectomy had lower cancer-specific and all-cause mortality than those who did not.67 An interaction model found lower all-cause mortality for all histologies after cytoreductive nephrectomy. Another retrospective study has investigated the outcomes of cytoreductive nephrectomy for clear cell and non–clear cell RCC patients who had impaired performance status.68 Only 37.5% of patients who had a low Eastern Cooperative Oncology Group (ECOG) performance score of 2 or 3 experienced an improvement in their performance status in the postoperative period and only 57.5% received postoperative systemic therapy. Median disease-specific survival for this subgroup was 6.6 months. The investigators did observe that a subset of patients with an ECOG performance status of 2 or 3 who were symptomatic from bone metastasis may have derived greater benefit from cytoreductive nephrectomy than patients who were symptomatic owing to visceral metastases (median disease-specific survival: 17.7 months and 2.1 months, respectively; P=.006).68 Collectively, these studies show that cytoreductive nephrectomy can be considered for non–clear cell RCC patients, especially when taking into account life expectancy, sites of metastases, and performance status.

In terms of metastasectomy for non–clear cell disease, there have been a few case reports that have demonstrated long-term survival after resection.69,70 Larger series of all subtypes of RCC have shown that metastasectomy may improve OS and potentially elicit cure, especially in the setting of solitary or oligometastasis.71,72 Several series of patients who underwent pulmonary metastasectomy have reported 5-year OS rates ranging from 38.5% to 83.3%.73-75

Thus, in the absence of standard systemic therapies, this approach may be appropriate for selected patients with minimal disease burden and slow progression, especially in subtypes that tend to be more clinically indolent, such as chromophobe RCC.

Summary and Future Directions 

The available prospective and retrospective data suggest that the targeted agents currently approved for clear cell RCC can have activity in non–clear cell RCC. The phase 3 study of temsirolimus demonstrated a similar degree of benefit, as did the expanded access trials evaluating sunitinib, sorafenib, and everolimus in more real-world, generalizable patient populations.5,18,19,31,33,34 In addition, despite their poor prognosis, CDC and RCC with sarcomatoid components appear to demonstrate some sensitivity to chemotherapy. Ongoing clinical trials may provide additional evidence for the role of combinations of the targeted therapies with cytotoxic chemotherapy in this subgroup of patients.

However, although these agents can be effective, they are in no way a “home run,” and we remain with a paucity of effective systemic options for metastatic non–clear cell RCC. Thus, clinical investigation of novel therapeutics remains imperative. One such area of intense investigation in multiple solid tumors is inhibition of immune checkpoints, which may be reasonable targets in non–clear cell RCC. T cells express receptors critical to the control of the immune response. Programmed death 1 (PD-1) is an inhibitory receptor expressed on immune cells, including effector T cells.76,77 One of its ligands, programmed death ligand 1 (PD-L1), can be expressed by multiple normal tissues and also by tumors, and may represent a mechanism by which these tumors elude the host immune system. When PD-L1 on the tumor binds to PD-1 on the effector T cell, it inactivates the T cell. Overexpression of PD-L1 has been linked to poor prognosis in various tumor types, including RCC.78 Antibodies targeting PD-1 or PD-L1 have achieved clinical benefit in several small studies evaluating RCC, albeit mostly in clear cell disease.79-81 Recently, Choueiri and colleagues observed variable PD-L1 expression rates in non–clear cell RCC with higher levels in CDC and translocation carcinomas at 20% and 30%, respectively, compared with 10% and 5.6% in papillary and chromophobe RCC.82 Higher PD-L1 expression correlated with worse outcomes in non–clear cell RCC and was more common on the immune cells than on the tumor cell membrane.82 Preliminary results from the ongoing phase 1 Genentech trial (NCT01375842) evaluating the PD-L1 antibody MPDL3280A observed an ORR of 17% and a 24-week PFS of 20% in 6 non–clear cell RCC patients.83 This activity suggests that targeting the PD-1 pathway in non–clear cell RCC could be effective.

Diverse epigenetic alterations, such as histone modification and DNA methylation, can be involved in cancer development and progression.84 In RCC, lower levels of histone methylation have been correlated with higher tumor grade and pathologic stage.85 Targeting enzymes important in these epigenetic mechanisms may reverse the alterations and control tumor growth. In vitro, histone deacetylase (HDAC) inhibitors such as vorinostat (Zolinza, Merck), suberoylanilide hydroxamic acid (SAHA), and valproic acid have demonstrated cytotoxicity in RCC cell lines and synergy with other agents like everolimus.86-88 In phase 1 and 2 studies, HDAC inhibitors alone or in combination with other drugs that are active in RCC, such as sorafenib, generally have been well tolerated in patients with RCC.89-91 Response rates in heavily pretreated patients have been modest; however, prolonged disease stabilization can be observed. Another epigenetic mechanism that may play a role in RCC is the silencing of tumor suppressor genes by hypermethylation of the promoter region.92 To reverse this silencing, demethylating agents such as azacitidine have been studied. In a phase 1 study of azacitidine plus the HDAC inhibitor valproic acid, 1 patient with rapidly progressive RCC experienced stable disease for 6 months.93 The combination of azacitidine and bevacizumab also is under investigation in RCC (NCT00934440).

In summary, although the non–clear cell histologies are often lumped together as a single entity, they are distinct subtypes that likely have very different pathogenic drivers requiring more individualized treatments. Their less frequent occurrence has made large-scale investigation difficult. We are at the forefront of identifying molecular drivers of non–clear cell RCC, and thorough understanding of these alterations is critical to the development of appropriate treatment strategies. As such, clinical trials should be offered up front to all patients with non–clear cell RCC. In the absence of currently available evidence-based treatments, genetic tumor profiling may also help guide patients to rational therapies.

Disclosures

Drs Valenca and Hirsch have declared no financial conflicts of interest. Dr Choueiri has served on the advisory boards of Novartis, Pfizer, GlaxoSmithKline, Merck, Bristol-Myers Squibb, and Bayer. Dr Harshman has served on the advisory boards of Genentech, Bristol-Myers Squibb, Aveo, and Pfizer, and receives research funding or support from Pfizer and Medivation.

References

1. Gnarra JR, Tory K, Weng Y, et al. Mutations of the VHL tumour suppressor gene in renal carcinoma. Nat Genet. 1994;7(1):85-90.

2. Schraml P, Struckmann K, Hatz F, et al. VHL mutations and their correlation with tumour cell proliferation, microvessel density, and patient prognosis in clear cell renal cell carcinoma. J Pathol. 2002;196(2):186-193.

3. Motzer RJ, Hutson TE, Tomczak P, et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med. 2007;356(2):115-124.

4. Escudier B, Eisen T, Stadler WM, et al. Sorafenib for treatment of renal cell carcinoma: final efficacy and safety results of the phase III treatment approaches in renal cancer global evaluation trial. J Clin Oncol. 2009;27(20):3312-3318.

5. Hudes G, Carducci M, Tomczak P, et al; Global ARCC Trial. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med. 2007;356(22):2271-2281.

6. Escudier B, Pluzanska A, Koralewski P, et al; AVOREN Trial investigators. Bevacizumab plus interferon alfa-2a for treatment of metastatic renal cell carcinoma: a randomised, double-blind phase III trial. Lancet. 2007;370(9605):2103-2111.

7. Sternberg CN, Davis ID, Mardiak J, et al. Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial. J Clin Oncol. 2010;28(6):1061-1068.

8. Rini BI, Escudier B, Tomczak P, et al. Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomised phase 3 trial. Lancet. 2011;378(9807):1931-1939.

9. Motzer RJ, Escudier B, Oudard S, et al; RECORD‐1 Study Group. Phase 3 trial of everolimus for metastatic renal cell carcinoma: final results and analysis of prognostic factors. Cancer. 2010;116(18):4256-4265.

10. Motzer RJ, Bacik J, Mariani T, Russo P, Mazumdar M, Reuter V. Treatment outcome and survival associated with metastatic renal cell carcinoma of non-clear-cell histology. J Clin Oncol. 2002;20(9):2376-2381.

11. Tannir NM, Plimack E, Ng C, et al. A phase 2 trial of sunitinib in patients with advanced non-clear cell renal cell carcinoma. Eur Urol. 2012;62(6):1013-1019.

12. Paglino C, Imarisio I, Ganini C, et al. Sunitinib in advanced metastatic non-clear cell renal cell carcinoma: a single institution retrospective study. Future Oncol. 2012;8(12):1605-1612.

13. Jacobsen J, Grankvist K, Rasmuson T, Bergh A, Landberg G, Ljungberg B. Expression of vascular endothelial growth factor protein in human renal cell carcinoma. BJU Int. 2004;93(3):297-302.

14. Ljungberg BJ, Jacobsen J, Rudolfsson SH, Lindh G, Grankvist K, Rasmuson T. Different vascular endothelial growth factor (VEGF), VEGF-receptor 1 and -2 mRNA expression profiles between clear cell and papillary renal cell carcinoma. BJU Int. 2006;98(3):661-667.

15. Choueiri TK, Plantade A, Elson P, et al. Efficacy of sunitinib and sorafenib in metastatic papillary and chromophobe renal cell carcinoma. J Clin Oncol. 2008;26(1):127-131.

16. Lee JL, Ahn JH, Lim HY, et al. Multicenter phase II study of sunitinib in patients with non-clear cell renal cell carcinoma. Ann Oncol. 2012;23(8):2108-2114.

17. Lee JL, Gottfried M, Maimon N, et al. Patients with metastatic papillary renal cell carcinoma (RCC) who may benefit from sunitinib therapy (tx): results from an international metastatic RCC database [ASCO abstract e15547]. J Clin Oncol. 2014;32(15)(suppl).

18. Stadler WM, Figlin RA, McDermott DF, et al; ARCCS Study Investigators. Safety and efficacy results of the advanced renal cell carcinoma sorafenib expanded access program in North America. Cancer. 2010;116(5):1272-1280.

19. Gore ME, Szczylik C, Porta C, et al. Safety and efficacy of sunitinib for metastatic renal-cell carcinoma: an expanded-access trial. Lancet Oncol. 2009;10(8):757-763.

20. Molina AM, Feldman DR, Ginsberg MS, et al. Phase II trial of sunitinib in patients with metastatic non-clear cell renal cell carcinoma. Invest New Drugs. 2012;30(1):335-340.

21. Ravaud A, Oudard S, Gravis-Mescam G et al. First-line sunitinib in type I and II papillary renal cell carcinoma (PRCC): SUPAP, a phase II study of the French Genito-Urinary Group (GETUG) and the Group of Early Phase trials (GEP) [ASCO abstract 5146]. J Clin Oncol. 2009;27(15)(suppl).

22. Choueiri TK, Lim ZD, Hirsch MS, et al. Vascular endothelial growth factor-targeted therapy for the treatment of adult metastatic Xp11.2 translocation renal cell carcinoma. Cancer. 2010;116(22):5219-5225.

23. Choueiri TK, Mosquera JM, Hirsch MS. A case of adult metastatic Xp11 translocation renal cell carcinoma treated successfully with sunitinib. Clin Genitourin Cancer. 2009;7(3):E93-E94.

24. Procopio G, Testa I, Iacovelli R, et al. Treatment of collecting duct carcinoma: current status and future perspectives. Anticancer Res. 2014;34(2):1027-1030.

25. Zhao RN, Nie LH, Gong R, et al. Active targeted therapy for metastatic collecting duct carcinoma of the kidney: a case report and review of the literature. Int Urol Nephrol. 2013;45(4):1017-1021.

26. Kunene V, Miscoria M, Pirrie S, Islam MR, Afshar M, Porfiri E. Sarcomatoid renal cell carcinoma: clinical outcome and survival after treatment with sunitinib. Clin Genitourin Cancer. 2014;12(4):251-255.

27. Golshayan AR, George S, Heng DY, et al. Metastatic sarcomatoid renal cell carcinoma treated with vascular endothelial growth factor-targeted therapy. J Clin Oncol. 2009;27(2):235-241.

28. Staehler M, Haseke N, Roosen A, et al. Sorafenib after combination therapy with gemcitabine plus doxorubicine in patients with sarcomatoid renal cell carcinoma: a prospective evaluation. Eur J Med Res. 2010;15(7):287-291.

29. Klatte T, Han KR, Said JW, et al. Pathobiology and prognosis of chromophobe renal cell carcinoma. Urol Oncol. 2008;26(6):604-609.

30. Lin F, Zhang PL, Yang XJ, Prichard JW, Lun M, Brown RE. Morphoproteomic and molecular concomitants of an overexpressed and activated mTOR pathway in renal cell carcinomas. Ann Clin Lab Sci. 2006;36(3):283-293.

31. Dutcher JP, de Souza P, McDermott D, et al. Effect of temsirolimus versus interferon-alpha on outcome of patients with advanced renal cell carcinoma of different tumor histologies. Med Oncol. 2009;26(2):202-209.

32. Koh Y, Lim HY, Ahn JH, et al. Phase II trial of everolimus for the treatment of nonclear-cell renal cell carcinoma. Ann Oncol. 2013;24(4):1026-1031.

33. Grünwald V, Karakiewicz PI, Bavbek SE, et al; REACT Study Group. An international expanded-access programme of everolimus: addressing safety and efficacy in patients with metastatic renal cell carcinoma who progress after initial vascular endothelial growth factor receptor-tyrosine kinase inhibitor therapy. Eur J Cancer. 2012;48(3):324-332.

34. Blank CU, Bono P, Larkin JM, et al. Safety and efficacy of everolimus in patients with non-clear cell renal cell carcinoma refractory to VEGF-targeted therapy: sub-group analysis of REACT [ASCO abstract 402^]. J Clin Oncol. 2012;30(5)(suppl).

35. Harshman LC, Kroeger N, Rha SY, et al. First-line mammalian target of rapamycin inhibition in metastatic renal cell carcinoma: an analysis of practice patterns from the International Metastatic Renal Cell Carcinoma Database Consortium. Clin Genitourin Cancer. 2014;12(5):335-340.

36. Motzer RJ, Barrios CH, Kim TM, et al. Phase II randomized trial comparing sequential first-line everolimus and second-line sunitinib versus first-line sunitinib and second-line everolimus in patients with metastatic renal cell carcinoma. J Clin Oncol. 2014;32(25):2765-2772.

37. Tannir NM, Jonasch E, Altinmakas E, et al. Everolimus versus sunitinib prospective evaluation in metastatic non-clear cell renal cell carcinoma (the ESPN Trial): a multicenter randomized phase 2 trial [ASCO abstract 4505]. J Clin Oncol. 2014;32(15)(suppl).

38. Pal SK, He M, Tong T, et al. RNA-seq reveals aurora kinase-driven mTOR pathway activation in patients with sarcomatoid metastatic renal cell carcinoma. Mol Cancer Res. 2015;13(1):130-137.

39. Voss MH, Bastos DA, Karlo CA, et al. Treatment outcome with mTOR inhibitors for metastatic renal cell carcinoma with nonclear and sarcomatoid histologies. Ann Oncol. 2014;25(3):663-668.

40. Harshman LC, Choueiri TK. Targeting the hepatocyte growth factor/c-Met signaling pathway in renal cell carcinoma. Cancer J. 2013;19(4):316-323.

41. Dharmawardana PG, Giubellino A, Bottaro DP. Hereditary papillary renal carcinoma type I. Curr Mol Med. 2004;4(8):855-868.

42. Albiges L, Guegan J, Le Formal A, et al. MET is a potential target across all papillary renal cell carcinomas: result from a large molecular study of pRCC with CGH array and matching gene expression array. Clin Cancer Res. 2014;20(13):3411-3421. 43. Nakaigawa N, Yao M, Baba M, et al. Inactivation of von Hippel-Lindau gene induces constitutive phosphorylation of MET protein in clear cell renal carcinoma. Cancer Res. 2006;66(7):3699-3705.

44. Choueiri TK, Vaishampayan U, Rosenberg JE, et al. Phase II and biomarker study of the dual MET/VEGFR2 inhibitor foretinib in patients with papillary renal cell carcinoma. J Clin Oncol. 2013;31(2):181-186.

45. Schöffski P, Garcia JA, Stadler WM, et al. A phase II study of the efficacy and safety of AMG 102 in patients with metastatic renal cell carcinoma. BJU Int. 2011;108(5):679-686.

46. Yap TA, Olmos D, Brunetto AT, et al. Phase I trial of a selective c-MET inhibitor ARQ 197 incorporating proof of mechanism pharmacodynamic studies. J Clin Oncol. 2011;29(10):1271-1279.

47. Rosen LS, Senzer N, Mekhail T, et al. A phase I dose-escalation study of tivantinib (ARQ 197) in adult patients with metastatic solid tumors. Clin Cancer Res. 2011;17(24):7754-7764.

48. Wagner AJ, Goldberg JM, Dubois SG, et al. Tivantinib (ARQ 197), a selective inhibitor of MET, in patients with microphthalmia transcription factor-associated tumors: results of a multicenter phase 2 trial. Cancer. 2012;118(23):5894-5902.

49. Davis IJ, McFadden AW, Zhang Y, et al. Identification of the receptor tyrosine kinase c-Met and its ligand, hepatocyte growth factor, as therapeutic targets in clear cell sarcoma. Cancer Res. 2010;70(2):639-645.

50. Choueiri TK, Pal SK, McDermott DF, et al. A phase I study of cabozantinib (XL184) in patients with renal cell cancer. Ann Oncol. 2014;25(8):1603-1608.

51. Yagoda A, Petrylak D, Thompson S. Cytotoxic chemotherapy for advanced renal cell carcinoma. Urol Clin North Am. 1993;20(2):303-321.

52. Tsimafeyeu I, Demidov L, Kharkevich G, et al. Phase II, multicenter, uncontrolled trial of single-agent capecitabine in patients with non-clear cell metastatic renal cell carcinoma. Am J Clin Oncol. 2012;35(3):251-254.

53. Oudard S, Banu E, Vieillefond A, et al; GETUG (Groupe d’Etudes des Tumeurs Uro-Génitales). 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(5):1698-1702.

54. Barrascout E, Beuselinck B, Ayllon J, et al. Complete remission of pulmonary metastases of Bellini duct carcinoma with cisplatin, gemcitabine and bevacizumab. Am J Case Rep. 2012;13:1-2.

55. Pécuchet N, Bigot F, Gachet J, et al. Triple combination of bevacizumab, gemcitabine and platinum salt in metastatic collecting duct carcinoma. Ann Oncol. 2013;24(12):2963-2967.

56. Haas NB, Lin X, Manola J, et al. A phase II trial of doxorubicin and gemcitabine in renal cell carcinoma with sarcomatoid features: ECOG 8802. Med Oncol. 2012;29(2):761-767.

57. Dutcher JP, Nanus D. Long-term survival of patients with sarcomatoid renal cell cancer treated with chemotherapy. Med Oncol. 2011;28(4):1530-1533.

58. Nanus DM, Garino A, Milowsky MI, Larkin M, Dutcher JP. Active chemotherapy for sarcomatoid and rapidly progressing renal cell carcinoma. Cancer. 2004;101(7):1545-1551.

59. Roubaud G, Gross-Goupil M, Wallerand H, de Clermont H, Dilhuydy MS, Ravaud A. Combination of gemcitabine and doxorubicin in rapidly progressive metastatic renal cell carcinoma and/or sarcomatoid renal cell carcinoma. Oncology. 2011;80(3-4):214-218.

60. Escudier B, Droz JP, Rolland F, et al; Genitourinary Group of the French Federation of Cancer Centers. Doxorubicin and ifosfamide in patients with metastatic sarcomatoid renal cell carcinoma: a phase II study of the Genitourinary Group of the French Federation of Cancer Centers. J Urol. 2002;168(3):959-961.

61. McKay R, Choueiri TK, Werner L, et al. A phase II trial of sunitinib and gemcitabine in sarcomatoid and/or poor-risk patients with metastatic renal cell carcinoma. Poster presented at: 2015 Genitourinary Cancers Symposium; February 26-28, 2015; Orlando, FL. Abstract 408.

62. Flanigan RC, Salmon SE, Blumenstein BA, et al. Nephrectomy followed by interferon alfa-2b compared with interferon alfa-2b alone for metastatic renal-cell cancer. N Engl J Med. 2001;345(23):1655-1659.

63. Mickisch GH, Garin A, van Poppel H, de Prijck L, Sylvester R; European Organisation for Research and Treatment of Cancer (EORTC) Genitourinary Group. Radical nephrectomy plus interferon-alfa-based immunotherapy compared with interferon alfa alone in metastatic renal-cell carcinoma: a randomised trial. Lancet. 2001;358(9286):966-970.

64. Bamias A, Tzannis K, Papatsoris A, et al. Prognostic significance of cytoreductive nephrectomy in patients with synchronous metastases from renal cell carcinoma treated with first-line sunitinib: a European multiinstitutional study. Clin Genitourin Cancer. 2014;12(5):373-383.

65. Heng DY, Wells JC, Rini BI, et al. Cytoreductive nephrectomy in patients with synchronous metastases from renal cell carcinoma: results from the International Metastatic Renal Cell Carcinoma Database Consortium. Eur Urol. 2014;66(4)704-710.

66. Heng DY, Xie W, Regan MM, et al. Prognostic factors for overall survival in patients with metastatic renal cell carcinoma treated with vascular endothelial growth factor-targeted agents: results from a large, multicenter study. J Clin Oncol. 2009;27(34):5794-5799.

67. Aizer AA, Urun Y, McKay RR, Kibel AS, Nguyen PL, Choueiri TK. Cytoreductive nephrectomy in patients with metastatic non-clear-cell renal cell carcinoma (RCC). BJU Int. 2014;113(5b):E67-E74.

68. Shuch B, La Rochelle JC, Wu J, et al. Performance status and cytoreductive nephrectomy: redefining management in patients with poor performance. Cancer. 2008;113(6):1324-1331.

69. Wu HY, Xu LW, Zhang YY, Yu YL, Li XD, Li GH. Metachronous contralateral testicular and bilateral adrenal metastasis of chromophobe renal cell carcinoma: a case report and review of the literature. J Zhejiang Univ Sci B. 2010;11(5):386-389.

70. Zhang X, Zhou ZH, Cai SW, Dong JH. Papillary carcinoma of the duodenum combined with right renal carcinoma: a case report. World J Surg Oncol. 2013;11(1):30.

71. De Lichtenberg TH, Hermann GG, Rørth M, et al. Overall survival after immunotherapy, tyrosine kinase inhibitors and surgery in treatment of metastatic renal cell cancer: outcome of 143 consecutive patients from a single centre. Scand J Urol. 2014;48(4):379-386. Epub ahead of print.

72. Kim DY, Karam JA, Wood CG. Role of metastasectomy for metastatic renal cell carcinoma in the era of targeted therapy. World J Urol. 2014;32(3):631-642.

73. Vidarsdottir H, Moller PH, Jonasson JG, Pfannschmidt J, Gudbjartsson T. Indications and surgical outcome following pulmonary metastasectomy: a nationwide study. Thorac Cardiovasc Surg. 2012;60(6):383-389.

74. Hornbech K, Ravn J, Steinbrüchel DA. Outcome after pulmonary metastasectomy: analysis of 5 years consecutive surgical resections 2002-2006. J Thorac Oncol. 2011;6(10):1733-1740.

75. Chen F, Fujinaga T, Shoji T, et al. Pulmonary resection for metastasis from renal cell carcinoma. Interact Cardiovasc Thorac Surg. 2008;7(5):825-828.

76. Freeman GJ, Long AJ, Iwai Y, et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med. 2000;192(7):1027-1034.

77. Latchman YE, Liang SC, Wu Y, et al. PD-L1-deficient mice show that PD-L1 on T cells, antigen-presenting cells, and host tissues negatively regulates T cells. Proc Natl Acad Sci USA. 2004;101(29):10691-10696.

78. Thompson RH, Dong H, Lohse CM, et al. PD-1 is expressed by tumor-infiltrating immune cells and is associated with poor outcome for patients with renal cell carcinoma. Clin Cancer Res. 2007;13(6):1757-1761.

79. Topalian SL, Sznol M, Brahmer JR. Nivolumab (anti-PD-1; BMS-936558; ONO-4538) in patients with advanced solid tumors: survival and long-term safety in a phase I trial [ASCO abstract 3002]. J Clin Oncol. 2013;31(15)(suppl).

80. McDermott DF, Sznol M, Sosman JA, et al. Immune correlates and long term follow up of a phase Ia study of MPDL3280A, an engineered PD-L1 antibody, in patients with metastatic renal cell carcinoma (mRCC) [ESMO abstract 8090]. Ann Oncol. 2014;25(4)(suppl):iv280.

81. Motzer RJ, Rini BI, McDermott DF, et al. Nivolumab for metastatic renal cell carcinoma (mRCC): Results of a randomized, dose-ranging phase II trial [ASCO abstract 5009]. J Clin Oncol. 2014;32(15)(suppl).

82. Choueiri TK, Fay AP, Gray KP, et al. PD-L1 expression in nonclear-cell renal cell carcinoma. Ann Oncol. 2014;25(11):2178-2184.

83. Cho DC, Sosman JA, Sznol M, et al. Clinical activity, safety, and biomarkers of MPDL3280A, an engineered PD-L1 antibody in patients with metastatic renal cell carcinoma (mRCC) [ASCO abstract 4505]. J Clin Oncol. 2013;31(15)(suppl).

84. Yao Y, Des Marais TL, Costa M. Chromatin Memory in the Development of Human Cancers. Gene Technol. 2014;3(2):114.

85. Rogenhofer S, Kahl P, Holzapfel S, VON Ruecker A, Mueller SC, Ellinger J. Decreased levels of histone H3K9me1 indicate poor prognosis in patients with renal cell carcinoma. Anticancer Res. 2012;32(3):879-886.

86. Kim MJ, Lee JS, Park SE, et al. Combination treatment of renal cell carcinoma with belinostat and 5-fluorouracil: a role for oxidative stress-induced DNA damage and HSP90-regulated thymidine synthase [published online November 27, 2014]. J Urol. doi:10.1016/j.juro.2014.11.091.

87. Juengel E, Nowaz S, Makarevi J, et al. HDAC-inhibition counteracts everolimus resistance in renal cell carcinoma in vitro by diminishing cdk2 and cyclin A. Mol Cancer. 2014;13(1):152.

88. Sato A, Asano T, Ito K, Sumitomo M, Asano T. Suberoylanilide hydroxamic acid (SAHA) combined with bortezomib inhibits renal cancer growth by enhancing histone acetylation and protein ubiquitination synergistically. BJU Int. 2012;109(8):1258-1268.

89. Nanus DM, Tagawa ST, Dutcher JP, et al. NCI 6896: A phase I trial of suberoylanilide hydroxamic acid (SAHA) and 13-cis retinoic acid in the treatment of patients with advanced renal cell carcinoma (RCC) [ASCO abstract 349]. J Clin Oncol. 2011;29(7)(suppl).

90. Zibelman MR, Wong YN, Malizzia L, et al. Overcoming mTOR resistance: results of a phase I study of the mTOR inhibitor ridaforolimus and the HDAC inhibitor vorinostat [ASCO abstract 4568]. J Clin Oncol. 2014;32(5)(suppl).

91. Butler CM, Laboccetta LT, Brisendine A, Keane TE, Sherman CA, Drabkin HA. Phase I trial of the HDAC inhibitor LBH589 in combination with sorafenib in patients with renal cell carcinoma, non-small cell lung cancer and soft tissue sarcomas [ASCO abstract 440]. J Clin Oncol. 2012;30(5)(suppl).

92. Ricketts CJ, Morris MR, Gentle D, et al. Methylation profiling and evaluation of demethylating therapy in renal cell carcinoma. Clin Epigenetics. 2013;5(1):16.

93. Braiteh F, Soriano A, Luis CH, et al. Phase I study of low-dose hypomethylating agent azacitidine (5-AC) combined with the histone deacetylase inhibitor valproic acid (VPA) in patients with advanced cancers [ASCO abstract 3060]. J Clin Oncol. 2006;24(18)(suppl).