This is an open access article under the terms of a license that permits non-commercial use, provided the original work is properly cited.  
© 2022 The Authors. Société Internationale d'Urologie Journal, published by the Société Internationale d'Urologie, Canada.

Key Words Competing Interests Article Information

Renal cancer, immunotherapy, systematic 
review

None declared. Received on May 9, 2022 
Accepted on June 12, 2022 
This article has been peer reviewed.

Soc Int Urol J. 2022;3(5):341–352

DOI: 10.48083/ WIXM2804

Adjuvant Systemic Treatment for Renal Cancer  
After Surgery: A Network Meta-Analysis

Niranjan J. Sathianathen,1,2 Marc A. Furrer,1 Christopher J. Weight,3  
Declan G. Murphy,4 Shilpa Gupta,5 Nathan Lawrentschuk1,2,4

1 Department of Urology, Royal Melbourne Hospital, Parkville, Victoria, Australia  2 Department of Surgery, University of Melbourne, Parkville, Victoria, Australia 
3 Department of Urology, Cleveland Clinic, Cleveland, United States  4 Department of Uro-Oncology, Peter MacCallum Cancer Centre, Parkville, Victoria, Australia  
5Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, United States

Abstract

Background Approximately 15% to 20% of patients will experience disease recurrence following surgical removal 
of renal cell carcinoma. A range of pharmacological agents is prescribed for metastatic renal cell carcinoma, but there 
are trials testing whether these have an earlier role in the adjuvant setting. We aim to assess the efficacy of adjuvant 
systemic treatment following surgery in patients with renal cell carcinoma and to determine the most effective 
treatment.

Methods The protocol for this review was published in PROSPERO (CRD42021281588). We searched multiple 
databases up to August 2021. We included only randomized trials of patients with renal cell carcinoma that had been 
completely resected. We included patients with locoregional nodal disease if it was surgically removed, and excluded 
all cases of metastatic disease. We included all adjuvant systemic therapies that were commenced within 90 days of 
renal surgery. A network meta-analysis was performed using a frequentist approach.

Results A total of 13 studies with 8103 patients were included for analysis. Only pembrolizumab (HR 0.74; 95%CI 
0.57 to 0.96) and pazopanib (HR 0.80; 95%CI 0.68 to 0.95) improved disease-free survival compared with observation. 
These 2 treatments were the 2 highest ranked comparisons with a P-score of 0.87 and 0.80. No agent improved overall 
survival. All agents increased the risk of severe adverse events compared with observation.

Conclusions Pembrolizumab and pazopanib were the only 2 adjuvant agents that improved time to disease 
recurrence compared with observation, with the former likely being the more efficacious. None of the treatments 
improved overall survival and almost all increased severe adverse events.

Introduction

There has been an increased incidence of renal cell carcinoma, especially in developed countries[1]. Most of these 
cancers are localized to the kidney at the time of presentation and are curable by surgery. However, approximately 
20% of patients will experience disease recurrence following surgery[2]. Overall prognosis for advanced disease is 
poor with a median survival time of 21 months after recurrence[3].

341SIUJ.ORG SIUJ  •  Volume 3, Number 5  •  September 2022

REVIEW

mailto:niranjan19%40gmail.com?subject=SIUJ
http://SIUJ.org


A range of pharmacological agents has been used 
to treat metastatic renal cell carcinoma (mRCC) with 
varying efficacy, including chemotherapy, immuno-
therapy, tyrosine kinase inhibitors, monoclonal anti-
body against circulating vascular endothelial growth 
factor, and mTOR inhibitors. A network meta-anal-
ysis found that combination immunotherapy likely 
represents the current best available treatment[4]. The 
European Association of Urology guidelines support 
this by suggesting that immunotherapy (including 
combinations) should be used as first-line treatment in 
this setting[5]. As immunotherapy has come to the fore-
front of mRCC management, there has been increasing 
interest in employing these treatments at earlier stages 
of disease. Many of the aforementioned treatments have 
been trialled in the adjuvant setting with varying results, 
and there are recent reports of use of adjuvant immu-
notherapy. However, these trials have primarily been 
conducted using observation or placebo as a compara-
tor arm, which has not permitted direct comparisons of 
active agents.

We therefore aimed to perform a systematic review 
and network-meta-analysis of systemic agents used in 
the adjuvant setting after surgery for kidney cancer.

Methods
We registered the protocol of this systematic review in 
PROSPERO (CRD42021281588). We searched multiple 
databases (MEDLINE , EMBASE , ScienceDirect, 
Cochrane Libraries, HTA database, and Web of 
Science) up to 20 August 2021, with a range of keywords 
associated with “renal carcinoma” and “adjuvant 
therapy.” We also searched the abstracts from leading 
urological and oncological meetings, including those 
of the European Association of Urology, American 
Urological Association, American Society of Clinical 
Oncology, and European Society of Medical Oncology 
in the last 5 years. We also searched trial registries such 
as ClinicalTrials.gov. We did not place any restriction 
on language or date of publication. We included only 
randomized studies.

Our population of interest was patients with RCC 
that had been completely resected. Surgical treatment 
included both radical and partial nephrectomy. We 
included patients with locoregional nodal disease if they 
underwent surgical removal at the time of kidney extir-
pation, ie, N+ cases were eligible. We included all histo-
logical subtypes of renal carcinoma. We excluded all 
patients with distant metastatic disease even if they had 
undergone metastectomy, ie, M1 cases were not eligible 
for inclusion.

We included all adjuvant systematic therapies that 
were commenced within 90 days of renal surgery. We 
excluded autologous vaccine-based treatments because 

they are not widely available in clinical practice. We did 
not include adjuvant radiotherapy. Control arms eligible 
for analysis were observation, placebo, and active treat-
ments, although we did not find any studies with the last.

Following our search, titles and abstracts were 
screened by 2 independent authors according to the 
inclusion/exclusion criteria. Full texts of relevant 
abstracts were then reviewed by 2 independent authors 
to confirm eligibility. Any disagreements were resolved 
by a third senior author. Data were then extracted inde-
pendently.

The efficacy outcomes of interest were disease-free 
survival (DFS), defined as time from randomization to 
disease recurrence (local or distant) and/or death; and 
overall survival (OS), defined as time from randomiza-
tion to death from any cause.

The safety outcome of interest was severe adverse 
events defined as incidence of grade III to V events per 
patient.

We also intended to perform subgroup analysis on 
the efficacy outcome according to histological subtype 
(clear-cell versus other subtypes) and nodal disease (no 
nodal disease [N0] versus nodal disease [N1]).

Statistical analysis
We first performed traditional pairwise meta-analysis 
of the included studies (data not shown). To do this, 
we applied the model proposed by Woods et al. by 
extracting hazard rates for DFS and OS and number 
of severe adverse events from each of the included 
studies[6].

We then performed a network meta-analysis of all 
included trials which enables indirect comparisons 
of treatments based on a common comparator arm. 
We adopted a frequentist approach and performed a 
fixed-effect consistency network meta-analysis. As a 
sensitivity analysis, we used the same approach with a 
random-effects model. We used P-scores that estimate 
the extent that one treatment is superior to another, 
averaged over all competing treatments, to determine 
which agent is the most efficacious.

All analyses were performed using RJAGS and R  
(R Foundation for Statistical Computing, Vienna, 
Austria) version 3.4. Risk of bias was performed accord-
ing to the Cochrane framework[7].

Results
Our search retrieved 4088 abstracts of which 41 
proceeded to full text review. After inclusion/exclusion 
criteria were applied, 13 studies were eligible and 
included for analysis (Online Supplementary Figure 1). 
The details of included studies are shown in Table 1.  

342 SIUJ  •  Volume 3, Number 5  •  September 2022 SIUJ.ORG

REVIEW

http://siuj.org
http://SIUJ.org


 The included trials tested a range of adjuvant treatments: 
axitinib[8], girentuximab[9], interferon-alpha[10–14], 
interleukin-2[10, 11], pazopanib[15], pembrolizumab[16], 
sorafenib[17,18], sunitinib[17,19], and thalidomide[20]. 
Two of the trials tested combination adjuvant therapies 
of interleukin-2+interferon-alpha[10] and interleukin-
2+interferon-alpha+5-f lurouracil[11]. The PROTECT 
trial that compared pazopanib with placebo included 
patients who received either 600 mg or 800 mg, and 
we included both in this analysis[15]. The trials were 
overall of moderate quality, and the detailed risk of bias 
classification can be found in Online Supplementary 
Table 1. We also found a further 6 trials in progress 
(Table 2).

Disease-free survival
All eligible studies reported on DFS and were included 
in this analysis of 8103 patients. Only the 2016 study 
by Haas et al. reported on direct comparison between 
active agents[17]. The forest plot of HRs compared with 
control arm for each agent is shown in Figure 1A. Only 
pembrolizumab (HR 0.74; 95% CI 0.57 to 0.96) and 
pazopanib (HR 0.80; 95% CI 0.68 to 0.95) prolonged DFS 
compared with observation. These 2 treatments were the 

FIGURE 1. 
Treatment versus control for (A) DFS and (B) OS

A

B

TABLE 1. 

Characteristics of included studies 
 

Author/ Year
Adjuvant  
treatment

Number of 
participants

Inclusion criteria

Number of 
participants 

with ≤T2 
disease, n 

Number of 
participants 
with clear-

cell RCC 

Number of 
participants 
with nodal 

disease 

Gross-Goupil et al. 
2018[8]

Axitinib: 5 mg BD  
up to 3 years

724

• ≥pT2 and/or N+

• Any Fuhrman grade

• ECOG performance  
status 0/1

80 NR 36

Chamie et al. 
2017[9]

Girentuximab: IV 50mg 
week 1 followed by IV 

20 mg week 2–24
864

Histologically confirmed 
ccRCC pT3/pT4Nx/N0M0 or 
pTanyN+M0 or pT1b/pT2Nx/
N0M0 with nuclear grade 3 

or greater

139 834 65

Passalacqua et al. 
2014[10]

IL-2 + IFN-a: IL-2 SC 1 
mil IU/m2 5 days per 
week for 4 weeks; 

INF-a SC 1.8 mil IU/m2 
on day 3 and 5 each 

week. Cycles repeated 
every 4 months for  

2 years and 6 months 
for 3 years

310

Partial or radical 
nephrectomy with no 

residual disease and free 
surgical margins: pT2-3b 

pN0-3 M0

182 254 12

NR: not reported

, Cont’d 

continued on page 344

343SIUJ.ORG SIUJ  •  Volume 3, Number 5  •  September 2022

Adjuvant Systemic Treatment for Renal Cancer After Surgery: A Network Meta-Analysis

http://siuj.org
http://siuj.org
http://SIUJ.org


TABLE 1. 

Characteristics of included studies 
 

Author/ Year
Adjuvant  
treatment

Number of 
participants

Inclusion criteria

Number of 
participants 

with ≤T2 
disease, n 

Number of 
participants 
with clear-

cell RCC 

Number of 
participants 
with nodal 

disease 

Aitchison et 
al.2014[11]

IL-2 + IFN-a + 5-FU: 
IL-2 SC 20 mil IU/m2 
on days 3-5 in weeks 
1 and 4 and SC 5 mil 

IU/m2 on days 1, 3 and 
5 in weeks 2 and 3; 

IFN-a SC 6 mil IU/m2 
in weeks 2 and 3 and 
increasing to SC 9 mil 
IU/m2 in weeks 5-8 

given on days 1, 3 and 
5; 5-FU IV 750mg/m2 
weekly in weeks 5-8

309

Histologically proven stage 
T3b, T3c, T4 tumour or any pT 
stage and nodal status pN1 

or 2, or any pT stage

Clinical N+ disease removed 
and had no metastatic 
disease or macroscopic 

residual disease as 
confirmed within 2–4 weeks 
prior to randomization by CT 

or MRI plus CXR

65 NR 49

Messing et al. 
2003[12]

Interferon α-NL: SC 5 
days every 3 weeks  

(3 mil IU/m2 day  
1, 5 mil IU/m2 day 2, 
20 mil IU/m2 day 3-5) 

up to 12 cycles

283

Unilateral, locally 
advanced (pT3-4a) and/
or node-positive renal 

cancer following radical 
nephrectomy

No disseminated disease

36 176 44

Pizzocaro et 
al.2001[13]

Interferon α: IM 6 mil 
IU 3 times per week 

for 6 months
247

Radical nephrectomy with 
suggested unilateral para-

aortic nodal dissection

Patients with pathologic 
stages II and III RCC (1987 
tumour-node-metastasis 
categories T3aN0M0 and 

T3bN0M0 or T2/3N1-3M0) 
were eligible for the study

16 NR 43

Hinotsu et 
al.2013[14]

Interferon α: IM 3-6 
mil IU 3 times per 
week for 1 year

107

Histopathological diagnosis 
of renal cell carcinoma

resection of the primary 
tumour by nephrectomy, for 
which open or laparoscopic 

surgery could have been 
selected and lymph node 
dissection was possible

no metastatic disease

40 82 5

NR: not reported

, Cont’d 

continued on page 345

344 SIUJ  •  Volume 3, Number 5  •  September 2022 SIUJ.ORG

REVIEW

http://SIUJ.org


TABLE 1. 

Characteristics of included studies 
 

Author/ Year
Adjuvant  
treatment

Number of 
participants

Inclusion criteria

Number of 
participants 

with ≤T2 
disease, n 

Number of 
participants 
with clear-

cell RCC 

Number of 
participants 
with nodal 

disease 

Motzer et 
al.2017[15]

Pazopanib: 600 mg or 
800 mg once daily

1538

Resected non-metastatic 
(M0) clear-cell or 

predominant clear-cell 
RCC histology within the 

following TNM classification 
and Fuhrman grades: pT2G3-

4N0, pT3-T4 GanyN0, or 
pTanyGanyN1

235 1057 90

Choueiri et al. 
2021[16]

Pembrolizumab: IV 
200 mg once every 3 
weeks up to 17 cycles

994

Histologically confirmed 
locoregional renal cell 

carcinoma with a clear-
cell component that is at 
high risk of recurrence (ie, 

tumour stage 2 with nuclear 
grade 4 or sarcomatoid 

differentiation, tumour stage 
3 or higher, regional lymph 
node metastasis, or stage 

M1 with NED)

Surgery (partial or 
radical nephrectomy or 
metastasectomy) with 

negative surgical margins

In those with M1 NED 
status, M1 disease was 

present in addition to the 
primary tumour at diagnosis, 

and metastases had to be 
completely resected at 

the time of nephrectomy 
or within 1 year after 

nephrectomy

86 994 62

Eisen et al. 
2020[18]

Sorafenib: 400 mg 
BD PO

1711

Histologically confirmed RCC

No evidence of residual 
macroscopic disease on 

postoperative CT scan after 
resection of RCC

604 1455 74

NR: not reported

, Cont’d 

continued on page 346

345SIUJ.ORG SIUJ  •  Volume 3, Number 5  •  September 2022

Adjuvant Systemic Treatment for Renal Cancer After Surgery: A Network Meta-Analysis

http://SIUJ.org
http://SIUJ.org


, Cont’d 

TABLE 1. 

Characteristics of included studies 
 

Author/ Year
Adjuvant  
treatment

Number of 
participants

Inclusion criteria

Number of 
participants 

with ≤T2 
disease, n 

Number of 
participants 
with clear-

cell RCC 

Number of 
participants 
with nodal 

disease 

Haas et al. 
2016[17]

Sunitinib: 50 mg  
PO daily for first  
28 days of each 

6-week cycle

1943

Histologically proven, 
completely resected high-
risk clear-cell or non-clear-
cell RCC within 12 weeks 
of removal of the primary 

tumour. High-risk features: 
pT1b G3–4 N0 (or pNX where 

clinically N0) M0 to  
T(any) G(any)  

N + (fully resected) M0

Sorafenib: 400 mg  
BD PO daily

NR 1541 NR

Ravaud et al. 
2016[19]

Sunitinib: 50 mg  
PO daily 4 weeks on, 
2 weeks off schedule 

for 1 year

615

Locoregional RCC (tumour 
stage 3 or higher, regional 
lymph node metastasis,  

or both)
Histologic confirmation  

of clear-cell RCC
The absence of macroscopic 

residual or metastatic 
disease after nephrectomy, 

as confirmed on blinded 
independent central review 

of CT images

NR 615 49

Margulis et al. 
2009[20]

Thalidomide:  
100 mg/day for  

2 weeks then 200 
mg/day for 2 weeks 

followed by  
300 mg/day

46

Completely resected locally 
advanced high-risk RCC, 
as defined by one of the 
following criteria: pT2 
(Fuhrman grade 3 or 4), 

pT3a-c, T4, or N1–2 disease 
resected to no evidence of 

residual disease
All tumour subtypes were 

eligible

7 34 13

NR: not reported

346 SIUJ  •  Volume 3, Number 5  •  September 2022 SIUJ.ORG

REVIEW

http://SIUJ.org


TABLE 2. 

Trials in progress 

Trial name/ 
number

Interventions Inclusion criteria Current progress
Estimated  

completion date

EVEREST
NCT01120249

Everolimus

Histologically or cytologically confirmed  
renal cell carcinoma

Considered pathologically either intermediate 
high-risk or very high-risk disease
No history of distant metastases

Have undergone a full surgical resection  
(radical nephrectomy or partial nephrectomy) 

including removal of all clinically positive nodes
No evidence of residual or metastatic renal  

cell cancer on CT scan of the chest, abdomen, 
and pelvis (all with oral and IV contrast) 

performed after nephrectomy

Active,  
not recruiting

October 2021

SPARC-1
NCT04028245

Spartalizumab and 
Canakinumab

Histologically confirmed clear-cell or 
predominantly clear-cell RCC

Non-metastatic (localized) RCC that is clinical 
stage T2 and above, or clinical N1 disease  

with any T stage
Scheduled to undergo either radical or  

partial nephrectomy

Recruiting December 2022

PROSPER
NCT03055013

Nivolumab – 
neo-adjuvant and 

adjuvant

Patients must have a renal mass consistent 
with a clinical stage ≥T2Nx renal cell carcinoma 
(RCC) or TanyN+ RCC for which radical or partial 

nephrectomy is planned
Patients must have no clinical or radiological 
evidence of distant metastases (M0) unless 
the presumed M1 disease is planned to be 
resected/definitively treated (eg, thermal 

ablation, stereotactic radiation)

Active,  
not recruiting

November 2023

IMmotion010
NCT03024996

Atezolizumab

Pathologically confirmed RCC with a  
component of either clear-cell histology 
or sarcomatoid histology that has not 

been previously treated in the adjuvant or 
neoadjuvant setting and classified as being at 

high risk of RCC recurrence
Radical or partial nephrectomy with 

lymphadenectomy in select participants
Absence of residual disease and absence  
of metastasis, as confirmed by a negative 

baseline CT of the pelvis, abdomen, and chest
Absence of brain metastasis, as confirmed  
by a negative CT with contrast or MRI scan  

of the brain

Active,  
not recruiting

February 2024

NR: not reported

continued on page 348

347SIUJ.ORG SIUJ  •  Volume 3, Number 5  •  September 2022

Adjuvant Systemic Treatment for Renal Cancer After Surgery: A Network Meta-Analysis

http://SIUJ.org


TABLE 2. 

Trials in progress 

Trial name/ 
number

Interventions Inclusion criteria Current progress
Estimated  

completion date

CheckMate 914
NCT03138512

Nivolumab
Nivolumab + 
ipilimumab

Kidney tumour has been completely resected 
with negative surgical margins obtained

Pathologic TNM staging meeting one of the 
following: pT2a, G3 or G4, N0 M0; pT2b, G any, 
N0 M0; pT3, (a, b, c), G any, N0 M0; pT4, G any, 

N0 M0; pT any, G any, N1 M0
Post-nephrectomy tumour shows RCC with a 
predominantly clear-cell histology, including 

participants with sarcomatoid features
Participants must have no clinical or radiological 

evidence of macroscopic residual disease or 
distant metastases after nephrectomy

Recruiting July 2025

RAMPART
NCT03288532

Durvalumab
Durvalumab + 
Tremelimumab

Histologically proven RCC (all cell types of 
RCC are eligible, except for pure oncocytoma, 

collecting duct, medullary and transitional 
cell cancer [TCC]); no evidence of residual 

macroscopic disease on postoperative CT scan 
after resection of RCC

Patients with microscopically positive resection 
margins after radical nephrectomy at the 

nephrectomy bed, renal vein, or inferior vena 
cava are eligible provided the postoperative  

CT scan shows no evidence of residual 
macroscopic disease

Recruiting December 2034

NR: not reported

2 highest ranked comparisons with a P-score of 0.87 and 
0.80, respectively. Thalidomide was the lowest ranked 
treatment with a P-score of 0.03. Interferon-alpha was 
inferior to axitinib, pazopanib, pembrolizumab and 
sunitinib. Comparisons of all treatments are shown in 
Table 3. These findings were the same in the sensitivity 
analysis when using a random-effects model (data not 
shown).

Overall survival
The OS analysis included 7063 patients from all the 
studies from above except Margulis et al. (thalidomide, 
2009)[20] and Choueiri et al. (pembrolizumab, 2021)
[16]. The forest plot of HRs compared with the control 
arm for each agent is shown in Figure 1B. None of 
the agents demonstrated a survival benefit compared 
with observation. Pazopanib was the highest ranked 
treatment with a P-score of 0.83. Comparisons of 
all treatments are shown in Online Supplementary 

Table 2. There was no difference between any of the 
treatment comparisons. These findings were the same 
in the sensitivity analysis when using a random-effects 
model (data not shown).

Severe adverse events
Data from 8 trials using the following interventions were 
included in the safety analysis: axitinib, girentuximab, 
interferon-alpha, pazopanib, pembrolizumab, sorafenib, 
su n it i n ib, a nd t ha l idom ide[8,9,12 ,15 –17,19, 20].  
The forest plots of ORs compared with control are shown 
in Online Supplementary Figure 2. All of the active 
treatments except girentuximab significantly increased 
the likelihood of severe adverse events compared 
with observation. These findings were the same in the 
sensitivity analysis when using a random-effects model.

Subgroup analyses
There were insufficient data to perform a network meta-
analysis on the planned subgroups.

, Cont’d 

348 SIUJ  •  Volume 3, Number 5  •  September 2022 SIUJ.ORG

REVIEW

http://siuj.org
http://siuj.org
http://siuj.org
http://SIUJ.org


Discussion
Ou r net work meta-a na lysis repor t fou nd t hat 
pembrolizumab is likely the most efficacious adjuvant 
agent in prolonging time to disease recurrence 
compared with other tyrosine k inase inhibitors, 
monoclonal antibody against circulating vascular 
endothelial growth factor, and/or chemotherapies. The 
only other therapy that was shown to improve DFS 
compared with observation was pazopanib. However, 
the absolute difference in recurrence-free survival at  
3 years was only 3% between pazopanib and placebo[15]. 
We used data from patients who received both 600 mg 
and 800 mg where there were discrepancies in the results 
related to dose. Although patients receiving the lower 
dose did not experience improved DFS, those receiving 
the higher dose were noted to have a prolonged disease-
free survival. Therefore, it is likely that the benefit of 
pazopanib 800 mg is greater than the overall estimates 
in this review. It should also be noted that the sunitinib 
did show an improvement in DFS in the S-TRAC trial, 
although the estimates from this meta-analysis were not 
significant when including the ECOG trial. The results 
from this network meta-analysis are consistent with 
those of previously published meta-analyses on 
the topic[21, 22]. Despite these positive findings in 
delaying disease recurrence, none of the treatments 
improved overall survival. We acknowledge that 
the overall survival data have not matured for most 
of the recent studies and that there may still be a 
benefit with adjuvant therapy. Importantly, there 
was an increased risk of severe adverse events with 
adjuvant treatment compared with observation.

It should be considered that there are several factors 
that impact the use of adjuvant treatment and choice 
of agent. This review represented a population of 
patients with locoregional renal cancer who had 
undergone surgery, but there are sub-populations 
within this group in whom treatment effect may 
differ. For example, we believed there may have 
been differences based on histological subtype and 
nodal status but were unable to perform the pre-
planned subgroup analyses due to a lack of data. 
The POLAR-01 trial reported that patients with 
N0 disease had better outcomes with combination 
IL-2 and IFN-alpha treatment than did those 
with N+ disease[10]. In contrast, the ATLAS trial 
demonstrated that patients with highest risk (pT3 

with grade ≥ 3 or pT4 and/or N+, any T, any grade) 
benefitted with axitinib treatment compared with 
those with low risk (pT2 or pT3 with grade ≤ 2) who 
had no difference in outcomes with axitinib[8]. 
The wider literature, especially in the metastatic 
setting, highlights the increasing use of molecular 
biomarkers to tailor treatment choices[23]. This 
will increase in importance as immunotherapies 
are used more in this setting. Therefore, patient 
selection is key in determining the benefit of 
adjuvant therapy and the choice of agent.

The findings of this meta-analysis should be 
contextualised within its limitations. As mentioned 
above, there is heterogeneity within the popula-
tions of the included studies, and we were unable to 
perform the pre-planned subgroup analyses. There 
were also individual study limitations, especially 
with respect to blinding, that may have introduced 
bias into the estimates. Additionally, we did not 
assess patient-reported outcomes, which is crit-
ical in determining whether adjuvant treatment 
improves quality in life[24]. Future studies will 
need to assess the cost-effectiveness of these treat-
ments because immunotherapies are expensive and 
thus may not be cost-effective[25]. Health econom-
ics studies of advanced RCC have reported that a 
significant decrease in the cost of immunotherapy 
is required for it to be cost-effective at generally 
accepted thresholds[26]. It is likely that these would 
be generalizable to the use of immunotherapy in the 
adjuvant setting as the absolute benefits of treatment 
are small, albeit statistically significant, and come at 
significant cost. Furthermore, this study will need 
to be updated following the publication of trials in 
progress.

Conclusions
Pembrolizumab and pazopanib were the only  
2 adjuvant agents that improved time to disease 
recurrence compared with observation, with the 
former likely being the more efficacious. None 
of the treatments improved overall survival, and 
almost all increased severe adverse events. While it 
is promising to see these agents show efficacy in this 
setting, the duration and cost of treatment also need 
to be considered when determining utility.

349SIUJ.ORG SIUJ  •  Volume 3, Number 5  •  September 2022

Adjuvant Systemic Treatment for Renal Cancer After Surgery: A Network Meta-Analysis

http://SIUJ.org


TABLE 3. 

Matrix comparing hazard ratios [confidence intervals] for DFS between all therapies 

Axitinib
1.15 

[0.87; 1.52]
1.11 

[0.79; 1.57]
0.97 

[0.57; 1.63]
0.97 

[0.65; 1.44]
1.45 

[1.00; 2.10]
0.92 

[0.67; 1.27]
0.85 

[0.58; 1.24]
1.12 

[0.82; 1.53]
1.07 

[0.78; 1.47]
2.69 

[1.04; 7.01]

0.87 
[0.66; 1.15]

Control
0.97 

[0.79; 1.19]
0.84 

[0.54; 1.31]
0.84 

[0.63; 1.12]
1.26 

[0.99; 1.61]
0.80 

[0.68; 0.95]
0.74 

[0.57; 0.96]
0.97

[0.84; 1.13]
0.93 

[0.80; 1.09]
2.34 

[0.94; 5.86]

0.90 
[0.64; 1.26]

1.03 
[0.84; 1.26]

Girentuximab
0.87 

[0.53; 1.41]
0.87 

[0.61; 1.23]
1.30 

[0.95; 1.79]
0.82  

[0.64; 1.07]
0.76 

[0.55; 1.06]
1.00 

[0.78; 1.29]
0.96 

[0.74; 1.24]
2.42 

[0.95; 6.17]

1.04 [
0.61; 1.75]

1.19 
[0.76; 1.85]

1.15 
[0.71; 1.88]

IL-2 + Interferon
1.00 

[0.59; 1.70]
1.50 

[0.90; 2.49]
0.95  

[0.59; 1.53]
0.88 

[0.53; 1.47]
1.16 

[0.73; 1.85]
1.11 

[0.69; 1.77]
2.79 

[1.01; 7.72]

1.04 
[0.70; 1.54]

1.19 
[0.89; 1.59]

1.15 
[0.81; 1.64]

1.00 
[0.59; 1.70]

IL-2 + Interferon +5-FU 
1.50 

[1.03; 2.19]
0.95 

[0.68; 1.33]
0.88 

[0.60; 1.30]
1.16 

[0.84; 1.60]
1.11 

[0.80; 1.54]
2.79  

[1.07; 7.28]

0.69 
[0.48; 1.00]

0.79 
[0.62; 1.01]

0.77
[0.56; 1.06]

0.67 
[0.40; 1.11]

0.67 
[0.46; 0.97]

Interferon-alpha
0.63  

[0.47; 0.85]
0.59 

[0.41; 0.84]
0.77 

[0.58; 1.03]
0.74 

[0.55; 0.99]
1.86 

[0.72; 4.80]

1.09 
[0.79; 1.50]

1.25 
[1.06; 1.48]

1.21 
[0.93; 1.57]

1.05 
[0.65; 1.69]

1.05 
[0.75; 1.46]

1.58 
[1.17; 2.12]

Pazopanib
0.92 

[0.68; 1.26]
1.22 

[0.98; 1.52]
1.16 

[0.93; 1.46]
2.93 

[1.16; 7.43]

1.18 
[0.80; 1.72]

1.35
[1.04; 1.75]

1.31 
[0.94; 1.82]

1.14 
[0.68; 1.90]

1.14 
[0.77; 1.67]

1.70 
[1.19; 2.44]

1.08 
[0.79; 1.47]

Pembrolizumab
1.32 

[0.98; 1.77]
1.26 

[0.93; 1.71]
3.17 

[1.22; 8.21]

0.89 
[0.65; 1.22]

1.03 
[0.89; 1.19]

1.00 
[0.78; 1.28]

0.86 
[0.54; 1.37]

0.86 
[0.62; 1.19]

1.29 
[0.97; 1.72]

0.82 
[0.66; 1.02]

0.76 
[0.56; 1.02]

Sorafenib
0.96 

[0.77; 1.18]
2.41 

[0.95; 6.08]

0.93 
[0.68; 1.28]

1.07 
[0.92; 1.26]

1.04
[0.81; 1.34]

0.90 
[0.56; 1.44]

0.90 
[0.65; 1.25]

1.35 
[1.01; 1.81]

0.86 
[0.68; 1.08]

0.79 
[0.59; 1.08]

1.05 
[0.85; 1.29]

Sunitinib
2.52 

[0.99; 6.37]

0.37 
[0.14; 0.97]

0.43 
[0.17; 1.07]

0.41 
[0.16; 1.06]

0.36 
[0.13; 0.99]

0.36 
[0.14; 0.94]

0.54 
[0.21; 1.39]

0.34 
[0.13; 0.87]

0.32 
[0.12; 0.82]

0.42 
[0.16; 1.05]

0.40 
[0.16; 1.01]

Thalidomide

Light blue shading shows superiority, and dark blue shading shows inferiority of the row compared with the column.

References

1. Capitanio U, Bensalah K, Bex A, Boorjian SA, Bray F, Coleman J, et 
al. Epidemiology of renal cell carcinoma. Eur Urol.2019;75(1):74-84.

2. Cindolo L, Patard JJ, Chiodini P, Schips L, Ficarra V, Tostain J, et al. 
Comparison of predictive accuracy of four prognostic models for 
nonmetastatic renal cell carcinoma after nephrectomy: a multicenter 
European study. Cancer.2005;104(7):1362-1371.

3. Eggener SE, Yossepowitch O, Pettus JA, Snyder ME, Motzer RJ, 
Russo P. Renal cell carcinoma recurrence after nephrectomy for 
localized disease: predicting survival from time of recurrence. J Clin 
Oncol.2006;24(19):3101-3106.

4. Riaz IB, He H, Ryu AJ, Siddiqi R, Naqvi SAA, Yao Y, et al. A living, 
interactive systematic review and network meta-analysis of first-line 
treatment of metastatic renal cell carcinoma. Eur Urol.2021.

5. Ljungberg B, Albiges L, Abu-Ghanem Y, Bensalah K, Dabestani S, 
Fernández-Pello S, et al. European Association of Urology Guidelines 
on renal cell carcinoma: the 2019 update. Eur Urol.2019;75(5):799-810.

6. Woods BS, Hawkins N, Scott DA. Network meta-analysis on the 
log-hazard scale, combining count and hazard ratio statistics 
accounting for multi-arm trials: a tutorial. BMC Med Res Methodol. 
2010;10:54.

7. Higgins JP, Savović J, Page MJ, Elbers RG, Sterne JA. Assessing risk of 
bias in a randomized trial. Cochrane Handbook for Systematic Reviews 
of Interventions.2019:205-228.

8. Gross-Goupil M, Kwon TG, Eto M, Ye D, Miyake H, Seo SI, et al. 
A xitinib versus placebo as an adjuvant treatment of renal cell 
carcinoma: results from the phase III, randomized ATLAS trial. Ann 
Oncol.2018;29(12):2371-2378.

350 SIUJ  •  Volume 3, Number 5  •  September 2022 SIUJ.ORG

REVIEW

http://SIUJ.org


9. Chamie K, Donin NM, Klöpfer P, Bevan P, Fall B, Wilhelm O, et al. 
Adjuvant weekly girentuximab following nephrectomy for high-risk 
renal cell carcinoma: The ARISER Randomized Clinical Trial. JAMA 
Oncol.2017;3(7):913-920.

10. Passalacqua R, Caminiti C, Buti S, Porta C, Camisa R, Braglia L, et 
al. Adjuvant low-dose interleukin-2 (IL-2) plus interferon-α (IFN-α) 
in operable renal cell carcinoma (RCC): a phase III, randomized, 
multicentre trial of the Italian Oncology Group for Clinical Research 
(GOIRC). J Immunother.2014;37(9):440-447.

11. Aitchison M, Bray CA, Van Poppel H, Sylvester R, Graham J, Innes C, 
et al. Adjuvant 5-flurouracil, alpha-interferon and interleukin-2 versus 
observation in patients at high risk of recurrence after nephrectomy 
for renal cell carcinoma: results of a phase III randomised European 
Organisation for Research and Treatment of Cancer (Genito-Urinary 
Cancers Group)/National Cancer Research Institute trial. Eur J 
Cancer.2014;50(1):70-77.

12. Messing EM, Manola J, Wilding G, Propert K, Fleischmann J, Crawford 
ED, et al. 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(7):1214-1222.

13. Pizzocaro G, Piva L, Colavita M, Ferri S, Artusi R, Boracchi P, 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(2):425-431.

14. Hinotsu S, Kawai K, Ozono S, Tsushima T, Tokuda N, Nomata 
K, et al. Randomized controlled study of natural interferon α as 
adjuvant treatment for stage II or III renal cell carcinoma. Int J Clin 
Oncol.2013;18(1):68-74.

15. Motzer RJ, Haas NB, Donskov F, Gross-Goupil M, Varlamov S, 
Kopyltsov E, et al. Randomized phase III trial of adjuvant pazopanib 
versus placebo after nephrectomy in patients with localized or locally 
advanced renal cell carcinoma. J Clin Oncol.2017;35(35):3916-3923.

TABLE 3. 

Matrix comparing hazard ratios [confidence intervals] for DFS between all therapies 

Axitinib
1.15 

[0.87; 1.52]
1.11 

[0.79; 1.57]
0.97 

[0.57; 1.63]
0.97 

[0.65; 1.44]
1.45 

[1.00; 2.10]
0.92 

[0.67; 1.27]
0.85 

[0.58; 1.24]
1.12 

[0.82; 1.53]
1.07 

[0.78; 1.47]
2.69 

[1.04; 7.01]

0.87 
[0.66; 1.15]

Control
0.97 

[0.79; 1.19]
0.84 

[0.54; 1.31]
0.84 

[0.63; 1.12]
1.26 

[0.99; 1.61]
0.80 

[0.68; 0.95]
0.74 

[0.57; 0.96]
0.97

[0.84; 1.13]
0.93 

[0.80; 1.09]
2.34 

[0.94; 5.86]

0.90 
[0.64; 1.26]

1.03 
[0.84; 1.26]

Girentuximab
0.87 

[0.53; 1.41]
0.87 

[0.61; 1.23]
1.30 

[0.95; 1.79]
0.82  

[0.64; 1.07]
0.76 

[0.55; 1.06]
1.00 

[0.78; 1.29]
0.96 

[0.74; 1.24]
2.42 

[0.95; 6.17]

1.04 [
0.61; 1.75]

1.19 
[0.76; 1.85]

1.15 
[0.71; 1.88]

IL-2 + Interferon
1.00 

[0.59; 1.70]
1.50 

[0.90; 2.49]
0.95  

[0.59; 1.53]
0.88 

[0.53; 1.47]
1.16 

[0.73; 1.85]
1.11 

[0.69; 1.77]
2.79 

[1.01; 7.72]

1.04 
[0.70; 1.54]

1.19 
[0.89; 1.59]

1.15 
[0.81; 1.64]

1.00 
[0.59; 1.70]

IL-2 + Interferon +5-FU 
1.50 

[1.03; 2.19]
0.95 

[0.68; 1.33]
0.88 

[0.60; 1.30]
1.16 

[0.84; 1.60]
1.11 

[0.80; 1.54]
2.79  

[1.07; 7.28]

0.69 
[0.48; 1.00]

0.79 
[0.62; 1.01]

0.77
[0.56; 1.06]

0.67 
[0.40; 1.11]

0.67 
[0.46; 0.97]

Interferon-alpha
0.63  

[0.47; 0.85]
0.59 

[0.41; 0.84]
0.77 

[0.58; 1.03]
0.74 

[0.55; 0.99]
1.86 

[0.72; 4.80]

1.09 
[0.79; 1.50]

1.25 
[1.06; 1.48]

1.21 
[0.93; 1.57]

1.05 
[0.65; 1.69]

1.05 
[0.75; 1.46]

1.58 
[1.17; 2.12]

Pazopanib
0.92 

[0.68; 1.26]
1.22 

[0.98; 1.52]
1.16 

[0.93; 1.46]
2.93 

[1.16; 7.43]

1.18 
[0.80; 1.72]

1.35
[1.04; 1.75]

1.31 
[0.94; 1.82]

1.14 
[0.68; 1.90]

1.14 
[0.77; 1.67]

1.70 
[1.19; 2.44]

1.08 
[0.79; 1.47]

Pembrolizumab
1.32 

[0.98; 1.77]
1.26 

[0.93; 1.71]
3.17 

[1.22; 8.21]

0.89 
[0.65; 1.22]

1.03 
[0.89; 1.19]

1.00 
[0.78; 1.28]

0.86 
[0.54; 1.37]

0.86 
[0.62; 1.19]

1.29 
[0.97; 1.72]

0.82 
[0.66; 1.02]

0.76 
[0.56; 1.02]

Sorafenib
0.96 

[0.77; 1.18]
2.41 

[0.95; 6.08]

0.93 
[0.68; 1.28]

1.07 
[0.92; 1.26]

1.04
[0.81; 1.34]

0.90 
[0.56; 1.44]

0.90 
[0.65; 1.25]

1.35 
[1.01; 1.81]

0.86 
[0.68; 1.08]

0.79 
[0.59; 1.08]

1.05 
[0.85; 1.29]

Sunitinib
2.52 

[0.99; 6.37]

0.37 
[0.14; 0.97]

0.43 
[0.17; 1.07]

0.41 
[0.16; 1.06]

0.36 
[0.13; 0.99]

0.36 
[0.14; 0.94]

0.54 
[0.21; 1.39]

0.34 
[0.13; 0.87]

0.32 
[0.12; 0.82]

0.42 
[0.16; 1.05]

0.40 
[0.16; 1.01]

Thalidomide

Light blue shading shows superiority, and dark blue shading shows inferiority of the row compared with the column.

351SIUJ.ORG SIUJ  •  Volume 3, Number 5  •  September 2022

Adjuvant Systemic Treatment for Renal Cancer After Surgery: A Network Meta-Analysis

http://SIUJ.org


16. Choueiri TK, Tomczak P, Park SH, Venugopal B, Ferguson T, Chang 
Y-H, et al. Adjuvant pembrolizumab after nephrectomy in renal-cell 
carcinoma. N Engl J Med.2021;385(8):683-694.

17. Haas NB, Manola J, Uzzo RG, Flaherty KT, Wood CG, Kane C, et al. 
Adjuvant sunitinib or sorafenib for high-risk, non-metastatic renal-cell 
carcinoma (ECOG-ACRIN E2805): a double-blind, placebo-controlled, 
randomised, phase 3 trial. Lancet.2016;387(10032):2008-2016.

18. Eisen T, Frangou E, Oza B, Ritchie AWS, Smith B, Kaplan R, et al. 
Adjuvant sorafenib for renal cell carcinoma at intermediate or high 
risk of relapse: results from the SORCE randomized phase III intergroup 
trial. J Clin Oncol.2020;38(34):4064-4075.

19. Ravaud A, Motzer RJ, Pandha HS, George DJ, Pantuck AJ, Patel 
A, et al. Adjuvant sunitinib in high-risk renal-cell carcinoma after 
nephrectomy. N Engl J Med.2016;375(23):2246-2254.

20. Margulis V, Matin SF, Tannir N, Tamboli P, Shen Y, Lozano M, et al. 
Randomized trial of adjuvant thalidomide versus observation in 
patients with completely resected high-risk renal cell carcinoma. 
Urology.2009;73(2):337-341.

21. Bai Y, Li S, Jia Z, Ding Y, Gu C, Yang J. Adjuvant therapy for locally 
advanced renal cell carcinoma: a meta-analysis and systematic review. 
Urol Oncol.2018;36(2):79.e1-e10.

22. Massari F, Di Nunno V, Mollica V, Graham J, Gatto L, Heng D. Adjuvant 
tyrosine kinase inhibitors in treatment of renal cell carcinoma: 
a meta-analysis of available clinical trials. Clin Genitourin Cancer. 
2019;17(2):e339-e344.

23. Pourmir I, Noel J, Simonaggio A, Oudard S, Vano YA. Update on the most 
promising biomarkers of response to immune checkpoint inhibitors in 
clear cell renal cell carcinoma. World J Urol.2021;39(5):1377-1385.

24. Efficace F, Fayers P, Pusic A, Cemal Y, Yanagawa J, Jacobs M, et 
al. Quality of patient-reported outcome reporting across cancer 
randomized controlled trials according to the CONSORT patient-
repor ted outcome extension: a pooled analysis of 557 trials. 
Cancer.2015;121(18):3335-3342.

25. Chien C-R, Geynisman DM, Kim B, Xu Y, Shih Y-CT. Economic 
burden of renal cell carcinoma-par t I: an updated review. 
Pharmacoeconomics.2019;37(3):301-331.

26. Watson TR, Gao X, Reynolds KL, Kong CY. Cost-effectiveness of 
pembrolizumab plus axitinib vs nivolumab plus ipilimumab as first-line 
treatment of advanced renal cell carcinoma in the US. JAMA Netw 
Open.2020;3(10):e2016144.

352 SIUJ  •  Volume 3, Number 5  •  September 2022 SIUJ.ORG

REVIEW

http://SIUJ.org