Skip to main content
American Journal of Physiology - Renal Physiology logoLink to American Journal of Physiology - Renal Physiology
. 2017 Apr 5;313(2):F145–F154. doi: 10.1152/ajprenal.00480.2016

Renal cell carcinoma: new insights and challenges for a clinician scientist

Roman Shingarev 1, Edgar A Jaimes 1,
PMCID: PMC5582896  PMID: 28381462

Abstract

There is a growing recognition of the complex interplay between renal cell cancer (RCC), kidney function, mechanical reduction of nephron mass, and systemic agents targeting the cancer. Earlier detection of RCC and rising life expectancy of cancer survivors places a greater emphasis on preservation of renal function after cancer resection and during systemic therapy. Unique adverse effects associated with RCC drugs not only help reveal cancer pathophysiology but also expand our knowledge of normal cell signaling and metabolism. In this review, we outline our current understanding of RCC biology and treatment, their bidirectional relationship with kidney function, and unmet research needs in this field.

Keywords: renal cell carcinoma genotyping, radical and partial nephrectomy, molecularly targeted therapy and immune checkpoint inhibition


renal cell carcinoma (RCC) is the most common malignancy originating in the kidney and the most lethal genitourinary cancer (110). It has been classically characterized by an asymptomatic disease course, with a late and highly variable presentation portending a poor survival prognosis (43). Although its management in the past involved primarily urologists and oncologists, more recently, changing trends in RCC epidemiology, new therapeutic options, and overall improvement in patients’ life expectancy has drawn in nephrologists in a multidisciplinary approach to this disease and generated interest in the emerging field of onco-nephrology.

Epidemiology and Diagnostic Evaluation

Approximately 65,000 new kidney cancers are diagnosed in the US annually. Clear cell RCC (ccRCC) comprises 70%, papillary and chromophobe RCC 25%, and the more rare tumors of the medullary and collecting system origin 5% of these cancers (Table 1) (31, 96). These diagnoses require surgical resection or percutaneous needle biopsy of the mass and must be viewed in the context of overall prevalence of solitary renal lesions >1 cm estimated to be 12–14% (18). The majority of these (70%) are simple cysts (21) and in most cases do not require further workup or followup.

Table 1.

Histopathological classification of renal cell carcinoma and selected clinical characteristics of its subtypes (see Refs. 16, 68, 93, and 115)

RCC Type Clear Cell Papillary Chromophobe Collecting Duct
Cell of origin Proximal tubular Proximal tubular Intercalated Collecting duct
Relative prevalence among RCC 80–90% 10–15% 4–5% <1%
Cancer-specific 10-yr survival 71% 91% 88% 34%
Prevalence of distant metastases on presentation 15% 3% 4% 32%
Recommended first-line therapy for advanced disease IL-2 immunotherapy Anti-VEGF Anti-VEGF Cisplatin and gemcitabine

RCC, renal cell carcinoma.

Reduction of glomerular filtration rate (GFR) is associated with progressively higher risk of RCC (20, 75), although a confounding of the shared risk factors, such as smoking, hypertension, diabetes, and obesity, or the altered metabolism and immunology associated with chronic kidney disease (CKD) is possible (15, 19, 60). Incidence of both simple cysts and RCC increases 10- to 100-fold in patients with end-stage renal disease (ESRD) and is proportionate to the dialysis dependence duration (9, 29). In the absence of screening guidelines, this has led to significant variability in screening protocols among nephrology practices. Ultrasonography is frequently employed to that end and can reliably identify simple cysts on the basis of the characteristic triad of findings, such as 1) round shape with smooth walls, 2) absence of echoes within the lesion, and 3) strong signal reflecting off the posterior cyst wall (25). Conversely, complex cysts with multiple thickened septa often containing calcifications and all solid lesions <4 cm enhancing with contrast on computer tomography qualify as small renal masses (SRMs) and warrant a referral to a urologist because of their high (70–80%) malignancy potential (37, 66, 81). The significant remainder of the SRMs is thus benign, which, together with the growing occurrence of incidental masses, has led to implementation of active surveillance with serial imaging as a safe initial approach to managing SRMs, especially in patients with high competing risks of death (1, 11). The same factors led to the emergence of tumor biopsy as an important adjunct to the diagnostic assessment of SRMs in unlikely surgical candidates. Although historically avoided in suspected RCC because of the risk of needle track seeding, more recently, SRM biopsy has been shown to be a safe method of obtaining an accurate diagnosis differentiating between RCC, renal metastasis from a distant malignancy, lymphoma, abscess, and benign cyst (2, 78). It also provides tissue for genomic and molecular marker analyses that can potentially further individualize treatment approach.

Basic Biology

Traditional classification of RCC is based on histomorphology of tumors, separating them into the three main types: clear cell (ccRCC), papillary (pRCC), and chromophobe (chRCC). Recent advances in the genomic profiling of RCC yielded further differentiation of tumor types, allowing for better understanding of their biology and greater specificity of treatment targets.

ccRCC

The most common type of renal cancer, ccRCC, originates from the proximal tubular cells. Genetic aberrations in ccRCC are broadly divided into those affecting the von Hippel-Lindau (VHL) gene, epigenetic regulators/chromatin remodeling genes, and disrupting phosphatidylinositol 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling.

Most cases are sporadic, and only 2–3% of ccRCC are accounted for by hereditary diseases, including VHL autosomal dominant syndrome, characterized by a germline mutation of the VHL tumor suppressor gene on chromosome 3p25 (73). Conversely, mutations or silencing of the same VHL gene are associated with >80% of sporadic ccRCC (71, 85). The best known function of VHL protein (pVHL) is polyubiquitination of several proteins, including hypoxia-inducible factor (HIF)-1 and -2, thereby marking them for degradation in the presence of normal tissue oxygenation (79). Under hypoxic conditions, HIF activates transcription of hypoxia-inducible genes, resulting in increased production of erythropoietin, vascular endothelial growth factor (VEGF), PDGFβ, and transforming growth factor-α (TGF-α) and several glycolytic enzymes (62, 76, 107). Failure of pVHL to reign in the activity of HIF in normoxia has crucial implications for the clinical behavior and metabolic features of ccRCC. Among them are erythropoietin-dependent erythrocytosis, marked angiogenesis, and cell proliferation mediated by TGFα and VEGF (62, 113) as well as aerobic glycolysis (Warburg effect) and immunosuppressive effects of increased tryptophan catabolism (125). The nature of the interactions between HIF and cellular metabolism is likely bidirectional, as shown by the inhibitory effects of the gluconeogenic enzyme fructose-1,6-bisphosphatase on HIF function (69). Although some studies showed an association between such metabolic abnormalities and tumor aggressiveness, metabolomic analyses have not yet become a part of clinical practice (10).

The second-most common type of mutation involves several epigenetic regulators also localized on 3p25 chromosome, including Polybromo 1 (PBRM1), BRCA-associated protein-1 (BAP1), and SET domain containing 1 (SETD2) genes involved in regulation of chromatin maintenance and remodeling. Generally, hypermethylation of promoter sites is associated with the tumors carrying the above mutations (with the exception of SETD1) and higher tumor grade, but their exact role in ccRCC pathogenesis is not well understood (26, 30, 45, 97, 122).

Genetic aberration of mTOR pathway proteins (PTEN, Akt, PIK3CA, and mTOR) involved in cell proliferation signaling have been identified in ≤28% of RCC patients (12). Deficiency in PTEN in particular is associated with more aggressive tumors (108). Together with the abnormal expression of focal adhesion kinase modulator of mTOR pathway, these genetic defects are present in >50% of ccRCC patients (12). As a part of the Cancer Genome Atlas project, less frequent mutations of genes involved in cell metabolism and division have been described, with some of them, such as p53 overexpression, associated with a worsening prognosis (12).

Papillary RCC

This second-most common RCC is also of proximal tubule origin but much less studied (13). Type 1 papillary RCC (pRCC) is characterized by MET proto-oncogene-activating mutations occurring in germ cells in the hereditary form of the disease and 10–20% of cases of somatic mutations of the sporadic (and most common) form of the disease (106). Additionally, trisomy 7 has been implicated in MET amplification in some cases of pRCC (65). Type 2 pRCC is associated with activation of the NRF2/antioxidant response element pathway due to the increased oxidative stress. Mutations in the fumarate hydratase (FH) gene in this disorder are associated with hereditary leiomyomatosis and renal cell carcinoma (HLRCC) (71, 116). Mutations in epigenetic modifier genes PBRM1, BAP1, and SETD2 have been described with this tumor type but seem to be less prevalent compared with ccRCC (65). Type 2 pRCC portends a considerably worse prognosis compared with type 1 pRCC (3, 86).

Chromophobe RCC

This rare cancer arising from the collecting duct and resembling benign oncocytoma comprises <5% of RCC and is commonly associated with whole chromosome losses and germline mutations in the folliculin gene (FLCN) found in the autosomal dominant Birt-Hogg-Dube syndrome (74, 117). The most common mutation in sporadic cases of chRCC involves downregulation of p53 tumor suppressor signaling and loss of function of PTEN, leading to PI3K-driven cell proliferation. ChRCC is more common in younger females and is typically the least aggressive of all RCC types, unless it is characterized by sarcomatous transformation (27).

Management

As more RCC are diagnosed at earlier stages, resection of smaller solitary tumors offers a cure for most patients (121). Similarly, advances in systemic therapies improve life expectancy in patients with advanced metastatic disease. Together, these positive trends highlight the importance of preserved renal function for RCC patients’ survival.

Surgical Strategies

Complete surgical tumor resection remains the standard of care for all localized RCC, whereas cytoreductive surgery improves the survival of patients with metastatic disease undergoing immunotherapy (83) and possibly molecularly targeted therapies (17, 23). Cryo- and radiofrequency ablation are associated with fewer perioperative complication profiles but higher cancer recurrence rates and are usually reserved for patients unfit for surgery (44, 57, 98). Radical and partial nephrectomies (RN and PN) seem to offer similar overall survival on the basis of a single randomized controlled trial (EORTC) (121) and the largest meta-analysis to date that included over 180,000 patients from 107 studies (98). Some cohort studies, however, demonstrated a more favorable renal functional prognosis associated with PN (49, 70, 126), although the absolute volume of resected parenchyma may not necessarily correlate with renal outcomes (42). In a meta-analysis from 2012, PN was associated with a 61% risk reduction for the incident severe CKD (61).

Postsurgical Sequelae

The overall prevalence of CKD stage 3 or higher is ∼50% after tumor resection (46) and is associated with cardiovascular complications, including death (124). Although significant proportion of RCC patients have CKD at baseline, nephrectomy-induced decline in GFR has been well described and attributed to the nephron loss. In recognition of this surgical CKD significance, there has been an increase in PN rates over the past decade. Despite this trend, RN remains indispensable for patients with larger tumors, and its effects are yet to be fully understood. In fact, a recent study demonstrated that larger tumor volume is associated with better postoperative renal function in patients undergoing RN (128). This finding suggests possible but unconfirmed significant compensatory hyperfiltration by the contralateral kidney preceding the actual RN. However, a similar phenomenon has been observed in living kidney donors (58), raising concerns about the validity of MDRD or CKD-EPI formulas for eGFR in staging the CKD in the settings of surgically induced nephron loss. There have been several attempts to employ three-dimensional image reconstruction to correlate tumor and renal volumes with eGFR (72). One interesting approach to estimating change in renal function due to tumor resection involves correcting eGFR by functional renal volume (eGFR/FRV), which emerged as an independent predictor of long-term postoperative renal function. However, the eGFR/FRV index remains to be validated in terms of long-term consequences of CKD defined in this particular fashion (48).

In addition to RN, preoperative hypertension, diabetes, reduced GFR, and albuminuria are the recognized risk factors for postoperative CKD (28, 48, 49). They are present frequently in RCC patients and are potentially modifiable. Albuminuria in particular has been given much attention in recent nephrology literature and has been established as a strong and independent predictor of adverse cardiovascular outcomes and mortality (40). Importantly, albuminuria is a manifestation of glomerular injury in many systemic and kidney diseases, including membranous and membranoproliferative glomerulonephritis (GN), focal segmental glomerulosclerosis (FSGS), IgA nephropathy, minimal change disease, amyloidosis, and crescentic GN, that are found in 16–43% of patients undergoing nephrectomies (6, 7, 47, 105) and may prove helpful in managing postoperative CKD. However, albuminuria is largely overlooked in both clinical urology practice and research, with most studies on proteinuria relying on semiquantitative methodology (48, 64). Moreover, only a minority of nonneoplastic tissue samples are formally examined (4, 105), squandering an opportunity for the safe establishment of a formal histopathological diagnosis for many patients that would benefit from a disease-specific or disease-modifying therapy immediately or later in life. Because CKD is a well-established risk factor for cardiovascular disease, all-cause mortality, and ESRD (38, 41), the 50%, 5-yr mortality of which exceeds that of RCC itself (22, 94), efforts of urologists, nephrologists, and pathologists should be coordinated and concentrated on minimizing the risks of GFR decline in the perioperative period in RCC patients.

Systemic Therapies

Systemic treatment is indicated for patients with unresectable or metastatic RCC (Fig. 1). IL-2 immunotherapy is historically the first-line treatment option intended to direct the immune system against the tumor. Because of its prohibitively high rate of severe toxicities, IL-2 treatment is currently offered only to a small proportion of exceptionally healthy RCC patients, in whom it may produce a durable response (63). Antiangiogenic drugs are the mainstay for the remaining patient majority and those whose disease progresses after immunotherapy, whereas immune checkpoint inhibitors may be offered as part of a clinical trial. Failure of antiangiogenic drugs in patients who were not exposed to immunotherapy warrants initiation of immune checkpoint inhibitors. Otherwise, a different antiangiogenic agent can be offered. Mechanistic target of rapamycin (mTOR) inhibitors may be considered as a third-line therapy or for patients with non-ccRCC.

Fig. 1.

Fig. 1.

Mechanisms of action of commonly used systemic agents in treatment of renal cell carcinoma (RCC). Multiple strategies have been developed for the treatment of advanced RCC. High-dose IL-2 is still the first-line treatment for the minority of patients with intact organ function. If available, protocol treatment with nivolumab is another preferred strategy aimed at activating an immune response against RCC. It can also be used for patients with progression of disease on molecularly targeted therapy that includes vascular endothelial growth factor (VEGF) antibody, bevacizumab, and multiple small-molecule tyrosine kinase inhibitors (TKI) active against VEGF receptor and, in the case of sorafenib, against members of the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinases (ERK) that promote endothelial cell proliferation and angiogenesis. Mammalian target of rapamycin (mTOR) inhibitors, including everolimus and temsirolimus, disrupt growth factor signaling via the phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt)/mTOR pathway regulating cell proliferation. At present, these agents are recommended for patients not responding to first-line anti-VEGF therapy.

The heterogeneic nature of RCC poses significant challenges in evaluating patient treatment responses. Recently expanding knowledge of ccRCC pathophysiology in particular continues to produce novel therapeutic agents tailored to the specific biological pathways described below.

Antiangiogenic Agents

The angiogenic response in RCC tissue characterized by “functional hypoxia” induced by VHL gene aberrations made this tumor a prime target for the first intravenous antiangiogenic drug, bevacizumab, that was approved for use in RCC in combination with immunomodulator interferon-α in 2009 (36, 102). This regimen is reported to be associated with the overall survival (OS) of ≤23 mo (34). Since then, several small molecule tyrosine kinase inhibitors with anti-VEGF activity and comparable efficacy have been developed for oral administration (35, 91, 101, 112). An extensively studied representative of this group, sunitinib, exerts its antiangiogenic and antiproliferative effects by inhibiting VEGF (1 and 2) and PDGF (α and β) receptors and is a common first-line agent for patients with prior exposure or contraindications to immunotherapy and is reported to prolong OS to 26 mo (90). Another recently approved first-line agent, pazopanib, has similar receptor targets and efficacy but somewhat better side effect profile compared with sunitinib (89). Axitinib is thought to be a more potent TKI with a wider range of receptor targets (VEGF1-3) compared with suntinib. Although it is a second-line agent, its dose adjustability can be potentially advantageous for balancing the incidence of side effects and maximizing clinical outcomes in select patients (101, 103). A distinguishing feature of another second-line agent, sorafenib, is its additional activity against Raf kinases that can be important for ≤50% of RCC patients with constitutive activation of the BRAF pathway (120). Both sorafenib and axitinib are associated with similar progression-free survival (53), but neither has been directly compared with sunitinib or pazopanib.

Among the side effects associated with antiangiogenic therapy, hypertension and proteinuria are frequently the reason for referral to a nephrologist. They have been well described, but the relationship between the two remains poorly understood (55, 77). Limited data from human kidney tissue and murine experimental models suggest the crucial role of podocyte-produced VEGF (33). Its selective inhibition results in the loss of glomerular endothelium fenestration, which promotes thrombotic microangiopathy and resultant hypertension (32). Additionally, hypertension is promoted by inactivation of VEGF-mediated endothelial nitric oxide synthase and prostacycline downregulation (104, 109). Unlike in rodents, proteinuria and hypertension in humans are much less uniform; both appear to be dose dependent and often diminish with dose reduction, treatment break, or addition of an antihypertensive agent, which is often an angiotensin-converting enzyme inhibitor (ACEi) (67, 103, 127). In some reports, incidents of anti-VEGF-induced hypertension have been described as markers or treatment responses and appear to be associated with prolonged patient survival; however, this relationship has been inconsistent (51, 84, 100). The choice of an antihypertensive agent has been addressed in several retrospective studies uniformly reporting better survival in patients treated with ACEi or angiotensin receptor blockers (ARB), highlighting the plausible role of angiotensin II in tumorigenesis (54, 59, 80). This effect held true for patients starting these agents before and after anti-VEGF treatment, although a significant proportion of the former group did not have the actual preexisting diagnosis of hypertension (80). Because significant creatinine elevation frequently leads to cancer therapy suspension or discontinuation, the optimal use of ACEi and ARB remains to be elucidated in the settings of RCC therapy frequently preceded by cytoreductive nephrectomy.

mTOR Inhibitors

Intravenous temsirolimus and oral everolimus are two derivatives of rapamycin most commonly used in patients with advanced metastatic RCC. Both agents form a complex with an intracellular protein FKPB-12, which blocks mTOR signaling. Disruption of the mTOR activation leads to G1 phase cell cycle arrest and HIF-1, HIF-2, and VEGF level reduction attenuating angiogenesis (82). These agents are commonly used for high-risk patients with nonclear cell RCC, confirmed PI3K pathway mutations, or those with disease refractory to VEGF-targeting drugs (50, 52, 88). Although generally well tolerated and described to have antifibrotic effects in renal tissue (8), mTOR inhibitors have been reported to be associated with AKI in renal transplant population (5) and cause significant glomerulosclerosis and proteinuria in animal models of reduced nephron volume (123). A possible explanation of these discordant treatment effects may be related to the dose of the drug and the predominant cell type (mesangial, endothelial, or epithelial) injured before mTOR therapy initiation (118).

Immune Checkpoint Inhibitors

Monoclonal antibodies against programmed cell death 1 (PD-1) protein have become an important option in the treatment of advanced RCC. PD-1 prevents activation of T cells by binding its target ligands PD-L1 and PD-L2, which are expressed on antigen-presenting cells; therefore, PD-1 inhibition is thought to augment the immune response against tumor cells (95). Compared with everolimus, intravenous nivolumab showed an improved overall survival of RCC patients, who had previously been treated with anti-VEGF drugs, and was approved by the FDA for RCC in 2015 (87). Unique toxicities of these agents termed immune-related adverse effects include colitis, hepatitis, interstitial nephritis, and inflammatory disorders of other organ systems that may limit therapy duration (92). In cases of biopsy-confirmed acute interstitial nephritis, renal dysfunction seems to readily resolve with immunotherapy discontinuation and short courses of glucocorticoids (24). With a rapid increase in the number of available therapeutic agents, multiple studies have been launched to compare the efficacy of combinations of drugs from different classes and their sequential use.

Conclusion and Future Directions

Significant stage migration of RCC at presentation due to earlier detection of smaller renal masses and greater life expectancy with novel treatment strategies associated with unique adverse effects offer fertile ground for research aimed at improving patient survival and understanding the molecular biology of RCC (Fig. 2). A significant proportion of RCC patients with metastatic disease at presentation calls for reliable screening biomarkers. Increased levels of markers of programmed cell death or altered angiogenesis such as cytokeratins or (PHD3) antibodies, respectively, have been detected in the serum of RCC patients (111, 114). Multiple biomarkers such as osteopontin, VEGF, and interleukins have been tested individually and in combinations to predict disease progression and response to different therapies with variable degrees of success (56, 99, 119). Metabolomic analysis of serum lipids and amino acids yielded promising preliminary results for RCC identification (39, 130). However, when applied to patients with resectable tumors, metabolomic analyses appear to be greatly impacted by the changes in body metabolism arising from the surgically reduced nephron volume (129). The underutilized opportunity to analyze nonneoplastic tissue removed during nephrectomy offers a prospect of discovering tissue biomarkers of postoperative CKD progression and adverse effects from systemic therapies. For example, discovery of immunoglobulin A nephropathy at the time of resection may inform oncologists against using everolimus in such patients with active glomerular inflammation that can be worsened by mTOR inhibition. Incident hypertension and proteinuria observed with anti-VEGF agents have not been adequately studied, and although their development often triggers treatment modifications, the rationale for treatment adjustment is not guided by the knowledge of long-term outcomes. In particular, the choice between ACEi and ARB for treatment of hypertension is currently overlooked but may be important because of the ARB’s preferential inhibition of angiotensin receptor (ATR) 2, which, unlike ATR1, mediates antiangiogenic effects (14). Common use of dipstick urinalysis in RCC patients is inadequate for accurate quantification of urine protein and may grossly underestimate the prevalence of thrombotic microangiopathy, its role in the development of anti-VEGF-induced hypertension, and the appropriate supportive therapy. The recently characterized genetic profiles of different histological types of RCC frequently overlap and may lead to reclassification of RCC while offering a more individualized approach to treatment strategy and establishing new treatment targets.

Fig. 2.

Fig. 2.

Research questions for future investigations in the areas of renal cell cancer diagnosis and treatment. CKD, chronic kidney disease; GFR, glomerular filtration rate; mTOR, mammalian target of rapamycin; RCC, renal cell carcinoma; VEGF, vascular endothelial growth factor.

The complexity of RCC calls for a collaborative effort of basic scientists and medical specialists directed at advancing and consolidating our knowledge of this tumor.

GRANTS

We acknowledge funding from National Institutes of Health/NCI Cancer Center Support Grant P30-CA-00874 and the Byrne Research Fund.

DISCLOSURES

No conflicts of interest, financial or otherwise, are declared by the authors.

AUTHOR CONTRIBUTIONS

R.S. and E.A.J. prepared figures; R.S. drafted manuscript; R.S. and E.A.J. approved final version of manuscript; E.A.J. edited and revised manuscript.

REFERENCES

  • 1.Abou Youssif T, Kassouf W, Steinberg J, Aprikian AG, Laplante MP, Tanguay S. Active surveillance for selected patients with renal masses: updated results with long-term follow-up. Cancer 110: 1010–1014, 2007. doi: 10.1002/cncr.22871. [DOI] [PubMed] [Google Scholar]
  • 2.Ahmad AE, Finelli A, Jewett MA. Surveillance of small renal masses. Urology 98: 8–13, 2016. doi: 10.1016/j.urology.2016.06.005. [DOI] [PubMed] [Google Scholar]
  • 3.Albiges L, Molinie V, Escudier B. Non-clear cell renal cell carcinoma: does the mammalian target of rapamycin represent a rational therapeutic target? Oncologist 17: 1051–1062, 2012. doi: 10.1634/theoncologist.2012-0038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Algaba F, Delahunt B, Berney DM, Camparo P, Compérat E, Griffiths D, Kristiansen G, Lopez-Beltran A, Martignoni G, Moch H, Montironi R, Varma M, Egevad L. Handling and reporting of nephrectomy specimens for adult renal tumours: a survey by the European Network of Uropathology. J Clin Pathol 65: 106–113, 2012. doi: 10.1136/jclinpath-2011-200339. [DOI] [PubMed] [Google Scholar]
  • 5.Barbari A, Maawad M, Kfoury Kassouf H, Kamel G. Mammalian target of rapamycin inhibitors and nephrotoxicity: fact or fiction. Exp Clin Transplant 13: 377–386, 2015. [PubMed] [Google Scholar]
  • 6.Beaufils H, Patte R, Aubert P, Camey M, Kuss R, Barbagelatta M, Chomette G. Renal immunopathology in renal cell carcinoma. Virchows Arch A Pathol Anat Histopathol 404: 87–97, 1984. doi: 10.1007/BF00704253. [DOI] [PubMed] [Google Scholar]
  • 7.Bijol V, Mendez GP, Hurwitz S, Rennke HG, Nosé V. Evaluation of the nonneoplastic pathology in tumor nephrectomy specimens: predicting the risk of progressive renal failure. Am J Surg Pathol 30: 575–584, 2006. doi: 10.1097/01.pas.0000194296.74097.87. [DOI] [PubMed] [Google Scholar]
  • 8.Bonegio RG, Fuhro R, Wang Z, Valeri CR, Andry C, Salant DJ, Lieberthal W. Rapamycin ameliorates proteinuria-associated tubulointerstitial inflammation and fibrosis in experimental membranous nephropathy. J Am Soc Nephrol 16: 2063–2072, 2005. doi: 10.1681/ASN.2004030180. [DOI] [PubMed] [Google Scholar]
  • 9.Brennan JF, Stilmant MM, Babayan RK, Siroky MB. Acquired renal cystic disease: implications for the urologist. Br J Urol 67: 342–348, 1991. doi: 10.1111/j.1464-410X.1991.tb15158.x. [DOI] [PubMed] [Google Scholar]
  • 10.Brooks SA, Khandani AH, Fielding JR, Lin W, Sills T, Lee Y, Arreola A, Milowsky MI, Wallen EM, Woods ME, Smith AB, Nielsen ME, Parker JS, Lalush DS, Rathmell WK. Alternate metabolic programs define regional variation of relevant biological features in renal cell carcinoma progression. Clin Cancer Res 22: 2950–2959, 2016. doi: 10.1158/1078-0432.CCR-15-2115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Brunocilla E, Borghesi M, Schiavina R, Palmieri F, Pernetti R, Monti C, Martorana G. Active surveillance for small renal masses diagnosed in elderly or comorbid patients: looking for the best treatment strategy. Actas Urol Esp 38: 1–6, 2014. doi: 10.1016/j.acuro.2013.04.012. [DOI] [PubMed] [Google Scholar]
  • 12.Creighton CJ, Morgan M, Gunaratne PH, Wheeler DA, Gibbs RA, Gordon Robertson A, Chu A, Beroukhim R, Cibulskis K, Signoretti S, Vandin Hsin-Ta Wu F, Raphael BJ, Verhaak RGW, Tamboli P, Torres-Garcia W, Akbani R, Weinstein JN, Reuter V, Hsieh JJ, Rose Brannon A, Ari Hakimi A, Jacobsen A, Ciriello G, Reva B, Ricketts CJ, Marston Linehan W, Stuart JM, Kimryn Rathmell W, Shen H, Laird PW, Muzny D, Davis C, Morgan M, Xi L, Chang K, Kakkar N, Treviño LR, Benton S, Reid JG, Morton D, Doddapaneni H, Han Y, Lewis L, Dinh H, Kovar C, Zhu Y, Santibanez J, Wang M, Hale W, Kalra D, Creighton CJ, Wheeler DA, Gibbs RA, Getz G, Cibulskis K, Lawrence MS, Sougnez C, Carter SL, Sivachenko A, Lichtenstein L, Stewart C, Voet D, Fisher S, Gabriel SB, Lander E, Beroukhim R, Schumacher SE, Tabak B, Saksena G, Onofrio RC, Carter SL, Cherniack AD, Gentry J, Ardlie K, Sougnez C, Getz G, Gabriel SB, Meyerson M, Gordon Robertson A, Chu A, Chun H-JE, Mungall AJ, Sipahimalani P, Stoll D, Ally A, Balasundaram M, Butterfield YSN, Carlsen R, Carter C, Chuah E, Coope RJN, Dhalla N, Gorski S, Guin R, Hirst C, Hirst M, Holt RA, Lebovitz C, Lee D, Li HI, Mayo M, Moore RA, Pleasance E, Plettner P, Schein JE, Shafiei A, Slobodan JR, Tam A, Thiessen N, Varhol RJ, Wye N, Zhao Y, Birol I, Jones SJM, Marra MA, Auman JT, Tan D, Jones CD, Hoadley KA, Mieczkowski PA, Mose LE, Jefferys SR, Topal MD, Liquori C, Turman YJ, Shi Y, Waring S, Buda E, Walsh J, Wu J, Bodenheimer T, Hoyle AP, Simons JV, Soloway MG, Balu S, Parker JS, Neil Hayes D, Perou CM, Kucherlapati R, Park P, Shen H, Triche T Jr, Weisenberger DJ, Lai PH, Bootwalla MS, Maglinte DT, Mahurkar S, Berman BP, Van Den Berg DJ, Cope L, Baylin SB, Laird PW, Creighton CJ, Wheeler DA, Getz G, Noble MS, DiCara D, Zhang H, Cho J, Heiman DI, Gehlenborg N, Voet D, Mallard W, Lin P, Frazer S, Stojanov P, Liu Y, Zhou L, Kim J, Lawrence MS, Chin L, Vandin F, Wu H-T, Raphael BJ, Benz C, Yau C, Reynolds SM, Shmulevich I, Verhaak RGW, Torres-Garcia W, Vegesna R, Kim H, Zhang W, Cogdell D, Jonasch E, Ding Z, Lu Y, Akbani R, Zhang N, Unruh AK, Casasent TD, Wakefield C, Tsavachidou D, Chin L, Mills GB, Weinstein JN, Jacobsen A, Rose Brannon A, Ciriello G, Schultz N, Ari Hakimi A, Reva B, Antipin Y, Gao J, Cerami E, Gross B, Arman Aksoy B, Sinha R, Weinhold N, Onur Sumer S, Taylor BS, Shen R, Ostrovnaya I, Hsieh JJ, Berger MF, Ladanyi M, Sander C, Fei SS, Stout A, Spellman PT, Rubin DL, Liu TT, Stuart JM, Ng S, Paull EO, Carlin D, Goldstein T, Waltman P, Ellrott K, Zhu J, Haussler D, Gunaratne PH, Xiao W, Shelton C, Gardner J, Penny R, Sherman M, Mallery D, Morris S, Paulauskis J, Burnett K, Shelton T, Signoretti S, Kaelin WG, Choueiri T, Atkins MB, Penny R, Burnett K, Mallery D, Curley E, Tickoo S, Reuter V, Kimryn Rathmell W, Thorne L, Boice L, Huang M, Fisher JC, Marston Linehan W, Vocke CD, Peterson J, Worrell R, Merino MJ, Schmidt LS, Tamboli P, Czerniak BA, Aldape KD, Wood CG, Boyd J, Weaver JE, Iacocca MV, Petrelli N, Witkin G, Brown J, Czerwinski C, Huelsenbeck-Dill L, Rabeno B, Myers J, Morrison C, Bergsten J, Eckman J, Harr J, Smith C, Tucker K, Anne Zach L, Bshara W, Gaudioso C, Morrison C, Dhir R, Maranchie J, Nelson J, Parwani A, Potapova O, Fedosenko K, Cheville JC, Houston Thompson R, Signoretti S, Kaelin WG, Atkins MB, Tickoo S, Reuter V, Marston Linehan W, Vocke CD, Peterson J, Merino MJ, Schmidt LS, Tamboli P, Mosquera JM, Rubin MA, Blute ML, Kimryn Rathmell W, Pihl T, Jensen M, Sfeir R, Kahn A, Chu A, Kothiyal P, Snyder E, Pontius J, Ayala B, Backus M, Walton J, Baboud J, Berton D, Nicholls M, Srinivasan D, Raman R, Girshik S, Kigonya P, Alonso S, Sanbhadti R, Barletta S, Pot D, Sheth M, Demchok JA, Davidsen T, Wang Z, Yang L, Tarnuzzer RW, Zhang J, Eley G, Ferguson ML, Mills Shaw KR, Guyer MS, Ozenberger BA, Sofia HJ; Cancer Genome Atlas Research Network . Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature 499: 43–49, 2013. doi: 10.1038/nature12222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Cancer Genome Atlas Research Network; Linehan WM, Spellman PT, Ricketts CJ, Creighton CJ, Fei SS, Davis C, Wheeler DA, Murray BA, Schmidt L, Vocke CD, Peto M, Al Mamun AA, Shinbrot E, Sethi A, Brooks S, Rathmell WK, Brooks AN, Hoadley KA, Robertson AG, Brooks D, Bowlby R, Sadeghi S, Shen H, Weisenberger DJ, Bootwalla M, Baylin SB, Laird PW, Cherniack AD, Saksena G, Haake S, Li J, Liang H, Lu Y, Mills GB, Akbani R, Leiserson MD, Raphael BJ, Anur P, Bottaro D, Albiges L, Barnabas N, Choueiri TK, Czerniak B, Godwin AK, Hakimi AA, Ho TH, Hsieh J, Ittmann M, Kim WY, Krishnan B, Merino MJ, Mills Shaw KR, Reuter VE, Reznik E, Shelley CS, Shuch B, Signoretti S, Srinivasan R, Tamboli P, Thomas G, Tickoo S, Burnett K, Crain D, Gardner J, Lau K, Mallery D, Morris S, Paulauskis JD, Penny RJ, Shelton C, Shelton WT, Sherman M, Thompson E, Yena P, Avedon MT, Bowen J, Gastier-Foster JM, Gerken M, Leraas KM, Lichtenberg TM, Ramirez NC, Santos T, Wise L, Zmuda E, Demchok JA, Felau I, Hutter CM, Sheth M, Sofia HJ, Tarnuzzer R, Wang Z, Yang L, Zenklusen JC, Zhang J, Ayala B, Baboud J, Chudamani S, Liu J, Lolla L, Naresh R, Pihl T, Sun Q, Wan Y, Wu Y, Ally A, Balasundaram M, Balu S, Beroukhim R, Bodenheimer T, Buhay C, Butterfield YS, Carlsen R, Carter SL, Chao H, Chuah E, Clarke A, Covington KR, Dahdouli M, Dewal N, Dhalla N, Doddapaneni HV, Drummond JA, Gabriel SB, Gibbs RA, Guin R, Hale W, Hawes A, Hayes DN, Holt RA, Hoyle AP, Jefferys SR, Jones SJ, Jones CD, Kalra D, Kovar C, Lewis L, Li J, Ma Y, Marra MA, Mayo M, Meng S, Meyerson M, Mieczkowski PA, Moore RA, Morton D, Mose LE, Mungall AJ, Muzny D, Parker JS, Perou CM, Roach J, Schein JE, Schumacher SE, Shi Y, Simons JV, Sipahimalani P, Skelly T, Soloway MG, Sougnez C, Tam A, Tan D, Thiessen N, Veluvolu U, Wang M, Wilkerson MD, Wong T, Wu J, Xi L, Zhou J, Bedford J, Chen F, Fu Y, Gerstein M, Haussler D, Kasaian K, Lai P, Ling S, Radenbaugh A, Van Den Berg D, Weinstein JN, Zhu J, Albert M, Alexopoulou I, Andersen JJ, Auman JT, Bartlett J, Bastacky S, Bergsten J, Blute ML, Boice L, Bollag RJ, Boyd J, Castle E, Chen YB, Cheville JC, Curley E, Davies B, DeVolk A, Dhir R, Dike L, Eckman J, Engel J, Harr J, Hrebinko R, Huang M, Huelsenbeck-Dill L, Iacocca M, Jacobs B, Lobis M, Maranchie JK, McMeekin S, Myers J, Nelson J, Parfitt J, Parwani A, Petrelli N, Rabeno B, Roy S, Salner AL, Slaton J, Stanton M, Thompson RH, Thorne L, Tucker K, Weinberger PM, Winemiller C, Zach LA, Zuna R. Comprehensive molecular characterization of papillary renal-cell carcinoma. N Engl J Med 374: 135–145, 2016. doi: 10.1056/NEJMoa1505917. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Carbajo-Lozoya J, Lutz S, Feng Y, Kroll J, Hammes HP, Wieland T. Angiotensin II modulates VEGF-driven angiogenesis by opposing effects of type 1 and type 2 receptor stimulation in the microvascular endothelium. Cell Signal 24: 1261–1269, 2012. doi: 10.1016/j.cellsig.2012.02.005. [DOI] [PubMed] [Google Scholar]
  • 15.Centers for Disease Control and Prevention (CDC) Prevalence of chronic kidney disease and associated risk factors–United States, 1999-2004. MMWR Morb Mortal Wkly Rep 56: 161–165, 2007. [PubMed] [Google Scholar]
  • 16.Choueiri TK, Plantade A, Elson P, Negrier S, Ravaud A, Oudard S, Zhou M, Rini BI, Bukowski RM, Escudier B. Efficacy of sunitinib and sorafenib in metastatic papillary and chromophobe renal cell carcinoma. J Clin Oncol 26: 127–131, 2008. doi: 10.1200/JCO.2007.13.3223. [DOI] [PubMed] [Google Scholar]
  • 17.Choueiri TK, Xie W, Kollmannsberger C, North S, Knox JJ, Lampard JG, McDermott DF, Rini BI, Heng DY. The impact of cytoreductive nephrectomy on survival of patients with metastatic renal cell carcinoma receiving vascular endothelial growth factor targeted therapy. J Urol 185: 60–66, 2011. doi: 10.1016/j.juro.2010.09.012. [DOI] [PubMed] [Google Scholar]
  • 18.Chow WH, Devesa SS, Warren JL, Fraumeni JF Jr. Rising incidence of renal cell cancer in the United States. JAMA 281: 1628–1631, 1999. doi: 10.1001/jama.281.17.1628. [DOI] [PubMed] [Google Scholar]
  • 19.Chow WH, Dong LM, Devesa SS. Epidemiology and risk factors for kidney cancer. Nat Rev Urol 7: 245–257, 2010. doi: 10.1038/nrurol.2010.46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Clark MA, Shikanov S, Raman JD, Smith B, Kaag M, Russo P, Wheat JC, Wolf JS Jr, Matin SF, Huang WC, Shalhav AL, Eggener SE. Chronic kidney disease before and after partial nephrectomy. J Urol 185: 43–48, 2011. doi: 10.1016/j.juro.2010.09.019. [DOI] [PubMed] [Google Scholar]
  • 21.Clayman RV, Surya V, Miller RP, Reinke DB, Fraley EE. Pursuit of the renal mass. Is ultrasound enough? Am J Med 77: 218–223, 1984. doi: 10.1016/0002-9343(84)90694-6. [DOI] [PubMed] [Google Scholar]
  • 22.Collins AJ, Kasiske B, Herzog C, Chavers B, Foley R, Gilbertson D, Grimm R, Liu J, Louis T, Manning W, Matas A, McBean M, Murray A, St Peter W, Xue J, Fan Q, Guo H, Li S, Li S, Roberts T, Snyder J, Solid C, Wang C, Weinhandl E, Arko C, Chen SC, Dalleska F, Daniels F, Dunning S, Ebben J, Frazier E, Johnson R, Sheets D, Forrest B, Berrini D, Constantini E, Everson S, Frederick P, Eggers P, Agodoa L; United States Renal Data System . Excerpts from the United States Renal Data System 2004 annual data report: atlas of end-stage renal disease in the United States. Am J Kidney Dis 45, Suppl 1: A5–A7, 2005. doi: 10.1053/j.ajkd.2004.10.009. [DOI] [PubMed] [Google Scholar]
  • 23.Conti SL, Thomas IC, Hagedorn JC, Chung BI, Chertow GM, Wagner TH, Brooks JD, Srinivas S, Leppert JT. Utilization of cytoreductive nephrectomy and patient survival in the targeted therapy era. Int J Cancer 134: 2245–2252, 2014. doi: 10.1002/ijc.28553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Cortazar FB, Marrone KA, Troxell ML, Ralto KM, Hoenig MP, Brahmer JR, Le DT, Lipson EJ, Glezerman IG, Wolchok J, Cornell LD, Feldman P, Stokes MB, Zapata SA, Hodi FS, Ott PA, Yamashita M, Leaf DE. Clinicopathological features of acute kidney injury associated with immune checkpoint inhibitors. Kidney Int 90: 638–647, 2016. doi: 10.1016/j.kint.2016.04.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Curry NS. Small renal masses (lesions smaller than 3 cm): imaging evaluation and management. AJR Am J Roentgenol 164: 355–362, 1995. doi: 10.2214/ajr.164.2.7839969. [DOI] [PubMed] [Google Scholar]
  • 26.Dalgliesh GL, Furge K, Greenman C, Chen L, Bignell G, Butler A, Davies H, Edkins S, Hardy C, Latimer C, Teague J, Andrews J, Barthorpe S, Beare D, Buck G, Campbell PJ, Forbes S, Jia M, Jones D, Knott H, Kok CY, Lau KW, Leroy C, Lin ML, McBride DJ, Maddison M, Maguire S, McLay K, Menzies A, Mironenko T, Mulderrig L, Mudie L, O’Meara S, Pleasance E, Rajasingham A, Shepherd R, Smith R, Stebbings L, Stephens P, Tang G, Tarpey PS, Turrell K, Dykema KJ, Khoo SK, Petillo D, Wondergem B, Anema J, Kahnoski RJ, Teh BT, Stratton MR, Futreal PA. Systematic sequencing of renal carcinoma reveals inactivation of histone modifying genes. Nature 463: 360–363, 2010. doi: 10.1038/nature08672. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Daugherty M, Blakely S, Shapiro O, Vourganti S, Mollapour M, Bratslavsky G. Chromophobe Renal Cell Carcinoma is the Most Common Nonclear Renal Cell Carcinoma in Young Women: Results from the SEER Database. J Urol 195: 847–851, 2016. doi: 10.1016/j.juro.2015.10.177. [DOI] [PubMed] [Google Scholar]
  • 28.Demirjian S, Lane BR, Derweesh IH, Takagi T, Fergany A, Campbell SC. Chronic kidney disease due to surgical removal of nephrons: relative rates of progression and survival. J Urol 192: 1057–1063, 2014. doi: 10.1016/j.juro.2014.04.016. [DOI] [PubMed] [Google Scholar]
  • 29.Denton MD, Magee CC, Ovuworie C, Mauiyyedi S, Pascual M, Colvin RB, Cosimi AB, Tolkoff-Rubin N. Prevalence of renal cell carcinoma in patients with ESRD pre-transplantation: a pathologic analysis. Kidney Int 61: 2201–2209, 2002. doi: 10.1046/j.1523-1755.2002.00374.x. [DOI] [PubMed] [Google Scholar]
  • 30.Duns G, Hofstra RM, Sietzema JG, Hollema H, van Duivenbode I, Kuik A, Giezen C, Jan O, Bergsma JJ, Bijnen H, van der Vlies P, van den Berg E, Kok K. Targeted exome sequencing in clear cell renal cell carcinoma tumors suggests aberrant chromatin regulation as a crucial step in ccRCC development. Hum Mutat 33: 1059–1062, 2012. doi: 10.1002/humu.22090. [DOI] [PubMed] [Google Scholar]
  • 31.Eble JN, Sauter G, Epstein JI, Sesterhenn IA. Pathology and Genetics of Tumours of the Urinary System and Male Genital Organs. Lyon, France: International Agency for Research on Cancer (IARC), 2004. [Google Scholar]
  • 32.Eremina V, Jefferson JA, Kowalewska J, Hochster H, Haas M, Weisstuch J, Richardson C, Kopp JB, Kabir MG, Backx PH, Gerber HP, Ferrara N, Barisoni L, Alpers CE, Quaggin SE. VEGF inhibition and renal thrombotic microangiopathy. N Engl J Med 358: 1129–1136, 2008. doi: 10.1056/NEJMoa0707330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Eremina V, Quaggin SE. The role of VEGF-A in glomerular development and function. Curr Opin Nephrol Hypertens 13: 9–15, 2004. doi: 10.1097/00041552-200401000-00002. [DOI] [PubMed] [Google Scholar]
  • 34.Escudier B, Bellmunt J, Négrier S, Bajetta E, Melichar B, Bracarda S, Ravaud A, Golding S, Jethwa S, Sneller V. Phase III trial of bevacizumab plus interferon alfa-2a in patients with metastatic renal cell carcinoma (AVOREN): final analysis of overall survival. J Clin Oncol 28: 2144–2150, 2010. doi: 10.1200/JCO.2009.26.7849. [DOI] [PubMed] [Google Scholar]
  • 35.Escudier B, Eisen T, Stadler WM, Szczylik C, Oudard S, Siebels M, Negrier S, Chevreau C, Solska E, Desai AA, Rolland F, Demkow T, Hutson TE, Gore M, Freeman S, Schwartz B, Shan M, Simantov R, Bukowski RM; TARGET Study Group . Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med 356: 125–134, 2007. doi: 10.1056/NEJMoa060655. [DOI] [PubMed] [Google Scholar]
  • 36.Escudier B, Pluzanska A, Koralewski P, Ravaud A, Bracarda S, Szczylik C, Chevreau C, Filipek M, Melichar B, Bajetta E, Gorbunova V, Bay JO, Bodrogi I, Jagiello-Gruszfeld A, Moore N; AVOREN Trial investigators . Bevacizumab plus interferon alfa-2a for treatment of metastatic renal cell carcinoma: a randomised, double-blind phase III trial. Lancet 370: 2103–2111, 2007. doi: 10.1016/S0140-6736(07)61904-7. [DOI] [PubMed] [Google Scholar]
  • 37.Frank I, Blute ML, Cheville JC, Lohse CM, Weaver AL, Zincke H. Solid renal tumors: an analysis of pathological features related to tumor size. J Urol 170: 2217–2220, 2003. doi: 10.1097/01.ju.0000095475.12515.5e. [DOI] [PubMed] [Google Scholar]
  • 38.Fried LF, Katz R, Sarnak MJ, Shlipak MG, Chaves PH, Jenny NS, Stehman-Breen C, Gillen D, Bleyer AJ, Hirsch C, Siscovick D, Newman AB. Kidney function as a predictor of noncardiovascular mortality. J Am Soc Nephrol 16: 3728–3735, 2005. doi: 10.1681/ASN.2005040384. [DOI] [PubMed] [Google Scholar]
  • 39.Gao H, Dong B, Liu X, Xuan H, Huang Y, Lin D. Metabonomic profiling of renal cell carcinoma: high-resolution proton nuclear magnetic resonance spectroscopy of human serum with multivariate data analysis. Anal Chim Acta 624: 269–277, 2008. doi: 10.1016/j.aca.2008.06.051. [DOI] [PubMed] [Google Scholar]
  • 40.Gerstein HC, Mann JF, Yi Q, Zinman B, Dinneen SF, Hoogwerf B, Hallé JP, Young J, Rashkow A, Joyce C, Nawaz S, Yusuf S; HOPE Study Investigators . Albuminuria and risk of cardiovascular events, death, and heart failure in diabetic and nondiabetic individuals. JAMA 286: 421–426, 2001. doi: 10.1001/jama.286.4.421. [DOI] [PubMed] [Google Scholar]
  • 41.Go AS, Chertow GM, Fan D, McCulloch CE, Hsu CY. Chronic kidney disease and the risks of death, cardiovascular events, and hospitalization. N Engl J Med 351: 1296–1305, 2004. doi: 10.1056/NEJMoa041031. [DOI] [PubMed] [Google Scholar]
  • 42.Golan S, Patel AR, Eggener SE, Shalhav AL. The volume of nonneoplastic parenchyma in a minimally invasive partial nephrectomy specimen: predictive factors and impact on renal function. J Endourol 28: 196–200, 2014. doi: 10.1089/end.2013.0486. [DOI] [PubMed] [Google Scholar]
  • 43.Gudbjartsson T, Thoroddsen A, Petursdottir V, Hardarson S, Magnusson J, Einarsson GV. Effect of incidental detection for survival of patients with renal cell carcinoma: results of population-based study of 701 patients. Urology 66: 1186–1191, 2005. doi: 10.1016/j.urology.2005.07.009. [DOI] [PubMed] [Google Scholar]
  • 44.Haddad RL, Patel MI, Vladica P, Kassouf W, Bladou F, Anidjar M. Percutaneous radiofrequency ablation of small renal tumors using CT-guidance: a review and its current role. Urol J 9: 629–638, 2012. [PubMed] [Google Scholar]
  • 45.Hakimi AA, Chen YB, Wren J, Gonen M, Abdel-Wahab O, Heguy A, Liu H, Takeda S, Tickoo SK, Reuter VE, Voss MH, Motzer RJ, Coleman JA, Cheng EH, Russo P, Hsieh JJ. Clinical and pathologic impact of select chromatin-modulating tumor suppressors in clear cell renal cell carcinoma. Eur Urol 63: 848–854, 2013. doi: 10.1016/j.eururo.2012.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Hemmelgarn BR, Zhang J, Manns BJ, James MT, Quinn RR, Ravani P, Klarenbach SW, Culleton BF, Krause R, Thorlacius L, Jain AK, Tonelli M; Alberta Kidney Disease Network . Nephrology visits and health care resource use before and after reporting estimated glomerular filtration rate. JAMA 303: 1151–1158, 2010. doi: 10.1001/jama.2010.303. [DOI] [PubMed] [Google Scholar]
  • 47.Henriksen KJ, Meehan SM, Chang A. Non-neoplastic renal diseases are often unrecognized in adult tumor nephrectomy specimens: a review of 246 cases. Am J Surg Pathol 31: 1703–1708, 2007. doi: 10.1097/PAS.0b013e31804ca63e. [DOI] [PubMed] [Google Scholar]
  • 48.Hosokawa Y, Tanaka N, Mibu H, Anai S, Torimoto K, Yoneda T, Hirayama A, Yoshida K, Hayashi Y, Hirao Y, Fujimoto K. Follow-up study of unilateral renal function after nephrectomy assessed by glomerular filtration rate per functional renal volume. World J Surg Oncol 12: 59, 2014. doi: 10.1186/1477-7819-12-59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Huang WC, Levey AS, Serio AM, Snyder M, Vickers AJ, Raj GV, Scardino PT, Russo P. Chronic kidney disease after nephrectomy in patients with renal cortical tumours: a retrospective cohort study. Lancet Oncol 7: 735–740, 2006. doi: 10.1016/S1470-2045(06)70803-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Hudes G, Carducci M, Tomczak P, Dutcher J, Figlin R, Kapoor A, Staroslawska E, Sosman J, McDermott D, Bodrogi I, Kovacevic Z, Lesovoy V, Schmidt-Wolf IG, Barbarash O, Gokmen E, O’Toole T, Lustgarten S, Moore L, Motzer RJ; Global ARCC Trial . Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med 356: 2271–2281, 2007. doi: 10.1056/NEJMoa066838. [DOI] [PubMed] [Google Scholar]
  • 51.Hurwitz HI, Douglas PS, Middleton JP, Sledge GW, Johnson DH, Reardon DA, Chen D, Rosen O. Analysis of early hypertension and clinical outcome with bevacizumab: results from seven phase III studies. Oncologist 18: 273–280, 2013. doi: 10.1634/theoncologist.2012-0339. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Hutson TE, Escudier B, Esteban E, Bjarnason GA, Lim HY, Pittman KB, Senico P, Niethammer A, Lu DR, Hariharan S, Motzer RJ. Randomized phase III trial of temsirolimus versus sorafenib as second-line therapy after sunitinib in patients with metastatic renal cell carcinoma. J Clin Oncol 32: 760–767, 2014. doi: 10.1200/JCO.2013.50.3961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Hutson TE, Lesovoy V, Al-Shukri S, Stus VP, Lipatov ON, Bair AH, Rosbrook B, Chen C, Kim S, Vogelzang NJ. Axitinib versus sorafenib as first-line therapy in patients with metastatic renal-cell carcinoma: a randomised open-label phase 3 trial. Lancet Oncol 14: 1287–1294, 2013. doi: 10.1016/S1470-2045(13)70465-0. [DOI] [PubMed] [Google Scholar]
  • 54.Izzedine H, Derosa L, Le Teuff G, Albiges L, Escudier B. Hypertension and angiotensin system inhibitors: impact on outcome in sunitinib-treated patients for metastatic renal cell carcinoma. Ann Oncol 26: 1128–1133, 2015. doi: 10.1093/annonc/mdv147. [DOI] [PubMed] [Google Scholar]
  • 55.Izzedine H, Massard C, Spano JP, Goldwasser F, Khayat D, Soria JC. VEGF signalling inhibition-induced proteinuria: Mechanisms, significance and management. Eur J Cancer 46: 439–448, 2010. doi: 10.1016/j.ejca.2009.11.001. [DOI] [PubMed] [Google Scholar]
  • 56.Jacobsen J, Grankvist K, Rasmuson T, Bergh A, Landberg G, Ljungberg B. Expression of vascular endothelial growth factor protein in human renal cell carcinoma. BJU Int 93: 297–302, 2004. doi: 10.1111/j.1464-410X.2004.04605.x. [DOI] [PubMed] [Google Scholar]
  • 57.Johnson DB, Solomon SB, Su LM, Matsumoto ED, Kavoussi LR, Nakada SY, Moon TD, Shingleton WB, Cadeddu JA. Defining the complications of cryoablation and radio frequency ablation of small renal tumors: a multi-institutional review. J Urol 172: 874–877, 2004. doi: 10.1097/01.ju.0000135833.67906.ec. [DOI] [PubMed] [Google Scholar]
  • 58.Kasiske BL, Anderson-Haag T, Israni AK, Kalil RS, Kimmel PL, Kraus ES, Kumar R, Posselt AA, Pesavento TE, Rabb H, Steffes MW, Snyder JJ, Weir MR. A prospective controlled study of living kidney donors: three-year follow-up. Am J Kidney Dis 66: 114–124, 2015. doi: 10.1053/j.ajkd.2015.01.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Keizman D, Huang P, Eisenberger MA, Pili R, Kim JJ, Antonarakis ES, Hammers H, Carducci MA. Angiotensin system inhibitors and outcome of sunitinib treatment in patients with metastatic renal cell carcinoma: a retrospective examination. Eur J Cancer 47: 1955–1961, 2011. doi: 10.1016/j.ejca.2011.04.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Kim SH, Lee SE, Hong SK, Jeong CW, Park YH, Kim YJ, Kang SH, Hong SH, Choi WS, Byun SS. Incidence and risk factors of chronic kidney disease in korean patients with t1a renal cell carcinoma before and after radical or partial nephrectomy. Jpn J Clin Oncol 43: 1243–1248, 2013. doi: 10.1093/jjco/hyt149. [DOI] [PubMed] [Google Scholar]
  • 61.Kim SP, Thompson RH, Boorjian SA, Weight CJ, Han LC, Murad MH, Shippee ND, Erwin PJ, Costello BA, Chow GK, Leibovich BC. Comparative effectiveness for survival and renal function of partial and radical nephrectomy for localized renal tumors: a systematic review and meta-analysis. J Urol 188: 51–57, 2012. doi: 10.1016/j.juro.2012.03.006. [DOI] [PubMed] [Google Scholar]
  • 62.Kim WY, Kaelin WG. Role of VHL gene mutation in human cancer. J Clin Oncol 22: 4991–5004, 2004. doi: 10.1200/JCO.2004.05.061. [DOI] [PubMed] [Google Scholar]
  • 63.Klapper JA, Downey SG, Smith FO, Yang JC, Hughes MS, Kammula US, Sherry RM, Royal RE, Steinberg SM, Rosenberg S. High-dose interleukin-2 for the treatment of metastatic renal cell carcinoma : a retrospective analysis of response and survival in patients treated in the surgery branch at the National Cancer Institute between 1986 and 2006. Cancer 113: 293–301, 2008. doi: 10.1002/cncr.23552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Klarenbach S, Moore RB, Chapman DW, Dong J, Braam B. Adverse renal outcomes in subjects undergoing nephrectomy for renal tumors: a population-based analysis. Eur Urol 59: 333–339, 2011. doi: 10.1016/j.eururo.2010.11.013. [DOI] [PubMed] [Google Scholar]
  • 65.Kovac M, Navas C, Horswell S, Salm M, Bardella C, Rowan A, Stares M, Castro-Giner F, Fisher R, de Bruin EC, Kovacova M, Gorman M, Makino S, Williams J, Jaeger E, Jones A, Howarth K, Larkin J, Pickering L, Gore M, Nicol DL, Hazell S, Stamp G, O’Brien T, Challacombe B, Matthews N, Phillimore B, Begum S, Rabinowitz A, Varela I, Chandra A, Horsfield C, Polson A, Tran M, Bhatt R, Terracciano L, Eppenberger-Castori S, Protheroe A, Maher E, El Bahrawy M, Fleming S, Ratcliffe P, Heinimann K, Swanton C, Tomlinson I. Recurrent chromosomal gains and heterogeneous driver mutations characterise papillary renal cancer evolution. Nat Commun 6: 6336, 2015. doi: 10.1038/ncomms7336. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Kutikov A, Fossett LK, Ramchandani P, Tomaszewski JE, Siegelman ES, Banner MP, Van Arsdalen KN, Wein AJ, Malkowicz SB. Incidence of benign pathologic findings at partial nephrectomy for solitary renal mass presumed to be renal cell carcinoma on preoperative imaging. Urology 68: 737–740, 2006. doi: 10.1016/j.urology.2006.04.011. [DOI] [PubMed] [Google Scholar]
  • 67.Langenberg MH, van Herpen CM, De Bono J, Schellens JH, Unger C, Hoekman K, Blum HE, Fiedler W, Drevs J, Le Maulf F, Fielding A, Robertson J, Voest EE. Effective strategies for management of hypertension after vascular endothelial growth factor signaling inhibition therapy: results from a phase II randomized, factorial, double-blind study of Cediranib in patients with advanced solid tumors. J Clin Oncol 27: 6152–6159, 2009. doi: 10.1200/JCO.2009.22.2273. [DOI] [PubMed] [Google Scholar]
  • 68.Leibovich BC, Lohse CM, Crispen PL, Boorjian SA, Thompson RH, Blute ML, Cheville JC. Histological subtype is an independent predictor of outcome for patients with renal cell carcinoma. J Urol 183: 1309–1316, 2010. doi: 10.1016/j.juro.2009.12.035. [DOI] [PubMed] [Google Scholar]
  • 69.Li B, Qiu B, Lee DS, Walton ZE, Ochocki JD, Mathew LK, Mancuso A, Gade TP, Keith B, Nissim I, Simon MC. Fructose-1,6-bisphosphatase opposes renal carcinoma progression. Nature 513: 251–255, 2014. doi: 10.1038/nature13557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Li L, Lau WL, Rhee CM, Harley K, Kovesdy CP, Sim JJ, Jacobsen S, Chang A, Landman J, Kalantar-Zadeh K. Risk of chronic kidney disease after cancer nephrectomy. Nat Rev Nephrol 10: 135–145, 2014. doi: 10.1038/nrneph.2013.273. [DOI] [PubMed] [Google Scholar]
  • 71.Linehan WM, Pinto PA, Bratslavsky G, Pfaffenroth E, Merino M, Vocke CD, Toro JR, Bottaro D, Neckers L, Schmidt LS, Srinivasan R. Hereditary kidney cancer: unique opportunity for disease-based therapy. Cancer 115, Suppl: 2252–2261, 2009. doi: 10.1002/cncr.24230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Liu W, Zhu Y, Zhu X, Yang G, Xu Y, Tang L. CT-based renal volume measurements: correlation with renal function in patients with renal tumours. Clin Radiol 70: 1445–1450, 2015. doi: 10.1016/j.crad.2015.09.005. [DOI] [PubMed] [Google Scholar]
  • 73.Lonser RR, Glenn GM, Walther M, Chew EY, Libutti SK, Linehan WM, Oldfield EH. von Hippel-Lindau disease. Lancet 361: 2059–2067, 2003. doi: 10.1016/S0140-6736(03)13643-4. [DOI] [PubMed] [Google Scholar]
  • 74.Lopez-Beltran A, Carrasco JC, Cheng L, Scarpelli M, Kirkali Z, Montironi R. 2009 update on the classification of renal epithelial tumors in adults. Int J Urol 16: 432–443, 2009. doi: 10.1111/j.1442-2042.2009.02302.x. [DOI] [PubMed] [Google Scholar]
  • 75.Lowrance WT, Ordoñez J, Udaltsova N, Russo P, Go AS. CKD and the risk of incident cancer. J Am Soc Nephrol 25: 2327–2334, 2014. doi: 10.1681/ASN.2013060604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Lum JJ, Bui T, Gruber M, Gordan JD, DeBerardinis RJ, Covello KL, Simon MC, Thompson CB. The transcription factor HIF-1α plays a critical role in the growth factor-dependent regulation of both aerobic and anaerobic glycolysis. Genes Dev 21: 1037–1049, 2007. doi: 10.1101/gad.1529107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Maitland ML, Bakris GL, Black HR, Chen HX, Durand JB, Elliott WJ, Ivy SP, Leier CV, Lindenfeld J, Liu G, Remick SC, Steingart R, Tang WH; Cardiovascular Toxicities Panel, Convened by the Angiogenesis Task Force of the National Cancer Institute Investigational Drug Steering Committee . Initial assessment, surveillance, and management of blood pressure in patients receiving vascular endothelial growth factor signaling pathway inhibitors. J Natl Cancer Inst 102: 596–604, 2010. doi: 10.1093/jnci/djq091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Marconi L, Dabestani S, Lam TB, Hofmann F, Stewart F, Norrie J, Bex A, Bensalah K, Canfield SE, Hora M, Kuczyk MA, Merseburger AS, Mulders PF, Powles T, Staehler M, Ljungberg B, Volpe A. Systematic Review and Meta-analysis of Diagnostic Accuracy of Percutaneous Renal Tumour Biopsy. Eur Urol 69: 660–673, 2016. doi: 10.1016/j.eururo.2015.07.072. [DOI] [PubMed] [Google Scholar]
  • 79.Maxwell PH, Wiesener MS, Chang GW, Clifford SC, Vaux EC, Cockman ME, Wykoff CC, Pugh CW, Maher ER, Ratcliffe PJ. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature 399: 271–275, 1999. doi: 10.1038/20459. [DOI] [PubMed] [Google Scholar]
  • 80.McKay RR, Rodriguez GE, Lin X, Kaymakcalan MD, Hamnvik OP, Sabbisetti VS, Bhatt RS, Simantov R, Choueiri TK. Angiotensin system inhibitors and survival outcomes in patients with metastatic renal cell carcinoma. Clin Cancer Res 21: 2471–2479, 2015. doi: 10.1158/1078-0432.CCR-14-2332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.McKiernan J, Yossepowitch O, Kattan MW, Simmons R, Motzer RJ, Reuter VE, Russo P. Partial nephrectomy for renal cortical tumors: pathologic findings and impact on outcome. Urology 60: 1003–1009, 2002. doi: 10.1016/S0090-4295(02)01967-2. [DOI] [PubMed] [Google Scholar]
  • 82.Meric-Bernstam F, Gonzalez-Angulo AM. Targeting the mTOR signaling network for cancer therapy. J Clin Oncol 27: 2278–2287, 2009. doi: 10.1200/JCO.2008.20.0766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Mickisch GH, Garin A, van Poppel H, de Prijck L, Sylvester R; European Organisation for Research and Treatment of Cancer (EORTC) Genitourinary Group . Radical nephrectomy plus interferon-alfa-based immunotherapy compared with interferon alfa alone in metastatic renal-cell carcinoma: a randomised trial. Lancet 358: 966–970, 2001. doi: 10.1016/S0140-6736(01)06103-7. [DOI] [PubMed] [Google Scholar]
  • 84.Mir O, Coriat R, Cabanes L, Ropert S, Billemont B, Alexandre J, Durand JP, Treluyer JM, Knebelmann B, Goldwasser F. An observational study of bevacizumab-induced hypertension as a clinical biomarker of antitumor activity. Oncologist 16: 1325–1332, 2011. doi: 10.1634/theoncologist.2010-0002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Moore LE, Nickerson ML, Brennan P, Toro JR, Jaeger E, Rinsky J, Han SS, Zaridze D, Matveev V, Janout V, Kollarova H, Bencko V, Navratilova M, Szeszenia-Dabrowska N, Mates D, Schmidt LS, Lenz P, Karami S, Linehan WM, Merino M, Chanock S, Boffetta P, Chow WH, Waldman FM, Rothman N. Von Hippel-Lindau (VHL) inactivation in sporadic clear cell renal cancer: associations with germline VHL polymorphisms and etiologic risk factors. PLoS Genet 7: e1002312, 2011. doi: 10.1371/journal.pgen.1002312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Motzer RJ, Bacik J, Mariani T, Russo P, Mazumdar M, Reuter V. Treatment outcome and survival associated with metastatic renal cell carcinoma of non-clear-cell histology. J Clin Oncol 20: 2376–2381, 2002. doi: 10.1200/JCO.2002.11.123. [DOI] [PubMed] [Google Scholar]
  • 87.Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, Tykodi SS, Sosman JA, Procopio G, Plimack ER, Castellano D, Choueiri TK, Gurney H, Donskov F, Bono P, Wagstaff J, Gauler TC, Ueda T, Tomita Y, Schutz FA, Kollmannsberger C, Larkin J, Ravaud A, Simon JS, Xu LA, Waxman IM, Sharma P; CheckMate 025 Investigators . Nivolumab versus everolimus in advanced renal-cell carcinoma. N Engl J Med 373: 1803–1813, 2015. doi: 10.1056/NEJMoa1510665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Motzer RJ, Escudier B, Oudard S, Hutson TE, Porta C, Bracarda S, Grünwald V, Thompson JA, Figlin RA, Hollaender N, Urbanowitz G, Berg WJ, Kay A, Lebwohl D, Ravaud A; RECORD-1 Study Group . Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet 372: 449–456, 2008. doi: 10.1016/S0140-6736(08)61039-9. [DOI] [PubMed] [Google Scholar]
  • 89.Motzer RJ, Hutson TE, Cella D, Reeves J, Hawkins R, Guo J, Nathan P, Staehler M, de Souza P, Merchan JR, Boleti E, Fife K, Jin J, Jones R, Uemura H, De Giorgi U, Harmenberg U, Wang J, Sternberg CN, Deen K, McCann L, Hackshaw MD, Crescenzo R, Pandite LN, Choueiri TK. Pazopanib versus sunitinib in metastatic renal-cell carcinoma. N Engl J Med 369: 722–731, 2013. doi: 10.1056/NEJMoa1303989. [DOI] [PubMed] [Google Scholar]
  • 90.Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Oudard S, Negrier S, Szczylik C, Pili R, Bjarnason GA, Garcia-del-Muro X, Sosman JA, Solska E, Wilding G, Thompson JA, Kim ST, Chen I, Huang X, Figlin RA. Overall survival and updated results for sunitinib compared with interferon alfa in patients with metastatic renal cell carcinoma. J Clin Oncol 27: 3584–3590, 2009. doi: 10.1200/JCO.2008.20.1293. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Motzer RJ, Hutson TE, Tomczak P, Michaelson MD, Bukowski RM, Rixe O, Oudard S, Negrier S, Szczylik C, Kim ST, Chen I, Bycott PW, Baum CM, Figlin RA. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med 356: 115–124, 2007. doi: 10.1056/NEJMoa065044. [DOI] [PubMed] [Google Scholar]
  • 92.Naidoo J, Page DB, Li BT, Connell LC, Schindler K, Lacouture ME, Postow MA, Wolchok JD. Toxicities of the anti-PD-1 and anti-PD-L1 immune checkpoint antibodies. Ann Oncol 26: 2375–2391, 2015. doi: 10.1093/annonc/mdv383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Oudard S, Banu E, Vieillefond A, Fournier L, Priou F, Medioni J, Banu A, Duclos B, Rolland F, Escudier B, Arakelyan N, Culine S; GETUG (Groupe d’Etudes des Tumeurs Uro-Génitales) . Prospective multicenter phase II study of gemcitabine plus platinum salt for metastatic collecting duct carcinoma: results of a GETUG (Groupe d’Etudes des Tumeurs Uro-Génitales) study. J Urol 177: 1698–1702, 2007. doi: 10.1016/j.juro.2007.01.063. [DOI] [PubMed] [Google Scholar]
  • 94.Pantuck AJ, Zisman A, Belldegrun AS. The changing natural history of renal cell carcinoma. J Urol 166: 1611–1623, 2001. doi: 10.1016/S0022-5347(05)65640-6. [DOI] [PubMed] [Google Scholar]
  • 95.Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer 12: 252–264, 2012. doi: 10.1038/nrc3239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Patard JJ, Leray E, Rioux-Leclercq N, Cindolo L, Ficarra V, Zisman A, De La Taille A, Tostain J, Artibani W, Abbou CC, Lobel B, Guillé F, Chopin DK, Mulders PF, Wood CG, Swanson DA, Figlin RA, Belldegrun AS, Pantuck AJ. Prognostic value of histologic subtypes in renal cell carcinoma: a multicenter experience. J Clin Oncol 23: 2763–2771, 2005. doi: 10.1200/JCO.2005.07.055. [DOI] [PubMed] [Google Scholar]
  • 97.Peña-Llopis S, Vega-Rubín-de-Celis S, Liao A, Leng N, Pavía-Jiménez A, Wang S, Yamasaki T, Zhrebker L, Sivanand S, Spence P, Kinch L, Hambuch T, Jain S, Lotan Y, Margulis V, Sagalowsky AI, Summerour PB, Kabbani W, Wong SW, Grishin N, Laurent M, Xie XJ, Haudenschild CD, Ross MT, Bentley DR, Kapur P, Brugarolas J. BAP1 loss defines a new class of renal cell carcinoma. Nat Genet 44: 751–759, 2012. doi: 10.1038/ng.2323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Pierorazio PM, Johnson MH, Patel HD, Sozio SM, Sharma R, Iyoha E, Bass EB, Allaf ME. Management of Renal Masses and Localized Renal Cancer: Systematic Review and Meta-Analysis. J Urol 196: 989–999, 2016. doi: 10.1016/j.juro.2016.04.081. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Porta C, Paglino C, Imarisio I, Ganini C, Sacchi L, Quaglini S, Giunta V, De Amici M. Changes in circulating pro-angiogenic cytokines, other than VEGF, before progression to sunitinib therapy in advanced renal cell carcinoma patients. Oncology 84: 115–122, 2013. doi: 10.1159/000342099. [DOI] [PubMed] [Google Scholar]
  • 100.Rini BI, Cohen DP, Lu DR, Chen I, Hariharan S, Gore ME, Figlin RA, Baum MS, Motzer RJ. Hypertension as a biomarker of efficacy in patients with metastatic renal cell carcinoma treated with sunitinib. J Natl Cancer Inst 103: 763–773, 2011. doi: 10.1093/jnci/djr128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Rini BI, Escudier B, Tomczak P, Kaprin A, Szczylik C, Hutson TE, Michaelson MD, Gorbunova VA, Gore ME, Rusakov IG, Negrier S, Ou YC, Castellano D, Lim HY, Uemura H, Tarazi J, Cella D, Chen C, Rosbrook B, Kim S, Motzer RJ. Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomised phase 3 trial. Lancet 378: 1931–1939, 2011. doi: 10.1016/S0140-6736(11)61613-9. [DOI] [PubMed] [Google Scholar]
  • 102.Rini BI, Halabi S, Rosenberg JE, Stadler WM, Vaena DA, Ou SS, Archer L, Atkins JN, Picus J, Czaykowski P, Dutcher J, Small EJ. 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. doi: 10.1200/JCO.2008.16.9847. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Rini BI, Wilding G, Hudes G, Stadler WM, Kim S, Tarazi J, Rosbrook B, Trask PC, Wood L, Dutcher JP. Phase II study of axitinib in sorafenib-refractory metastatic renal cell carcinoma. J Clin Oncol 27: 4462–4468, 2009. doi: 10.1200/JCO.2008.21.7034. [DOI] [PubMed] [Google Scholar]
  • 104.Robinson ES, Khankin EV, Choueiri TK, Dhawan MS, Rogers MJ, Karumanchi SA, Humphreys BD. Suppression of the nitric oxide pathway in metastatic renal cell carcinoma patients receiving vascular endothelial growth factor-signaling inhibitors. Hypertension 56: 1131–1136, 2010. doi: 10.1161/HYPERTENSIONAHA.110.160481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Salvatore SP, Cha EK, Rosoff JS, Seshan SV. Nonneoplastic renal cortical scarring at tumor nephrectomy predicts decline in kidney function. Arch Pathol Lab Med 137: 531–540, 2013. doi: 10.5858/arpa.2012-0070-OA. [DOI] [PubMed] [Google Scholar]
  • 106.Schmidt L, Duh FM, Chen F, Kishida T, Glenn G, Choyke P, Scherer SW, Zhuang Z, Lubensky I, Dean M, Allikmets R, Chidambaram A, Bergerheim UR, Feltis JT, Casadevall C, Zamarron A, Bernues M, Richard S, Lips CJ, Walther MM, Tsui LC, Geil L, Orcutt ML, Stackhouse T, Lipan J, Slife L, Brauch H, Decker J, Niehans G, Hughson MD, Moch H, Storkel S, Lerman MI, Linehan WM, Zbar B. Germline and somatic mutations in the tyrosine kinase domain of the MET proto-oncogene in papillary renal carcinomas. Nat Genet 16: 68–73, 1997. doi: 10.1038/ng0597-68. [DOI] [PubMed] [Google Scholar]
  • 107.Schödel J, Grampp S, Maher ER, Moch H, Ratcliffe PJ, Russo P, Mole DR. Hypoxia, Hypoxia-inducible Transcription Factors, and Renal Cancer. Eur Urol 69: 646–657, 2016. doi: 10.1016/j.eururo.2015.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Shin Lee J, Seok Kim H, Bok Kim Y, Cheol Lee M, Soo Park C. Expression of PTEN in renal cell carcinoma and its relation to tumor behavior and growth. J Surg Oncol 84: 166–172, 2003. doi: 10.1002/jso.10302. [DOI] [PubMed] [Google Scholar]
  • 109.Sica DA. Angiogenesis inhibitors and hypertension: an emerging issue. J Clin Oncol 24: 1329–1331, 2006. doi: 10.1200/JCO.2005.04.5740. [DOI] [PubMed] [Google Scholar]
  • 110.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin 66: 7–30, 2016. doi: 10.3322/caac.21332. [DOI] [PubMed] [Google Scholar]
  • 111.Skinnider BF, Folpe AL, Hennigar RA, Lim SD, Cohen C, Tamboli P, Young A, de Peralta-Venturina M, Amin MB. Distribution of cytokeratins and vimentin in adult renal neoplasms and normal renal tissue: potential utility of a cytokeratin antibody panel in the differential diagnosis of renal tumors. Am J Surg Pathol 29: 747–754, 2005. doi: 10.1097/01.pas.0000163362.78475.63. [DOI] [PubMed] [Google Scholar]
  • 112.Sternberg CN, Davis ID, Mardiak J, Szczylik C, Lee E, Wagstaff J, Barrios CH, Salman P, Gladkov OA, Kavina A, Zarbá JJ, Chen M, McCann L, Pandite L, Roychowdhury DF, Hawkins RE. Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial. J Clin Oncol 28: 1061–1068, 2010. doi: 10.1200/JCO.2009.23.9764. [DOI] [PubMed] [Google Scholar]
  • 113.Sufan RI, Jewett MA, Ohh M. The role of von Hippel-Lindau tumor suppressor protein and hypoxia in renal clear cell carcinoma. Am J Physiol Renal Physiol 287: F1–F6, 2004. doi: 10.1152/ajprenal.00424.2003. [DOI] [PubMed] [Google Scholar]
  • 114.Tanaka T, Kitamura H, Torigoe T, Hirohashi Y, Sato E, Masumori N, Sato N, Tsukamoto T. Autoantibody against hypoxia-inducible factor prolyl hydroxylase-3 is a potential serological marker for renal cell carcinoma. J Cancer Res Clin Oncol 137: 789–794, 2011. doi: 10.1007/s00432-010-0940-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Tokuda N, Naito S, Matsuzaki O, Nagashima Y, Ozono S, Igarashi T; Japanese Society of Renal Cancer . Collecting duct (Bellini duct) renal cell carcinoma: a nationwide survey in Japan. J Urol 176: 40–43, 2006. doi: 10.1016/S0022-5347(06)00502-7. [DOI] [PubMed] [Google Scholar]
  • 116.Toro JR, Nickerson ML, Wei MH, Warren MB, Glenn GM, Turner ML, Stewart L, Duray P, Tourre O, Sharma N, Choyke P, Stratton P, Merino M, Walther MM, Linehan WM, Schmidt LS, Zbar B. Mutations in the fumarate hydratase gene cause hereditary leiomyomatosis and renal cell cancer in families in North America. Am J Hum Genet 73: 95–106, 2003. doi: 10.1086/376435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Toro JR, Wei MH, Glenn GM, Weinreich M, Toure O, Vocke C, Turner M, Choyke P, Merino MJ, Pinto PA, Steinberg SM, Schmidt LS, Linehan WM. BHD mutations, clinical and molecular genetic investigations of Birt-Hogg-Dubé syndrome: a new series of 50 families and a review of published reports. J Med Genet 45: 321–331, 2008. doi: 10.1136/jmg.2007.054304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Torras J, Herrero-Fresneda I, Gulias O, Flaquer M, Vidal A, Cruzado JM, Lloberas N, Franquesa M, Grinyó JM. Rapamycin has dual opposing effects on proteinuric experimental nephropathies: is it a matter of podocyte damage? Nephrol Dial Transplant 24: 3632–3640, 2009. doi: 10.1093/ndt/gfp367. [DOI] [PubMed] [Google Scholar]
  • 119.Tran HT, Liu Y, Zurita AJ, Lin Y, Baker-Neblett KL, Martin AM, Figlin RA, Hutson TE, Sternberg CN, Amado RG, Pandite LN, Heymach JV. 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. doi: 10.1016/S1470-2045(12)70241-3. [DOI] [PubMed] [Google Scholar]
  • 120.Turner KJ, Moore JW, Jones A, Taylor CF, Cuthbert-Heavens D, Han C, Leek RD, Gatter KC, Maxwell PH, Ratcliffe PJ, Cranston D, Harris AL. Expression of hypoxia-inducible factors in human renal cancer: relationship to angiogenesis and to the von Hippel-Lindau gene mutation. Cancer Res 62: 2957–2961, 2002. [PubMed] [Google Scholar]
  • 121.Van Poppel H, Da Pozzo L, Albrecht W, Matveev V, Bono A, Borkowski A, Colombel M, Klotz L, Skinner E, Keane T, Marreaud S, Collette S, Sylvester R. 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. doi: 10.1016/j.eururo.2010.12.013. [DOI] [PubMed] [Google Scholar]
  • 122.Varela I, Tarpey P, Raine K, Huang D, Ong CK, Stephens P, Davies H, Jones D, Lin ML, Teague J, Bignell G, Butler A, Cho J, Dalgliesh GL, Galappaththige D, Greenman C, Hardy C, Jia M, Latimer C, Lau KW, Marshall J, McLaren S, Menzies A, Mudie L, Stebbings L, Largaespada DA, Wessels LF, Richard S, Kahnoski RJ, Anema J, Tuveson DA, Perez-Mancera PA, Mustonen V, Fischer A, Adams DJ, Rust A, Chan-on W, Subimerb C, Dykema K, Furge K, Campbell PJ, Teh BT, Stratton MR, Futreal PA. Exome sequencing identifies frequent mutation of the SWI/SNF complex gene PBRM1 in renal carcinoma. Nature 469: 539–542, 2011. doi: 10.1038/nature09639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Vogelbacher R, Wittmann S, Braun A, Daniel C, Hugo C. The mTOR inhibitor everolimus induces proteinuria and renal deterioration in the remnant kidney model in the rat. Transplantation 84: 1492–1499, 2007. doi: 10.1097/01.tp.0000282866.92367.99. [DOI] [PubMed] [Google Scholar]
  • 124.Weight CJ, Larson BT, Fergany AF, Gao T, Lane BR, Campbell SC, Kaouk JH, Klein EA, Novick AC. Nephrectomy induced chronic renal insufficiency is associated with increased risk of cardiovascular death and death from any cause in patients with localized cT1b renal masses. J Urol 183: 1317–1323, 2010. doi: 10.1016/j.juro.2009.12.030. [DOI] [PubMed] [Google Scholar]
  • 125.Wettersten HI, Hakimi AA, Morin D, Bianchi C, Johnstone ME, Donohoe DR, Trott JF, Aboud OA, Stirdivant S, Neri B, Wolfert R, Stewart B, Perego R, Hsieh JJ, Weiss RH. Grade-Dependent Metabolic Reprogramming in Kidney Cancer Revealed by Combined Proteomics and Metabolomics Analysis. Cancer Res 75: 2541–2552, 2015. doi: 10.1158/0008-5472.CAN-14-1703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Yap SA, Finelli A, Urbach DR, Tomlinson GA, Alibhai SM. Partial nephrectomy for the treatment of renal cell carcinoma (RCC) and the risk of end-stage renal disease (ESRD). BJU Int 115: 897–906, 2015. doi: 10.1111/bju.12883. [DOI] [PubMed] [Google Scholar]
  • 127.Yeh J, Frieze D, Martins R, Carr L. Clinical utility of routine proteinuria evaluation in treatment decisions of patients receiving bevacizumab for metastatic solid tumors. Ann Pharmacother 44: 1010–1015, 2010. doi: 10.1345/aph.1M670. [DOI] [PubMed] [Google Scholar]
  • 128.Zabor EC, Furberg H, Mashni J, Lee B, Jaimes EA, Russo P. Factors Associated with Recovery of Renal Function following Radical Nephrectomy for Kidney Neoplasms. Clin J Am Soc Nephrol 11: 101–107, 2016. doi: 10.2215/CJN.04070415. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Zheng H, Ji J, Zhao L, Chen M, Shi A, Pan L, Huang Y, Zhang H, Dong B, Gao H. Prediction and diagnosis of renal cell carcinoma using nuclear magnetic resonance-based serum metabolomics and self-organizing maps. Oncotarget 7: 59189–59198, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Zira AN, Theocharis SE, Mitropoulos D, Migdalis V, Mikros E. (1)H NMR metabonomic analysis in renal cell carcinoma: a possible diagnostic tool. J Proteome Res 9: 4038–4044, 2010. doi: 10.1021/pr100226m. [DOI] [PubMed] [Google Scholar]

Articles from American Journal of Physiology - Renal Physiology are provided here courtesy of American Physiological Society

RESOURCES