You are on page 1of 13

Seminar

Renal cancer
Umberto Capitanio, Francesco Montorsi

The diagnosis and management of renal cell carcinoma have changed remarkably rapidly. Although the incidence of Published Online
renal cell carcinoma has been increasing, survival has improved substantially. As incidental diagnosis of small August 26, 2015
http://dx.doi.org/10.1016/
indolent cancers has become more frequent, active surveillance, robot-assisted nephron-sparing surgical techniques, S0140-6736(15)00046-X
and minimally invasive procedures, such as thermal ablation, have gained popularity. Despite progression in cancer
Department of Urology,
control and survival, locally advanced disease and distant metastases are still diagnosed in a notable proportion of Vita-Salute San Raffaele
patients. An integrated management strategy that includes surgical debulking and systemic treatment with well University (U Capitanio MD)
established targeted biological drugs has improved the care of patients. Nevertheless, uncertainties, controversies, and Division of Experimental
Oncology, URI, Urological
and research questions remain. Further advances are expected from translational and clinical studies. Research Institute, Renal
Cancer Unit
Introduction Pathophysiology and genetics (Prof F Montorsi FRCS),
Abdominal imaging is used for many different medical Renal cell carcinoma comprises a heterogeneous group of IRCCS San Raffaele Scientific
Institute, Milan, Italy
disorders (eg, hypertension, diabetes) and, therefore, cancers with different genetic and molecular alterations
Correspondence to:
incidental detection of renal masses is becoming underlying the many documented histological subtypes.18
Dr Umberto Capitanio,
increasingly common. A renal mass might be merely a Clear-cell, papillary (types 1 and 2), and chromophobe are Department of Urology,
simple renal cyst that requires no treatment or follow-up,1 the most common solid renal cell carcinomas within the San Raffaele Hospital,
but in a notable proportion of cases masses are benign kidney and account for 85–90% of all renal malignancies.18 Via Olgettina 60,
Milan 20132, Italy
renal lesions (eg, angiomyolipomas or oncocytomas) or Less common cancers include papillary adenoma, capitanio.umberto@hsr.it
malignant renal cell carcinomas that might require multilocular cystic clear-cell carcinoma, hybrid oncocytic
additional procedures or interventions.2 Although most chromophobe tumour, carcinoma of the collecting ducts
incidentally detected lesions are small low-grade tumours, of Bellini, renal medullary carcinoma, carcinoma
up to 17% of all renal cell carcinomas have distant associated with neuroblastoma, and mucinous tubular
metastases at the time of diagnosis.3 In this heterogeneous and spindle-cell carcinoma.18 The International Society of
clinical setting, developments in molecular biology, Urological Pathology Vancouver Consensus Statement
diagnostic techniques, surgery, and medical oncology are added five more epithelial tumour subtypes of renal cell
revolutionising the approach to this disease. carcinoma: tubulocystic, acquired cystic disease-
associated, clear-cell tubulopapillary, microphthalmia
Epidemiology family translocation, and hereditary leiomyomatosis–
In 2013, renal cell carcinoma was diagnosed in more renal cell carcinoma syndrome-associated.19
than 350 000 people worldwide, which made it the Although most renal masses are initially confined to
seventh most common site for tumours, and this cancer one organ, early haematogenic dissemination is
is associated with more than 140 000 deaths per year.4 responsible for metastatic disease at the time of
Incidence of renal cell carcinoma varies worldwide, diagnosis. The most frequent sites of distant metastases
being higher in developed countries than in developing are lungs, bone, and brain, but adrenal glands,
countries (appendix). Incidence predominates in men, contralateral kidney, and liver might be involved.20 See Online for appendix
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 Search strategy and selection criteria
reported over the past three decades,3,5 although these We identified data for this Seminar through searches of
might be skewed by disease stage migration.6–8 In Europe, PubMed and Web of Science. We included papers in core
mortality from renal cell carcinoma peaked at 4·8 per clinical journals that described studies in adults and were
100 000 in 1990–94 and had declined to 4·1 per published in English from 2004 to 2014. Search terms
100 000 (−13%) in 2000–04.5 In the USA, 5-year relative included “renal cancer” or “kidney cancer” in combination
survival rates at diagnosis increased from 50% in 1975–77 with the terms “epidemiology”, “genetics”, “pathophysiology”,
to 57% in 1987–89, and reached 73% in 2003–09.3 “diagnosis”, “biopsy”, “treatment”, “surgery”, “active
Established risk factors include smoking tobacco,9 surveillance”, “medical therapy”, or “targeted therapy”. We also
hypertension,10–12 and obesity.13,14 These associations might searched cited references from selected articles and previous
be biased by the probability that such patients undergo reviews to identify further relevant papers not retrieved by
routine imaging, which could increase the likelihood of this search, including those outside the time period of the
incidental detection of renal masses. Several other initial search. We also searched for updates on the main
associations have been suggested, for example, a features of renal cell carcinoma, with particular attention paid
protective effect of alcohol consumption,15 a negative to reports focusing on novel treatments, renal biopsy, new
effect of red meat consumption,16 and occupational imaging technologies, and minimally invasive techniques.
exposure to carcinogens,17 but the data are unclear.

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 Diagnosis


both forms are generally associated with structural With the expansion of routine imaging for many disorders,
alterations of the short arm of chromosome 3.21 patients with renal cell carcinoma are increasingly being
Translocation tumours are increasingly being identified identified by chance. Only 30% of patients are diagnosed
in adolescents and young adults.18 Many gene variants on the basis of symptoms. At all stages, renal cell
have been associated with renal cell carcinoma. Somatic carcinoma might produce multiple hormone-like or
mutations or epigenetic alterations of VHL, a tumour- cytokine-like biologically active products that lead to
suppressor gene, are seen in more than 80% of clear-cell clinically important paraneoplastic syndromes (table 1).31
subtypes. Specifically, the VHL tumour suppressor The findings of routine diagnostic blood tests might be
protein is important in cellular oxygen sensing by altered by renal cell carcinoma, but no change is
targeting hypoxia-inducible factors for ubiquitination pathognomonic. Physical examination has a limited role
and proteasomal degradation.22 Sequencing studies have in diagnosis of renal cell carcinoma, but the presence of
identified other driver genes that are involved in the a palpable abdominal mass, new-onset varicocele, or
pathogenesis of renal cell carcinoma, including PBRM1,23 lower extremity oedema should always prompt additional
BAP1,23 SETD2,24 TCEB1,24 and KDM5C.25 Studies imaging assessments for retroperitoneal neoplasia.
analysing the effects of these mutations on clinical
outcomes, however, are limited by small sample sizes, Standard imaging
short-term follow-up, and lack of assessment of multiple Although renal cell carcinoma is frequently detected by
markers in the same cohort.24,25 Hereditary forms of renal abdominal ultrasound scanning, limitations in specificity
cell carcinoma include von Hippel-Lindau syndrome and accuracy make it necessary to use CT or MRI to
(VHL 3p25–26),26 hereditary papillary renal cell confirm suspicious findings. The main goals of imaging
carcinoma (MET 7q31–34),27 Birt-Hogg-Dubé syndrome are to characterise the mass and possible abdominal
(FLCN 17p11),28 hereditary leiomyomatosis (FH 1q42–43),29 metastases, tumour extension, and venous involvement
and tuberous sclerosis (TSC1 9q34 or TSC2 16p13).30 for staging (appendix). Imaging might also be useful to

Occurrence among Proposed causes


patients with
symptomatic RCC
Local symptoms
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
Paraneoplastic disorders
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
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 Less frequent Non-specific hepatitis associated with a prominent lymphocytic infiltrate, raised
absence of liver metastases (Stauffer’s concentrations of interleukin-6 in serum
syndrome)
Hypercalcaemia Less frequent Osteolytic metastatic involvement of bone, production of parathyroid-hormone-
like 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, Anecdotal Abnormal production of 1,25-dihydroxycholecalciferol, renin, erythropoietin,
amyloidosis, vascular thrombosis, Cushing’s prostaglandins, parathyroid-hormone-like peptides, lupus-type anticoagulant,
syndrome, protein enteropathy, human chorionic gonadotropin, insulin, various cytokines or inflammatory
galactorrhoea, gynaecomastia, decreased mediators
libido, hirsutism, amenorrhoea, chills,
necrotising myopathy, immune
thrombocytopenic purpura

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 tumours, detection of small renal lesions44 and
if active management is planned (eg, bilateral renal accumulating evidence that surgically induced chronic
function, renal vascular supply, retroperitoneal anatomy). kidney disease can increase patients’ morbidity45 have led
If malignant renal cell carcinoma is suspected, additional to more conservative approaches.46 Specifically, nephron-
imaging (eg, thoracic and brain CT scan, total-body bone sparing surgery, active surveillance, and minimally
scan) can be considered in symptomatic patients or in invasive techniques46 have been introduced into daily
cases of bulky abdominal disease.32–35 clinical practice. These approaches limit invasiveness,
iatrogenic renal function impairment, and overtreatment.
New imaging technologies
New technologies for cancer detection and character- Active surveillance
isation are being investigated for renal cell carcinoma; for Management of localised renal cell carcinoma must take
example, advanced MRI techniques, such as diffusion- into account competing causes of mortality, particularly
weighted and perfusion-weighted imaging, are being in elderly patients.47 This consideration has led to the
explored for the assessment of renal masses.36 Although increasing use of active surveillance46 to avoid surgically
PET might be useful for the diagnosis and follow-up of induced short-term and medium-term complications.
renal cell carcinoma, this technique is not yet part of the The rationale for active surveillance is that roughly 20%
standard strategy.37 Some studies have suggested the use of small renal masses harbour benign final pathology
of combined ¹²⁴iodine PET and CT for the detection of and the median growth rate is usually slow
labelled antibodies to the carbonic anhydrase IX antigen, (2–3 mm/year).48 Moreover, the risk of metastatic
which is widely expressed in more than 90% of clear-cell progression is very low (less than 1%), with virtually no
renal cell carcinomas but not in the normal kidney.38,39 risk of cancer-specific mortality in well selected
patients.49–51 Biopsy might be useful before active
Emerging role of renal biopsy surveillance is started. Active surveillance has been
The use of renal biopsy has been limited by concerns proposed for larger tumours to determine tumour
about accuracy and safety, and in the past has had little growth kinetics, especially if a patient has severe
effect on treatment decisions because the standard competing risks and limited life expectancy.52 Absolute
treatment for solid renal masses was upfront removal.40 cutoffs for tumour size and growth rate that should
Increased active surveillance for small renal masses and prompt intervention during active surveillance are not
individualised targeted therapy for patients with well defined. In various proposed protocols, imaging at
metastases, however, have pushed the scientific 3 and 6 months initially, every 6 months during the next
community to improve expertise in biopsy performance 2–3 years, and annually thereafter is suggested, but
and pathological interpretation.41 Consequently, renal cumulative radiation risk and increased costs are being
biopsy is now used to establish the diagnosis of assessed.49,50,53 Data are needed from large cohorts with
radiologically indeterminate renal masses, obtain long-term follow-up to better characterise clinical
histology of incidentally detected renal masses in patients indications and standardise protocols.
who are potential candidates for active surveillance, and
to select the most suitable targeted therapy for patients Minimally invasive techniques
with metastatic renal tumours.42 Although surgery still represents the standard of care for
Accuracy of biopsy in detecting malignancy ranges from renal cell carcinoma, the use of minimally invasive
38% to 100%.41 In one of the largest study cohorts reported techniques to treat incidentally detected small tumours
(n=345), histological subtype and Fuhrman grade were has been increasing. Cryotherapy and radiofrequency
correctly assessed at biopsy in 88% and 64% of cases, ablation (RFA) were initially contemplated only for
respectively.43 Renal biopsy has low associated morbidity. patients with one kidney or for those deemed unfit to
Contemporary series have revealed overall complication undergo a major procedure. Since preliminary reports
rates (mainly bleeding, infection, and arteriovenous showed acceptable cancer control,37,54–57 the clinical
fistula) in 0·3–5·3% of cases. Tumour seeding throughout indications for these procedures has been extending. For
the biopsy tract, which had been reported anecdotally in example, 8% of US patients are treated with cryotherapy
the past, is no longer a risk with modern biopsy and RFA, compared with 4% in 1998.58
techniques.42 Although renal biopsy has a definite and Renal ablative treatment uses the cell-destroying
expanding role in the assessment and treatment of renal properties of temperature (hot or cold) to cause apoptosis
masses, it remains substantially underused.40 in cancer cells.54 RFA induces thermal damage (50–120°C)
through frictional heating resulting from ionic oscillation
Management by a high-frequency alternating current (375–500 kHz).59
Notwithstanding advances in the understanding of renal Cryotherapy stimulates tumour cell death directly by
cell carcinoma biology, surgery remains the mainstay of damaging cell membranes and organelles and indirectly
curative treatment. Although radical nephrectomy was by initiating vascular compromise through thrombosis
historically the standard of care for management of renal 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 Although many retrospective reports have suggested a
cryotherapy are renal bleeding or abscess formation or benefit in overall survival for patients treated with
extrarenal effects in the bowel, pleura, spleen, pancreas, nephron-sparing surgery relative to radical
and vasculature. nephrectomy,67,69 no improvements with nephron-sparing
Most studies of outcomes after renal tumour techniques were found in a comparison of the two
cryoablation consist of single-institution retrospective techniques.70 However, many flaws undermined the study
reports with low numbers of patients and short follow-up. (eg, low recruitment, selection and verification biases,
An analysis of the Mayo Clinic Renal Tumor Registry that and being done at the beginning of the nephron-sparing
included more than 1800 patients with primary cT1N0M0 learning curve).70 Further efforts are needed to improve
renal tumours showed similar local recurrence-free understanding of which patients might benefit from
survival for patients treated with nephron-sparing nephron-sparing surgery in terms of overall survival.
surgery, RFA, or cryotherapy (p=0·4). By contrast, International guidelines include standard indications
metastases-free survival was significantly better after for nephron-sparing surgery: absolute, indicating
nephron-sparing surgery (p=0·005) and cryoablation patients with only one anatomical or functional kidney;
(p=0·02) than with RFA.60 Although this comparison relative, indicating patients with a functioning opposite
included a large number of cases, its retrospective nature, kidney that is affected by a disorder that might impair
the differing baseline characteristics between studies, and renal function in the future or with hereditary forms of
other unmeasurable confounders make it difficult to renal cell carcinoma who are at increased risk of
draw conclusions about cancer control.61 A meta-analysis developing a tumour in the contralateral kidney; and
by Kunkle and colleagues56 suggested that recurrence-free elective, indicating localised unilateral renal cell
survival might be worse for RFA and cryoablation than carcinoma with a healthy contralateral kidney.37,58
for partial nephrectomy, but showed no differences for Although nephron-sparing surgery has been proposed
metastasis-free survival. Conversely, a systematic review for use even for locally advanced disease and metastatic
and meta-analysis of six clinical trials (one prospective spread,71,72 further data are required before a conservative
randomised and five retrospective cohort trials) showed approach can be approved in those settings.
that recurrence-free survival and cancer-specific survival Surgical excision of tumours is done by kidney
were similar for patients treated with surgery or RFA, mobilisation, resection of the tumour (with or without a
with less postoperative decline in estimated glomerular rim of normal parenchyma, according to the anatomical
filtration rate in the RFA group.55 The overall complication and tumour features), and final reconstruction
rate was significantly lower in the RFA group (7% vs 11%; (renorrhaphy; figure 1). During resection of the tumour,
relative risk 0·55, 95% CI 0·31–0·97, p=0·04). The groups the main renal artery can be clamped temporarily (hilar
had similar local recurrence rates of 3–4% (0·92, clamping) to minimise blood loss. Sustained hilar
0·4–2·14, p=0·8) and disease-free survival (hazard ratio clamping might cause impairment of renal function and,
1·04, 95% CI 0·48–2·24, p=0·9).55 therefore, limited duration of ischaemia is suggested,
although the functional long-term consequences are
Surgery controversial.73–75 If technically possible, techniques
Nephron-sparing surgery without clamping or selective intraparenchymal
Although radical nephrectomy involves removal of the clamping (eg, clamping of only a branch of the main
entire kidney, with nephron-sparing surgery only the artery) should be considered.76
tumour is excised and as much of the normal renal Nephron-sparing surgery should be preferred unless
parenchyma as possible is maintained (figure 1). Although pre-existing disorders are likely to decrease the positive
initially reserved for imperative situations, according to effect of a conservative approach, such as frailty or short
international guidelines, nephron-sparing surgery is now life expectancy. Other risk factors are low surgical
the treatment of choice whenever a healthy part of kidney expertise or volume of relevant surgeries, excessive
can safely be spared. Nephron-sparing surgery has been ischaemia time, use of non-conservative haemostasis
used increasingly since observations suggested techniques, or large or anatomically complex renal
oncological control similar to radical nephrectomy62–64 but masses with a low percentage of nephrons that can be
with the additional benefit of renal preservation.65 spared. In such situations, the potential benefit in
Moreover, a long-term protective effect has been functional outcomes might be jeopardised. Conservative
suggested for nephron-sparing surgery relative to radical surgical techniques require substantial technical
nephrectomy for risk of cardiovascular events after expertise and might be associated with increased risk of
surgery (eg, new-onset hypertension, coronary artery haemorrhage (3%) and urinary leakage (4%).77
disease, vasculopathy, and cerebrovascular disease).45,66,67 Consequently, nephron-sparing surgery is still underused
Because patients with organ-confined renal cell carcinoma for patients with renal cell carcinoma, especially in non-
that has been surgically treated are usually long-term academic hospitals.78–80
cancer survivors (85–96% cancer-specific survival 10 years If the aim of the surgery is the preservation of the
after surgery68), these functional outcomes are important. normal renal parenchyma, the choice of surgical approach

4 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 nephron-sparing surgery, which was the standard
characteristics and the surgeon’s expertise. Tumour approach until the early 2000s, is now used only in cases
features that dictate the feasibility of nephron-sparing where there are anatomical difficulties (eg, urinary tract
surgery versus radical nephrectomy include diameter, invasion or hilar or larger tumours) or for which
location, depth, and proximity to hilar vessels and the minimally invasive techniques are unavailable.
urinary collecting system. The three foremost anatomical A meta-analysis of non-randomised studies including
scores (PADUA,81 RENAL,82 and C-index83) may be used to 2240 patients showed that robot-assisted compared with
assess those features before surgery, with the aim of laparoscopic nephron-sparing surgery was associated
improving selection of patients, surgical management, with reduced rates of conversion to open surgery (relative
research reporting, and outcome prediction.81–83 risk 0·4, 95% CI 0·2–0·9, p=0·02) and radical surgery
Robot-assisted nephron-sparing surgery is gaining (0·19, 0·1–0·5, p=0·0006).85 Additionally, robot-assisted
popularity because challenging cases can be approached nephron-sparing surgery resulted in significantly shorter
with a minimally invasive procedure (figure 1). In warm ischaemia, (weighted mean difference 3 min,
hospitals that have acquired surgical robotic systems, 95% CI 1–5, p=0·005), although the clinical relevance of
treatment with nephron-sparing surgery has increased this outcome was marginal, smaller change in estimated
by 16–35% relative to hospitals without such technology.84 glomerular filtration rate (standardised mean difference
Conversely, laparoscopic nephron-sparing surgery might 0·2, 0·02–0·3, p=0·03), and shorter length of stay in
be challenging and is usually limited to simpler cases or hospital (weighted mean difference 0·2 days, 0·1–0·4,
done by experienced surgeons (appendix). Open 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

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 intermediate-
risk 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
Figure 2: CT scan of a patient aged 55 years with a bulky right kidney tumour and caval thrombus invading dictated primarily by the extent of the thrombus. When
the right atrium confined to the renal vein, minimum modification of the
Images were taken before nephrectomy and venous thrombectomy, and are two slices from the same scan. Initial
standard surgical approach is required. As the thrombus
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 extends cranially, opening of the vena cava, liver
dyspnoea were secondary to initial heart failure and abnormal production of prostaglandins, cytokines, and mobilisation, opening of the right atrium, and
inflammatory mediators by the tumour. cardiopulmonary bypass might be necessary.

operative outcomes, including complications with Metastatic disease at diagnosis


Clavien-Dindo classification grades 1–2 (relative risk 1·1, Historically, patients with metastatic clear-cell renal
95% CI 0·9–1·3, p=06) and 3–5 (0·9, 0·6–1·5, p=0·8).85 cell carcinoma were treated with systemic therapy based
on immune modulators, mainly interferon α and
Radical nephrectomy interleukin-2, but outcomes were only slightly improved.96
If sparing the parenchyma is not possible and the aim of In a few cases these agents have led to T-cell-mediated
the surgery is removal of the entire kidney, open radical tumour regression secondary to enhancement in
nephrectomy has been replaced in most cases by lymphocyte mitogenesis, lymphocyte cytotoxicity, and
laparoscopic nephrectomy, which is associated with activity of lymphokine-activated and natural killer cells.97
shorter stay in hospital, less perioperative blood loss, Immunotherapy with high-dose interleukin 2 has been
fewer analgesic requirements, and shorter convalescence associated with durable complete response in less than
time.86,87 Although randomised studies assessing 10% of patients and has been associated with severe
oncological outcomes have never been done, similar treatment-related toxic effects.97 Subsequently, combined
oncological outcomes for laparoscopic and open radical data from 99 study groups show an overall chance of
nephrectomy are suggested.86,87 Robot-assisted radical partial or complete remission in only 13% of patients,
nephrectomy results in increased medical expense compared with 3–4% in placebo or non-immunotherapy
without improving morbidity compared with laparoscopic control groups.98 Aldesleukin and interferon α (associated
procedures,88 but might still be worth considering for with bevacizumab) remain the only immune-modulating
selected cases (eg, when retroperitoneal lymphadenectomy drugs approved for selected cases of metastatic renal cell
is thought to be required). carcinoma.57
Several non-randomised studies had suggested a
Lymph node dissection, adrenalectomy, and venous response to systemic therapy and improved survival in
thrombectomy patients who had undergone nephrectomy and, therefore,
Adrenalectomy was historically viewed as an unavoidable two randomised clinical trials were planned at the end of
part of radical procedures. Removal of the ipsilateral 1980s to test the effect of surgery on survival in patients
adrenal gland, however, is now optional if no macroscopic with metastatic renal cell carcinoma. 331 patients overall
invasion is suggested by preoperative imaging or found were randomly assigned treatment with nephrectomy
during surgery.37,57,89 plus interferon α-2b or interferon α-2b alone.99,100 A
Irrespective of T stage, patients who have renal cell combined analysis of the findings showed a 31% decrease
carcinoma with nodal invasion (pN1) have poor cancer- in the risk of death with surgery compared with
specific survival (20–30% at 3 years after surgery).90 interferon α-2b alone (median survival 13·6 months,
Although lymphadenectomy provides the most accurate 95% CI 9·7–17·4 vs 7·8 months, 5·9–9·7).101 Several

6 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 Administration for management of renal cell carcinoma
of nephrectomy: reduction of disease burden; decrease of (table 2, figure 3). Sunitinib and sorafenib were associated
immunosuppression and improved clinical response to significant benefits in progression-free survival and
cytokines; decreased growth-factor secretion (mainly overall survival compared with placebo in two phase 3
VEGF); and inhibited tumour microenvironment trials,103,104 as was, temsirolimus in high-risk patients
secondary to surgically induced chronic metabolic when compared with IFN monotherapy.110 In contrast, in
acidosis.102 patients with metastatic renal cell carcinoma who
Over the past decade, the main advance in the progressed after treatment with sunitinib, overall survival
treatment of metastatic renal cell carcinoma has been the was shorter with temsirolimus than with sorafenib.111
development of treatments that affect specific targets in Everolimus was associated with a significant benefit in
biological pathways. Agents targeting the VEGF/PDGFR/ progression-free survival relative to placebo in patients
mTOR pathway continue to be the mainstays of with metastatic renal cell carcinoma who progressed
treatment (table 2). Although improved target specificity after sunitinib or sorafenib.112 Everolimus has been
has lessened the risk of toxic effects associated with these approved by the US Food and Drug Administration as
drugs, durable complete responses remain the exception. the first drug to be used specifically to treat non-
Seven targeted drugs have been approved by European cancerous kidney tumours (renal angiomyolipomas) in
Medicines Agency and the US Food and Drug patients with tuberous sclerosis complex.30

Mechanism Administration route Setting Treatment Main findings


Sunitinib PDGFR and VEGFR Capsules Treatment naive Sunitinib vs interferon α PFS, 11 months (95% CI 10–12) vs 5 months (95% CI 4–6),
inhibitor (n=750) HR 0·42 (95% CI 0·32–0·54), p<0·001103
Sorafenib PDGFR, VEGFR-2, and Capsules Second-line therapy Sorafenib vs placebo PFS, 5·5 vs 2·8 months, HR 0·44 (95% CI 0·35–0·55),
MAPK/MEK/ (n=903) p<0·00001;104 OS, 17·8 vs 15·2 months, HR 0·74 (95% CI
ERK inhibitor 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 Intravenous injection Treatment naive Bevacizumab plus interferon PFS, 10·2 vs 5·4 months, HR 0·63 (95% CI 0·52–0·75), p<0·001;105
humanised (n=649) vs interferon plus placebo OS, 23·3 vs 21·3 months, HR 0·91 (95% CI 0·76–1·10), p=0·33106†
monoclonal antibody
directed against
VEGF-A
Treatment naive 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
(n=732) 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
Temsirolimus mTOR inhibitor Intravenous injection Treatment naive Temsirolimus plus PFS, 9·1 vs 9·3 months, HR 1·1 (95% CI 0·9–1·3), p=0·8;109
(791) bevacizumab vs interferon OS, 25·8 vs 25·5 months, HR 1·0, p=0·6109
plus bevacizumab
Treatment naive Temsirolimus vs interferon PFS, 3·8 vs 1·9 months, p<0·0001; OS, 10·9 vs 7·3 months,
(poor-risk, n=626) HR 0·73 (95% CI 0·58–0·92), p=0·008110
Second line after Temsirolimus vs sorafenib PFS, 4·3 months (95% CI 4·0–5·4) vs 3·9 months
sunitinib (n=512) (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
Everolimus mTOR inhibitor Capsules Second-line therapy Everolimus vs placebo PFS, 4·0 months (95% CI 3·7–5·5) vs 1·9 months (95% CI
(n=410) 1·8–1·9), HR 0·30 (95% CI 0·22–0·40), p<0·0001112
Pazopanib VEGFR/PDGFR/c-Kit Capsules Treatment naive Pazopanib vs placebo PFS, 11·1 vs 2·8 months, HR 0·40 (95% CI 0·27–0·60),
inhibitor (n=233) 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 Pazopanib vs sunitinib PFS, 8·4 months (95% CI 8·3–10·9) vs 9·5 months (95% CI
(n=1110) 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 Pazopanib vs placebo PFS, 7·4 vs 4·2 months, HR 0·54 (95% CI 0·35–0·84), p<0·0001;113
(n=202) 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
Axitinib VEGFR (1–3), PDGFR, Capsules Treatment naive Axitinib vs sorafenib PFS, 10·1 month (95% CI 7·2–12·1) vs 6·5 months (95% CI
and c-Kit inhibitors with mRCC (n=288) 4·7–8·3), HR 0·77 (95% CI 0·56–1·05), p=0·038117
Second-line therapy Axitinib vs sorafenib PFS, 6·7 months (95% CI 6·3–8·6) vs 4·7 months
for mRCC (n=723) (95% CI 4·6–5·6), HR 0·66 (95% CI 0·54–0·81), p<0·0001118

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

Immune modulation Blood vessel

Interferon α
NK cell T cell

Sunitinib
PDGFR Sorafenib
CTLA4 Pazopanib
Aldesleukin

Angiogenesis
PD-1
(Ipilimumab))
(Rilotumumab) Axitinib
(Nivolumab)
FGF
PDGF
Paracrine
PD-L1 growth
HGF FGFR
stimulation
VEGFR
(Brivanib)
MET/HGFR (Dovitinib Temsirolimus VEGF
AKT Everolimus
(Erlotinib)
Bevacizumab
PI3K mTORC1–2
PTEN
EGF
HIF1α Autocrine
growth stimulation
Nucleus
EGFR

VHL
MAPK/ERK TGFα
pathway Gene expression,
cell growth, proliferation, survival

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/ Bevacizumab is a recombinant humanised monoclonal


PDGFR/c-Kit pathway that has shown a significant benefit antibody directed against VEGF. In two phase 3 trials of
in progression-free survival compared with placebo in bevacizumab plus interferon compared with interferon
treatment-naive patients (table 2).113 The difference in final plus placebo105 or interferon alone,106 bevacizumab was
overall survival between patients treated with pazopanib superior in terms of progression-free survival. One of the
or placebo was not significant, partly because of early and studies was unblinded because a clinically meaningful
frequent crossover from placebo to pazopanib.113 In a significant benefit was found in an interim analysis.105 In
phase 3 trial, similar efficacy was found with pazopanib the final overall survival analyses of both trials, however,
and sunitinib, with slightly lower incidence of toxic effects no significant differences were found between treatment
(palmar-plantar erythrodysaesthesia and fatigue) but with groups (table 2), probably because of the confounding
higher incidence of liver function test abnormalities.115,116 A effect of crossover after the interim analysis and the use
randomised crossover trial revealed a significant with other targeted drugs as salvage therapies in the
preference for pazopanib over sunitinib by patients and interferon groups. 107,108
physicians (70% vs 22%, p<0·001),119 mainly because of a The introduction of targeted therapies has set a new
better adverse event profile for fatigue, improved quality benchmark for the treatment of metastatic renal cell
of life,119 and reduced cost.120 carcinoma, but most patients included in trials have had
Axitinib is a potent, selective, second-generation clear-cell metastatic disease. The results, therefore, might
inhibitor of VEGFR 1, 2, and 3. In patients with treatment- not be universally applicable to all histological types. A
naive metastatic renal cell carcinoma, progression-free contemporary analysis of patients with metastatic renal
survival was not significantly increased with axitinib cell carcinoma treated with VEGF-targeting agents showed
compared with sorafenib (table 2), although adverse median overall survival of 18·8 months (95% CI 17·6–21·4)
events of any grade were less common in the axitinib with a remarkable peak at 43·2 months for patients in the
group by an average of 10%.117 Conversely, among patients most favourable risk group (appendix).121 The most
treated with a cytokine-containing regimen before informative clinical variables to predict the outlook of
axitinib, progression-free survival was longer than in patients with metastatic renal cell carcinoma treated with
those who received sorafenib (table 2).118 targeted therapies include haemoglobin (p<0·0001),

8 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 the use of immune checkpoint inhibitors. These
performance status (p<0·0001), time from initial diagnosis treatments work by targeting molecules that serve as
to the start of treatment (p=0·009), absolute neutrophil checks and balances on immune responses. By blocking
count (p<0·0001), and platelet count (p=0·012).122 inhibitory molecules or activating stimulatory molecules,
The most common treatment-related adverse events it is hoped that pre-existing anticancer immune
associated with targeted therapies for metastatic renal responses will be restored. Nivolumab, for instance, is a
cell carcinoma are fatigue, hypertension, nausea, fully human IgG4 monoclonal antibody that binds to the
diarrhoea, dysphonia, and palmar-plantar erythro- PD-1 receptor and restores T-cell immune activity. This
dysaesthesia.123 Grade 3 or 4 adverse events are expected drug has shown promising results in the treatment of
in around a third of patients, with profiles improving metastatic renal cell carcinoma.127 Many other new drugs
slightly for the new-generation targeted therapies (eg, are being investigated, and tivozanib128,129 dovitinib,130
pazopanib).123 Clinical and translational studies are cabozantinib,131 erlotinib,132 and sirolimus133 are being
needed to identify the phenotypic predictors of response tested as potential treatments (figure 3). Additionally,
for each of the drugs. two phase 3 studies are assessing the role of cytoreductive
nephrectomy in the context of novel systemic therapies
Controversies, uncertainties, and research and the best treatment sequencing for surgery and
questions targeted drugs (NCT01099423).134 The role of adjuvant
Despite progress in the understanding of renal cell therapy after surgery is also being investigated. Past
carcinoma, uncertainties, controversies, and research attempts to prove clinical benefit with adjuvant
questions remain. Advances are expected over the next immunotherapy (interferon α and interleukin-2) system-
few years from translational and clinical studies. Novel atically failed.135–137 Five phase 3 trials including more
tissue and genetic markers are expected to improve than 6000 patients with non-metastatic renal cell
accuracy of renal biopsy in characterising histology and carcinoma are being done to assess the role of targeted
disease aggressiveness at diagnosis (indolent mass vs therapies in the adjuvant setting,138 no benefit has yet
clinically relevant malignancy). This approach could be been found with adjuvant VEGF inhibitors (sorafenib or
crucial to the selection of patients who will benefit from sunitinib) in patients at high risk of recurrence
active surveillance and other non-surgical approaches. (NCT00326898).
Data are too immature to validate universal use of Surgical metastasectomy has been gaining popularity
minimally invasive techniques (cryotherapy and RFA) for as part of a multimodal approach to treating patients
renal cell carcinoma. Only case series, unmatched with oligometastatic renal cell carcinoma and has shown
retrospective studies, and chart reviews with short follow- acceptable morbidity in selected patients.139–141 Partial or
up are available. Longer follow-up data and well designed complete removal of the metastatic lesions was associated
comparative trials are needed to confirm oncological with improved overall survival, especially in patients with
results, standardise techniques, and to recommend adequate performance and functional statuses.139–142
alternatives to surgery, especially for small renal masses. Patients are being recruited into randomised clinical
In terms of the potential use of radiotherapy as part of trials to assess the efficacy of a multimodal approach
a multimodal approach, renal cell carcinoma had been based on targeted therapy after surgical metastasectomy
viewed as a radioresistant tumour because of poor (NCT00918775 and NCT01444807).
outcomes with low-dose radiotherapy.124 This notion has
been challenged by emerging evidence that stereotactic Conclusions
body radiotherapy with a high-fraction dose has an effect Renal cell carcinoma provides one of the best examples
in primary tumours and oligometastatic disease.124 in oncology of how innovative basic research, novel
Randomised trials of radiotherapy regimens and clinical findings, and improved surgical techniques can
radiotherapy combined with targeted drugs are awaited converge to improve the care of patients. Alternative
to confirm the potential clinical implications. management strategies, such as active surveillance or
Medical and integrated therapies, identification of new cryotherapy, can be considered for an increasing number
target pathways, and optimum sequencing and of patients. For patients who are candidates for surgical
combination of existing targeted agents are areas of excision, the development of minimally invasive
active study.125 Molecular markers that seem to be techniques, such as robot-assisted laparoscopic nephron-
predictive of response to sunitinib in patients with sparing surgery, has decreased the risk of surgically
metastatic renal cell carcinoma have been identified. induced functional impairment. Additionally, extended
Prediction is based on molecular subtypes and suggests understanding of biological pathways and novel targeted
that tailored and personalised treatment is feasible.126 therapies has led to improved survival outcomes in
Upcoming results of ongoing trials testing novel drugs patients with metastatic disease. Further developments
will increase the choice for urologists and oncologists in translational research and findings from trials are
treating locally advanced and metastatic disease. A expected to integrate surgical and systemic treatments to
promising avenue of clinical research, for example, is 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 21 Srinivasan R, Ricketts CJ, Sourbier C, Linehan WM. New strategies


Both authors did the literature search, wrote the drafts, and reviewed the in renal cell carcinoma: targeting the genetic and metabolic basis of
contents of the paper. disease. Clin Cancer Res 2015; 21: 10–17.
22 Moore LE, Nickerson ML, Brennan P, et al. Von Hippel-Lindau
Declaration of interests (VHL) inactivation in sporadic clear cell renal cancer: associations
UC has received consultancy and lecturer honoraria and FM has with germline VHL polymorphisms and etiologic risk factors.
received research support, consultancy fees, and lecturer honoraria from PLoS Genet 2011; 7: e1002312.
Recordati. 23 Brugarolas J. PBRM1 and BAP1 as novel targets for renal cell
Acknowledgments carcinoma. Cancer J 2013; 19: 324–32.
We thank Jeni Crockett-Holme for editorial support and Nicola Spreafico 24 Cancer Genome Atlas Research Network. Comprehensive molecular
for graphics support. characterization of clear cell renal cell carcinoma. Nature 2013;
499: 43–49.
References 25 Ibragimova I, Maradeo ME, Dulaimi E, Cairns P. Aberrant
1 Ellimoottil C, Greco KA, Hart S, et al. New modalities for evaluation promoter hypermethylation of PBRM1, BAP1, SETD2, KDM6A and
and surveillance of complex renal cysts. J Urol 2014; 192: 1604–11. other chromatin-modifying genes is absent or rare in clear cell
2 Shah S, Watnick T, Atta MG. Not all renal cysts are created equal. RCC. Epigenetics 2013; 8: 486–93.
Lancet 2010; 376: 1024. 26 Gossage L, Eisen T, Maher ER. VHL, the story of a tumour
3 Siegel R, Ma J, Zou Z, Jemal A. Cancer statistics, 2014. suppressor gene. Nat Rev Cancer 2015; 15: 55–64.
CA Cancer J Clin 2014; 64: 9–29. 27 Dharmawardana PG, Giubellino A, Bottaro DP. Hereditary papillary
4 Ferlay JSI, Ervik M, Dikshit R, et al. GLOBOCAN 2012 v1.0, cancer renal carcinoma type I. Curr Mol Med 2004; 4: 855–68.
incidence and mortality worldwide: IARC CancerBase No. 11. 2013. 28 Perdeaux E, Solly J. Birt-Hogg-Dubé syndrome. JAMA 2013;
http://globocan.iarc.fr (accessed Jan 1, 2015). 309: 1460.
5 Levi F, Ferlay J, Galeone C, et al. The changing pattern of kidney 29 Shuch B, Ricketts CJ, Vocke CD, et al. Adrenal nodular hyperplasia
cancer incidence and mortality in Europe. BJU Int 2008; in hereditary leiomyomatosis and renal cell cancer. J Urol 2013;
101: 949–58. 189: 430–35.
6 Chow WH, Linehan WM, Devesa SS. Re: rising incidence of small 30 Sooriakumaran P, Anderson CJ. Everolimus for renal
renal masses: a need to reassess treatment effect. angiomyolipoma in tuberous sclerosis. Lancet 2013; 381: 783–85.
J Natl Cancer Inst 2007; 99: 569–70. 31 Sacco E, Pinto F, Sasso F, et al. Paraneoplastic syndromes in
7 Hollingsworth JM, Miller DC, Daignault S, Hollenbeck BK. Rising patients with urological malignancies. Urol Int 2009; 83: 1–11.
incidence of small renal masses: a need to reassess treatment effect. 32 Hofbauer SL, de Martino M, Seemann C, et al. Associations
J Natl Cancer Inst 2006; 98: 1331–34. between presenting symptoms, clinicopathological parameters, and
8 Welch HG, Schwartz LM, Woloshin S. Are increasing 5-year prognosis in a contemporary series of patients with renal cell
survival rates evidence of success against cancer? JAMA 2000; carcinoma. Korean J Urol 2014; 55: 505–10.
283: 2975–78. 33 Ljungberg BBK, Bex A, Canfield S, et al. Guidelines on renal cell
9 Theis RP, Dolwick Grieb SM, Burr D, Siddiqui T, Asal NR. carcinoma, update. April 2014. http://uroweb.org/wp-content/
Smoking, environmental tobacco smoke, and risk of renal cell uploads/10-Renal-Cell-Carcinoma_LR.pdf (accessed July 15, 2015).
cancer: a population-based case-control study. BMC Cancer 2008; 34 National Comprehensive Cancer Network. NCCN Clinical Practice
8: 387. Guidelines in Oncology: Kidney Cancer 2. 2014. http://www.nccn.
10 Deckers IA, van den Brandt PA, van Engeland M, et al. org/professionals/physician_gls/pdf/kidney.pdf (accessed
Polymorphisms in genes of the renin-angiotensin-aldosterone July 15, 2015).
system and renal cell cancer risk: interplay with hypertension and 35 American Urological Association. Follow-up for clinically localized
intakes of sodium, potassium and fluid. Int J Cancer 2015; renal neoplasms: AUA guideline 2013. April 2013. https://www.
136: 1104–16. auanet.org/common/pdf/education/clinical-guidance/Renal-
11 Zucchetto A, Dal Maso L, Tavani A, et al. History of treated Cancer-Followup.pdf (accessed July 15, 2015).
hypertension and diabetes mellitus and risk of renal cell cancer. 36 Hilton S, Jones LP. Recent advances in imaging cancer of the
Ann Oncol 2007; 18: 596–600. kidney and urinary tract. Surg Oncol Clin N Am 2014; 23: 863–910.
12 Corrao G, Scotti L, Bagnardi V, Sega R. Hypertension, 37 Ljungberg B, Bensalah K, Canfield S, et al. EAU guidelines on renal
antihypertensive therapy and renal-cell cancer: a meta-analysis. cell carcinoma: 2014 update. Eur Urol 2015; 67: 913–24.
Curr Drug Saf 2007; 2: 125–33.
38 Muselaers CH, Boerman OC, Oosterwijk E, Langenhuijsen JF,
13 Gati A, Kouidhi S, Marrakchi R, et al. Obesity and renal cancer: Role Oyen WJ, Mulders PF. Indium-111-labeled girentuximab
of adipokines in the tumor-immune system conflict. immunoSPECT as a diagnostic tool in clear cell renal cell
Oncoimmunology 2014; 3: e27810. carcinoma. Eur Urol 2013; 63: 1101–06.
14 Bergström A, Hsieh CC, Lindblad P, Lu CM, Cook NR, Wolk A. 39 Divgi CR, Uzzo RG, Gatsonis C, et al. Positron emission
Obesity and renal cell cancer—a quantitative review. tomography/computed tomography identification of clear cell renal
Br J Cancer 2001; 85: 984–90. cell carcinoma: results from the REDECT trial. J Clin Oncol 2013;
15 Karami S, Daugherty SE, Purdue MP. A prospective study of 31: 187–94.
alcohol consumption and renal cell carcinoma risk. 40 Tomaszewski JJ, Uzzo RG, Smaldone MC. Heterogeneity and renal
Int J Cancer 2015; 137: 238–42. mass biopsy: a review of its role and reliability.
16 Rohrmann S, Linseisen J, Overvad K, et al. Meat and fish Cancer Biol Med 2014; 11: 162–72.
consumption and the risk of renal cell carcinoma in the European 41 Ball MW, Bezerra SM, Gorin MA, et al. Grade heterogeneity in
prospective investigation into cancer and nutrition. small renal masses: potential implications for renal mass biopsy.
Int J Cancer 2015; 136: E423–31. J Urol 2015; 193: 36–40.
17 Ljungberg B, Campbell SC, Choi HY, et al. The epidemiology of 42 Volpe A, Finelli A, Gill IS, et al. Rationale for percutaneous biopsy
renal cell carcinoma. Eur Urol 2011; 60: 615–21. and histologic characterisation of renal tumours. Eur Urol 2012;
18 Shuch B, Amin A, Armstrong AJ, et al. Understanding pathologic 62: 491–504.
variants of renal cell carcinoma: distilling therapeutic opportunities 43 Leveridge MJ, Finelli A, Kachura JR, et al. Outcomes of small renal
from biologic complexity. Eur Urol 2015; 67: 85–97. mass needle core biopsy, nondiagnostic percutaneous biopsy, and
19 Srigley JR, Delahunt B, Eble JN, et al, and the ISUP Renal Tumor the role of repeat biopsy. Eur Urol 2011; 60: 578–84.
Panel. The International Society of Urological Pathology (ISUP) 44 Baillargeon-Gagné S, Jeldres C, Lughezzani G, et al. A comparative
Vancouver classification of renal neoplasia. Am J Surg Pathol 2013; population-based analysis of the rate of partial vs radical
37: 1469–89. nephrectomy for clinically localized renal cell carcinoma.
20 Bianchi M, Sun M, Jeldres C, et al. Distribution of metastatic sites BJU Int 2010; 105: 359–64.
in renal cell carcinoma: a population-based analysis.
Ann Oncol 2012; 23: 973–80.

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


Seminar

45 Capitanio U, Terrone C, Antonelli A, et al. Nephron-sparing 67 Huang WC, Elkin EB, Levey AS, Jang TL, Russo P. Partial
techniques independently decrease the risk of cardiovascular events nephrectomy versus radical nephrectomy in patients with small
relative to radical nephrectomy in patients with a T1a-T1b renal mass renal tumors—is there a difference in mortality and cardiovascular
and normal preoperative renal function. Eur Urol 2015; 67: 683–89. outcomes? J Urol 2009; 181: 55–61.
46 Sun M, Abdollah F, Bianchi M, et al. Treatment management of 68 Karakiewicz PI, Briganti A, Chun FK, et al. Multi-institutional
small renal masses in the 21st century: a paradigm shift. validation of a new renal cancer-specific survival nomogram.
Ann Surg Oncol 2012; 19: 2380–87. J Clin Oncol 2007; 25: 1316–22.
47 Kutikov A, Egleston BL, Wong YN, Uzzo RG. Evaluating overall 69 Zini L, Perrotte P, Capitanio U, et al. Radical versus partial
survival and competing risks of death in patients with localized nephrectomy: effect on overall and noncancer mortality.
renal cell carcinoma using a comprehensive nomogram. Cancer 2009; 115: 1465–71.
J Clin Oncol 2010; 28: 311–17. 70 Terrone C, Volpe A. Can emerging level 1 evidence “discourage”
48 Mehrazin R, Smaldone MC, Egleston B, et al. Is anatomic elective nephron-sparing surgery for small renal tumors?
complexity associated with renal tumor growth kinetics under active Eur Urol 2011; 59: 553–55.
surveillance? Urol Oncol 2015; 33: 167.e7–12. 71 Babaian KN, Merrill MM, Matin S, et al. Partial nephrectomy in the
49 Rendon RA. Active surveillance as the preferred management setting of metastatic renal cell carcinoma. J Urol 2014; 192: 36–42.
option for small renal masses. Can Urol Assoc J 2010; 4: 136–38. 72 Capitanio U, Zini L, Perrotte P, et al. Cytoreductive partial
50 Pierorazio PM, Johnson MH, Ball MW, et al. Five-year analysis of a nephrectomy does not undermine cancer control in metastatic renal
multi-institutional prospective clinical trial of delayed intervention and cell carcinoma: a population-based study. Urology 2008; 72: 1090–95.
surveillance for small renal masses: the DISSRM registry. Eur Urol 73 Volpe A, Blute ML, Ficarra V, et al. Renal ischemia and function
2015; published online Feb 16. DOI:10.1016/j.eururo.2015.02.001. after partial nephrectomy: a collaborative review of the literature.
51 Crispen PL, Viterbo R, Fox EB, Greenberg RE, Chen DY, Uzzo RG. Eur Urol 2015; 68: 61–74.
Delayed intervention of sporadic renal masses undergoing active 74 Parekh DJ, Weinberg JM, Ercole B, et al. Tolerance of the human kidney
surveillance. Cancer 2008; 112: 1051–57. to isolated controlled ischemia. J Am Soc Nephrol 2013; 24: 506–17.
52 Mehrazin R, Smaldone MC, Kutikov A, et al. Growth kinetics and 75 Simmons MN, Hillyer SP, Lee BH, Fergany AF, Kaouk J,
short-term outcomes of cT1b and cT2 renal masses under active Campbell SC. Functional recovery after partial nephrectomy: effects
surveillance. J Urol 2014; 192: 659–64. of volume loss and ischemic injury. J Urol 2012; 187: 1667–73.
53 Jewett MA, Mattar K, Basiuk J, et al. Active surveillance of small 76 Funahashi Y, Hattori R, Yamamoto T, Kamihira O, Kato K, Gotoh M.
renal masses: progression patterns of early stage kidney cancer. Ischemic renal damage after nephron-sparing surgery in patients
Eur Urol 2011; 60: 39–44. with normal contralateral kidney. Eur Urol 2009; 55: 209–15.
54 Hoffmann NE, Bischof JC. The cryobiology of cryosurgical injury. 77 Van Poppel H, Da Pozzo L, Albrecht W, et al, and the European
Urology 2002; 60 (suppl 1): 40–49. Organization for Research and Treatment of Cancer (EORTC), and
55 Katsanos K, Mailli L, Krokidis M, McGrath A, Sabharwal T, Adam A. the National Cancer Institute of Canada Clinical Trials Group
Systematic review and meta-analysis of thermal ablation versus (NCIC CTG), and the Southwest Oncology Group (SWOG), and the
surgical nephrectomy for small renal tumours. Eastern Cooperative Oncology Group (ECOG). A prospective
Cardiovasc Intervent Radiol 2014; 37: 427–37. randomized EORTC intergroup phase 3 study comparing the
56 Kunkle DA, Egleston BL, Uzzo RG. Excise, ablate or observe: the complications of elective nephron-sparing surgery and radical
small renal mass dilemma--a meta-analysis and review. J Urol 2008; nephrectomy for low-stage renal cell carcinoma. Eur Urol 2007;
179: 1227–33. 51: 1606–15.
57 Motzer RJ, Jonasch E, Agarwal N, et al. Kidney Cancer, Version 78 Hollenbeck BK, Taub DA, Miller DC, Dunn RL, Wei JT. National
3.2015: featured updates to the NCCN guidelines. utilization trends of partial nephrectomy for renal cell carcinoma:
J Natl Compr Canc Netw 2015; 13: 151–59. a case of underutilization? Urology 2006; 67: 254–59.
58 Woldrich JM, Palazzi K, Stroup SP, et al. Trends in the surgical 79 Miller DC, Hollingsworth JM, Hafez KS, Daignault S,
management of localized renal masses: thermal ablation, partial Hollenbeck BK. Partial nephrectomy for small renal masses:
and radical nephrectomy in the USA, 1998–2008. BJU Int 2013; an emerging quality of care concern? J Urol 2006; 175: 853–57.
111: 1261–68. 80 Thompson RH, Kaag M, Vickers A, et al. Contemporary use of
59 Gervais DA, McGovern FJ, Arellano RS, McDougal WS, Mueller PR. partial nephrectomy at a tertiary care center in the United States.
Radiofrequency ablation of renal cell carcinoma: part 1, Indications, J Urol 2009; 181: 993–97.
results, and role in patient management over a 6-year period and 81 Ficarra V, Novara G, Secco S, et al. Preoperative aspects and
ablation of 100 tumors. AJR Am J Roentgenol 2005; 185: 64–71. dimensions used for an anatomical (PADUA) classification of renal
60 Thompson RH, Atwell T, Schmit G, et al. Comparison of Partial tumours in patients who are candidates for nephron-sparing
Nephrectomy and Percutaneous Ablation for cT1 Renal Masses. surgery. Eur Urol 2009; 56: 786–93.
Eur Urol 2015; 67: 252–59. 82 Kutikov A, Uzzo RG. The R.E.N.A.L. nephrometry score: a
61 Kutikov A, Smaldone MC, Uzzo RG. Focal therapy for treatment of comprehensive standardized system for quantitating renal tumor
the small renal mass: dealer’s choice or a therapeutic gamble? size, location and depth. J Urol 2009; 182: 844–53.
Eur Urol 2015; 67: 260–61. 83 Simmons MN, Ching CB, Samplaski MK, Park CH, Gill IS. Kidney
62 Crépel M, Jeldres C, Sun M, et al. A population-based comparison tumor location measurement using the C index method.
of cancer-control rates between radical and partial nephrectomy for J Urol 2010; 183: 1708–13.
T1A renal cell carcinoma. Urology 2010; 76: 883–88. 84 Sivarajan G, Taksler GB, Walter D, Gross CP, Sosa RE, Makarov DV.
63 Crépel M, Jeldres C, Perrotte P, et al. Nephron-sparing surgery is The Effect of the Diffusion of the Surgical Robot on the Hospital-
equally effective to radical nephrectomy for T1BN0M0 renal cell level Utilization of Partial Nephrectomy. Med Care 2015; 53: 71–78.
carcinoma: a population-based assessment. Urology 2010; 75: 271–75. 85 Choi JE, You JH, Kim DK, Rha KH, Lee SH. Comparison of
64 Van Poppel H, Da Pozzo L, Albrecht W, et al. A prospective, perioperative outcomes between robotic and laparoscopic partial
randomised EORTC intergroup phase 3 study comparing the nephrectomy: a systematic review and meta-analysis. Eur Urol 2015;
oncologic outcome of elective nephron-sparing surgery and radical 67: 891–901.
nephrectomy for low-stage renal cell carcinoma. Eur Urol 2011; 86 Gratzke C, Seitz M, Bayrle F, et al. Quality of life and perioperative
59: 543–52. outcomes after retroperitoneoscopic radical nephrectomy (RN),
65 Scosyrev E, Messing EM, Sylvester R, Campbell S, Van Poppel H. open RN and nephron-sparing surgery in patients with renal cell
Renal function after nephron-sparing surgery versus radical carcinoma. BJU Int 2009; 104: 470–75.
nephrectomy: results from EORTC randomized trial 30904. 87 Hemal AK, Kumar A, Kumar R, Wadhwa P, Seth A, Gupta NP.
Eur Urol 2014; 65: 372–77. Laparoscopic versus open radical nephrectomy for large renal tumors:
66 Lane BR, Campbell SC, Demirjian S, Fergany AF. Surgically a long-term prospective comparison. J Urol 2007; 177: 862–66.
induced chronic kidney disease may be associated with a lower risk 88 Asimakopoulos AD, Miano R, Annino F, et al. Robotic radical
of progression and mortality than medical chronic kidney disease. nephrectomy for renal cell carcinoma: a systematic review.
J Urol 2013; 189: 1649–55. 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 Bekema HJ, MacLennan S, Imamura M, et al. Systematic review of 110 Hudes G, Carducci M, Tomczak P, et al, and the Global ARCC Trial.
adrenalectomy and lymph node dissection in locally advanced renal Temsirolimus, interferon alfa, or both for advanced renal-cell
cell carcinoma. Eur Urol 2013; 64: 799–810. carcinoma. N Engl J Med 2007; 356: 2271–81.
90 Capitanio U, Becker F, Blute ML, et al. Lymph node dissection in 111 Hutson TE, Escudier B, Esteban E, et al. Randomized phase III trial
renal cell carcinoma. Eur Urol 2011; 60: 1212–20. of temsirolimus versus sorafenib as second-line therapy after
91 Blom JH, van Poppel H, Maréchal JM, et al, and the EORTC sunitinib in patients with metastatic renal cell carcinoma.
Genitourinary Tract Cancer Group. Radical nephrectomy with and J Clin Oncol 2014; 32: 760–67.
without lymph-node dissection: final results of European 112 Motzer RJ, Escudier B, Oudard S, et al, and the RECORD-1 Study
Organization for Research and Treatment of Cancer (EORTC) Group. Efficacy of everolimus in advanced renal cell carcinoma:
randomized phase 3 trial 30881. Eur Urol 2009; 55: 28–34. a double-blind, randomised, placebo-controlled phase III trial.
92 Capitanio UB, Becker F, Blute F, et al. Lymph node dissection in Lancet 2008; 372: 449–56.
renal cell carcinoma. Eur Urol 2011; 60: 1212–20. 113 Sternberg CN, Davis ID, Mardiak J, et al. Pazopanib in locally
93 Bex A, Vermeeren L, de Windt G, Prevoo W, Horenblas S, Olmos RA. advanced or metastatic renal cell carcinoma: results of a
Feasibility of sentinel node detection in renal cell carcinoma: a pilot randomized phase III trial. J Clin Oncol 2010; 28: 1061–68.
study. Eur J Nucl Med Mol Imaging 2010; 37: 1117–23. 114 Sternberg CN, Hawkins RE, Wagstaff J, et al. A randomised,
94 Bex A, Vermeeren L, Meinhardt W, Prevoo W, Horenblas S, double-blind phase III study of pazopanib in patients with advanced
Valdés Olmos RA. Intraoperative sentinel node identification and and/or metastatic renal cell carcinoma: final overall survival results
sampling in clinically node-negative renal cell carcinoma: initial and safety update. Eur J Cancer 2013; 49: 1287–96.
experience in 20 patients. World J Urol 2011; 29: 793–99. 115 Motzer RJ, Hutson TE, Cella D, et al. Pazopanib versus sunitinib in
95 Haferkamp A, Bastian PJ, Jakobi H, et al. Renal cell carcinoma with metastatic renal-cell carcinoma. N Engl J Med 2013; 369: 722–31.
tumor thrombus extension into the vena cava: prospective 116 Motzer RJ, Hutson TE, McCann L, Deen K, Choueiri TK. Overall
long-term followup. J Urol 2007; 177: 1703–08. survival in renal-cell carcinoma with pazopanib versus sunitinib.
96 Bellmunt J, Puente J, Garcia de Muro J, Lainez N, Rodríguez C, N Engl J Med 2014; 370: 1769–70.
Duran I. SEOM clinical guidelines for the treatment of renal cell 117 Hutson TE, Lesovoy V, Al-Shukri S, et al. Axitinib versus sorafenib
carcinoma. Clin Transl Oncol 2014; 16: 1043–50. as first-line therapy in patients with metastatic renal-cell carcinoma:
97 Marincola FM, White DE, Wise AP, Rosenberg SA. Combination a randomised open-label phase 3 trial. Lancet Oncol 2013;
therapy with interferon alfa-2a and interleukin-2 for the treatment 14: 1287–94.
of metastatic cancer. J Clin Oncol 1995; 13: 1110–22. 118 Rini BI, Escudier B, Tomczak P, et al. Comparative effectiveness of
98 Coppin C, Porzsolt F, Awa A, Kumpf J, Coldman A, Wilt T. axitinib versus sorafenib in advanced renal cell carcinoma (AXIS):
Immunotherapy for advanced renal cell cancer. a randomised phase 3 trial. Lancet 2011; 378: 1931–39.
Cochrane Database Syst Rev 2005; 1: CD001425. 119 Escudier B, Porta C, Bono P, et al. Randomized, controlled, double-
99 Flanigan RC, Salmon SE, Blumenstein BA, et al. Nephrectomy blind, cross-over trial assessing treatment preference for pazopanib
followed by interferon alfa-2b compared with interferon alfa-2b versus sunitinib in patients with metastatic renal cell carcinoma:
alone for metastatic renal-cell cancer. N Engl J Med 2001; PISCES Study. J Clin Oncol 2014; 32: 1412–18.
345: 1655–59. 120 Delea TE, Amdahl J, Diaz J, Nakhaipour HR, Hackshaw MD.
100 Mickisch GH, Garin A, van Poppel H, de Prijck L, Sylvester R, and Cost-effectiveness of pazopanib versus sunitinib for renal cancer in
the European Organisation for Research and Treatment of Cancer the United States. J Manag Care Spec Pharm 2015;
(EORTC) Genitourinary Group. Radical nephrectomy plus 121 Heng DY, Xie W, Regan MM, et al. External validation and
interferon-alfa-based immunotherapy compared with interferon comparison with other models of the International Metastatic
alfa alone in metastatic renal-cell carcinoma: a randomised trial. Renal-Cell Carcinoma Database Consortium prognostic model:
Lancet 2001; 358: 966–70. a population-based study. Lancet Oncol 2013; 14: 141–48.
101 Flanigan RC, Mickisch G, Sylvester R, Tangen C, Van Poppel H, 122 Heng DY, Xie W, Regan MM, et al. Prognostic factors for overall
Crawford ED. Cytoreductive nephrectomy in patients with survival in patients with metastatic renal cell carcinoma treated with
metastatic renal cancer: a combined analysis. J Urol 2004; vascular endothelial growth factor-targeted agents: results from a
171: 1071–76. large, multicenter study. J Clin Oncol 2009; 27: 5794–99.
102 Rini BI, Campbell SC. The evolving role of surgery for advanced 123 Ruiz JN, Belum VR, Creel P, Cohn A, Ewer M, Lacouture ME.
renal cell carcinoma in the era of molecular targeted therapy. Current practices in the management of adverse events associated
J Urol 2007; 177: 1978–84. with targeted therapies for advanced renal cell carcinoma: a national
103 Motzer RJ, Hutson TE, Tomczak P, et al. Sunitinib versus interferon survey of oncologists. Clin Genitourin Cancer 2014; 12: 341–47.
alfa in metastatic renal-cell carcinoma. N Engl J Med 2007; 356: 115–24. 124 De Meerleer G, Khoo V, Escudier B, et al. Radiotherapy for
104 Escudier B, Eisen T, Stadler WM, et al. Sorafenib for treatment of renal-cell carcinoma. Lancet Oncol 2014; 15: e170–77.
renal cell carcinoma: final efficacy and safety results of the phase III 125 Singh P, Agarwal N, Pal SK. Sequencing systemic therapies for
treatment approaches in renal cancer global evaluation trial. metastatic kidney cancer. Curr Treat Options Oncol 2015; 16: 316.
J Clin Oncol 2009; 27: 3312–18. 126 Beuselinck B, Job S, Becht E, et al. Molecular subtypes of clear cell
105 Escudier B, Pluzanska A, Koralewski P, et al, and the AVOREN Trial renal cell carcinoma are associated with sunitinib response in the
investigators. Bevacizumab plus interferon alfa-2a for treatment of metastatic setting. Clin Cancer Res 2015; 21: 1329–39.
metastatic renal cell carcinoma: a randomised, double-blind 127 Motzer RJ, Rini BI, McDermott DF, et al. Nivolumab for metastatic
phase III trial. Lancet 2007; 370: 2103–11. renal cell carcinoma: results of a randomized phase II trial.
106 Escudier B, Bellmunt J, Négrier S, et al. Phase III trial of J Clin Oncol 2015; 33: 1430–37.
bevacizumab plus interferon alfa-2a in patients with metastatic 128 Motzer RJ, Nosov D, Eisen T, et al. Tivozanib versus sorafenib as initial
renal cell carcinoma (AVOREN): final analysis of overall survival. targeted therapy for patients with metastatic renal cell carcinoma:
J Clin Oncol 2010; 28: 2144–50. results from a phase III trial. J Clin Oncol 2013; 31: 3791–99.
107 Rini BI, Halabi S, Rosenberg JE, et al. Bevacizumab plus interferon 129 Mehta A, Sonpavde G, Escudier B. Tivozanib for the treatment of
alfa compared with interferon alfa monotherapy in patients with renal cell carcinoma: results and implications of the TIVO-1 trial.
metastatic renal cell carcinoma: CALGB 90206. J Clin Oncol 2008; Future Oncol 2014; 10: 1819–26.
26: 5422–28. 130 Motzer RJ, Porta C, Vogelzang NJ, et al. Dovitinib versus sorafenib
108 Rini BI, Halabi S, Rosenberg JE, et al. Phase III trial of for third-line targeted treatment of patients with metastatic renal
bevacizumab plus interferon alfa versus interferon alfa cell carcinoma: an open-label, randomised phase 3 trial.
monotherapy in patients with metastatic renal cell carcinoma: final Lancet Oncol 2014; 15: 286–96.
results of CALGB 90206. J Clin Oncol 2010; 28: 2137–43. 131 Vaishampayan U. Cabozantinib as a novel therapy for renal cell
109 Rini BI, Bellmunt J, Clancy J, et al. Randomized phase III trial of carcinoma. Curr Oncol Rep 2013; 15: 76–82.
temsirolimus and bevacizumab versus interferon alfa and 132 Gordon MS, Hussey M, Nagle RB, et al. Phase II study of erlotinib
bevacizumab in metastatic renal cell carcinoma: INTORACT trial. in patients with locally advanced or metastatic papillary histology
J Clin Oncol 2014; 32: 752–59. renal cell cancer: SWOG S0317. J Clin Oncol 2009; 27: 5788–93.

12 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 138 Lam JS, Shvarts O, Belldegrun AS. Kidney cancer management in
metatastic renal cell carcinoma. J Clin Oncol 2008; 26: 3457–60. 2004: an update for the practicing general urologist.
134 EORTC. Randomized phase III trial comparing presurgical Curr Urol Rep 2004; 5: 1–3.
sunitinib followed by nephrectomy and sunitinib versus 139 Kim DY, Karam JA, Wood CG. Role of metastasectomy for
nephrectomy followed by sunitinib in patients with synchronous metastatic renal cell carcinoma in the era of targeted therapy.
metastatic renal cell carcinoma. 2010. http://www.eortc.be/protoc/ World J Urol 2014; 32: 631–42.
Details.asp?Protocol=30073&T=2010 (accessed July 15, 2015). 140 Breau RH, Blute ML. Surgery for renal cell carcinoma metastases.
135 Messing EM, Manola J, Wilding G, et al, and the Eastern Curr Opin Urol 2010; 20: 375–81.
Cooperative Oncology Group/Intergroup trial. Phase III study of 141 Russo P, O’Brien MF. Surgical intervention in patients with
interferon alfa-NL as adjuvant treatment for resectable renal cell metastatic renal cancer: metastasectomy and cytoreductive
carcinoma: an Eastern Cooperative Oncology Group/Intergroup nephrectomy. Urol Clin North Am 2008; 35: 679–86, viii.
trial. J Clin Oncol 2003; 21: 1214–22. 142 Dabestani S, Marconi L, Hofmann F, et al. Local treatments for
136 Pizzocaro G, Piva L, Colavita M, et al. Interferon adjuvant to radical metastases of renal cell carcinoma: a systematic review.
nephrectomy in Robson stages II and III renal cell carcinoma: Lancet Oncol 2014; 15: e549–61.
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.

www.thelancet.com Published online August 26, 2015 http://dx.doi.org/10.1016/S0140-6736(15)00046-X 13

You might also like