Targeted therapy for renal cell carcinoma: The next lap


1 Department of Medical Oncology, National Cancer Centre Singapore, Singapore 169610, Singapore
2 Department of Medical Oncology, National Cancer Centre Singapore, Singapore 169610; Institute of Bioengineering and Nanotechnology, Singapore 138669, Singapore

Date of Submission 14-Oct-2013
Date of Acceptance 15-Dec-2013
Date of Web Publication 20-Feb-2014

Correspondence Address:

Min-Han Tan
Department of Medical Oncology, National Cancer Centre Singapore, Singapore 169610; Institute of Bioengineering and Nanotechnology, Singapore 138669
Singapore.

Source of Support: None, Conflict of Interest: R. K. is an advisory board member for Glaxo.Smith.Kline. M. H. T. has filed patents for predictive biomarkers for kidney cancer.

 
 

DOI: 10.4103/1477-3163.127638

Abstract

Advances in rationally targeted therapeutics over the last decade have transformed the clinical care of advanced kidney cancer. While oncologists consolidate the gains of the wave of new agents, comprising a panoply of anti-vascular endothelial growth factor multi-targeted tyrosine kinase inhibitors and inhibitors of the mammalian target of rapamycin (mTOR), there is an increasing sense that a plateau has been reached in the short term. It is sobering that all currently approved targeted therapies have not yielded durable remissions and remain palliative in intent. In the context of recent insights in kidney cancer biology, we review promising ongoing and future approaches for kidney cancer therapeutics aimed toward forging new paths in the systemic management of renal cell carcinoma. Broadly, candidate agents for such innovative strategies include immune check-point inhibitors, anti-cancer stem cell agents, next-generation anti-vascular endothelial growth factor receptor and anti-mTOR agents as well as more investigational agents in the preclinical and early clinical development settings.

Keywords: Immunotherapy, renal cell carcinoma, targeted therapy, tyrosine kinase inhibitors.

How to cite this article:
Kanesvaran R, Tan MH. Targeted therapy for renal cell carcinoma: The next lap. J Carcinog 2014;13:3

 

How to cite this URL:
Kanesvaran R, Tan MH. Targeted therapy for renal cell carcinoma: The next lap. J Carcinog [serial online] 2014 [cited 2021 Oct 18];13:3. Available from: https://carcinogenesis.com/text.asp?2014/13/1/3/127638


Introduction

Advances in targeted therapy has wrought a transformation in the clinical care of metastatic renal cell carcinoma (RCC), with the US Food and Drug Administration (FDA) approval of a series of agents active against the vascular endothelial growth factor (VEGF) and mammalian target of rapamycin (mTOR) pathways over the last decade, including sorafenib, [1],[2] sunitinib, [3] bevacizumab, [4] temsirolimus, [5] everolimus, [6],[7] pazopanib [8],[9] and axitinib. [10],[11] With the benefit of several years of clinical experience, it is recognized that while these agents clearly represented useful advances over traditional interferon-based therapies, none of these new agents have demonstrated durable long-term complete remissions in metastatic RCC, as has been possible in about 7% of individuals using high-dose interleukin (IL)-2 infusions. [12] Further, newer agents have generally shown only marginal benefits over the established predecessors in the same drug classes, raising concerns about a dearth of innovation. The maturation of this approach is represented through increasing discussions over optimal sequences and combinations of such therapies. The identification of novel therapies is therefore of interest for transcending the current therapeutic plateau. We review here recent insights on both clinical and laboratory fronts, focusing on new directions in rationally designed targeted therapy, including targeted immunotherapy. Given the importance of developing new directions beyond the current state of the art of management, this review will favor discussions of efficacy over toxicity.

Biology

Epithelial RCC is a heterogeneous disease comprising multiple entities. Several major histological subtypes are recognized, including clear cell renal cell carcinoma (ccRCC) (~75%), papillary RCC (~15%) and chromophobe RCC (5%), with mixed tumors also being commonly seen. Most recently in late 2013, the International Society of Urological Pathology proposed the Vancouver Classification of Renal Neoplasia [13] recognizing rarer variants such as tubulocystic RCC, acquired cystic disease-associated RCC, clear cell (tubulo) papillary RCC, the MiT family translocation RCCs (in particular t (6;11) RCC) and hereditary leiomyomatosis RCC syndrome-associated RCC. Each individual entity has distinct germline and somatic genetic and molecular expression programs, [14] serving as a potential basis for developing precision medicine guided approaches to diagnostic and therapeutic. We review here several key biological insights in the last 5 years which are likely to underpin upcoming investigational drug development for RCC.

Progress in genetics

The key insight for ccRCC biology was the identification of germline von Hippel-Lindau (VHL) mutations underpinning the VHL syndrome, a hereditary multi-tumor syndrome in the early 1990s, [15] and the corresponding identification of somatic VHL mutations in sporadic ccRCC. Long recognized since the 19 th century, the myriad manifestations of VHL syndrome included ccRCC, craniospinal and ophthalmic hemangioblastoma, pheochromocytoma, pancreatic and renal cysts. VHL is regarded as a gatekeeper gene of ccRCC [16] in view of its relatively high somatic mutation prevalence (30-60%) in ccRCC and the existence of somatic mutations in non-malignant renal cysts adjacent to the tumor. Over 90% of ccRCC tumors show loss of one arm of chromosome 3p, resulting in allelic loss of VHL. pVHL mediates the ubiquitination-mediated pathway of hypoxia inducible factor (HIF) degradation, with mutant pVHL forms resulting in excess HIF and consequently, upregulated angiogenesis and glucose transport. While each of these downstream pathways is of therapeutic interest, angiogenesis inhibition in RCC has taken center stage since 2006 with the demonstration of clinical efficacy of tyrosine-kinase inhibitors and monoclonal antibodies in advanced RCC.

Several other germline alterations have been implicated in RCC (MET activating mutations in hereditary papillary RCC, FH mutations in hereditary leiomyomatosis-RCC syndrome, FLCN mutations in Birt-Hogg-Dube syndrome), but unlike VHL, these germline mutations are rarely identified in sporadic tumors. Hence, whereas it is commonly assumed that similar pathways are dysregulated in sporadic renal tumors, it is less clear that therapeutic strategies aimed at these genetic alterations and downstream pathways represent an optimal strategy.

In recent years, next-generation sequencing techniques have provided further insights into the genetics of RCC, highlighting several key biological themes through identification of somatic mutations of genes involved in sporadic RCC, particularly ccRCC. Notably, high frequencies of truncating somatic mutations in genes involved in chromatin modification have been demonstrated in several studies. [17],[18],[19] In particular, involved genes included histone modifying genes PBRM1SETD2KDM5CKDM6A and BAP1,[20],[21] and genes involved in the ubiquitin-mediated proteolysis pathway. [21] PBRM1 encodes the BAF180 protein, a subunit of the SWI/SNF complex which targets chromatin. BAP1 encodes for BRCA1 associated protein-1, which is involved in histone deubiquitination. This body of work has been received with significant interest as animal models have by and large been unable to directly recapitulate the link between VHL mutation and carcinogenesis, [22] suggesting that additional genetic “hits” are required for renal carcinogenesis to occur. In the above studies, deoxyribonucleic acid (DNA) repair has also been identified as a possible theme in ccRCC, with mutations identified in PMS1WRN and NBN, which are genes encoding DNA repair enzymes. It is expected that over the next few years, the molecular pathways dysregulated by these recently discovered somatic mutations will be unraveled.

Additional recent comprehensive molecular characterization studies of ccRCC have been conducted in the USA [23] and Japan, [24] integrating techniques such as whole-exome sequencing, whole-genome sequencing, ribonucleic acid (RNA) sequencing, array-based expression, micro-RNA profiling and methylation analyses. These have generally confirmed the findings of the earlier next-generation sequencing studies, but several themes were in addition highlighted, including recurrent mutations of PI3K-AKT-mTOR pathway; expression studies demonstrated metabolic derangement in aggressive ccRCC where genes involved in the pentose phosphate pathway and the glutamine pathway were overexpressed, with down regulation of genes expressed in the tricarboxylic acid cycle. The Japanese group additionally identified hotspot TCEB1 mutations in a small proportion of samples (8/240) mutually exclusive with VHL mutations. These mutations prevented elongin C-pVHL binding, with consequent HIF accumulation. While translation into therapies will take time, these insights into novel biological themes will certainly serve as the foundation for a fruitful clinical research program aimed at identifying newer therapeutic approaches.

Tumor heterogeneity and single cell analysis

Recent landmark articles studying tumor heterogeneity have provided considerable insight into the complex architecture of ccRCC. The presence of intratumor heterogeneity has been elegantly and directly demonstrated using spatially separated samples obtained from primary and metastatic RCC tumors, [25] as well as through single cell analysis using the high throughput sequencing approaches. [26] Practically, this work provides important guidance on therapeutic failure through intratumor heterogeneity – in essence, given the variation of mutations spatially and between cells, the sobering concern that rationally targeted therapy based on targeting mutant pathways in the cancer cell will be fundamentally limited in curative potential is a real problem that has to be confronted. From a more basic point of view, these studies showing extensive intratumor heterogeneity as well as convergent phenotypic mutational evolution highlight that most next-generation sequencing approaches today utilizing bulk tissue may underestimate the diversity of mutations and expression profiles within a single tumor. It therefore provokes the question whether approaches that indirectly target cancer through tumor microenvironment or the immune system may have superior outcomes; certainly the observation that high-dose IL-2 is of curative potential in metastatic RCC [12] supports this idea.

Cancer stem cell markers

The CSC hypothesis suggests that tumors are sustained by a subpopulation of cells, known as CSCs or tumor-initiating cells, which are able to initiate and renew tumor recapitulating the tumor of origin. [27] In ccRCC, a putative marker CD105, also known as endoglin, has been reported to distinguish a rare subpopulation of cells that exhibit CSC properties. [28],[29] These CD105-positive cells demonstrate clonal growth, express stem cell markers and are able to recapitulate tumors in immunosuppressed mice. Importantly, these cells release microvesicles that are proangiogenic and enhance lung metastases, suggesting a role in the formation of the pre-metastatic niche. [30] IL-15 has been identified as a possible agent to target these cells for differentiation. [31] Other markers have been investigated as putative markers for CSCs, including CD133, [32] and the side population using the Hoescht 33342 dye efflux study, [33] but these have not reliably demonstrated CSC properties. Overall, it is suggestive that CD105 may be of potential value as a novel target though there are potential concerns of off-target effects.

Therapeutic Strategies

Many new agents are being evaluated for the treatment of metastatic RCC, which may be broadly and somewhat arbitrarily classified into “canonical” and “emerging” approaches, depending on whether the agent has activity in previously established biological pathways of RCC management. Canonical approaches would include small molecule tyrosine kinase inhibition, with special focus on the VEGF and PDGF pathways, vascular endothelial growth factor receptor (VEGFR) blockade and mTOR inhibition. Emerging approaches would include agents at the preclinical and early clinical stages focused on novel pathways in RCC. Clinical trials that are currently open are summarized in [Table 1].

Table 1: Novel rationally targeted agents currently under investigation in RCC*

Click here to view


Canonical approaches

A slew of tyrosine kinase inhibitors

There have been an abundance of anti-VEGFR small molecules approved for RCC therapeutics, including sunitinib, [3] sorafenib, [1] pazopanib [9] and axitinib. [11] Indeed, while there are some differences in terms of toxicities between the agents in clinical practice, there has been little evidence to suggest clear advantages in efficacy, with pazopanib being most recently demonstrated to be non-inferior to sunitinib in terms of survival in the first-line management of metastatic RCC. [34] It is not clear that comparisons of differences in survival between Phase 3 trials are meaningful for deriving information on efficacy. While comparison studies are useful to guide daily practice, it is also true that such trials of drugs in the same class are unlikely to yield major concrete gains based on the current state of the art. Currently, many such agents are being investigated in Phase II and III trials, including dovitinib, tivozanib, cedirafenib, regorafenib, vandetanib and more. In what may be a herald for the future for Phase III trials for agents in this class, tivozanib was recently rejected by the U.S. FDA for concerns over overall survival relative to sorafenib, [35] and dovitinib failed to demonstrate progression-free survival over sorafenib. [36] It may be noted that chemical modification through drug encapsulation may be an interesting strategy to modify the properties of drugs, including efficacy and toxicities, allowing for a more favorable biodistribution. While liposomal formulations of cytotoxics such as doxorubicin and paclitaxel are commonly used in the clinical practice, similar methods for the TKIs are still in development, [37] and outcomes in the human setting have not been established.

Anti-VEGF/VEGFR blockade

Bevacizumab was the first in the class of anti-VEGFR monoclonal antibodies to be approved for use in metastatic RCC in combination with interferon. [38] Several other related agents are under investigation, including ziv-aflibercept (a soluble decoy receptor that binds to VEGF) and ramicirumab (a fully human IgG1 mAb targeting VEGFR-2) are currently under evaluation in Phase II studies (NCT00357760 and NCT00515697). [39]

The many splendored PI3K-AKT-mTOR pathway

The molecular signaling pathway of PI3K/AKT/mTOR [40] has been regularly implicated in RCC, where most recently, recurrent pathway mutations have been identified in comprehensive next generation sequencing studies as described above. [23],[24] Of interest, it should be noted that in the Japanese analysis, [24] although 26% of cases had mutations involving the PI3K-AKT-mTOR pathway, the most commonly mutated gene in this pathway mTOR had only a mutant prevalence of 5.7%. In addition, other than the known tumor suppressor genes (PTEN, tuberous sclerosis complex 1 [TSC1] and TSC2), the remaining mutations in other genes were not truncating in nature. The canonical pathway is activated by the binding of growth factors/ligands such as insulin-like growth factor or fibroblast growth factor (FGF) to their respective receptors leading to the recruitment of PI3K. This initiates a cascade of events starting with the conversion of phosphatidylinositol-4,5-phosphate (PIP2) to PIP3 which in turn activates AKT. Activated AKT indirectly activates mTOR through a phosphorylation of a number of targets downstream. Once activated, mTOR affects cell growth, proliferation, angiogenesis and metabolism. [41] mTOR exerts its effects through 2 different multiprotein signaling complexes, mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2). [41] mTORC1 is activated by the inhibition of TSC2, which subsequently activates ribosomal S6 kinase and eukaryotic initiation factor 4E-binding protein 1 (4E-BP1). In cancer cells, 4E-BP1 phosphorylation results in translation initiation. Activation of the mTORC1 downstream targets results in modulation of the activity of cell cycle regulating proteins like HIF, FGF, VEGF, STAT3, cyclin D and c-Myc. [42],[43],[44] Activated mTORC2 phosphorylates the hydrophobic motif of the AGC kinase family thereby inducing AKT activation. [45] Strong expression of various mTOR pathway proteins (PI3K, p-AKT, p-MTOR, p-70S6K) is observed in RCC relative to non-neoplastic kidney. [46],[47] Beyond temsirolimus and everolimus, there have been a slew of second generation mTOR inhibitors that have been developed and are currently being evaluated in clinical trials. Therapies developed against this pathway target it at several levels. These include selective inhibitors of mTOR itself, AKT, PI3K individually or combination of mTOR/PI3K inhibitors. A number of new mTOR inhibitors like AZD8055 have shown promising activity in pre-clinical studies [48] and have entered Phase I trials. [49] Amongst the PI3K inhibitors, BKM120 has shown good preclinical activity and is being evaluated in by itself and also in combination with drugs like bevacizumab in metastatic RCC patients who had failed anti-VEGFR therapy (NCT01239342). A dual inhibitor of PI3K and mTOR (NVP-BEZ235) has demonstrated better in vitro antiproliferative effects compared to rapamycin [50] and is currently in Phase I/II trials (NCT01453595). All this activity suggests that the PI3K-AKT-mTOR pathway remains of high interest from the perspective of drug discovery, particularly in the context of patients who have developed resistance to anti-VEGFR therapies, [51] consistent with the current fundamental insights afforded by next generation sequencing as described above.

Emerging therapeutics

Immune reinvigoration through programmed death-1 and programmed death ligand-1 inhibition

PD-1 and PD-L1 inhibitors are an exciting new class of agents targeting cancer cells via an immune modulated mechanism (“immune checkpoint blockade”). [52] The PD-1 pathway is an important tumor-evasion mechanism, with the two principal components of the PD-1 pathway comprising PD-1 (CD279), an inhibitory receptor expressed on the surface of activated T cells, B cells and myeloid cells and PD-L1, which is expressed on cancer cells. When PD-1 and PD-L1 are bound in a complex, T cell proliferation and survival is inhibited. Conversely, PD-1 blockade leads to auto-reactive T cell formation. [53] PD-L1 expression has been associated with poor prognosis in both primary and metastatic RCC. [54],[55],[56] Thus, targeting either PD-1 or PD-L1 may stimulate the immune system and enhance tumor-specific cytotoxicity of T-cells. Currently, anti-PD-1 inhibition with nivolumab (BMS-936558) is already being evaluated in a Phase III study in metastatic RCC (NCT01668784). The basis for this was derived from observations of durable responses for nivolumab in heavily pretreated metastatic RCC approximating 27%, [57] as well as early data showing reasonable safety profiles, even after long term continuous dosing. [58] Currently, PD-L1 antibody activity is also being actively investigated, with potentially exciting outcomes. In two recent reports, durable responses (including complete responses) were also observed in metastatic RCC with PD-L1 antibody (MPDL3280A). [59],[60] Potential tumor biomarkers of efficacy have been identified for anti-PD-L1 antibody such as PD-L1 status and low IL-17 expression. These biomarkers may provide guidance as to further mechanistic evaluation. [59] The continuous rather than bimodal expression of these proteins represents a challenge for PD-L1 as a clinical-grade biomarker. [56] Overall, toxicities of these agents seem fairly limited and these very exciting results show the significant potential for targeted immunotherapy to transform RCC management in the short term. A major area for future development may be the adjunctive use of stimulatory agents for anti-tumor immunity, given the recent discovery of mechanisms of immune escape through PD-L1 mediated immune down regulation. [61] Indeed, multiple studies are currently ongoing exploring combinations of these agents with dendritic cell vaccines (NCT01441765), tyrosine kinase inhibitors or ipilumumab (NCT01472081). [62]

Anti-CSC approaches

The identification of CD105/endoglin as a potential CSC marker in ccRCC is an important advance, [28] and the biology of this has been discussed above. The evaluation of anti-CD105/endoglin antibodies in RCC is a logical extension of this work. A Phase II trial (NCT01727089) evaluating TRC105, a chimeric IgG1 monoclonal antibody that binds CD105 (endoglin), is currently recruiting patients with RCC. In a Phase I trial, dose-limiting toxicities was essentially hypoproliferative anemia, [63] which is a likely off-target effect arising from the expression of CD105 in hematopoietic stem cells. [64]

Anti-angiogenesis as a common endpoint

Beyond anti-VEGFR TKIs and antibody blockade, inhibition of other pathways is being considered toward a common endpoint of anti-angiogenesis. Vascular disrupting agents (VDA) recognize and disrupt tumor blood supply by targeting dysmorphic endothelial cells and pericytes on the tumor vasculature. Compared to the anti-VEGFR and mTOR inhibitors, VDAs exert a cytotoxic rather than cytostatic effect. Small molecule VDAs are either flavonoids or tubulin binding agents. An example of such a drug is BNC105, which is a tubulin polymerization inhibitor. A multicenter Phase I/II clinical trial testing the combination of BNC105 and everolimus in the second-line treatment of mRCC has been recently completed (NCT01034631).

Inhibition of the angiopoietin/TIE2 pathway through the use of agents such as trebananib (AMG 386) is also being considered both alone and in combination with other agents including sorafenib, [65] sunitinib and temsirolimus (NCT01548482). Currently, AMG 386 does not appear to improve progression-free survival when used in combination with sorafenib versus sorafenib alone. [65]

The Notch pathway which mediates angiogenesis has been identified as another potential target in anticancer drug development. A Notch signaling pathway inhibitor, R04929097, is currently in use in a Phase I trial in mRCC patients that have failed anti-VEGF therapy (NCT01141569).

Antibody drug conjugates

ADCs represent a novel method in the treatment of mRCC. Such conjugates have received significant attention since positive results in breast cancer were reported for TDM-1, [66] a conjugate of the monoclonal antibody trastuzumab and the cytotoxic maytansine. These ADCs are composed of a monoclonal antibody that can bind to specific target receptors (antigens) on the RCC cell and a conjugated cytotoxic payload. The main target of this antibody is CD70 that is expressed on these cells. An example of this drug antibody conjugate is MDX-1203 which is conjugated to rachelmycin (CC1065) (prodrug), which exerts an alkylating action on adenine in dividing cells resulting in cell death. [67] This conjugate is currently under evaluation in advanced RCC patients (NCT00944905) and results are pending. Other anti-CD70 agents currently being investigated include MDX-1411 (NCT00656734) and SGN-75 (NCT01015911).

Carbonic anhydrase IX targeting

Radio-labeled antibodies are also being evaluated in mRCC patients, particularly through targeting of CAIX, a cell surface antigen. This cell surface antigen is highly expressed in RCC but is absent in normal renal epithelium. [68] G250 is a murine monoclonal antibody that targets CAIX and a chimeric version of this antibody, cG250 has shown antibody dependent cellular toxicity against RCC cells in vitro.[69] This antibody has also been tested in preclinical trials in combination with interferon gamma with promising synergistic tumoricidal activity noted. [70] A Phase II study has been conducted, [71] and a study of cG250 conjugated to yttrium-90 is currently ongoing (NCT00199875).

Histone deacetylase inhibitors

Of late a new class of drugs called histone deacetylase (HDAC) inhibitors have come to the forefront of cancer therapeutics. This was after the discovery that aberrant HDAC activity play an important role in carcinogenesis. [72] Its activity seems related to the recruitment of the BcL-2 family of genes. HDAC inhibitors that have been tested in metastatic RCC include LBH589 (panobinostat) and entinostat. In a recent phase II trial of patients that have had at least one prior TKI or mTOR inhibitor, panobinostat was well-tolerated but with no objective responses noted. [73]

Other candidate pathways

There are several candidate pathways of strong interest in RCC as identified in the pre-clinical setting. Signal transducer and activator of transcription 3, also known as STAT3, is a transcription factor which in humans is encoded by the STAT3 gene. It is involved in several signaling pathways that regulate cell survival and proliferation and it is aberrantly activated in RCC. [74] A STAT3 inhibitor (WP1066) has demonstrated antiproliferative activity in RCC cell lines and in vivo on murine xenografts. [75] Similarly, aurora kinases, oncogenic serine-threonine kinases that regulate the cell cycle, are key regulators of mitosis and have been found to be overexpressed in RCC. [76] Preclinical work has demonstrated that inhibition of these pathways through VX680 (a pan-aurora kinase inhibitor) similarly leads to significant cell death in RCC in vitro and in animal models, with additional anti-angiogenic effects. [76]

Conclusion

The systemic management of RCC has matured in recent years, with the development of a slew of rationally targeted therapies focusing on inhibition of the VEGF and mTOR pathways. The primary therapeutic approach to metastatic RCC remains palliative in nature, with the notable exception of a limited subset of patients experiencing durable complete remissions with high-dose IL-2. Beyond consolidating past gains through improved combinations and sequencing of existing agents, several promising leads for rationally targeted treatment have opened-up new directions for exploration and clinical investigation, with the hope of continuing to advance clinical care.

 References

1. Escudier B, Eisen T, Stadler WM, Szczylik C, Oudard S, Siebels M, et al. Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med 2007;356:125-34.  Back to cited text no. 1
[PUBMED]    
2. Escudier B, Eisen T, Stadler WM, Szczylik C, Oudard S, Staehler M, 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;27:3312-8.  Back to cited text no. 2
[PUBMED]    
3. Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Rixe O, et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med 2007;356:115-24.  Back to cited text no. 3
[PUBMED]    
4. Escudier B, Pluzanska A, Koralewski P, Ravaud A, Bracarda S, Szczylik C, et al. Bevacizumab plus interferon alfa-2a for treatment of metastatic renal cell carcinoma: A randomised, double-blind phase III trial. Lancet 2007;370:2103-11.  Back to cited text no. 4
[PUBMED]    
5. Hudes G, Carducci M, Tomczak P, Dutcher J, Figlin R, Kapoor A, et al. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med 2007;356:2271-81.  Back to cited text no. 5
[PUBMED]    
6. Motzer RJ, Escudier B, Oudard S, Hutson TE, Porta C, Bracarda S, et al. Phase 3 trial of everolimus for metastatic renal cell carcinoma: Final results and analysis of prognostic factors. Cancer 2010;116:4256-65.  Back to cited text no. 6
[PUBMED]    
7. Motzer RJ, Escudier B, Oudard S, Hutson TE, Porta C, Bracarda S, et al. Efficacy of everolimus in advanced renal cell carcinoma: A double-blind, randomised, placebo-controlled phase III trial. Lancet 2008;372:449-56.  Back to cited text no. 7
[PUBMED]    
8. Sternberg CN, Hawkins RE, Wagstaff J, Salman P, Mardiak J, Barrios CH, et al. A randomised, double-blind phase III study of pazopanib in patients with advanced and/or metastatic renal cell carcinoma: Final overall survival results and safety update. Eur J Cancer 2013;49:1287-96.  Back to cited text no. 8
[PUBMED]    
9. Sternberg CN, Davis ID, Mardiak J, Szczylik C, Lee E, Wagstaff J, et al. Pazopanib in locally advanced or metastatic renal cell carcinoma: Results of a randomized phase III trial. J Clin Oncol 2010;28:1061-8.  Back to cited text no. 9
[PUBMED]    
10. Motzer RJ, Escudier B, Tomczak P, Hutson TE, Michaelson MD, Negrier S, et al. Axitinib versus sorafenib as second-line treatment for advanced renal cell carcinoma: Overall survival analysis and updated results from a randomised phase 3 trial. Lancet Oncol 2013;14:552-62.  Back to cited text no. 10
    
11. Rini BI, Escudier B, Tomczak P, Kaprin A, Szczylik C, Hutson TE, et al. Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): A randomised phase 3 trial. Lancet 2011;378:1931-9.  Back to cited text no. 11
[PUBMED]    
12. Fyfe G, Fisher RI, Rosenberg SA, Sznol M, Parkinson DR, Louie AC. Results of treatment of 255 patients with metastatic renal cell carcinoma who received high-dose recombinant interleukin-2 therapy. J Clin Oncol 1995;13:688-96.  Back to cited text no. 12
[PUBMED]    
13. Srigley JR, Delahunt B, Eble JN, Egevad L, Epstein JI, Grignon D, et al. The International Society of Urological Pathology (ISUP) Vancouver classification of renal neoplasia. Am J Surg Pathol 2013;37:1469-89.  Back to cited text no. 13
[PUBMED]    
14. Tan MH, Rogers CG, Cooper JT, Ditlev JA, Maatman TJ, Yang X, et al. Gene expression profiling of renal cell carcinoma. Clin Cancer Res 2004;10:6315S-21.  Back to cited text no. 14
[PUBMED]    
15. Latif F, Tory K, Gnarra J, Yao M, Duh FM, Orcutt ML, et al. Identification of the von Hippel-Lindau disease tumor suppressor gene. Science 1993;260:1317-20.  Back to cited text no. 15
[PUBMED]    
16. Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA Jr, Kinzler KW. Cancer genome landscapes. Science 2013;339:1546-58.  Back to cited text no. 16
[PUBMED]    
17. Varela I, Tarpey P, Raine K, Huang D, Ong CK, Stephens P, et al. Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma. Nature 2011;469:539-42.  Back to cited text no. 17
[PUBMED]    
18. Dalgliesh GL, Furge K, Greenman C, Chen L, Bignell G, Butler A, et al. Systematic sequencing of renal carcinoma reveals inactivation of histone modifying genes. Nature 2010;463:360-3.  Back to cited text no. 18
[PUBMED]    
19. van Haaften G, Dalgliesh GL, Davies H, Chen L, Bignell G, Greenman C, et al. Somatic mutations of the histone H3K27 demethylase gene UTX in human cancer. Nat Genet 2009;41:521-3.  Back to cited text no. 19
[PUBMED]    
20. Peña-Llopis S, Vega-Rubín-de-Celis S, Liao A, Leng N, Pavía-Jiménez A, Wang S, et al. BAP1 loss defines a new class of renal cell carcinoma. Nat Genet 2012;44:751-9.  Back to cited text no. 20
    
21. Guo G, Gui Y, Gao S, Tang A, Hu X, Huang Y, et al. Frequent mutations of genes encoding ubiquitin-mediated proteolysis pathway components in clear cell renal cell carcinoma. Nat Genet 2011;44:17-9.  Back to cited text no. 21
[PUBMED]    
22. Rankin EB, Tomaszewski JE, Haase VH. Renal cyst development in mice with conditional inactivation of the von Hippel-Lindau tumor suppressor. Cancer Res 2006;66:2576-83.  Back to cited text no. 22
[PUBMED]    
23. Cancer Genome Atlas Research Network. Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature 2013;499:43-9.  Back to cited text no. 23
[PUBMED]    
24. Sato Y, Yoshizato T, Shiraishi Y, Maekawa S, Okuno Y, Kamura T, et al. Integrated molecular analysis of clear-cell renal cell carcinoma. Nat Genet 2013;45:860-7.  Back to cited text no. 24
[PUBMED]    
25. Gerlinger M, Rowan AJ, Horswell S, Larkin J, Endesfelder D, Gronroos E, et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N Engl J Med 2012;366:883-92.  Back to cited text no. 25
[PUBMED]    
26. Xu X, Hou Y, Yin X, Bao L, Tang A, Song L, et al. Single-cell exome sequencing reveals single-nucleotide mutation characteristics of a kidney tumor. Cell 2012;148:886-95.  Back to cited text no. 26
[PUBMED]    
27. Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci U S A 2003;100:3983-8.  Back to cited text no. 27
[PUBMED]    
28. Bussolati B, Bruno S, Grange C, Ferrando U, Camussi G. Identification of a tumor-initiating stem cell population in human renal carcinomas. FASEB J 2008;22:3696-705.  Back to cited text no. 28
[PUBMED]    
29. Bussolati B, Brossa A, Camussi G. Resident stem cells and renal carcinoma. Int J Nephrol 2011;2011:286985.  Back to cited text no. 29
[PUBMED]    
30. Grange C, Tapparo M, Collino F, Vitillo L, Damasco C, Deregibus MC, et al. Microvesicles released from human renal cancer stem cells stimulate angiogenesis and formation of lung premetastatic niche. Cancer Res 2011;71:5346-56.  Back to cited text no. 30
[PUBMED]    
31. Azzi S, Bruno S, Giron-Michel J, Clay D, Devocelle A, Croce M, et al. Differentiation therapy: Targeting human renal cancer stem cells with interleukin 15. J Natl Cancer Inst 2011;103:1884-98.  Back to cited text no. 31
[PUBMED]    
32. Bruno S, Bussolati B, Grange C, Collino F, Graziano ME, Ferrando U, et al. CD133+renal progenitor cells contribute to tumor angiogenesis. Am J Pathol 2006;169:2223-35.  Back to cited text no. 32
[PUBMED]    
33. Addla SK, Brown MD, Hart CA, Ramani VA, Clarke NW. Characterization of the Hoechst 33342 side population from normal and malignant human renal epithelial cells. Am J Physiol Renal Physiol 2008;295:F680-7.  Back to cited text no. 33
[PUBMED]    
34. Motzer RJ, Hutson TE, Cella D, Reeves J, Hawkins R, Guo J, et al. Pazopanib versus sunitinib in metastatic renal-cell carcinoma. N Engl J Med 2013;369:722-31.  Back to cited text no. 34
[PUBMED]    
35. Motzer RJ, Nosov D, Eisen T, Bondarenko I, Lesovoy V, Lipatov O, et al. Tivozanib versus sorafenib as initial targeted therapy for patients with metastatic renal cell carcinoma: Results from a phase III trial. J Clin Oncol 2013;31:3791-9.  Back to cited text no. 35
[PUBMED]    
36. Carroll J. Novartis buries a PhIII kidney cancer failure behind Q2 successes. 2013. http://www.fiercebiotech.com/story/novartis-buries-phiii-kidney-cancer-failure-behind-q2-successes/2013-07-19.  Back to cited text no. 36
    
37. Maitani Y, Saito H, Seishi Y, Iwase Y, Yamauchi T, Higashiyama K, et al. A combination of liposomal sunitinib plus liposomal irinotecan and liposome co-loaded with two drugs enhanced antitumor activity in PC12-bearing mouse. J Drug Target 2012;20:873-82.  Back to cited text no. 37
[PUBMED]    
38. Rini BI, Halabi S, Rosenberg JE, Stadler WM, Vaena DA, Archer L, et al. Phase III trial of bevacizumab plus interferon alfa versus interferon alfa monotherapy in patients with metastatic renal cell carcinoma: Final results of CALGB 90206. J Clin Oncol 2010;28:2137-43.  Back to cited text no. 38
[PUBMED]    
39. Tew WP, Gordon M, Murren J, Dupont J, Pezzulli S, Aghajanian C, et al. Phase 1 study of aflibercept administered subcutaneously to patients with advanced solid tumors. Clin Cancer Res 2010;16:358-66.  Back to cited text no. 39
[PUBMED]    
40. Gibbons JJ, Abraham RT, Yu K. Mammalian target of rapamycin: Discovery of rapamycin reveals a signaling pathway important for normal and cancer cell growth. Semin Oncol 2009;36 Suppl 3:S3-17.  Back to cited text no. 40
    
41. Wullschleger S, Loewith R, Hall MN. TOR signaling in growth and metabolism. Cell 2006;124:471-84.  Back to cited text no. 41
[PUBMED]    
42. Hudes GR. Targeting mTOR in renal cell carcinoma. Cancer 2009;115:2313-20.  Back to cited text no. 42
[PUBMED]    
43. Guertin DA, Sabatini DM. Defining the role of mTOR in cancer. Cancer Cell 2007;12:9-22.  Back to cited text no. 43
[PUBMED]    
44. Azim H, Azim HA Jr, Escudier B. Targeting mTOR in cancer: Renal cell is just a beginning. Target Oncol 2010;5:269-80.  Back to cited text no. 44
[PUBMED]    
45. Sarbassov DD, Guertin DA, Ali SM, Sabatini DM. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science 2005;307:1098-101.  Back to cited text no. 45
[PUBMED]    
46. Abou Youssif T, Fahmy MA, Koumakpayi IH, Ayala F, Al Marzooqi S, Chen G, et al. The mammalian target of rapamycin pathway is widely activated without PTEN deletion in renal cell carcinoma metastases. Cancer 2011;117:290-300.  Back to cited text no. 46
[PUBMED]    
47. Pantuck AJ, Seligson DB, Klatte T, Yu H, Leppert JT, Moore L, et al. Prognostic relevance of the mTOR pathway in renal cell carcinoma: Implications for molecular patient selection for targeted therapy. Cancer 2007;109:2257-67.  Back to cited text no. 47
[PUBMED]    
48. Chresta CM, Davies BR, Hickson I, Harding T, Cosulich S, Critchlow SE, et al. AZD8055 is a potent, selective, and orally bioavailable ATP-competitive mammalian target of rapamycin kinase inhibitor with in vitro and in vivo antitumor activity. Cancer Res 2010;70:288-98.  Back to cited text no. 48
[PUBMED]    
49. Asahina H, Nokihara H, Yamamoto N, Yamada Y, Tamura Y, Honda K, et al. Safety and tolerability of AZD8055 in Japanese patients with advanced solid tumors; a dose-finding phase I study. Invest New Drugs 2013;31:677-84.  Back to cited text no. 49
[PUBMED]    
50. Cho DC, Cohen MB, Panka DJ, Collins M, Ghebremichael M, Atkins MB, et al. The efficacy of the novel dual PI3-kinase/mTOR inhibitor NVP-BEZ235 compared with rapamycin in renal cell carcinoma. Clin Cancer Res 2010;16:3628-38.  Back to cited text no. 50
[PUBMED]    
51. Serova M, de Gramont A, Tijeras-Raballand A, Dos Santos C, Riveiro ME, Slimane K, et al. Benchmarking effects of mTOR, PI3K, and dual PI3K/mTOR inhibitors in hepatocellular and renal cell carcinoma models developing resistance to sunitinib and sorafenib. Cancer Chemother Pharmacol 2013;71:1297-307.  Back to cited text no. 51
[PUBMED]    
52. Brahmer JR, Drake CG, Wollner I, Powderly JD, Picus J, Sharfman WH, et al. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: Safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol 2010;28:3167-75.  Back to cited text no. 52
[PUBMED]    
53. Zha Yy, Blank C, Gajewski TF. Negative regulation of T-cell function by PD-1. Crit Rev Immunol 2004;24:229-37.  Back to cited text no. 53
[PUBMED]    
54. Thompson RH, Kuntz SM, Leibovich BC, Dong H, Lohse CM, Webster WS, et al. Tumor B7-H1 is associated with poor prognosis in renal cell carcinoma patients with long-term follow-up. Cancer Res 2006;66:3381-5.  Back to cited text no. 54
[PUBMED]    
55. Thompson RH, Gillett MD, Cheville JC, Lohse CM, Dong H, Webster WS, et al. Costimulatory molecule B7-H1 in primary and metastatic clear cell renal cell carcinoma. Cancer 2005;104:2084-91.  Back to cited text no. 55
[PUBMED]    
56. Thompson RH, Gillett MD, Cheville JC, Lohse CM, Dong H, Webster WS, et al. Costimulatory B7-H1 in renal cell carcinoma patients: Indicator of tumor aggressiveness and potential therapeutic target. Proc Natl Acad Sci U S A 2004;101:17174-9.  Back to cited text no. 56
[PUBMED]    
57. Topalian SL, Hodi FS, Brahmer JR, Gettinger SN, Smith DC, McDermott DF, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012;366:2443-54.  Back to cited text no. 57
[PUBMED]    
58. Drake CG, McDermott DF, Sznol M, Choueiri TK, Kluger HM, Powderly JD, et al. Survival, safety, and response duration results of nivolumab (Anti-PD-1; BMS-936558; ONO-4538) in a phase I trial in patients with previously treated metastatic renal cell carcinoma (mRCC): Long-term patient follow-up. J Clin Oncol 31, 2013 (suppl; abstr 4514) 2013;31:4514.  Back to cited text no. 58
    
59. Cho DC, Sosman JA, Sznol M, Gordon MS, Hollebecque A, Hamid O, et al. Clinical activity, safety, and biomarkers of MPDL3280A, an engineered PD-L1 antibody in patients with metastatic renal cell carcinoma (mRCC). J Clin Oncol 31, 2013 (suppl; abstr 4505) 2013;31:4505.  Back to cited text no. 59
    
60. Herbst RS, Gordon MS, Fine GD, Sosman JA, Soria JC, Hamid O, et al. A study of MPDL3280A, an engineered PD-L1 antibody in patients with locally advanced or metastatic tumors. J Clin Oncol 31, 2013 (suppl; abstr 3000) 2013;31:3000.  Back to cited text no. 60
    
61. Taube JM, Anders RA, Young GD, Xu H, Sharma R, McMiller TL, et al. Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci Transl Med 2012;4:127ra37.  Back to cited text no. 61
[PUBMED]    
62. Amin A, Ernstoff MS, Infante JR, Heng DY, Rini BI, Plimack ER, et al. A phase I study of nivolumab (anti-PD-1; BMS-936558; ONO-4538) in combination with sunitinib, pazopanib, or ipilimumab in patients (pts) with metastatic renal cell carcinoma (mRCC). J Clin Oncol 31, 2013 (suppl; abstr TPS4593) 2013;31:TPS4593.  Back to cited text no. 62
    
63. Rosen LS, Hurwitz HI, Wong MK, Goldman J, Mendelson DS, Figg WD, et al. A phase I first-in-human study of TRC105 (Anti-Endoglin Antibody) in patients with advanced cancer. Clin Cancer Res 2012;18:4820-9.  Back to cited text no. 63
[PUBMED]    
64. Pierelli L, Bonanno G, Rutella S, Marone M, Scambia G, Leone G. CD105 (endoglin) expression on hematopoietic stem/progenitor cells. Leuk Lymphoma 2001;42:1195-206.  Back to cited text no. 64
[PUBMED]    
65. Rini B, Szczylik C, Tannir NM, Koralewski P, Tomczak P, Deptala A, et al. AMG 386 in combination with sorafenib in patients with metastatic clear cell carcinoma of the kidney: A randomized, double-blind, placebo-controlled, phase 2 study. Cancer 2012;118:6152-61.  Back to cited text no. 65
[PUBMED]    
66. Verma S, Miles D, Gianni L, Krop IE, Welslau M, Baselga J, et al. Trastuzumab emtansine for HER2-positive advanced breast cancer. N Engl J Med 2012;367:1783-91.  Back to cited text no. 66
[PUBMED]    
67. Law CL, Gordon KA, Toki BE, Yamane AK, Hering MA, Cerveny CG, et al. Lymphocyte activation antigen CD70 expressed by renal cell carcinoma is a potential therapeutic target for anti-CD70 antibody-drug conjugates. Cancer Res 2006;66:2328-37.  Back to cited text no. 67
[PUBMED]    
68. Oosterwijk E, Ruiter DJ, Hoedemaeker PJ, Pauwels EK, Jonas U, Zwartendijk J, et al. Monoclonal antibody G 250 recognizes a determinant present in renal-cell carcinoma and absent from normal kidney. Int J Cancer 1986;38:489-94.  Back to cited text no. 68
[PUBMED]    
69. Liu Z, Smyth FE, Renner C, Lee FT, Oosterwijk E, Scott AM. Anti-renal cell carcinoma chimeric antibody G250: Cytokine enhancement of in vitro antibody-dependent cellular cytotoxicity. Cancer Immunol Immunother 2002;51:171-7.  Back to cited text no. 69
[PUBMED]    
70. Bauer S, Oosterwijk-Wakka JC, Adrian N, Oosterwijk E, Fischer E, Wüest T, et al. Targeted therapy of renal cell carcinoma: Synergistic activity of cG250-TNF and IFNg. Int J Cancer 2009;125:115-23.  Back to cited text no. 70
    
71. Bleumer I, Knuth A, Oosterwijk E, Hofmann R, Varga Z, Lamers C, et al. A phase II trial of chimeric monoclonal antibody G250 for advanced renal cell carcinoma patients. Br J Cancer 2004;90:985-90.  Back to cited text no. 71
[PUBMED]    
72. Shao Y, Gao Z, Marks PA, Jiang X. Apoptotic and autophagic cell death induced by histone deacetylase inhibitors. Proc Natl Acad Sci U S A 2004;101:18030-5.  Back to cited text no. 72
[PUBMED]    
73. Hainsworth JD, Infante JR, Spigel DR, Arrowsmith ER, Boccia RV, Burris HA. A phase II trial of panobinostat, a histone deacetylase inhibitor, in the treatment of patients with refractory metastatic renal cell carcinoma. Cancer Invest 2011;29:451-5.  Back to cited text no. 73
[PUBMED]    
74. Niu G, Wright KL, Huang M, Song L, Haura E, Turkson J, et al. Constitutive Stat3 activity up-regulates VEGF expression and tumor angiogenesis. Oncogene 2002;21:2000-8.  Back to cited text no. 74
[PUBMED]    
75. Horiguchi A, Asano T, Kuroda K, Sato A, Asakuma J, Ito K, et al. STAT3 inhibitor WP1066 as a novel therapeutic agent for renal cell carcinoma. Br J Cancer 2010;102:1592-9.  Back to cited text no. 75
[PUBMED]    
76. Li Y, Zhang ZF, Chen J, Huang D, Ding Y, Tan MH, et al. VX680/MK-0457, a potent and selective Aurora kinase inhibitor both tumor and endothelial cells in clear cell renal cell carcinoma. Am J Transl Res 2010;2:296-308.  Back to cited text no. 76
[PUBMED]    


 Authors

Dr. Ravindran Kanesvaran: Department of Medical Oncology, National Cancer Centre Singapore, Singapore 169610, Republic of Singapore Dr. Min‑Han Tan: Department of Medical Oncology, National Cancer Centre Singapore, Singapore 169610, Republic of Singapore; Institute of Bioengineering and Nanotechnology, Singapore 138669, Republic of Singapore.

Tables

 

[Table 1]