Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Primer
  • Published:

Renal cell carcinoma

Abstract

Renal cell carcinoma (RCC) denotes cancer originated from the renal epithelium and accounts for >90% of cancers in the kidney. The disease encompasses >10 histological and molecular subtypes, of which clear cell RCC (ccRCC) is most common and accounts for most cancer-related deaths. Although somatic VHL mutations have been described for some time, more-recent cancer genomic studies have identified mutations in epigenetic regulatory genes and demonstrated marked intra-tumour heterogeneity, which could have prognostic, predictive and therapeutic relevance. Localized RCC can be successfully managed with surgery, whereas metastatic RCC is refractory to conventional chemotherapy. However, over the past decade, marked advances in the treatment of metastatic RCC have been made, with targeted agents including sorafenib, sunitinib, bevacizumab, pazopanib and axitinib, which inhibit vascular endothelial growth factor (VEGF) and its receptor (VEGFR), and everolimus and temsirolimus, which inhibit mechanistic target of rapamycin complex 1 (mTORC1), being approved. Since 2015, agents with additional targets aside from VEGFR have been approved, such as cabozantinib and lenvatinib; immunotherapies, such as nivolumab, have also been added to the armamentarium for metastatic RCC. Here, we provide an overview of the biology of RCC, with a focus on ccRCC, as well as updates to complement the current clinical guidelines and an outline of potential future directions for RCC research and therapy.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Distinct subtypes of renal cell carcinoma.
Figure 2: Global kidney cancer incidence.
Figure 3: VHL inactivation in clear cell renal cell carcinoma and its implication in targeted therapy.
Figure 4: Cancer evolution and tumour heterogeneity in clear cell renal cell carcinoma.
Figure 5: Stages of kidney cancer and recommended treatments.
Figure 6: Indications for radical nephrectomy.
Figure 7: Therapeutic evolution and survival outcome of metastatic clear cell renal cell carcinoma through the four different eras.
Figure 8: Treatment algorithms for renal cell carcinoma.

Similar content being viewed by others

References

  1. Eble, J. N., Sauter, G., Epstein, J. I. & Sesterhenn, I. A. (eds) World Health Organization Classification Of Tumours (IARC Press, 2004).

    Google Scholar 

  2. Kovacs, G. et al. The Heidelberg classification of renal cell tumours. J. Pathol. 183, 131–133 (1997). This is the first consensus classification of RCC.

    Google Scholar 

  3. Lopez-Beltran, A., Scarpelli, M., Montironi, R. & Kirkali, Z. 2004 WHO classification of the renal tumors of the adults. Eur. Urol. 49, 798–805 (2006).

    Google Scholar 

  4. Srigley, J. R. et al. The International Society of Urological Pathology (ISUP) Vancouver classification of renal neoplasia. Am. J. Surg. Pathol. 37, 1469–1489 (2013).

    Google Scholar 

  5. Moch, H., Cubilla, A. L., Humphrey, P. A., Reuter, V. E. & Ulbright, T. M. The 2016 WHO classification of tumours of the urinary system and male genital organs — part A: renal, penile, and testicular tumours. Eur. Urol. 70, 93–105 (2016). This is the current consensus on RCC classification.

    Google Scholar 

  6. Chen, F. J. et al. Multilevel genomics-based taxonomy of renal cell carcinoma. Cell Rep. 14, 2476–2489 (2016). This is a comprehensive genomic analysis of three major RCC subtypes (KIRC, KICH and KIRP), characterized by The Cancer Genome Atlas kidney cancer working groups.

    Google Scholar 

  7. Cancer Genome Atlas Research Network. Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature 499, 43–49 (2013). This paper presents the most comprehensive genomic analysis of ccRCC and confirmed the loss of VHL and 3p as truncal events.

    Google Scholar 

  8. Cancer Genome Atlas Research Network et al. Comprehensive molecular characterization of papillary renal-cell carcinoma. N. Engl. J. Med. 374, 135–145 (2016).

    Google Scholar 

  9. Davis, C. F. et al. The somatic genomic landscape of chromophobe renal cell carcinoma. Cancer Cell 26, 319–330 (2014).

    Google Scholar 

  10. Chen, Y. B. et al. Molecular analysis of aggressive renal cell carcinoma with unclassified histology reveals distinct subsets. Nat. Commun. 7, 13131 (2016).

    Google Scholar 

  11. Rini, B. I., Campbell, S. C. & Escudier, B. Renal cell carcinoma. Lancet 373, 1119–1132 (2009).

    Google Scholar 

  12. Vera-Badillo, F. E. et al. Systemic therapy for non-clear cell renal cell carcinomas: a systematic review and meta-analysis. Eur. Urol. 67, 740–749 (2015).

    Google Scholar 

  13. Kroeger, N. et al. Metastatic non-clear cell renal cell carcinoma treated with targeted therapy agents: characterization of survival outcome and application of the International mRCC Database Consortium criteria. Cancer 119, 2999–3006 (2013).

    Google Scholar 

  14. Escudier, B. et al. Renal cell carcinoma: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 27, v58–v68 (2016).

    Google Scholar 

  15. Motzer, R. J. et al. Kidney cancer, version 3. 2015. J. Natl Compr. Canc. Netw. 13, 151–159 (2015).

    Google Scholar 

  16. Sankin, A., Hakimi, A. A., Hsieh, J. J. & Molina, A. M. Metastatic non-clear cell renal cell carcinoma: an evidence based review of current treatment strategies. Front. Oncol. 5, 67 (2015).

    Google Scholar 

  17. Gerlinger, M. et al. Intratumor heterogeneity and branched evolution revealed by multiregion sequencing. N. Engl. J. Med. 366, 883–892 (2012). This paper demonstrates the highly heterogeneous nature of somatic mutations in ccRCC through multiregion sequencing, which opened up the field of intra-tumour heterogeneity and convergent evolution through mutating the same gene within a given tumour.

    Google Scholar 

  18. Gerlinger, M. et al. Genomic architecture and evolution of clear cell renal cell carcinomas defined by multiregion sequencing. Nat. Genet. 46, 225–233 (2014).

    Google Scholar 

  19. Sankin, A. et al. The impact of genetic heterogeneity on biomarker development in kidney cancer assessed by multiregional sampling. Cancer Med. 3, 1485–1492 (2014).

    Google Scholar 

  20. Hsieh, J. J. et al. Genomic biomarkers of a randomized trial comparing first-line everolimus and sunitinib in patients with metastatic renal cell carcinoma. Eur. Urol. 71, 405–414 (2017). This paper reports on the prevalent mutations in metastatic ccRCC tumours and the prognostic and predictive values of molecular classification of ccRCC based on genomic mutations in patients treated with targeted therapies in a randomized trial.

    Google Scholar 

  21. Molina, A. M. et al. Sunitinib objective response in metastatic renal cell carcinoma: analysis of 1059 patients treated on clinical trials. Eur. J. Cancer 50, 351–358 (2014).

    Google Scholar 

  22. Motzer, R. J. 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. 32, 2765–2772 (2014).

    Google Scholar 

  23. Hsieh, J. J. et al. Overcome tumor heterogeneity-imposed therapeutic barriers through convergent genomic biomarker discovery: a braided cancer river model of kidney cancer. Semin. Cell Dev. Biol.http://dx.doi.org/10.1016/j.semcdb.2016.09.002 (2016).

  24. Ljungberg, B. et al. EAU guidelines on renal cell carcinoma: 2014 update. Eur. Urol. 67, 913–924 (2015).

    Google Scholar 

  25. El Dib, R., Touma, N. J. & Kapoor, A. Cryoablation versus radiofrequency ablation for the treatment of renal cell carcinoma: a meta-analysis of case series studies. BJU Int. 110, 510–516 (2012).

    Google Scholar 

  26. Pierorazio, P. M. et al. Five-year analysis of a multi-institutional prospective clinical trial of delayed intervention and surveillance for small renal masses: the DISSRM registry. Eur. Urol. 68, 408–415 (2015).

    Google Scholar 

  27. Frank, I. et al. An outcome prediction model for patients with clear cell renal cell carcinoma treated with radical nephrectomy based on tumor stage, size, grade and necrosis: the SSIGN score. J. Urol. 168, 2395–2400 (2002). This paper reports on the first commonly employed outcome prediction model (SSIGN) for patients with localized ccRCC.

    Google Scholar 

  28. Patard, J. J. et al. Use of the University of California Los Angeles integrated staging system to predict survival in renal cell carcinoma: an international multicenter study. J. Clin. Oncol. 22, 3316–3322 (2004).

    Google Scholar 

  29. Wolff, I. et al. Do we need new high-risk criteria for surgically treated renal cancer patients to improve the outcome of future clinical trials in the adjuvant setting? Results of a comprehensive analysis based on the multicenter CORONA database. Eur. J. Surg. Oncol. 42, 744–750 (2016).

    Google Scholar 

  30. Meskawi, M. et al. A review of integrated staging systems for renal cell carcinoma. Eur. Urol. 62, 303–314 (2012).

    Google Scholar 

  31. Motzer, R. J., Hutson, T. E., McCann, L., Deen, K. & Choueiri, T. K. Overall survival in renal-cell carcinoma with pazopanib versus sunitinib. N. Engl. J. Med. 370, 1769–1770 (2014).

    Google Scholar 

  32. Ferlay, J. et al. GLOBOCAN 2012 v1. 0, Cancer incidence and mortality worldwide: IARC CancerBase No. 11. 2013. GLOBOCANhttp://globocan.iarc.fr (accessed on 4 Aug 2016).

  33. Torre, L. A. et al. Global cancer statistics, 2012. CA Cancer J. Clin. 65, 87–108 (2015).

    Google Scholar 

  34. Global Burden of Disease Cancer Collaboration et al. The Global Burden of Cancer 2013. JAMA Oncol. 1, 505–527 (2015).

    Google Scholar 

  35. Siegel, R. L., Miller, K. D. & Jemal, A. Cancer statistics, 2016. CA Cancer J. Clin. 66, 7–30 (2016).

    Google Scholar 

  36. Ferlay, J. et al. Cancer incidence and mortality patterns in Europe: estimates for 40 countries in 2012. Eur. J. Cancer 49, 1374–1403 (2013).

    Google Scholar 

  37. Shuch, B. et al. Defining early-onset kidney cancer: implications for germline and somatic mutation testing and clinical management. J. Clin. Oncol. 32, 431–437 (2014).

    Google Scholar 

  38. Nielsen, S. M. et al. Von Hippel–Lindau disease: genetics and role of genetic counseling in a multiple neoplasia syndrome. J. Clin. Oncol. 34, 2172–2181 (2016).

    Google Scholar 

  39. Haas, N. B. & Nathanson, K. L. Hereditary kidney cancer syndromes. Adv. Chronic Kidney Dis. 21, 81–90 (2014).

    Google Scholar 

  40. Adeniran, A. J., Shuch, B. & Humphrey, P. A. Hereditary renal cell carcinoma syndromes: clinical, pathologic, and genetic features. Am. J. Surg. Pathol. 39, e1–e18 (2015).

    Google Scholar 

  41. Znaor, A., Lortet-Tieulent, J., Laversanne, M., Jemal, A. & Bray, F. International variations and trends in renal cell carcinoma incidence and mortality. Eur. Urol. 67, 519–530 (2015).

    Google Scholar 

  42. Sun, M. et al. Age-adjusted incidence, mortality, and survival rates of stage-specific renal cell carcinoma in North America: a trend analysis. Eur. Urol. 59, 135–141 (2011).

    Google Scholar 

  43. Hakimi, A. A. et al. An epidemiologic and genomic investigation into the obesity paradox in renal cell carcinoma. J. Natl Cancer Inst. 105, 1862–1870 (2013).

    Google Scholar 

  44. McLaughlin, J., Lipworth, L., Tarone, R. & Blot, W. in Cancer Epidemiology and Prevention (eds Schottenfeld, D. & Fraumeni, J. F. ) 1087–1100 (Oxford Univ. Press, 2006).

    Google Scholar 

  45. Benichou, J., Chow, W. H., McLaughlin, J. K., Mandel, J. S. & Fraumeni, J. F. Jr. Population attributable risk of renal cell cancer in Minnesota. Am. J. Epidemiol. 148, 424–430 (1998).

    Google Scholar 

  46. Chow, W. H., Dong, L. M. & Devesa, S. S. Epidemiology and risk factors for kidney cancer. Nat. Rev. Urol. 7, 245–257 (2010).

    Google Scholar 

  47. Faramawi, M. F., Johnson, E., Fry, M. W., Sall, M. & Zhou, Y. Consumption of different types of meat and the risk of renal cancer: meta-analysis of case–control studies. Cancer Causes Control 18, 125–133 (2007).

    Google Scholar 

  48. Lee, J. E. et al. Fat, protein, and meat consumption and renal cell cancer risk: a pooled analysis of 13 prospective studies. J. Natl Cancer Inst. 100, 1695–1706 (2008).

    Google Scholar 

  49. Setiawan, V. W., Stram, D. O., Nomura, A. M., Kolonel, L. N. & Henderson, B. E. Risk factors for renal cell cancer: the multiethnic cohort. Am. J. Epidemiol. 166, 932–940 (2007).

    Google Scholar 

  50. Tavani, A. et al. Lifetime physical activity and the risk of renal cell cancer. Int. J. Cancer 120, 1977–1980 (2007).

    Google Scholar 

  51. Bergstrom, A. et al. Physical activity and risk of renal cell cancer. Int. J. Cancer 92, 155–157 (2001).

    Google Scholar 

  52. Karami, S. et al. Family history of cancer and renal cell cancer risk in Caucasians and African Americans. Br. J. Cancer 102, 1676–1680 (2010).

    Google Scholar 

  53. Kaelin, W. G. Von Hippel–Lindau disease. Annu. Rev. Pathol. 2, 145–173 (2007).

    Google Scholar 

  54. Gnarra, J. R. et al. Mutations of the VHL tumour suppressor gene in renal carcinoma. Nat. Genet. 7, 85–90 (1994).

    Google Scholar 

  55. Purdue, M. P. et al. Genome-wide association study of renal cell carcinoma identifies two susceptibility loci on 2p21 and 11q13.3. Nat. Genet. 43, 60–65 (2011).

    Google Scholar 

  56. Wu, X. et al. A genome-wide association study identifies a novel susceptibility locus for renal cell carcinoma on 12p11.23. Hum. Mol. Genet. 21, 456–462 (2012).

    Google Scholar 

  57. Henrion, M. et al. Common variation at 2q22.3 (ZEB2) influences the risk of renal cancer. Hum. Mol. Genet. 22, 825–831 (2013).

    Google Scholar 

  58. Gudmundsson, J. et al. A common variant at 8q24.21 is associated with renal cell cancer. Nat. Commun. 4, 2776 (2013).

    Google Scholar 

  59. Schodel, J. et al. Common genetic variants at the 11q13.3 renal cancer susceptibility locus influence binding of HIF to an enhancer of cyclin D1 expression. Nat. Genet. 44, 420–425 (2012).

    Google Scholar 

  60. Bigot, P. et al. Functional characterization of the 12p12.1 renal cancer-susceptibility locus implicates BHLHE41. Nat. Commun. 7, 12098 (2016).

    Google Scholar 

  61. Hakimi, A. A., Pham, C. G. & Hsieh, J. J. A clear picture of renal cell carcinoma. Nat. Genet. 45, 849–850 (2013).

    Google Scholar 

  62. Linehan, W. M., Srinivasan, R. & Schmidt, L. S. The genetic basis of kidney cancer: a metabolic disease. Nat. Rev. Urol. 7, 277–285 (2010). This paper proposes RCC as a metabolic disease with mutations concentrating on metabolic pathways.

    Google Scholar 

  63. Masson, N. & Ratcliffe, P. J. Hypoxia signaling pathways in cancer metabolism: the importance of co-selecting interconnected physiological pathways. Cancer Metab. 2, 3 (2014).

    Google Scholar 

  64. Semenza, G. L. HIF-1 mediates metabolic responses to intratumoral hypoxia and oncogenic mutations. J. Clin. Invest. 123, 3664–3671 (2013). This paper presents an updated, comprehensive view of HIF1 function in tumour biology.

    Google Scholar 

  65. Hakimi, A. A. et al. An integrated metabolic atlas of clear cell renal cell carcinoma. Cancer Cell 29, 104–116 (2016). This paper reports the largest global metabolomics study of ccRCC and demonstrates its prognostic values.

    Google Scholar 

  66. Reuter, V. E. & Tickoo, S. K. Differential diagnosis of renal tumours with clear cell histology. Pathology 42, 374–383 (2010).

    Google Scholar 

  67. Lee, C. H. et al. Bevacizumab monotherapy as salvage therapy for advanced clear cell renal cell carcinoma pretreated with targeted drugs. Clin. Genitourin. Cancer 14, 56–62 (2016).

    Google Scholar 

  68. Kapitsinou, P. P. & Haase, V. H. The VHL tumor suppressor and HIF: insights from genetic studies in mice. Cell Death Differ. 15, 650–659 (2008).

    Google Scholar 

  69. Wei, E. Y. & Hsieh, J. J. A river model to map convergent cancer evolution and guide therapy in RCC. Nat. Rev. Urol. 12, 706–712 (2015). This paper proposes a novel cancer evolution model of ccRCC through gene–pathway–phenotype convergence.

  70. Pena-Llopis, S. et al. BAP1 loss defines a new class of renal cell carcinoma. Nat. Genet. 44, 751–759 (2012).

    Google Scholar 

  71. Hakimi, A. A. et al. Clinical and pathologic impact of select chromatin-modulating tumor suppressors in clear cell renal cell carcinoma. Eur. Urol. 63, 848–854 (2013).

    Google Scholar 

  72. Sato, Y. et al. Integrated molecular analysis of clear-cell renal cell carcinoma. Nat. Genet. 45, 860–867 (2013).

    Google Scholar 

  73. Xu, J. et al. Mechanistically distinct cancer-associated mTOR activation clusters predict sensitivity to rapamycin. J. Clin. Invest. 126, 3526–3540 (2016).

    Google Scholar 

  74. Grabiner, B. C. et al. A diverse array of cancer-associated MTOR mutations are hyperactivating and can predict rapamycin sensitivity. Cancer Discov. 4, 554–563 (2014). Together with reference 73, these two papers demonstrate that activating mutations in MTOR are common genetic events in ccRCC.

    Google Scholar 

  75. Voss, M. H. et al. Tumor genetic analyses of patients with metastatic renal cell carcinoma and extended benefit from mTOR inhibitor therapy. Clin. Cancer Res. 20, 1955–1964 (2014). This paper demonstrates the association between mutations in the core mTORC1-regulatory pathway and long-term therapeutic benefit from rapalogues.

  76. Kwiatkowski, D. J. et al. Mutations in TSC1, TSC2, and MTOR are associated with response to rapalogs in patients with metastatic renal cell carcinoma. Clin. Cancer Res. 22, 2445–2452 (2016).

    Google Scholar 

  77. Hakimi, A. A. et al. Adverse outcomes in clear cell renal cell carcinoma with mutations of 3p21 epigenetic regulators BAP1 and SETD2: a report by MSKCC and the KIRC TCGA Research Network. Clin. Cancer Res. 19, 3259–3267 (2013).

    Google Scholar 

  78. Kapur, P. et al. Effects on survival of BAP1 and PBRM1 mutations in sporadic clear-cell renal-cell carcinoma: a retrospective analysis with independent validation. Lancet Oncol. 14, 159–167 (2013). Together with reference 77, these two papers demonstrate the prognostic values of PBRM1, BAP1 and SETD2 mutations in ccRCC.

    Google Scholar 

  79. Nam, S. J., Lee, C., Park, J. H. & Moon, K. C. Decreased PBRM1 expression predicts unfavorable prognosis in patients with clear cell renal cell carcinoma. Urol. Oncol. 33, 340.e9–340.e16 (2015).

    Google Scholar 

  80. Manley, B. J. et al. Integration of recurrent somatic mutations with clinical outcomes: a pooled analysis of 1049 patients with clear cell renal cell carcinoma. Eur. Urol. Focushttp://dx.doi.org/10.1016/j.euf.2016.06.015 (2016).

  81. Nowell, P. C. The clonal evolution of tumor cell populations. Science 194, 23–28 (1976).

    Google Scholar 

  82. Venkatesan, S. & Swanton, C. Tumor evolutionary principles: how intratumor heterogeneity influences cancer treatment and outcome. Am. Soc. Clin. Oncol. 35, e141–e149 (2016).

    Google Scholar 

  83. Kanu, N. et al. SETD2 loss-of-function promotes renal cancer branched evolution through replication stress and impaired DNA repair. Oncogene 34, 5699–5708 (2015).

    Google Scholar 

  84. Gulati, S. et al. Systematic evaluation of the prognostic impact and intratumour heterogeneity of clear cell renal cell carcinoma biomarkers. Eur. Urol. 66, 936–948 (2014).

    Google Scholar 

  85. Beuselinck, B. et al. Molecular subtypes of clear cell renal cell carcinoma are associated with sunitinib response in the metastatic setting. Clin. Cancer Res. 21, 1329–1339 (2015).

    Google Scholar 

  86. Brooks, S. A. et al. ClearCode34: a prognostic risk predictor for localized clear cell renal cell carcinoma. Eur. Urol. 66, 77–84 (2014).

    Google Scholar 

  87. Senbabaoglu, Y. et al. Tumor immune microenvironment characterization in clear cell renal cell carcinoma identifies prognostic and immunotherapeutically relevant messenger RNA signatures. Genome Biol. 17, 231 (2016).

    Google Scholar 

  88. Geissler, K. et al. Immune signature of tumor infiltrating immune cells in renal cancer. Oncoimmunology 4, e985082 (2015).

    Google Scholar 

  89. Sinha, R. et al. Evaluating renal cell carcinoma cell lines as tumor models. Nat. Commun. (in press). This paper presents the most comprehensive in silico analysis of publically available RCC cell lines.

  90. Winter, S. et al. Methylomes of renal cell lines and tumors or metastases differ significantly with impact on pharmacogenes. Sci. Rep. 6, 29930 (2016).

    Google Scholar 

  91. Huang, D. et al. Sunitinib acts primarily on tumor endothelium rather than tumor cells to inhibit the growth of renal cell carcinoma. Cancer Res. 70, 1053–1062 (2010). This paper provides clear evidence that sunitinib acts primarily on the tumour endothelium using cancer cell lines.

    Google Scholar 

  92. Huang, D. et al. Interleukin-8 mediates resistance to antiangiogenic agent sunitinib in renal cell carcinoma. Cancer Res. 70, 1063–1071 (2010).

    Google Scholar 

  93. Zhou, L. et al. Targeting MET and AXL overcomes resistance to sunitinib therapy in renal cell carcinoma. Oncogene 35, 2687–2697 (2016).

    Google Scholar 

  94. Dong, Y. et al. Development of a clear cell renal cell carcinoma xenograft model: a case for the use of biopsy tissue over surgical tissue. J. Urol. 195, e916 (2016). Using patient-derived xenograft ccRCC models and tumour cells from patients with known primary response or resistance to sunitinib, this paper confirms that the principal therapeutic effect derived from sunitinib is through targeting tumour blood vessels.

    Google Scholar 

  95. Wang, S. S. et al. Bap1 is essential for kidney function and cooperates with Vhl in renal tumorigenesis. Proc. Natl Acad. Sci. USA 111, 16538–16543 (2014).

    Google Scholar 

  96. Palapattu, G. S., Kristo, B. & Rajfer, J. Paraneoplastic syndromes in urologic malignancy: the many faces of renal cell carcinoma. Rev. Urol. 4, 163–170 (2002).

    Google Scholar 

  97. Low, G., Huang, G., Fu, W., Moloo, Z. & Girgis, S. Review of renal cell carcinoma and its common subtypes in radiology. World J. Radiol. 8, 484–500 (2016).

    Google Scholar 

  98. Karlo, C. A. et al. Radiogenomics of clear cell renal cell carcinoma: associations between CT imaging features and mutations. Radiology 270, 464–471 (2014). This is the first radiogenomics study of ccRCC.

    Google Scholar 

  99. Motzer, R. J., Bacik, J., Murphy, B. A., Russo, P. & Mazumdar, M. Interferon-alfa as a comparative treatment for clinical trials of new therapies against advanced renal cell carcinoma. J. Clin. Oncol. 20, 289–296 (2002). This paper presents the value of Memorial Sloan Kettering Cancer Center risk factors in stratifying patients with metastatic kidney cancer.

    Google Scholar 

  100. Heng, D. Y. et al. External validation and comparison with other models of the International Metastatic Renal-Cell Carcinoma Database Consortium prognostic model: a population-based study. Lancet Oncol. 14, 141–148 (2013).

    Google Scholar 

  101. Linehan, W. M., Srinivasan, R. & Garcia, J. A. Non-clear cell renal cancer: disease-based management and opportunities for targeted therapeutic approaches. Semin. Oncol. 40, 511–520 (2013).

    Google Scholar 

  102. Marconi, L. et al. Systematic review and meta-analysis of diagnostic accuracy of percutaneous renal tumour biopsy. Eur. Urol. 69, 660–673 (2016).

    Google Scholar 

  103. Tickoo, S. K. & Reuter, V. E. Differential diagnosis of renal tumors with papillary architecture. Adv. Anat. Pathol. 18, 120–132 (2011).

    Google Scholar 

  104. Moch, H. An overview of renal cell cancer: pathology and genetics. Semin. Cancer Biol. 23, 3–9 (2013).

    Google Scholar 

  105. Algaba, F. et al. Current pathology keys of renal cell carcinoma. Eur. Urol. 60, 634–643 (2011).

    Google Scholar 

  106. Fuhrman, S. A., Lasky, L. C. & Limas, C. Prognostic significance of morphologic parameters in renal cell carcinoma. Am. J. Surg. Pathol. 6, 655–663 (1982). This paper reports and establishes the nuclear grading of RCC.

    Google Scholar 

  107. Cheville, J. C., Blute, M. L., Zincke, H., Lohse, C. M. & Weaver, A. L. Stage pT1 conventional (clear cell) renal cell carcinmoa: pathological features associated with cancer specific survival. J. Urol. 166, 453–456 (2001).

    Google Scholar 

  108. Grignon, D. J. & Che, M. Clear cell renal cell carcinoma. Clin. Lab. Med. 25, 305–316 (2005).

    Google Scholar 

  109. de Peralta-Venturina, M. et al. Sarcomatoid differentiation in renal cell carcinoma: a study of 101 cases. Am. J. Surg. Pathol. 25, 275–284 (2001).

    Google Scholar 

  110. Bi, M. et al. Genomic characterization of sarcomatoid transformation in clear cell renal cell carcinoma. Proc. Natl Acad. Sci. USA 113, 2170–2175 (2016).

    Google Scholar 

  111. Malouf, G. G. et al. Genomic characterization of renal cell carcinoma with sarcomatoid dedifferentiation pinpoints recurrent genomic alterations. Eur. Urol. 70, 348–357 (2016).

    Google Scholar 

  112. Manley, B. J. & Hsieh, J. J. Sarcomatoid renal cell carcinoma: genomic insights from sequencing of matched sarcomatous and carcinomatous components. Transl Cancer Res. 5, S160–S165 (2016).

    Google Scholar 

  113. Thompson, I. M. The evaluation of microscopic hematuria: a population-based study. J. Urol. 138, 1189–1190 (1987).

    Google Scholar 

  114. Zhang, L. et al. iTRAQ-based quantitative proteomic analysis reveals potential early diagnostic markers of clear-cell renal cell carcinoma. Biosci. Trends 10, 210–219 (2016).

    Google Scholar 

  115. Morrissey, J. J. et al. Evaluation of urine aquaporin-1 and perilipin-2 concentrations as biomarkers to screen for renal cell carcinoma: a prospective cohort study. JAMA Oncol. 1, 204–212 (2015).

    Google Scholar 

  116. Tosaka, A. et al. Incidence and properties of renal masses and asymptomatic renal cell carcinoma detected by abdominal ultrasonography. J. Urol. 144, 1097–1099 (1990).

    Google Scholar 

  117. Gudbjartsson, T. et al. A population-based familial aggregation analysis indicates genetic contribution in a majority of renal cell carcinomas. Int. J. Cancer 100, 476–479 (2002).

    Google Scholar 

  118. Hunt, J. D., van der Hel, O. L., McMillan, G. P., Boffetta, P. & Brennan, P. Renal cell carcinoma in relation to cigarette smoking: meta-analysis of 24 studies. Int. J. Cancer 114, 101–108 (2005).

    Google Scholar 

  119. Theis, R. P., Dolwick Grieb, S. M., Burr, D., Siddiqui, T. & Asal, N. R. Smoking, environmental tobacco smoke, and risk of renal cell cancer: a population-based case–control study. BMC Cancer 8, 387 (2008).

    Google Scholar 

  120. Renehan, A. G., Tyson, M., Egger, M., Heller, R. F. & Zwahlen, M. Body-mass index and incidence of cancer: a systematic review and meta-analysis of prospective observational studies. Lancet 371, 569–578 (2008).

    Google Scholar 

  121. Adams, K. F. et al. Body size and renal cell cancer incidence in a large US cohort study. Am. J. Epidemiol. 168, 268–277 (2008).

    Google Scholar 

  122. Luo, J. et al. Body size, weight cycling, and risk of renal cell carcinoma among postmenopausal women: the Women's Health Initiative (United States). Am. J. Epidemiol. 166, 752–759 (2007).

    Google Scholar 

  123. Albiges, L. et al. Body mass index and metastatic renal cell carcinoma: clinical and biological correlations. J. Clin. Oncol. 34, 3655–3663 (2016).

    Google Scholar 

  124. Chow, W. H., Gridley, G., Fraumeni, J. F. Jr & Jarvholm, B. Obesity, hypertension, and the risk of kidney cancer in men. N. Engl. J. Med. 343, 1305–1311 (2000).

    Google Scholar 

  125. Gago-Dominguez, M., Yuan, J. M., Castelao, J. E., Ross, R. K. & Yu, M. C. Regular use of analgesics is a risk factor for renal cell carcinoma. Br. J. Cancer 81, 542–548 (1999).

    Google Scholar 

  126. McCredie, M., Stewart, J. H. & Day, N. E. Different roles for phenacetin and paracetamol in cancer of the kidney and renal pelvis. Int. J. Cancer 53, 245–249 (1993).

    Google Scholar 

  127. Dhote, R., Thiounn, N., Debre, B. & Vidal-Trecan, G. Risk factors for adult renal cell carcinoma. Urol. Clin. North Am. 31, 237–247 (2004).

    Google Scholar 

  128. Khurana, V., Caldito, G. & Ankem, M. Statins might reduce risk of renal cell carcinoma in humans: case–control study of 500,000 veterans. Urology 71, 118–122 (2008).

    Google Scholar 

  129. Stewart-Merrill, S. B. et al. Oncologic surveillance after surgical resection for renal cell carcinoma: a novel risk-based approach. J. Clin. Oncol. 33, 4151–4157 (2015).

    Google Scholar 

  130. Ravaud, A. et al. Adjuvant sunitinib in high-risk renal-cell carcinoma after nephrectomy. N. Engl. J. Med. 375, 2246–2254 (2016).

    Google Scholar 

  131. Haas, N. B. et al. Adjuvant sunitinib or sorafenib for high-risk, non-metastatic renal-cell carcinoma (ECOG-ACRIN E2805): a double-blind, placebo-controlled, randomised, phase 3 trial. Lancet 387, 2008–2016 (2016).

    Google Scholar 

  132. Negrier, S. et al. Prognostic factors of survival and rapid progression in 782 patients with metastatic renal carcinomas treated by cytokines: a report from the Groupe Francais d’Immunotherapie. Ann. Oncol. 13, 1460–1468 (2002).

    Google Scholar 

  133. Heng, D. Y. 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. 27, 5794–5799 (2009).

    Google Scholar 

  134. Bex, A., Ljungberg, B., van Poppel, H., Powles, T. & European Association of Urology. The role of cytoreductive nephrectomy: European Association of Urology recommendations in 2016. Eur. Urol. 70, 901–905 (2016).

    Google Scholar 

  135. Huang, W. C. et al. Chronic kidney disease after nephrectomy in patients with renal cortical tumours: a retrospective cohort study. Lancet Oncol. 7, 735–740 (2006). This paper reports on the increased incidence of chronic kidney disease, leading to the ensuing trials comparing radical and partial nephrectomy in RCC.

    Google Scholar 

  136. Scosyrev, E., Messing, E. M., Sylvester, R., Campbell, S. & Van Poppel, H. Renal function after nephron-sparing surgery versus radical nephrectomy: results from EORTC randomized trial 30904. Eur. Urol. 65, 372–377 (2014).

    Google Scholar 

  137. Pignot, G. et al. Nephron-sparing surgery is superior to radical nephrectomy in preserving renal function benefit even when expanding indications beyond the traditional 4- cm cutoff. Urol. Oncol. 32, 1024–1030 (2014).

    Google Scholar 

  138. Van Poppel, H. et al. A prospective, randomised EORTC intergroup phase 3 study comparing the oncologic outcome of elective nephron-sparing surgery and radical nephrectomy for low-stage renal cell carcinoma. Eur. Urol. 59, 543–552 (2011). This is the first randomized phase III trial comparing the oncological outcome between partial and radical nephrectomy.

    Google Scholar 

  139. MacLennan, S. et al. Systematic review of oncological outcomes following surgical management of localised renal cancer. Eur. Urol. 61, 972–993 (2012).

    Google Scholar 

  140. Weight, C. J. et al. Partial nephrectomy is associated with improved overall survival compared to radical nephrectomy in patients with unanticipated benign renal tumours. Eur. Urol. 58, 293–298 (2010).

    Google Scholar 

  141. Sun, M. et al. A non-cancer-related survival benefit is associated with partial nephrectomy. Eur. Urol. 61, 725–731 (2012).

    Google Scholar 

  142. Klatte, T. et al. A literature review of renal surgical anatomy and surgical strategies for partial nephrectomy. Eur. Urol. 68, 980–992 (2015).

    Google Scholar 

  143. Crestani, A. et al. Introduction to small renal tumours and prognostic indicators. Int. J. Surg. 36, 495–503 (2016).

    Google Scholar 

  144. Aboumarzouk, O. M. et al. Robotic versus laparoscopic partial nephrectomy: a systematic review and meta-analysis. Eur. Urol. 62, 1023–1033 (2012).

    Google Scholar 

  145. Zhang, X. et al. Comparison of peri-operative outcomes of robot-assisted versus laparoscopic partial nephrectomy: a meta-analysis. BJU Int. 112, 1133–1142 (2013).

    Google Scholar 

  146. Ficarra, V. et al. A multicentre matched-pair analysis comparing robot-assisted versus open partial nephrectomy. BJU Int. 113, 936–941 (2014).

    Google Scholar 

  147. Wu, Z. et al. Robotic versus open partial nephrectomy: a systematic review and meta-analysis. PLoS ONE 9, e94878 (2014).

    Google Scholar 

  148. Marszalek, M. et al. Positive surgical margins after nephron-sparing surgery. Eur. Urol. 61, 757–763 (2012).

    Google Scholar 

  149. MacLennan, S. et al. Systematic review of perioperative and quality-of-life outcomes following surgical management of localised renal cancer. Eur. Urol. 62, 1097–1117 (2012).

    Google Scholar 

  150. Blom, J. H. et al. Radical nephrectomy with and without lymph-node dissection: final results of European Organization for Research and Treatment of Cancer (EORTC) randomized phase 3 trial 30881. Eur. Urol. 55, 28–34 (2009).

    Google Scholar 

  151. Hanna, N. et al. Survival analyses of patients with metastatic renal cancer treated with targeted therapy with or without cytoreductive nephrectomy: a National Cancer Data Base study. J. Clin. Oncol. 34, 3267–3275 (2016).

    Google Scholar 

  152. Lane, B. R., Tobert, C. M. & Riedinger, C. B. Growth kinetics and active surveillance for small renal masses. Curr. Opin. Urol. 22, 353–359 (2012).

    Google Scholar 

  153. Campbell, S. C. et al. Guideline for management of the clinical T1 renal mass. J. Urol. 182, 1271–1279 (2009).

    Google Scholar 

  154. Escudier, B. et al. Sorafenib in advanced clear-cell renal-cell carcinoma. N. Engl. J. Med. 356, 125–134 (2007). This pivotal trial led to the approval of the first anti-VEGFR targeted therapy sorafenib for metastatic ccRCC.

    Google Scholar 

  155. Motzer, R. J. et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N. Engl. J. Med. 356, 115–124 (2007). This pivotal trial establishes sunitinib as the first-line treatment for ccRCC, which greatly improved the survival outcome.

    Google Scholar 

  156. Motzer, R. J., McCann, L. & Deen, K. Pazopanib versus sunitinib in renal cancer. N. Engl. J. Med. 369, 1970 (2013).

    Google Scholar 

  157. Rini, B. I. et al. Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomised phase 3 trial. Lancet 378, 1931–1939 (2011).

    Google Scholar 

  158. Motzer, R. J. et al. Lenvatinib, everolimus, and the combination in patients with metastatic renal cell carcinoma: a randomised, phase 2, open-label, multicentre trial. Lancet Oncol. 16, 1473–1482 (2015).

    Google Scholar 

  159. Choueiri, T. K. et al. Cabozantinib versus everolimus in advanced renal-cell carcinoma. N. Engl. J. Med. 373, 1814–1823 (2015).

    Google Scholar 

  160. Escudier, B. et al. Bevacizumab plus interferon alfa-2a for treatment of metastatic renal cell carcinoma: a randomised, double-blind phase III trial. Lancet 370, 2103–2111 (2007).

    Google Scholar 

  161. Rini, B. I. et al. Bevacizumab plus interferon alfa compared with interferon alfa monotherapy in patients with metastatic renal cell carcinoma: CALGB 90206. J. Clin. Oncol. 26, 5422–5428 (2008).

    Google Scholar 

  162. Motzer, R. J. et al. Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet 372, 449–456 (2008).

    Google Scholar 

  163. Hudes, G. et al. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N. Engl. J. Med. 356, 2271–2281 (2007).

    Google Scholar 

  164. McDermott, D. F. et al. Randomized phase III trial of high-dose interleukin-2 versus subcutaneous interleukin-2 and interferon in patients with metastatic renal cell carcinoma. J. Clin. Oncol. 23, 133–141 (2005). This paper reports on a randomized phase III trial, which demonstrated a 23% response rate for high-dose IL-2.

    Google Scholar 

  165. Motzer, R. J. et al. Nivolumab versus everolimus in advanced renal-cell carcinoma. N. Engl. J. Med. 373, 1803–1813 (2015). This pivotal trial led the approval of the immune checkpoint inhibitor nivolumab, which is an anti-PD1 antibody, for the treatment of metastatic RCC.

    Google Scholar 

  166. Cella, D. Quality of life in patients with metastatic renal cell carcinoma: the importance of patient-reported outcomes. Cancer Treat. Rev. 35, 733–737 (2009).

    Google Scholar 

  167. Cella, D. et al. Quality of life in patients with advanced renal cell carcinoma given nivolumab versus everolimus in CheckMate 025: a randomised, open-label, phase 3 trial. Lancet Oncol. 17, 994–1003 (2016).

    Google Scholar 

  168. Donskov, F. et al. Sunitinib-associated hypertension and neutropenia as efficacy biomarkers in metastatic renal cell carcinoma patients. Br. J. Cancer 113, 1571–1580 (2015).

    Google Scholar 

  169. Funakoshi, T., Lee, C. H. & Hsieh, J. J. A systematic review of predictive and prognostic biomarkers for VEGF-targeted therapy in renal cell carcinoma. Cancer Treat. Rev. 40, 533–547 (2014).

    Google Scholar 

  170. Tran, H. T. et al. Prognostic or predictive plasma cytokines and angiogenic factors for patients treated with pazopanib for metastatic renal-cell cancer: a retrospective analysis of phase 2 and phase 3 trials. Lancet Oncol. 13, 827–837 (2012).

    Google Scholar 

  171. Voss, M. H. et al. Circulating biomarkers and outcome from a randomised phase II trial of sunitinib versus everolimus for patients with metastatic renal cell carcinoma. Br. J. Cancer 114, 642–649 (2016).

    Google Scholar 

  172. Armstrong, A. J., George, D. J. & Halabi, S. Serum lactate dehydrogenase predicts for overall survival benefit in patients with metastatic renal cell carcinoma treated with inhibition of mammalian target of rapamycin. J. Clin. Oncol. 30, 3402–3407 (2012).

    Google Scholar 

  173. Fay, A. P. et al. Whole-exome sequencing in two extreme phenotypes of response to VEGF-targeted therapies in patients with metastatic clear cell renal cell carcinoma. J. Natl Compr. Canc. Netw. 14, 820–824 (2016).

    Google Scholar 

  174. Ho, T. H. et al. Correlation between molecular subclassifications of clear cell renal cell carcinoma and targeted therapy response. Eur. Urol. Focus 2, 204–209 (2016).

    Google Scholar 

  175. Hsieh, J. et al. Identification of efficacy biomarkers in a large metastatic renal cell carcinoma (mRCC) cohort through next generation sequencing (NGS): results from RECORD-3. J. Clin. Oncol. 33, abstr. 4509 (2015).

    Google Scholar 

  176. Hammers, H. J. et al. Expanded cohort results from CheckMate 016: a phase I study of nivolumab in combination with ipilimumab in metastatic renal cell carcinoma (mRCC). J. Clin. Oncol. 33, abstr. 4516 (2015).

    Google Scholar 

  177. Amin, A. et al. Survival with AGS-003, an autologous dendritic cell-based immunotherapy, in combination with sunitinib in unfavorable risk patients with advanced renal cell carcinoma (RCC): phase 2 study results. J. Immunother. Cancer 3, 14 (2015).

    Google Scholar 

  178. Chen, W. et al. Targeting renal cell carcinoma with a HIF-2 antagonist. Nature 539, 112–117 (2016).

    Google Scholar 

  179. Cho, H. et al. On-target efficacy of a HIF2alpha antagonist in preclinical kidney cancer models. Nature 539, 107–111 (2016).

    Google Scholar 

  180. Wettersten, H. I. et al. Grade-dependent metabolic reprogramming in kidney cancer revealed by combined proteomics and metabolomics analysis. Cancer Res. 75, 2541–2552 (2015).

    Google Scholar 

  181. Rodrigues, D. et al. Renal cell carcinoma: a critical analysis of metabolomic biomarkers emerging from current model systems. Transl Res. 180, 1–11 (2017).

    Google Scholar 

  182. Harding, J. J. et al. Safety and tolerability of increasing doses of CB-839, a first-in-class, orally administered small molecule inhibitor of glutaminase, in solid tumors. ASCOhttp://meetinglibrary.asco.org/content/109133?media=sl (2015).

  183. Beatty, G. L. et al. Phase I study of the safety, pharmacokinetics (PK), and pharmacodynamics (PD) of the oral inhibitor of indoleamine 2,3-dioxygenase (IDO1) INCB024360 in patients (pts) with advanced malignancies. ASCOhttp://meetinglibrary.asco.org/content/83930?media=vm (2013).

  184. Tartour, E. et al. Angiogenesis and immunity: a bidirectional link potentially relevant for the monitoring of antiangiogenic therapy and the development of novel therapeutic combination with immunotherapy. Cancer Metastasis Rev. 30, 83–95 (2011).

    Google Scholar 

  185. Remark, R. et al. Characteristics and clinical impacts of the immune environments in colorectal and renal cell carcinoma lung metastases: influence of tumor origin. Clin. Cancer Res. 19, 4079–4091 (2013).

    Google Scholar 

  186. Giraldo, N. A. et al. Orchestration and prognostic significance of immune checkpoints in the microenvironment of primary and metastatic renal cell cancer. Clin. Cancer Res. 21, 3031–3040 (2015).

    Google Scholar 

  187. Fridman, W. H., Pages, F., Sautes-Fridman, C. & Galon, J. The immune contexture in human tumours: impact on clinical outcome. Nat. Rev. Cancer 12, 298–306 (2012).

    Google Scholar 

  188. Schumacher, T. N. & Schreiber, R. D. Neoantigens in cancer immunotherapy. Science 348, 69–74 (2015).

    Google Scholar 

  189. Demaria, S. et al. Ionizing radiation inhibition of distant untreated tumors (abscopal effect) is immune mediated. Int. J. Radiat. Oncol. Biol. Phys. 58, 862–870 (2004).

    Google Scholar 

  190. Janowitz, T., Welsh, S. J., Zaki, K., Mulders, P. & Eisen, T. Adjuvant therapy in renal cell carcinoma-past, present, and future. Semin. Oncol. 40, 482–491 (2013).

    Google Scholar 

  191. Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646–674 (2011).

    Google Scholar 

  192. Bozic, I. et al. Evolutionary dynamics of cancer in response to targeted combination therapy. eLife 2, e00747 (2013).

    Google Scholar 

  193. Molina, A. M. et al. Phase 1 trial of everolimus plus sunitinib in patients with metastatic renal cell carcinoma. Cancer 118, 1868–1876 (2012).

    Google Scholar 

  194. Voss, M. H. et al. Phase II trial and correlative genomic analysis of everolimus plus bevacizumab in advanced non-clear cell renal cell carcinoma. J. Clin. Oncol. 4, 3846–3853 (2016).

    Google Scholar 

  195. Hsieh, J. J. & Cheng, E. H. A braided cancer river connects tumor heterogeneity and precision medicine. Clin. Transl Med. 5, 42 (2016).

    Google Scholar 

  196. Powles, T. et al. Randomized open-label phase II trial of apitolisib (GDC-0980), a novel inhibitor of the PI3K/mammalian target of rapamycin pathway, versus everolimus in patients with metastatic renal cell carcinoma. J. Clin. Oncol. 34, 1660–1668 (2016).

    Google Scholar 

  197. Carlo, M. I. et al. A phase Ib study of BEZ235, a dual inhibitor of phosphatidylinositol 3-kinase (PI3K) and mammalian target of rapamycin (mTOR), in patients with advanced renal cell carcinoma. Oncologist 21, 787–788 (2016).

    Google Scholar 

  198. Bergers, G. & Hanahan, D. Modes of resistance to anti-angiogenic therapy. Nat. Rev. Cancer 8, 592–603 (2008).

    Google Scholar 

  199. Nakaigawa, N. et al. Inactivation of von Hippel–Lindau gene induces constitutive phosphorylation of MET protein in clear cell renal carcinoma. Cancer Res. 66, 3699–3705 (2006).

    Google Scholar 

  200. Mizukami, Y. et al. Induction of interleukin-8 preserves the angiogenic response in HIF-1alpha-deficient colon cancer cells. Nat. Med. 11, 992–997 (2005).

    Google Scholar 

  201. Albiges, L. et al. A systematic review of sequencing and combinations of systemic therapy in metastatic renal cancer. Eur. Urol. 67, 100–110 (2015).

    Google Scholar 

  202. Zaretsky, J. M. et al. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N. Engl. J. Med. 375, 819–829 (2016).

    Google Scholar 

  203. Massari, F. et al. PD-1 blockade therapy in renal cell carcinoma: current studies and future promises. Cancer Treat. Rev. 41, 114–121 (2015).

    Google Scholar 

  204. Armstrong, A. J. et al. Everolimus versus sunitinib for patients with metastatic non-clear cell renal cell carcinoma (ASPEN): a multicentre, open-label, randomised phase 2 trial. Lancet Oncol. 17, 378–388 (2016).

    Google Scholar 

  205. Tannir, N. M. et al. Everolimus versus sunitinib prospective evaluation in metastatic non-clear cell renal cell carcinoma (ESPN): a randomized multicenter phase 2 trial. Eur. Urol. 69, 866–874 (2016).

    Google Scholar 

  206. Dutcher, J. P. et al. Effect of temsirolimus versus interferon-alpha on outcome of patients with advanced renal cell carcinoma of different tumor histologies. Med. Oncol. 26, 202–209 (2009).

    Google Scholar 

  207. Yates, L. R. & Campbell, P. J. Evolution of the cancer genome. Nat. Rev. Genet. 13, 795–806 (2012).

    Google Scholar 

  208. Shuch, B. et al. Understanding pathologic variants of renal cell carcinoma: distilling therapeutic opportunities from biologic complexity. Eur. Urol. 67, 85–97 (2015).

    Google Scholar 

  209. Beaumont, J. L. et al. Patient-reported outcomes in a phase III study of everolimus versus placebo in patients with metastatic carcinoma of the kidney that has progressed on vascular endothelial growth factor receptor tyrosine kinase inhibitor therapy. Oncologist 16, 632–640 (2011).

    Google Scholar 

  210. Yang, S., de Souza, P., Alemao, E. & Purvis, J. Quality of life in patients with advanced renal cell carcinoma treated with temsirolimus or interferon-alpha. Br. J. Cancer 102, 1456–1460 (2010).

    Google Scholar 

Download references

Acknowledgements

J.J.H. is supported by the J. Randall & Kathleen L. MacDonald Family Research Fund, the Tom and Mila Tuttle Family Research Fund, the Jill and Rafic Dahan Family Research Fund, and the Jill and Jeffrey Weiss Family Research Fund for the cure of metastatic kidney cancer. J.L. is supported by the National Institute of Health Research (NIHR) Royal Marsden/Institute of Cancer Research Biomedical Research Centre for Cancer. M.P.P. is supported by the Intramural Research Program of the National Cancer Institute, US National Institutes of Health.

Author information

Authors and Affiliations

Authors

Contributions

Introduction (J.J.H.); Epidemiology (M.P.P.); Mechanisms/pathophysiology (S.S., J.J.H. and C.S.); Diagnosis screening and prevention (L.A. and M.S.); Management (V.F. and J.L.); Quality of life (D.Y.H.); Outlook (J.J.H.); Overview of Primer (J.J.H.).

Corresponding author

Correspondence to James J. Hsieh.

Ethics declarations

Competing interests

J.J.H. is a consultant for Novartis, Eisai and Chugai and has received research funding from Pfizer, Novartis, Eisai and Cancer Genomics Inc. C.S. is a consultant for Roche, Pfizer, Boehringer Ingelheim, Novartis, Celgene, Servler, Eli Lilly and GlaxoSmithKline, and owns stock options from Achilles Therapeutics, Epic Biosciences, Grail and ApoGen Biotech. L.A. is a consultant for Pfizer, Novartis, Sanofi, Amgen, Bristol-Myers Squibb, Bayer and Cerulean, and has received research funding from Pfizer and Novartis. M.S. is a consultant for Pfizer, Bristol-Myers Squibb, Ipsen, Exelixis, Eisai, Roche, Novartis and Astellas. D.Y.H. is a consultant for Pfizer, Novartis and Bristol-Myers Squibb. J.L. has received research funding from Novartis, Pfizer, Bristol-Myers Squibb and Merck Sharp & Dohme. M.P.P., S.S. and V.F. declare no competing interests.

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hsieh, J., Purdue, M., Signoretti, S. et al. Renal cell carcinoma. Nat Rev Dis Primers 3, 17009 (2017). https://doi.org/10.1038/nrdp.2017.9

Download citation

  • Published:

  • DOI: https://doi.org/10.1038/nrdp.2017.9

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer