Renal cancer

Renal cancer

Seminar Renal cancer Umberto Capitanio, Francesco Montorsi The diagnosis and management of renal cell carcinoma have changed remarkably rapidly. Alt...

2MB Sizes 2 Downloads 10 Views

Seminar

Renal cancer Umberto Capitanio, Francesco Montorsi

The diagnosis and management of renal cell carcinoma have changed remarkably rapidly. Although the incidence of renal cell carcinoma has been increasing, survival has improved substantially. As incidental diagnosis of small indolent cancers has become more frequent, active surveillance, robot-assisted nephron-sparing surgical techniques, and minimally invasive procedures, such as thermal ablation, have gained popularity. Despite progression in cancer control and survival, locally advanced disease and distant metastases are still diagnosed in a notable proportion of patients. An integrated management strategy that includes surgical debulking and systemic treatment with well established targeted biological drugs has improved the care of patients. Nevertheless, uncertainties, controversies, and research questions remain. Further advances are expected from translational and clinical studies.

Introduction

Pathophysiology and genetics

Abdominal imaging is used for many different medical disorders (eg, hypertension, diabetes) and, therefore, incidental detection of renal masses is becoming increasingly common. A renal mass might be merely a simple renal cyst that requires no treatment or follow-up,1 but in a notable proportion of cases masses are benign renal lesions (eg, angiomyolipomas or oncocytomas) or malignant renal cell carcinomas that might require additional procedures or interventions.2 Although most incidentally detected lesions are small low-grade tumours, up to 17% of all renal cell carcinomas have distant metastases at the time of diagnosis.3 In this heterogeneous clinical setting, developments in molecular biology, diagnostic techniques, surgery, and medical oncology are revolutionising the approach to this disease.

Renal cell carcinoma comprises a heterogeneous group of cancers with different genetic and molecular alterations underlying the many documented histological subtypes.18 Clear-cell, papillary (types 1 and 2), and chromophobe are the most common solid renal cell carcinomas within the kidney and account for 85–90% of all renal malignancies.18 Less common cancers include papillary adenoma, multilocular cystic clear-cell carcinoma, hybrid oncocytic chromophobe tumour, carcinoma of the collecting ducts of Bellini, renal medullary carcinoma, carcinoma associated with neuroblastoma, and mucinous tubular and spindle-cell carcinoma.18 The International Society of Urological Pathology Vancouver Consensus Statement added five more epithelial tumour subtypes of renal cell carcinoma: tubulocystic, acquired cystic diseaseassociated, clear-cell tubulopapillary, microphthalmia family translocation, and hereditary leiomyomatosis– renal cell carcinoma syndrome-associated.19 Although most renal masses are initially confined to one organ, early haematogenic dissemination is responsible for metastatic disease at the time of diagnosis. The most frequent sites of distant metastases are lungs, bone, and brain, but adrenal glands, contralateral kidney, and liver might be involved.20

Epidemiology In 2013, renal cell carcinoma was diagnosed in more than 350 000 people worldwide, which made it the seventh most common site for tumours, and this cancer is associated with more than 140 000 deaths per year.4 Incidence of renal cell carcinoma varies worldwide, being higher in developed countries than in developing countries (appendix). Incidence predominates in men, with the male-to-female ratio being 1·5:1·0, and peaks at age 60–70 years.5 Despite increased incidence overall, improvements in relative survival rates have been reported over the past three decades,3,5 although these might be skewed by disease stage migration.6–8 In Europe, mortality from renal cell carcinoma peaked at 4·8 per 100 000 in 1990–94 and had declined to 4·1 per 100 000 (−13%) in 2000–04.5 In the USA, 5-year relative survival rates at diagnosis increased from 50% in 1975–77 to 57% in 1987–89, and reached 73% in 2003–09.3 Established risk factors include smoking tobacco,9 hypertension,10–12 and obesity.13,14 These associations might be biased by the probability that such patients undergo routine imaging, which could increase the likelihood of incidental detection of renal masses. Several other associations have been suggested, for example, a protective effect of alcohol consumption,15 a negative effect of red meat consumption,16 and occupational exposure to carcinogens,17 but the data are unclear.

Published Online August 26, 2015 http://dx.doi.org/10.1016/ S0140-6736(15)00046-X Department of Urology, Vita-Salute San Raffaele University (U Capitanio MD) and Division of Experimental Oncology, URI, Urological Research Institute, Renal Cancer Unit (Prof F Montorsi FRCS), IRCCS San Raffaele Scientific Institute, Milan, Italy Correspondence to: Dr Umberto Capitanio, Department of Urology, San Raffaele Hospital, Via Olgettina 60, Milan 20132, Italy [email protected]

See Online for appendix

Search strategy and selection criteria We identified data for this Seminar through searches of PubMed and Web of Science. We included papers in core clinical journals that described studies in adults and were published in English from 2004 to 2014. Search terms included “renal cancer” or “kidney cancer” in combination with the terms “epidemiology”, “genetics”, “pathophysiology”, “diagnosis”, “biopsy”, “treatment”, “surgery”, “active surveillance”, “medical therapy”, or “targeted therapy”. We also searched cited references from selected articles and previous reviews to identify further relevant papers not retrieved by this search, including those outside the time period of the initial search. We also searched for updates on the main features of renal cell carcinoma, with particular attention paid to reports focusing on novel treatments, renal biopsy, new imaging technologies, and minimally invasive techniques.

www.thelancet.com Published online August 26, 2015 http://dx.doi.org/10.1016/S0140-6736(15)00046-X

1

Seminar

Renal cell carcinoma can be sporadic or hereditary, but both forms are generally associated with structural alterations of the short arm of chromosome 3.21 Translocation tumours are increasingly being identified in adolescents and young adults.18 Many gene variants have been associated with renal cell carcinoma. Somatic mutations or epigenetic alterations of VHL, a tumoursuppressor gene, are seen in more than 80% of clear-cell subtypes. Specifically, the VHL tumour suppressor protein is important in cellular oxygen sensing by targeting hypoxia-inducible factors for ubiquitination and proteasomal degradation.22 Sequencing studies have identified other driver genes that are involved in the pathogenesis of renal cell carcinoma, including PBRM1,23 BAP1,23 SETD2,24 TCEB1,24 and KDM5C.25 Studies analysing the effects of these mutations on clinical outcomes, however, are limited by small sample sizes, short-term follow-up, and lack of assessment of multiple markers in the same cohort.24,25 Hereditary forms of renal cell carcinoma include von Hippel-Lindau syndrome (VHL 3p25–26),26 hereditary papillary renal cell carcinoma (MET 7q31–34),27 Birt-Hogg-Dubé syndrome (FLCN 17p11),28 hereditary leiomyomatosis (FH 1q42–43),29 and tuberous sclerosis (TSC1 9q34 or TSC2 16p13).30

Diagnosis With the expansion of routine imaging for many disorders, patients with renal cell carcinoma are increasingly being identified by chance. Only 30% of patients are diagnosed on the basis of symptoms. At all stages, renal cell carcinoma might produce multiple hormone-like or cytokine-like biologically active products that lead to clinically important paraneoplastic syndromes (table 1).31 The findings of routine diagnostic blood tests might be altered by renal cell carcinoma, but no change is pathognomonic. Physical examination has a limited role in diagnosis of renal cell carcinoma, but the presence of a palpable abdominal mass, new-onset varicocele, or lower extremity oedema should always prompt additional imaging assessments for retroperitoneal neoplasia.

Standard imaging Although renal cell carcinoma is frequently detected by abdominal ultrasound scanning, limitations in specificity and accuracy make it necessary to use CT or MRI to confirm suspicious findings. The main goals of imaging are to characterise the mass and possible abdominal metastases, tumour extension, and venous involvement for staging (appendix). Imaging might also be useful to

Occurrence among patients with symptomatic RCC

Proposed causes

Acute or chronic flank pain

Common

Urinary system obstruction or infiltration, invasion of adjacent organs, retroperitoneal space-occupying mass

Gross haematuria

Less frequent

Urinary system infiltration

Palpable abdominal mass

Less frequent

Retroperitoneal space-occupying mass

Varicocele

Rare

Increased venous pressure due to space-occupying mass or venous thrombus

Hypertension

Common

Increased production of renin directly by tumour; compression or encasement of the renal artery or its branches causing renal artery stenosis; or arteriovenous fistula within the tumour, polycythaemia, hypercalcaemia, ureteral obstruction, and increased intracranial pressure associated with cerebral metastases

Anaemia

Common

Bleeding, abnormal production of prostaglandins, cytokines, and inflammatory mediators

Local symptoms

Paraneoplastic disorders

Cachexia, weight loss

Common

Abnormal production of prostaglandins, cytokines, and inflammatory mediators

Pyrexia

Less frequent

Abnormal production of prostaglandins, cytokines, and inflammatory mediators

Raised hepatic enzyme concentrations in absence of liver metastases (Stauffer’s syndrome)

Less frequent

Non-specific hepatitis associated with a prominent lymphocytic infiltrate, raised concentrations of interleukin-6 in serum

Hypercalcaemia

Less frequent

Osteolytic metastatic involvement of bone, production of parathyroid-hormonelike peptides, tumour-derived 1,25-dihydroxycholecalciferol, and prostaglandins

Polycythaemia

Rare

Increased production of erythropoietin directly by the tumour or by the adjacent parenchyma in response to hypoxia induced by tumour growth

Hypoglycaemia, neuromyopathy, amyloidosis, vascular thrombosis, Cushing’s syndrome, protein enteropathy, galactorrhoea, gynaecomastia, decreased libido, hirsutism, amenorrhoea, chills, necrotising myopathy, immune thrombocytopenic purpura

Anecdotal

Abnormal production of 1,25-dihydroxycholecalciferol, renin, erythropoietin, prostaglandins, parathyroid-hormone-like peptides, lupus-type anticoagulant, human chorionic gonadotropin, insulin, various cytokines or inflammatory mediators

RCC=renal cell carcinoma.

Table 1: Most frequent local symptoms and paraneoplastic disorders associated with RCC

2

www.thelancet.com Published online August 26, 2015 http://dx.doi.org/10.1016/S0140-6736(15)00046-X

Seminar

assess additional characteristics that could be important if active management is planned (eg, bilateral renal function, renal vascular supply, retroperitoneal anatomy). If malignant renal cell carcinoma is suspected, additional imaging (eg, thoracic and brain CT scan, total-body bone scan) can be considered in symptomatic patients or in cases of bulky abdominal disease.32–35

tumours, detection of small renal lesions44 and accumulating evidence that surgically induced chronic kidney disease can increase patients’ morbidity45 have led to more conservative approaches.46 Specifically, nephronsparing surgery, active surveillance, and minimally invasive techniques46 have been introduced into daily clinical practice. These approaches limit invasiveness, iatrogenic renal function impairment, and overtreatment.

New imaging technologies New technologies for cancer detection and characterisation are being investigated for renal cell carcinoma; for example, advanced MRI techniques, such as diffusionweighted and perfusion-weighted imaging, are being explored for the assessment of renal masses.36 Although PET might be useful for the diagnosis and follow-up of renal cell carcinoma, this technique is not yet part of the standard strategy.37 Some studies have suggested the use of combined ¹²⁴iodine PET and CT for the detection of labelled antibodies to the carbonic anhydrase IX antigen, which is widely expressed in more than 90% of clear-cell renal cell carcinomas but not in the normal kidney.38,39

Emerging role of renal biopsy The use of renal biopsy has been limited by concerns about accuracy and safety, and in the past has had little effect on treatment decisions because the standard treatment for solid renal masses was upfront removal.40 Increased active surveillance for small renal masses and individualised targeted therapy for patients with metastases, however, have pushed the scientific community to improve expertise in biopsy performance and pathological interpretation.41 Consequently, renal biopsy is now used to establish the diagnosis of radiologically indeterminate renal masses, obtain histology of incidentally detected renal masses in patients who are potential candidates for active surveillance, and to select the most suitable targeted therapy for patients with metastatic renal tumours.42 Accuracy of biopsy in detecting malignancy ranges from 38% to 100%.41 In one of the largest study cohorts reported (n=345), histological subtype and Fuhrman grade were correctly assessed at biopsy in 88% and 64% of cases, respectively.43 Renal biopsy has low associated morbidity. Contemporary series have revealed overall complication rates (mainly bleeding, infection, and arteriovenous fistula) in 0·3–5·3% of cases. Tumour seeding throughout the biopsy tract, which had been reported anecdotally in the past, is no longer a risk with modern biopsy techniques.42 Although renal biopsy has a definite and expanding role in the assessment and treatment of renal masses, it remains substantially underused.40

Management Notwithstanding advances in the understanding of renal cell carcinoma biology, surgery remains the mainstay of curative treatment. Although radical nephrectomy was historically the standard of care for management of renal

Active surveillance Management of localised renal cell carcinoma must take into account competing causes of mortality, particularly in elderly patients.47 This consideration has led to the increasing use of active surveillance46 to avoid surgically induced short-term and medium-term complications. The rationale for active surveillance is that roughly 20% of small renal masses harbour benign final pathology and the median growth rate is usually slow (2–3 mm/year).48 Moreover, the risk of metastatic progression is very low (less than 1%), with virtually no risk of cancer-specific mortality in well selected patients.49–51 Biopsy might be useful before active surveillance is started. Active surveillance has been proposed for larger tumours to determine tumour growth kinetics, especially if a patient has severe competing risks and limited life expectancy.52 Absolute cutoffs for tumour size and growth rate that should prompt intervention during active surveillance are not well defined. In various proposed protocols, imaging at 3 and 6 months initially, every 6 months during the next 2–3 years, and annually thereafter is suggested, but cumulative radiation risk and increased costs are being assessed.49,50,53 Data are needed from large cohorts with long-term follow-up to better characterise clinical indications and standardise protocols.

Minimally invasive techniques Although surgery still represents the standard of care for renal cell carcinoma, the use of minimally invasive techniques to treat incidentally detected small tumours has been increasing. Cryotherapy and radiofrequency ablation (RFA) were initially contemplated only for patients with one kidney or for those deemed unfit to undergo a major procedure. Since preliminary reports showed acceptable cancer control,37,54–57 the clinical indications for these procedures has been extending. For example, 8% of US patients are treated with cryotherapy and RFA, compared with 4% in 1998.58 Renal ablative treatment uses the cell-destroying properties of temperature (hot or cold) to cause apoptosis in cancer cells.54 RFA induces thermal damage (50–120°C) through frictional heating resulting from ionic oscillation by a high-frequency alternating current (375–500 kHz).59 Cryotherapy stimulates tumour cell death directly by damaging cell membranes and organelles and indirectly by initiating vascular compromise through thrombosis of small vessels,54 which leads to coagulative necrosis of

www.thelancet.com Published online August 26, 2015 http://dx.doi.org/10.1016/S0140-6736(15)00046-X

3

Seminar

the tissue. Potential complications after RFA and cryotherapy are renal bleeding or abscess formation or extrarenal effects in the bowel, pleura, spleen, pancreas, and vasculature. Most studies of outcomes after renal tumour cryoablation consist of single-institution retrospective reports with low numbers of patients and short follow-up. An analysis of the Mayo Clinic Renal Tumor Registry that included more than 1800 patients with primary cT1N0M0 renal tumours showed similar local recurrence-free survival for patients treated with nephron-sparing surgery, RFA, or cryotherapy (p=0·4). By contrast, metastases-free survival was significantly better after nephron-sparing surgery (p=0·005) and cryoablation (p=0·02) than with RFA.60 Although this comparison included a large number of cases, its retrospective nature, the differing baseline characteristics between studies, and other unmeasurable confounders make it difficult to draw conclusions about cancer control.61 A meta-analysis by Kunkle and colleagues56 suggested that recurrence-free survival might be worse for RFA and cryoablation than for partial nephrectomy, but showed no differences for metastasis-free survival. Conversely, a systematic review and meta-analysis of six clinical trials (one prospective randomised and five retrospective cohort trials) showed that recurrence-free survival and cancer-specific survival were similar for patients treated with surgery or RFA, with less postoperative decline in estimated glomerular filtration rate in the RFA group.55 The overall complication rate was significantly lower in the RFA group (7% vs 11%; relative risk 0·55, 95% CI 0·31–0·97, p=0·04). The groups had similar local recurrence rates of 3–4% (0·92, 0·4–2·14, p=0·8) and disease-free survival (hazard ratio 1·04, 95% CI 0·48–2·24, p=0·9).55

Surgery Nephron-sparing surgery Although radical nephrectomy involves removal of the entire kidney, with nephron-sparing surgery only the tumour is excised and as much of the normal renal parenchyma as possible is maintained (figure 1). Although initially reserved for imperative situations, according to international guidelines, nephron-sparing surgery is now the treatment of choice whenever a healthy part of kidney can safely be spared. Nephron-sparing surgery has been used increasingly since observations suggested oncological control similar to radical nephrectomy62–64 but with the additional benefit of renal preservation.65 Moreover, a long-term protective effect has been suggested for nephron-sparing surgery relative to radical nephrectomy for risk of cardiovascular events after surgery (eg, new-onset hypertension, coronary artery disease, vasculopathy, and cerebrovascular disease).45,66,67 Because patients with organ-confined renal cell carcinoma that has been surgically treated are usually long-term cancer survivors (85–96% cancer-specific survival 10 years after surgery68), these functional outcomes are important. 4

Although many retrospective reports have suggested a benefit in overall survival for patients treated with nephron-sparing surgery relative to radical nephrectomy,67,69 no improvements with nephron-sparing techniques were found in a comparison of the two techniques.70 However, many flaws undermined the study (eg, low recruitment, selection and verification biases, and being done at the beginning of the nephron-sparing learning curve).70 Further efforts are needed to improve understanding of which patients might benefit from nephron-sparing surgery in terms of overall survival. International guidelines include standard indications for nephron-sparing surgery: absolute, indicating patients with only one anatomical or functional kidney; relative, indicating patients with a functioning opposite kidney that is affected by a disorder that might impair renal function in the future or with hereditary forms of renal cell carcinoma who are at increased risk of developing a tumour in the contralateral kidney; and elective, indicating localised unilateral renal cell carcinoma with a healthy contralateral kidney.37,58 Although nephron-sparing surgery has been proposed for use even for locally advanced disease and metastatic spread,71,72 further data are required before a conservative approach can be approved in those settings. Surgical excision of tumours is done by kidney mobilisation, resection of the tumour (with or without a rim of normal parenchyma, according to the anatomical and tumour features), and final reconstruction (renorrhaphy; figure 1). During resection of the tumour, the main renal artery can be clamped temporarily (hilar clamping) to minimise blood loss. Sustained hilar clamping might cause impairment of renal function and, therefore, limited duration of ischaemia is suggested, although the functional long-term consequences are controversial.73–75 If technically possible, techniques without clamping or selective intraparenchymal clamping (eg, clamping of only a branch of the main artery) should be considered.76 Nephron-sparing surgery should be preferred unless pre-existing disorders are likely to decrease the positive effect of a conservative approach, such as frailty or short life expectancy. Other risk factors are low surgical expertise or volume of relevant surgeries, excessive ischaemia time, use of non-conservative haemostasis techniques, or large or anatomically complex renal masses with a low percentage of nephrons that can be spared. In such situations, the potential benefit in functional outcomes might be jeopardised. Conservative surgical techniques require substantial technical expertise and might be associated with increased risk of haemorrhage (3%) and urinary leakage (4%).77 Consequently, nephron-sparing surgery is still underused for patients with renal cell carcinoma, especially in nonacademic hospitals.78–80 If the aim of the surgery is the preservation of the normal renal parenchyma, the choice of surgical approach

www.thelancet.com Published online August 26, 2015 http://dx.doi.org/10.1016/S0140-6736(15)00046-X

Seminar

A

2 1

B

C

D

Figure 1: Procedures in robot-assisted nephron-sparing surgery to remove renal cell carcinoma (A) In robot-assisted surgery, instead of directly moving the instruments, the surgeon performs the normal movements associated with the surgery, and the robotic arms make those movements and use end-effectors and manipulators to perform the actual surgery on the patient. One arm is dedicated to the laparoscope and the two others hold forceps, monopolar curved scissors, a cautery hook, and a large needle driver. The patient is positioned in a modified flank position. Port configuration can vary based on tumour location to optimise the working angles. Surgical excision of the tumour is done by (B) kidney mobilisation, (C) tumour resection (with or without a rim of normal parenchyma according to anatomical and tumour features), and (D) final reconstruction (renorrhaphy).

(open, laparoscopic, or robotic) depends largely on tumour characteristics and the surgeon’s expertise. Tumour features that dictate the feasibility of nephron-sparing surgery versus radical nephrectomy include diameter, location, depth, and proximity to hilar vessels and the urinary collecting system. The three foremost anatomical scores (PADUA,81 RENAL,82 and C-index83) may be used to assess those features before surgery, with the aim of improving selection of patients, surgical management, research reporting, and outcome prediction.81–83 Robot-assisted nephron-sparing surgery is gaining popularity because challenging cases can be approached with a minimally invasive procedure (figure 1). In hospitals that have acquired surgical robotic systems, treatment with nephron-sparing surgery has increased by 16–35% relative to hospitals without such technology.84 Conversely, laparoscopic nephron-sparing surgery might be challenging and is usually limited to simpler cases or done by experienced surgeons (appendix). Open

nephron-sparing surgery, which was the standard approach until the early 2000s, is now used only in cases where there are anatomical difficulties (eg, urinary tract invasion or hilar or larger tumours) or for which minimally invasive techniques are unavailable. A meta-analysis of non-randomised studies including 2240 patients showed that robot-assisted compared with laparoscopic nephron-sparing surgery was associated with reduced rates of conversion to open surgery (relative risk 0·4, 95% CI 0·2–0·9, p=0·02) and radical surgery (0·19, 0·1–0·5, p=0·0006).85 Additionally, robot-assisted nephron-sparing surgery resulted in significantly shorter warm ischaemia, (weighted mean difference 3 min, 95% CI 1–5, p=0·005), although the clinical relevance of this outcome was marginal, smaller change in estimated glomerular filtration rate (standardised mean difference 0·2, 0·02–0·3, p=0·03), and shorter length of stay in hospital (weighted mean difference 0·2 days, 0·1–0·4, p=0·004).85 Moreover, no differences were noted in

www.thelancet.com Published online August 26, 2015 http://dx.doi.org/10.1016/S0140-6736(15)00046-X

5

Seminar

Figure 2: CT scan of a patient aged 55 years with a bulky right kidney tumour and caval thrombus invading the right atrium Images were taken before nephrectomy and venous thrombectomy, and are two slices from the same scan. Initial presentation was a newly diagnosed right varicocele, followed by fatigue and dyspnoea. The right varicocele was secondary to a venous thrombus occluding the vena cava and its branch, the right spermatic vein. Fatigue and dyspnoea were secondary to initial heart failure and abnormal production of prostaglandins, cytokines, and inflammatory mediators by the tumour.

operative outcomes, including complications with Clavien-Dindo classification grades 1–2 (relative risk 1·1, 95% CI 0·9–1·3, p=06) and 3–5 (0·9, 0·6–1·5, p=0·8).85

Radical nephrectomy If sparing the parenchyma is not possible and the aim of the surgery is removal of the entire kidney, open radical nephrectomy has been replaced in most cases by laparoscopic nephrectomy, which is associated with shorter stay in hospital, less perioperative blood loss, fewer analgesic requirements, and shorter convalescence time.86,87 Although randomised studies assessing oncological outcomes have never been done, similar oncological outcomes for laparoscopic and open radical nephrectomy are suggested.86,87 Robot-assisted radical nephrectomy results in increased medical expense without improving morbidity compared with laparoscopic procedures,88 but might still be worth considering for selected cases (eg, when retroperitoneal lymphadenectomy is thought to be required).

Lymph node dissection, adrenalectomy, and venous thrombectomy Adrenalectomy was historically viewed as an unavoidable part of radical procedures. Removal of the ipsilateral adrenal gland, however, is now optional if no macroscopic invasion is suggested by preoperative imaging or found during surgery.37,57,89 Irrespective of T stage, patients who have renal cell carcinoma with nodal invasion (pN1) have poor cancerspecific survival (20–30% at 3 years after surgery).90 Although lymphadenectomy provides the most accurate 6

and reliable staging of lymph node status, the value of lymphadenectomy in renal cell carcinoma remains controversial. A European Organisation for Research and Treatment of Cancer randomised trial showed no survival advantage for low-risk patients treated with nephrectomy plus lymphadenectomy versus nephrectomy only,91 although several retrospective studies have suggested a potential benefit with lymphadenectomy in intermediaterisk and high-risk patients.92 Sentinel node biopsy is not an option for renal cell carcinoma at present.93,94 Around 10% of patients with renal cell carcinoma have venous tumour thrombosis in the renal vein, inferior vena cava, or extending up to the right atrium (figure 2). Although prognosis is worse in such cases and surgery might be challenging and associated with an increased complication rate, a case–cohort study suggested that survival was better with surgical management than with non-surgical management.95 Operative management is dictated primarily by the extent of the thrombus. When confined to the renal vein, minimum modification of the standard surgical approach is required. As the thrombus extends cranially, opening of the vena cava, liver mobilisation, opening of the right atrium, and cardiopulmonary bypass might be necessary.

Metastatic disease at diagnosis Historically, patients with metastatic clear-cell renal cell carcinoma were treated with systemic therapy based on immune modulators, mainly interferon α and interleukin-2, but outcomes were only slightly improved.96 In a few cases these agents have led to T-cell-mediated tumour regression secondary to enhancement in lymphocyte mitogenesis, lymphocyte cytotoxicity, and activity of lymphokine-activated and natural killer cells.97 Immunotherapy with high-dose interleukin 2 has been associated with durable complete response in less than 10% of patients and has been associated with severe treatment-related toxic effects.97 Subsequently, combined data from 99 study groups show an overall chance of partial or complete remission in only 13% of patients, compared with 3–4% in placebo or non-immunotherapy control groups.98 Aldesleukin and interferon α (associated with bevacizumab) remain the only immune-modulating drugs approved for selected cases of metastatic renal cell carcinoma.57 Several non-randomised studies had suggested a response to systemic therapy and improved survival in patients who had undergone nephrectomy and, therefore, two randomised clinical trials were planned at the end of 1980s to test the effect of surgery on survival in patients with metastatic renal cell carcinoma. 331 patients overall were randomly assigned treatment with nephrectomy plus interferon α-2b or interferon α-2b alone.99,100 A combined analysis of the findings showed a 31% decrease in the risk of death with surgery compared with interferon α-2b alone (median survival 13·6 months, 95% CI 9·7–17·4 vs 7·8 months, 5·9–9·7).101 Several

www.thelancet.com Published online August 26, 2015 http://dx.doi.org/10.1016/S0140-6736(15)00046-X

Seminar

hypotheses were formed to explain the observed benefits of nephrectomy: reduction of disease burden; decrease of immunosuppression and improved clinical response to cytokines; decreased growth-factor secretion (mainly VEGF); and inhibited tumour microenvironment secondary to surgically induced chronic metabolic acidosis.102 Over the past decade, the main advance in the treatment of metastatic renal cell carcinoma has been the development of treatments that affect specific targets in biological pathways. Agents targeting the VEGF/PDGFR/ mTOR pathway continue to be the mainstays of treatment (table 2). Although improved target specificity has lessened the risk of toxic effects associated with these drugs, durable complete responses remain the exception. Seven targeted drugs have been approved by European Medicines Agency and the US Food and Drug

Administration for management of renal cell carcinoma (table 2, figure 3). Sunitinib and sorafenib were associated significant benefits in progression-free survival and overall survival compared with placebo in two phase 3 trials,103,104 as was, temsirolimus in high-risk patients when compared with IFN monotherapy.110 In contrast, in patients with metastatic renal cell carcinoma who progressed after treatment with sunitinib, overall survival was shorter with temsirolimus than with sorafenib.111 Everolimus was associated with a significant benefit in progression-free survival relative to placebo in patients with metastatic renal cell carcinoma who progressed after sunitinib or sorafenib.112 Everolimus has been approved by the US Food and Drug Administration as the first drug to be used specifically to treat noncancerous kidney tumours (renal angiomyolipomas) in patients with tuberous sclerosis complex.30

Mechanism

Administration route

Setting

Treatment

Main findings

Sunitinib

PDGFR and VEGFR inhibitor

Capsules

Treatment naive (n=750)

Sunitinib vs interferon α

PFS, 11 months (95% CI 10–12) vs 5 months (95% CI 4–6), HR 0·42 (95% CI 0·32–0·54), p<0·001103

Sorafenib

PDGFR, VEGFR-2, and MAPK/MEK/ ERK inhibitor

Capsules

Second-line therapy (n=903)

Sorafenib vs placebo

PFS, 5·5 vs 2·8 months, HR 0·44 (95% CI 0·35–0·55), p<0·00001;104 OS, 17·8 vs 15·2 months, HR 0·74 (95% CI 0·74–1·04), p=0·146;104 OS after censoring,* 17·8 vs 14·3 months, HR 0·78 (95% CI 0·62–0·97), p=0·029104

Bevacizumab

Recombinant humanised monoclonal antibody directed against VEGF-A

Intravenous injection

Treatment naive (n=649)

Bevacizumab plus interferon PFS, 10·2 vs 5·4 months, HR 0·63 (95% CI 0·52–0·75), p<0·001;105 vs interferon plus placebo OS, 23·3 vs 21·3 months, HR 0·91 (95% CI 0·76–1·10), p=0·33106†

Treatment naive (n=732)

Bevacizumab plus interferon PFS, 8·5 vs 5·2, HR 0·72 (95% CI 0·61–0·83), p<0·001;107 OS, 18·3 vs interferon alone (95% CI 16·5–22·5) vs 17·4 (95% CI 14·4–20·0), HR 0·86 (95% CI 0·73–1·01), p=0·069108

Treatment naive (791)

Temsirolimus plus bevacizumab vs interferon plus bevacizumab

PFS, 9·1 vs 9·3 months, HR 1·1 (95% CI 0·9–1·3), p=0·8;109 OS, 25·8 vs 25·5 months, HR 1·0, p=0·6109

Treatment naive (poor-risk, n=626)

Temsirolimus vs interferon

PFS, 3·8 vs 1·9 months, p<0·0001; OS, 10·9 vs 7·3 months, HR 0·73 (95% CI 0·58–0·92), p=0·008110

Second line after sunitinib (n=512)

Temsirolimus vs sorafenib

PFS, 4·3 months (95% CI 4·0–5·4) vs 3·9 months (95% CI 2·8–4·2), HR 0·87 (95% CI 0·71–1·07), p=0·19;111 OS, 12·3 months (95% CI 10·1–14·8) vs 16·6 months (95% CI 13·6–18·7), HR 1·31 (95% CI 1·05–1·63), p=0·01111

Temsirolimus

mTOR inhibitor

Intravenous injection

Everolimus

mTOR inhibitor

Capsules

Second-line therapy (n=410)

Everolimus vs placebo

PFS, 4·0 months (95% CI 3·7–5·5) vs 1·9 months (95% CI 1·8–1·9), HR 0·30 (95% CI 0·22–0·40), p<0·0001112

Pazopanib

VEGFR/PDGFR/c-Kit inhibitor

Capsules

Treatment naive (n=233)

Pazopanib vs placebo

PFS, 11·1 vs 2·8 months, HR 0·40 (95% CI 0·27–0·60), p<0·0001;113 OS, 22·9 months (95% CI 17·6–25·4) vs 23·5 months (95% CI 12·0–34·3), HR 1·01 (95% CI 0·72–1·42)114

Treatment naive (n=1110)

Pazopanib vs sunitinib

PFS, 8·4 months (95% CI 8·3–10·9) vs 9·5 months (95% CI 8·3–11·1);115 OS, 28·3 months (95% CI 26·0–35·5) vs 29·1 months (95% CI 25·4–33·1), HR 0·92 (95% CI 0·79–1·06), p=0·24116

Cytokine refractory (n=202)

Pazopanib vs placebo

PFS, 7·4 vs 4·2 months, HR 0·54 (95% CI 0·35–0·84), p<0·0001;113 OS, 22·7 months (95% CI 19·3–28·3) vs 18·7 months (95% CI 14·2–26·3), HR 0·82 (95% CI 0·57–1·16)114

Treatment naive with mRCC (n=288)

Axitinib vs sorafenib

PFS, 10·1 month (95% CI 7·2–12·1) vs 6·5 months (95% CI 4·7–8·3), HR 0·77 (95% CI 0·56–1·05), p=0·038117

Second-line therapy for mRCC (n=723)

Axitinib vs sorafenib

PFS, 6·7 months (95% CI 6·3–8·6) vs 4·7 months (95% CI 4·6–5·6), HR 0·66 (95% CI 0·54–0·81), p<0·0001118

Axitinib

VEGFR (1–3), PDGFR, and c-Kit inhibitors

Capsules

PFS=progression-free survival. HR=hazard ratio. OS=overall survival. mRCC=metastatic renal cell carcinoma. *After crossover of placebo patients. †Unblinded after interim analysis and crossover and use of other antineoplastic therapies allowed.

Table 2: Phase 3 trials of targeted therapies approved for management of mRCC

www.thelancet.com Published online August 26, 2015 http://dx.doi.org/10.1016/S0140-6736(15)00046-X

7

Seminar

Blood vessel

Immune modulation NK cell

Interferon α

T cell Sunitinib Sorafenib Pazopanib

PDGFR CTLA4 Aldesleukin

Angiogenesis PD-1 (Ipilimumab)) (Rilotumumab)

FGF

PDGF PD-L1

FGFR

HGF

Axitinib

(Nivolumab) Paracrine growth stimulation

VEGFR (Brivanib) (Dovitinib

MET/HGFR

AKT

Temsirolimus Everolimus

VEGF

(Erlotinib) PI3K PTEN

EGF

Bevacizumab

mTORC1–2 HIF1α

Autocrine growth stimulation

Nucleus EGFR MAPK/ERK pathway

VHL Gene expression, cell growth, proliferation, survival

TGFα

Tumour cell

Figure 3: Biological pathways related to development of renal cell carcinoma and treatment targets NK=natural killer.

Pazopanib is an oral inhibitor of the VEGFR/ PDGFR/c-Kit pathway that has shown a significant benefit in progression-free survival compared with placebo in treatment-naive patients (table 2).113 The difference in final overall survival between patients treated with pazopanib or placebo was not significant, partly because of early and frequent crossover from placebo to pazopanib.113 In a phase 3 trial, similar efficacy was found with pazopanib and sunitinib, with slightly lower incidence of toxic effects (palmar-plantar erythrodysaesthesia and fatigue) but with higher incidence of liver function test abnormalities.115,116 A randomised crossover trial revealed a significant preference for pazopanib over sunitinib by patients and physicians (70% vs 22%, p<0·001),119 mainly because of a better adverse event profile for fatigue, improved quality of life,119 and reduced cost.120 Axitinib is a potent, selective, second-generation inhibitor of VEGFR 1, 2, and 3. In patients with treatmentnaive metastatic renal cell carcinoma, progression-free survival was not significantly increased with axitinib compared with sorafenib (table 2), although adverse events of any grade were less common in the axitinib group by an average of 10%.117 Conversely, among patients treated with a cytokine-containing regimen before axitinib, progression-free survival was longer than in those who received sorafenib (table 2).118 8

Bevacizumab is a recombinant humanised monoclonal antibody directed against VEGF. In two phase 3 trials of bevacizumab plus interferon compared with interferon plus placebo105 or interferon alone,106 bevacizumab was superior in terms of progression-free survival. One of the studies was unblinded because a clinically meaningful significant benefit was found in an interim analysis.105 In the final overall survival analyses of both trials, however, no significant differences were found between treatment groups (table 2), probably because of the confounding effect of crossover after the interim analysis and the use with other targeted drugs as salvage therapies in the interferon groups. 107,108 The introduction of targeted therapies has set a new benchmark for the treatment of metastatic renal cell carcinoma, but most patients included in trials have had clear-cell metastatic disease. The results, therefore, might not be universally applicable to all histological types. A contemporary analysis of patients with metastatic renal cell carcinoma treated with VEGF-targeting agents showed median overall survival of 18·8 months (95% CI 17·6–21·4) with a remarkable peak at 43·2 months for patients in the most favourable risk group (appendix).121 The most informative clinical variables to predict the outlook of patients with metastatic renal cell carcinoma treated with targeted therapies include haemoglobin (p<0·0001),

www.thelancet.com Published online August 26, 2015 http://dx.doi.org/10.1016/S0140-6736(15)00046-X

Seminar

serum corrected calcium (p=0·0006), Karnofsky performance status (p<0·0001), time from initial diagnosis to the start of treatment (p=0·009), absolute neutrophil count (p<0·0001), and platelet count (p=0·012).122 The most common treatment-related adverse events associated with targeted therapies for metastatic renal cell carcinoma are fatigue, hypertension, nausea, diarrhoea, dysphonia, and palmar-plantar erythrodysaesthesia.123 Grade 3 or 4 adverse events are expected in around a third of patients, with profiles improving slightly for the new-generation targeted therapies (eg, pazopanib).123 Clinical and translational studies are needed to identify the phenotypic predictors of response for each of the drugs.

Controversies, uncertainties, and research questions Despite progress in the understanding of renal cell carcinoma, uncertainties, controversies, and research questions remain. Advances are expected over the next few years from translational and clinical studies. Novel tissue and genetic markers are expected to improve accuracy of renal biopsy in characterising histology and disease aggressiveness at diagnosis (indolent mass vs clinically relevant malignancy). This approach could be crucial to the selection of patients who will benefit from active surveillance and other non-surgical approaches. Data are too immature to validate universal use of minimally invasive techniques (cryotherapy and RFA) for renal cell carcinoma. Only case series, unmatched retrospective studies, and chart reviews with short followup are available. Longer follow-up data and well designed comparative trials are needed to confirm oncological results, standardise techniques, and to recommend alternatives to surgery, especially for small renal masses. In terms of the potential use of radiotherapy as part of a multimodal approach, renal cell carcinoma had been viewed as a radioresistant tumour because of poor outcomes with low-dose radiotherapy.124 This notion has been challenged by emerging evidence that stereotactic body radiotherapy with a high-fraction dose has an effect in primary tumours and oligometastatic disease.124 Randomised trials of radiotherapy regimens and radiotherapy combined with targeted drugs are awaited to confirm the potential clinical implications. Medical and integrated therapies, identification of new target pathways, and optimum sequencing and combination of existing targeted agents are areas of active study.125 Molecular markers that seem to be predictive of response to sunitinib in patients with metastatic renal cell carcinoma have been identified. Prediction is based on molecular subtypes and suggests that tailored and personalised treatment is feasible.126 Upcoming results of ongoing trials testing novel drugs will increase the choice for urologists and oncologists treating locally advanced and metastatic disease. A promising avenue of clinical research, for example, is

the use of immune checkpoint inhibitors. These treatments work by targeting molecules that serve as checks and balances on immune responses. By blocking inhibitory molecules or activating stimulatory molecules, it is hoped that pre-existing anticancer immune responses will be restored. Nivolumab, for instance, is a fully human IgG4 monoclonal antibody that binds to the PD-1 receptor and restores T-cell immune activity. This drug has shown promising results in the treatment of metastatic renal cell carcinoma.127 Many other new drugs are being investigated, and tivozanib128,129 dovitinib,130 cabozantinib,131 erlotinib,132 and sirolimus133 are being tested as potential treatments (figure 3). Additionally, two phase 3 studies are assessing the role of cytoreductive nephrectomy in the context of novel systemic therapies and the best treatment sequencing for surgery and targeted drugs (NCT01099423).134 The role of adjuvant therapy after surgery is also being investigated. Past attempts to prove clinical benefit with adjuvant immunotherapy (interferon α and interleukin-2) systematically failed.135–137 Five phase 3 trials including more than 6000 patients with non-metastatic renal cell carcinoma are being done to assess the role of targeted therapies in the adjuvant setting,138 no benefit has yet been found with adjuvant VEGF inhibitors (sorafenib or sunitinib) in patients at high risk of recurrence (NCT00326898). Surgical metastasectomy has been gaining popularity as part of a multimodal approach to treating patients with oligometastatic renal cell carcinoma and has shown acceptable morbidity in selected patients.139–141 Partial or complete removal of the metastatic lesions was associated with improved overall survival, especially in patients with adequate performance and functional statuses.139–142 Patients are being recruited into randomised clinical trials to assess the efficacy of a multimodal approach based on targeted therapy after surgical metastasectomy (NCT00918775 and NCT01444807).

Conclusions Renal cell carcinoma provides one of the best examples in oncology of how innovative basic research, novel clinical findings, and improved surgical techniques can converge to improve the care of patients. Alternative management strategies, such as active surveillance or cryotherapy, can be considered for an increasing number of patients. For patients who are candidates for surgical excision, the development of minimally invasive techniques, such as robot-assisted laparoscopic nephronsparing surgery, has decreased the risk of surgically induced functional impairment. Additionally, extended understanding of biological pathways and novel targeted therapies has led to improved survival outcomes in patients with metastatic disease. Further developments in translational research and findings from trials are expected to integrate surgical and systemic treatments to enhance cancer control.

www.thelancet.com Published online August 26, 2015 http://dx.doi.org/10.1016/S0140-6736(15)00046-X

9

Seminar

Contributors Both authors did the literature search, wrote the drafts, and reviewed the contents of the paper. Declaration of interests UC has received consultancy and lecturer honoraria and FM has received research support, consultancy fees, and lecturer honoraria from Recordati. Acknowledgments We thank Jeni Crockett-Holme for editorial support and Nicola Spreafico for graphics support. References 1 Ellimoottil C, Greco KA, Hart S, et al. New modalities for evaluation and surveillance of complex renal cysts. J Urol 2014; 192: 1604–11. 2 Shah S, Watnick T, Atta MG. Not all renal cysts are created equal. Lancet 2010; 376: 1024. 3 Siegel R, Ma J, Zou Z, Jemal A. Cancer statistics, 2014. CA Cancer J Clin 2014; 64: 9–29. 4 Ferlay JSI, Ervik M, Dikshit R, et al. GLOBOCAN 2012 v1.0, cancer incidence and mortality worldwide: IARC CancerBase No. 11. 2013. http://globocan.iarc.fr (accessed Jan 1, 2015). 5 Levi F, Ferlay J, Galeone C, et al. The changing pattern of kidney cancer incidence and mortality in Europe. BJU Int 2008; 101: 949–58. 6 Chow WH, Linehan WM, Devesa SS. Re: rising incidence of small renal masses: a need to reassess treatment effect. J Natl Cancer Inst 2007; 99: 569–70. 7 Hollingsworth JM, Miller DC, Daignault S, Hollenbeck BK. Rising incidence of small renal masses: a need to reassess treatment effect. J Natl Cancer Inst 2006; 98: 1331–34. 8 Welch HG, Schwartz LM, Woloshin S. Are increasing 5-year survival rates evidence of success against cancer? JAMA 2000; 283: 2975–78. 9 Theis RP, Dolwick Grieb SM, Burr D, Siddiqui T, Asal NR. Smoking, environmental tobacco smoke, and risk of renal cell cancer: a population-based case-control study. BMC Cancer 2008; 8: 387. 10 Deckers IA, van den Brandt PA, van Engeland M, et al. Polymorphisms in genes of the renin-angiotensin-aldosterone system and renal cell cancer risk: interplay with hypertension and intakes of sodium, potassium and fluid. Int J Cancer 2015; 136: 1104–16. 11 Zucchetto A, Dal Maso L, Tavani A, et al. History of treated hypertension and diabetes mellitus and risk of renal cell cancer. Ann Oncol 2007; 18: 596–600. 12 Corrao G, Scotti L, Bagnardi V, Sega R. Hypertension, antihypertensive therapy and renal-cell cancer: a meta-analysis. Curr Drug Saf 2007; 2: 125–33. 13 Gati A, Kouidhi S, Marrakchi R, et al. Obesity and renal cancer: Role of adipokines in the tumor-immune system conflict. Oncoimmunology 2014; 3: e27810. 14 Bergström A, Hsieh CC, Lindblad P, Lu CM, Cook NR, Wolk A. Obesity and renal cell cancer—a quantitative review. Br J Cancer 2001; 85: 984–90. 15 Karami S, Daugherty SE, Purdue MP. A prospective study of alcohol consumption and renal cell carcinoma risk. Int J Cancer 2015; 137: 238–42. 16 Rohrmann S, Linseisen J, Overvad K, et al. Meat and fish consumption and the risk of renal cell carcinoma in the European prospective investigation into cancer and nutrition. Int J Cancer 2015; 136: E423–31. 17 Ljungberg B, Campbell SC, Choi HY, et al. The epidemiology of renal cell carcinoma. Eur Urol 2011; 60: 615–21. 18 Shuch B, Amin A, Armstrong AJ, et al. Understanding pathologic variants of renal cell carcinoma: distilling therapeutic opportunities from biologic complexity. Eur Urol 2015; 67: 85–97. 19 Srigley JR, Delahunt B, Eble JN, et al, and the ISUP Renal Tumor Panel. The International Society of Urological Pathology (ISUP) Vancouver classification of renal neoplasia. Am J Surg Pathol 2013; 37: 1469–89. 20 Bianchi M, Sun M, Jeldres C, et al. Distribution of metastatic sites in renal cell carcinoma: a population-based analysis. Ann Oncol 2012; 23: 973–80.

10

21

22

23 24

25

26 27 28 29

30 31 32

33

34

35

36 37 38

39

40

41

42

43

44

Srinivasan R, Ricketts CJ, Sourbier C, Linehan WM. New strategies in renal cell carcinoma: targeting the genetic and metabolic basis of disease. Clin Cancer Res 2015; 21: 10–17. Moore LE, Nickerson ML, Brennan P, et al. Von Hippel-Lindau (VHL) inactivation in sporadic clear cell renal cancer: associations with germline VHL polymorphisms and etiologic risk factors. PLoS Genet 2011; 7: e1002312. Brugarolas J. PBRM1 and BAP1 as novel targets for renal cell carcinoma. Cancer J 2013; 19: 324–32. Cancer Genome Atlas Research Network. Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature 2013; 499: 43–49. Ibragimova I, Maradeo ME, Dulaimi E, Cairns P. Aberrant promoter hypermethylation of PBRM1, BAP1, SETD2, KDM6A and other chromatin-modifying genes is absent or rare in clear cell RCC. Epigenetics 2013; 8: 486–93. Gossage L, Eisen T, Maher ER. VHL, the story of a tumour suppressor gene. Nat Rev Cancer 2015; 15: 55–64. Dharmawardana PG, Giubellino A, Bottaro DP. Hereditary papillary renal carcinoma type I. Curr Mol Med 2004; 4: 855–68. Perdeaux E, Solly J. Birt-Hogg-Dubé syndrome. JAMA 2013; 309: 1460. Shuch B, Ricketts CJ, Vocke CD, et al. Adrenal nodular hyperplasia in hereditary leiomyomatosis and renal cell cancer. J Urol 2013; 189: 430–35. Sooriakumaran P, Anderson CJ. Everolimus for renal angiomyolipoma in tuberous sclerosis. Lancet 2013; 381: 783–85. Sacco E, Pinto F, Sasso F, et al. Paraneoplastic syndromes in patients with urological malignancies. Urol Int 2009; 83: 1–11. Hofbauer SL, de Martino M, Seemann C, et al. Associations between presenting symptoms, clinicopathological parameters, and prognosis in a contemporary series of patients with renal cell carcinoma. Korean J Urol 2014; 55: 505–10. Ljungberg BBK, Bex A, Canfield S, et al. Guidelines on renal cell carcinoma, update. April 2014. http://uroweb.org/wp-content/ uploads/10-Renal-Cell-Carcinoma_LR.pdf (accessed July 15, 2015). National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology: Kidney Cancer 2. 2014. http://www.nccn. org/professionals/physician_gls/pdf/kidney.pdf (accessed July 15, 2015). American Urological Association. Follow-up for clinically localized renal neoplasms: AUA guideline 2013. April 2013. https://www. auanet.org/common/pdf/education/clinical-guidance/RenalCancer-Followup.pdf (accessed July 15, 2015). Hilton S, Jones LP. Recent advances in imaging cancer of the kidney and urinary tract. Surg Oncol Clin N Am 2014; 23: 863–910. Ljungberg B, Bensalah K, Canfield S, et al. EAU guidelines on renal cell carcinoma: 2014 update. Eur Urol 2015; 67: 913–24. Muselaers CH, Boerman OC, Oosterwijk E, Langenhuijsen JF, Oyen WJ, Mulders PF. Indium-111-labeled girentuximab immunoSPECT as a diagnostic tool in clear cell renal cell carcinoma. Eur Urol 2013; 63: 1101–06. Divgi CR, Uzzo RG, Gatsonis C, et al. Positron emission tomography/computed tomography identification of clear cell renal cell carcinoma: results from the REDECT trial. J Clin Oncol 2013; 31: 187–94. Tomaszewski JJ, Uzzo RG, Smaldone MC. Heterogeneity and renal mass biopsy: a review of its role and reliability. Cancer Biol Med 2014; 11: 162–72. Ball MW, Bezerra SM, Gorin MA, et al. Grade heterogeneity in small renal masses: potential implications for renal mass biopsy. J Urol 2015; 193: 36–40. Volpe A, Finelli A, Gill IS, et al. Rationale for percutaneous biopsy and histologic characterisation of renal tumours. Eur Urol 2012; 62: 491–504. Leveridge MJ, Finelli A, Kachura JR, et al. Outcomes of small renal mass needle core biopsy, nondiagnostic percutaneous biopsy, and the role of repeat biopsy. Eur Urol 2011; 60: 578–84. Baillargeon-Gagné S, Jeldres C, Lughezzani G, et al. A comparative population-based analysis of the rate of partial vs radical nephrectomy for clinically localized renal cell carcinoma. BJU Int 2010; 105: 359–64.

www.thelancet.com Published online August 26, 2015 http://dx.doi.org/10.1016/S0140-6736(15)00046-X

Seminar

45

46

47

48

49 50

51

52

53

54 55

56

57

58

59

60

61

62

63

64

65

66

Capitanio U, Terrone C, Antonelli A, et al. Nephron-sparing techniques independently decrease the risk of cardiovascular events relative to radical nephrectomy in patients with a T1a-T1b renal mass and normal preoperative renal function. Eur Urol 2015; 67: 683–89. Sun M, Abdollah F, Bianchi M, et al. Treatment management of small renal masses in the 21st century: a paradigm shift. Ann Surg Oncol 2012; 19: 2380–87. Kutikov A, Egleston BL, Wong YN, Uzzo RG. Evaluating overall survival and competing risks of death in patients with localized renal cell carcinoma using a comprehensive nomogram. J Clin Oncol 2010; 28: 311–17. Mehrazin R, Smaldone MC, Egleston B, et al. Is anatomic complexity associated with renal tumor growth kinetics under active surveillance? Urol Oncol 2015; 33: 167.e7–12. Rendon RA. Active surveillance as the preferred management option for small renal masses. Can Urol Assoc J 2010; 4: 136–38. Pierorazio PM, Johnson MH, Ball MW, 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 2015; published online Feb 16. DOI:10.1016/j.eururo.2015.02.001. Crispen PL, Viterbo R, Fox EB, Greenberg RE, Chen DY, Uzzo RG. Delayed intervention of sporadic renal masses undergoing active surveillance. Cancer 2008; 112: 1051–57. Mehrazin R, Smaldone MC, Kutikov A, et al. Growth kinetics and short-term outcomes of cT1b and cT2 renal masses under active surveillance. J Urol 2014; 192: 659–64. Jewett MA, Mattar K, Basiuk J, et al. Active surveillance of small renal masses: progression patterns of early stage kidney cancer. Eur Urol 2011; 60: 39–44. Hoffmann NE, Bischof JC. The cryobiology of cryosurgical injury. Urology 2002; 60 (suppl 1): 40–49. Katsanos K, Mailli L, Krokidis M, McGrath A, Sabharwal T, Adam A. Systematic review and meta-analysis of thermal ablation versus surgical nephrectomy for small renal tumours. Cardiovasc Intervent Radiol 2014; 37: 427–37. Kunkle DA, Egleston BL, Uzzo RG. Excise, ablate or observe: the small renal mass dilemma--a meta-analysis and review. J Urol 2008; 179: 1227–33. Motzer RJ, Jonasch E, Agarwal N, et al. Kidney Cancer, Version 3.2015: featured updates to the NCCN guidelines. J Natl Compr Canc Netw 2015; 13: 151–59. Woldrich JM, Palazzi K, Stroup SP, et al. Trends in the surgical management of localized renal masses: thermal ablation, partial and radical nephrectomy in the USA, 1998–2008. BJU Int 2013; 111: 1261–68. Gervais DA, McGovern FJ, Arellano RS, McDougal WS, Mueller PR. Radiofrequency ablation of renal cell carcinoma: part 1, Indications, results, and role in patient management over a 6-year period and ablation of 100 tumors. AJR Am J Roentgenol 2005; 185: 64–71. Thompson RH, Atwell T, Schmit G, et al. Comparison of Partial Nephrectomy and Percutaneous Ablation for cT1 Renal Masses. Eur Urol 2015; 67: 252–59. Kutikov A, Smaldone MC, Uzzo RG. Focal therapy for treatment of the small renal mass: dealer’s choice or a therapeutic gamble? Eur Urol 2015; 67: 260–61. Crépel M, Jeldres C, Sun M, et al. A population-based comparison of cancer-control rates between radical and partial nephrectomy for T1A renal cell carcinoma. Urology 2010; 76: 883–88. Crépel M, Jeldres C, Perrotte P, et al. Nephron-sparing surgery is equally effective to radical nephrectomy for T1BN0M0 renal cell carcinoma: a population-based assessment. Urology 2010; 75: 271–75. Van Poppel H, Da Pozzo L, Albrecht W, 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 2011; 59: 543–52. Scosyrev E, Messing EM, Sylvester R, Campbell S, Van Poppel H. Renal function after nephron-sparing surgery versus radical nephrectomy: results from EORTC randomized trial 30904. Eur Urol 2014; 65: 372–77. Lane BR, Campbell SC, Demirjian S, Fergany AF. Surgically induced chronic kidney disease may be associated with a lower risk of progression and mortality than medical chronic kidney disease. J Urol 2013; 189: 1649–55.

67

68

69

70

71 72

73

74 75

76

77

78

79

80

81

82

83

84

85

86

87

88

Huang WC, Elkin EB, Levey AS, Jang TL, Russo P. Partial nephrectomy versus radical nephrectomy in patients with small renal tumors—is there a difference in mortality and cardiovascular outcomes? J Urol 2009; 181: 55–61. Karakiewicz PI, Briganti A, Chun FK, et al. Multi-institutional validation of a new renal cancer-specific survival nomogram. J Clin Oncol 2007; 25: 1316–22. Zini L, Perrotte P, Capitanio U, et al. Radical versus partial nephrectomy: effect on overall and noncancer mortality. Cancer 2009; 115: 1465–71. Terrone C, Volpe A. Can emerging level 1 evidence “discourage” elective nephron-sparing surgery for small renal tumors? Eur Urol 2011; 59: 553–55. Babaian KN, Merrill MM, Matin S, et al. Partial nephrectomy in the setting of metastatic renal cell carcinoma. J Urol 2014; 192: 36–42. Capitanio U, Zini L, Perrotte P, et al. Cytoreductive partial nephrectomy does not undermine cancer control in metastatic renal cell carcinoma: a population-based study. Urology 2008; 72: 1090–95. Volpe A, Blute ML, Ficarra V, et al. Renal ischemia and function after partial nephrectomy: a collaborative review of the literature. Eur Urol 2015; 68: 61–74. Parekh DJ, Weinberg JM, Ercole B, et al. Tolerance of the human kidney to isolated controlled ischemia. J Am Soc Nephrol 2013; 24: 506–17. Simmons MN, Hillyer SP, Lee BH, Fergany AF, Kaouk J, Campbell SC. Functional recovery after partial nephrectomy: effects of volume loss and ischemic injury. J Urol 2012; 187: 1667–73. Funahashi Y, Hattori R, Yamamoto T, Kamihira O, Kato K, Gotoh M. Ischemic renal damage after nephron-sparing surgery in patients with normal contralateral kidney. Eur Urol 2009; 55: 209–15. Van Poppel H, Da Pozzo L, Albrecht W, et al, and the European Organization for Research and Treatment of Cancer (EORTC), and the National Cancer Institute of Canada Clinical Trials Group (NCIC CTG), and the Southwest Oncology Group (SWOG), and the Eastern Cooperative Oncology Group (ECOG). A prospective randomized EORTC intergroup phase 3 study comparing the complications of elective nephron-sparing surgery and radical nephrectomy for low-stage renal cell carcinoma. Eur Urol 2007; 51: 1606–15. Hollenbeck BK, Taub DA, Miller DC, Dunn RL, Wei JT. National utilization trends of partial nephrectomy for renal cell carcinoma: a case of underutilization? Urology 2006; 67: 254–59. Miller DC, Hollingsworth JM, Hafez KS, Daignault S, Hollenbeck BK. Partial nephrectomy for small renal masses: an emerging quality of care concern? J Urol 2006; 175: 853–57. Thompson RH, Kaag M, Vickers A, et al. Contemporary use of partial nephrectomy at a tertiary care center in the United States. J Urol 2009; 181: 993–97. Ficarra V, Novara G, Secco S, et al. Preoperative aspects and dimensions used for an anatomical (PADUA) classification of renal tumours in patients who are candidates for nephron-sparing surgery. Eur Urol 2009; 56: 786–93. Kutikov A, Uzzo RG. The R.E.N.A.L. nephrometry score: a comprehensive standardized system for quantitating renal tumor size, location and depth. J Urol 2009; 182: 844–53. Simmons MN, Ching CB, Samplaski MK, Park CH, Gill IS. Kidney tumor location measurement using the C index method. J Urol 2010; 183: 1708–13. Sivarajan G, Taksler GB, Walter D, Gross CP, Sosa RE, Makarov DV. The Effect of the Diffusion of the Surgical Robot on the Hospitallevel Utilization of Partial Nephrectomy. Med Care 2015; 53: 71–78. Choi JE, You JH, Kim DK, Rha KH, Lee SH. Comparison of perioperative outcomes between robotic and laparoscopic partial nephrectomy: a systematic review and meta-analysis. Eur Urol 2015; 67: 891–901. Gratzke C, Seitz M, Bayrle F, et al. Quality of life and perioperative outcomes after retroperitoneoscopic radical nephrectomy (RN), open RN and nephron-sparing surgery in patients with renal cell carcinoma. BJU Int 2009; 104: 470–75. Hemal AK, Kumar A, Kumar R, Wadhwa P, Seth A, Gupta NP. Laparoscopic versus open radical nephrectomy for large renal tumors: a long-term prospective comparison. J Urol 2007; 177: 862–66. Asimakopoulos AD, Miano R, Annino F, et al. Robotic radical nephrectomy for renal cell carcinoma: a systematic review. BMC Urol 2014; 14: 75.

www.thelancet.com Published online August 26, 2015 http://dx.doi.org/10.1016/S0140-6736(15)00046-X

11

Seminar

89

90 91

92 93

94

95

96

97

98

99

100

101

102

103 104

105

106

107

108

109

12

Bekema HJ, MacLennan S, Imamura M, et al. Systematic review of adrenalectomy and lymph node dissection in locally advanced renal cell carcinoma. Eur Urol 2013; 64: 799–810. Capitanio U, Becker F, Blute ML, et al. Lymph node dissection in renal cell carcinoma. Eur Urol 2011; 60: 1212–20. Blom JH, van Poppel H, Maréchal JM, et al, and the EORTC Genitourinary Tract Cancer Group. 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 2009; 55: 28–34. Capitanio UB, Becker F, Blute F, et al. Lymph node dissection in renal cell carcinoma. Eur Urol 2011; 60: 1212–20. Bex A, Vermeeren L, de Windt G, Prevoo W, Horenblas S, Olmos RA. Feasibility of sentinel node detection in renal cell carcinoma: a pilot study. Eur J Nucl Med Mol Imaging 2010; 37: 1117–23. Bex A, Vermeeren L, Meinhardt W, Prevoo W, Horenblas S, Valdés Olmos RA. Intraoperative sentinel node identification and sampling in clinically node-negative renal cell carcinoma: initial experience in 20 patients. World J Urol 2011; 29: 793–99. Haferkamp A, Bastian PJ, Jakobi H, et al. Renal cell carcinoma with tumor thrombus extension into the vena cava: prospective long-term followup. J Urol 2007; 177: 1703–08. Bellmunt J, Puente J, Garcia de Muro J, Lainez N, Rodríguez C, Duran I. SEOM clinical guidelines for the treatment of renal cell carcinoma. Clin Transl Oncol 2014; 16: 1043–50. Marincola FM, White DE, Wise AP, Rosenberg SA. Combination therapy with interferon alfa-2a and interleukin-2 for the treatment of metastatic cancer. J Clin Oncol 1995; 13: 1110–22. Coppin C, Porzsolt F, Awa A, Kumpf J, Coldman A, Wilt T. Immunotherapy for advanced renal cell cancer. Cochrane Database Syst Rev 2005; 1: CD001425. Flanigan RC, Salmon SE, Blumenstein BA, et al. Nephrectomy followed by interferon alfa-2b compared with interferon alfa-2b alone for metastatic renal-cell cancer. N Engl J Med 2001; 345: 1655–59. Mickisch GH, Garin A, van Poppel H, de Prijck L, Sylvester R, and the European Organisation for Research and Treatment of Cancer (EORTC) Genitourinary Group. Radical nephrectomy plus interferon-alfa-based immunotherapy compared with interferon alfa alone in metastatic renal-cell carcinoma: a randomised trial. Lancet 2001; 358: 966–70. Flanigan RC, Mickisch G, Sylvester R, Tangen C, Van Poppel H, Crawford ED. Cytoreductive nephrectomy in patients with metastatic renal cancer: a combined analysis. J Urol 2004; 171: 1071–76. Rini BI, Campbell SC. The evolving role of surgery for advanced renal cell carcinoma in the era of molecular targeted therapy. J Urol 2007; 177: 1978–84. Motzer RJ, Hutson TE, Tomczak P, et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med 2007; 356: 115–24. Escudier B, Eisen T, Stadler WM, et al. Sorafenib for treatment of renal cell carcinoma: final efficacy and safety results of the phase III treatment approaches in renal cancer global evaluation trial. J Clin Oncol 2009; 27: 3312–18. Escudier B, Pluzanska A, Koralewski P, et al, and the AVOREN Trial investigators. Bevacizumab plus interferon alfa-2a for treatment of metastatic renal cell carcinoma: a randomised, double-blind phase III trial. Lancet 2007; 370: 2103–11. Escudier B, Bellmunt J, Négrier S, et al. 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 2010; 28: 2144–50. Rini BI, Halabi S, Rosenberg JE, et al. Bevacizumab plus interferon alfa compared with interferon alfa monotherapy in patients with metastatic renal cell carcinoma: CALGB 90206. J Clin Oncol 2008; 26: 5422–28. Rini BI, Halabi S, Rosenberg JE, et al. Phase III trial of bevacizumab plus interferon alfa versus interferon alfa monotherapy in patients with metastatic renal cell carcinoma: final results of CALGB 90206. J Clin Oncol 2010; 28: 2137–43. Rini BI, Bellmunt J, Clancy J, et al. Randomized phase III trial of temsirolimus and bevacizumab versus interferon alfa and bevacizumab in metastatic renal cell carcinoma: INTORACT trial. J Clin Oncol 2014; 32: 752–59.

110 Hudes G, Carducci M, Tomczak P, et al, and the Global ARCC Trial. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med 2007; 356: 2271–81. 111 Hutson TE, Escudier B, Esteban E, et al. Randomized phase III trial of temsirolimus versus sorafenib as second-line therapy after sunitinib in patients with metastatic renal cell carcinoma. J Clin Oncol 2014; 32: 760–67. 112 Motzer RJ, Escudier B, Oudard S, et al, and the RECORD-1 Study Group. Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled phase III trial. Lancet 2008; 372: 449–56. 113 Sternberg CN, Davis ID, Mardiak J, et al. Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized phase III trial. J Clin Oncol 2010; 28: 1061–68. 114 Sternberg CN, Hawkins RE, Wagstaff J, et al. A randomised, double-blind phase III study of pazopanib in patients with advanced and/or metastatic renal cell carcinoma: final overall survival results and safety update. Eur J Cancer 2013; 49: 1287–96. 115 Motzer RJ, Hutson TE, Cella D, et al. Pazopanib versus sunitinib in metastatic renal-cell carcinoma. N Engl J Med 2013; 369: 722–31. 116 Motzer RJ, Hutson TE, McCann L, Deen K, Choueiri TK. Overall survival in renal-cell carcinoma with pazopanib versus sunitinib. N Engl J Med 2014; 370: 1769–70. 117 Hutson TE, Lesovoy V, Al-Shukri S, et al. Axitinib versus sorafenib as first-line therapy in patients with metastatic renal-cell carcinoma: a randomised open-label phase 3 trial. Lancet Oncol 2013; 14: 1287–94. 118 Rini BI, Escudier B, Tomczak P, et al. Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): a randomised phase 3 trial. Lancet 2011; 378: 1931–39. 119 Escudier B, Porta C, Bono P, et al. Randomized, controlled, doubleblind, cross-over trial assessing treatment preference for pazopanib versus sunitinib in patients with metastatic renal cell carcinoma: PISCES Study. J Clin Oncol 2014; 32: 1412–18. 120 Delea TE, Amdahl J, Diaz J, Nakhaipour HR, Hackshaw MD. Cost-effectiveness of pazopanib versus sunitinib for renal cancer in the United States. J Manag Care Spec Pharm 2015; 121 Heng DY, Xie W, Regan MM, 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 2013; 14: 141–48. 122 Heng DY, Xie W, Regan MM, et al. Prognostic factors for overall survival in patients with metastatic renal cell carcinoma treated with vascular endothelial growth factor-targeted agents: results from a large, multicenter study. J Clin Oncol 2009; 27: 5794–99. 123 Ruiz JN, Belum VR, Creel P, Cohn A, Ewer M, Lacouture ME. Current practices in the management of adverse events associated with targeted therapies for advanced renal cell carcinoma: a national survey of oncologists. Clin Genitourin Cancer 2014; 12: 341–47. 124 De Meerleer G, Khoo V, Escudier B, et al. Radiotherapy for renal-cell carcinoma. Lancet Oncol 2014; 15: e170–77. 125 Singh P, Agarwal N, Pal SK. Sequencing systemic therapies for metastatic kidney cancer. Curr Treat Options Oncol 2015; 16: 316. 126 Beuselinck B, Job S, Becht E, et al. Molecular subtypes of clear cell renal cell carcinoma are associated with sunitinib response in the metastatic setting. Clin Cancer Res 2015; 21: 1329–39. 127 Motzer RJ, Rini BI, McDermott DF, et al. Nivolumab for metastatic renal cell carcinoma: results of a randomized phase II trial. J Clin Oncol 2015; 33: 1430–37. 128 Motzer RJ, Nosov D, Eisen T, et al. Tivozanib versus sorafenib as initial targeted therapy for patients with metastatic renal cell carcinoma: results from a phase III trial. J Clin Oncol 2013; 31: 3791–99. 129 Mehta A, Sonpavde G, Escudier B. Tivozanib for the treatment of renal cell carcinoma: results and implications of the TIVO-1 trial. Future Oncol 2014; 10: 1819–26. 130 Motzer RJ, Porta C, Vogelzang NJ, et al. Dovitinib versus sorafenib for third-line targeted treatment of patients with metastatic renal cell carcinoma: an open-label, randomised phase 3 trial. Lancet Oncol 2014; 15: 286–96. 131 Vaishampayan U. Cabozantinib as a novel therapy for renal cell carcinoma. Curr Oncol Rep 2013; 15: 76–82. 132 Gordon MS, Hussey M, Nagle RB, et al. Phase II study of erlotinib in patients with locally advanced or metastatic papillary histology renal cell cancer: SWOG S0317. J Clin Oncol 2009; 27: 5788–93.

www.thelancet.com Published online August 26, 2015 http://dx.doi.org/10.1016/S0140-6736(15)00046-X

Seminar

133 Brugarolas J, Lotan Y, Watumull L, Kabbani W. Sirolimus in metatastic renal cell carcinoma. J Clin Oncol 2008; 26: 3457–60. 134 EORTC. Randomized phase III trial comparing presurgical sunitinib followed by nephrectomy and sunitinib versus nephrectomy followed by sunitinib in patients with synchronous metastatic renal cell carcinoma. 2010. http://www.eortc.be/protoc/ Details.asp?Protocol=30073&T=2010 (accessed July 15, 2015). 135 Messing EM, Manola J, Wilding G, et al, and the Eastern Cooperative Oncology Group/Intergroup trial. Phase III study of interferon alfa-NL as adjuvant treatment for resectable renal cell carcinoma: an Eastern Cooperative Oncology Group/Intergroup trial. J Clin Oncol 2003; 21: 1214–22. 136 Pizzocaro G, Piva L, Colavita M, et al. Interferon adjuvant to radical nephrectomy in Robson stages II and III renal cell carcinoma: a multicentric randomized study. J Clin Oncol 2001; 19: 425–31. 137 Clark JI, Atkins MB, Urba WJ, et al. Adjuvant high-dose bolus interleukin-2 for patients with high-risk renal cell carcinoma: a cytokine working group randomized trial. J Clin Oncol 2003; 21: 3133–40.

138 Lam JS, Shvarts O, Belldegrun AS. Kidney cancer management in 2004: an update for the practicing general urologist. Curr Urol Rep 2004; 5: 1–3. 139 Kim DY, Karam JA, Wood CG. Role of metastasectomy for metastatic renal cell carcinoma in the era of targeted therapy. World J Urol 2014; 32: 631–42. 140 Breau RH, Blute ML. Surgery for renal cell carcinoma metastases. Curr Opin Urol 2010; 20: 375–81. 141 Russo P, O’Brien MF. Surgical intervention in patients with metastatic renal cancer: metastasectomy and cytoreductive nephrectomy. Urol Clin North Am 2008; 35: 679–86, viii. 142 Dabestani S, Marconi L, Hofmann F, et al. Local treatments for metastases of renal cell carcinoma: a systematic review. Lancet Oncol 2014; 15: e549–61.

www.thelancet.com Published online August 26, 2015 http://dx.doi.org/10.1016/S0140-6736(15)00046-X

13