Background: The relationship between adiposity and renal cell carcinoma is poorly understood. Prior studies have suggested body mass index (BMI) may be associated with indolent disease.

Methods: We reviewed the clinicopathologic records of 845 patients across 14 countries who were enrolled in a prospective, placebo-controlled study of adjuvant girentuximab treatment for high-risk renal cell carcinoma. Clinical features analyzed included age, gender, race, BMI, and performance status. BMI was stratified into <25 kg/m2, 25.0–29.9 kg/m2, 30.0–34.9 kg/m2, and ≥35 kg/m2. We examined the association of BMI with stage and survival using logistic and Cox regression analyses, respectively.

Results: 845 patients were included for analysis. The majority (72%) were overweight/obese. There was an inverse relationship between BMI and lymph node involvement (P = 0.04). Obesity was associated with improved disease-free and overall survival (log rank <0.01 for both). When compared with normal weight subjects, those with a BMI 30–34.9 [HR 0.50; 95% confidence interval (CI) 0.31–0.81] and BMI ≥35 (HR 0.24; 95% CI 0.09–0.60) had significantly improved overall survival. A trend towards improved disease-free survival was found among subjects with BMI 30–34.9 (HR 0.77; 95% CI 0.56–1.05) and ≥35 (HR 0.74; 95% CI, 0.48–1.15).

Conclusions: In a prospective cohort of nephrectomized patients with high-risk disease, obesity is associated with lower risk of lymphatic spread and improved overall survival.

Impact: This is the first study utilizing data from a prospective randomized trial reporting an association between obesity and improved overall survival for patients with clear cell renal cell carcinoma. Cancer Epidemiol Biomarkers Prev; 25(9); 1326–32. ©2016 AACR.

This article is featured in Highlights of This Issue, p. 1279

The relationship between adiposity and renal cell carcinoma (RCC) is poorly understood. Virtually all epidemiologic studies, including case–control and cohort studies from Asia, Europe, and the United States have identified obesity or high relative weight as a risk factor for the development of RCC (1), making obesity one of the few well-established risk factors for RCC. Various putative biological mechanisms have been hypothesized to explain the relationship between increased adiposity and RCC carcinogenesis, including chronic tissue hypoxia, altered hormonal milieu within adipose tissue, and immune dysfunction (2, 3). Indeed, natural and synthetic estrogens are known to induce renal neoplasms in Syrian hamsters with an incidence approaching 100% (4). Some have estimated almost 30% of RCC may be attributable to increased adiposity (5).

The rising prevalence of obesity in the United States over the past 4 decades (6, 7) has largely been paralleled by increases in the incidence of predominantly small, asymptomatic low-stage RCC (8), resulting in an overall downward stage migration. Similar parallel increases in obesity and low-stage RCC have been observed in countries with relatively low rates of body mass index (BMI), such as Japan and South Korea (9–14). While much of this increase likely results from the detection of incidental, small, asymptomatic tumors by imaging studies performed for unrelated reasons, some have questioned whether other factors may be contributing to the rise of RCC. However, despite the downward stage migration in RCC and seemingly appropriate increases in treatment, attendant decreases in advanced stage disease or mortality from RCC have not been realized (8, 15). Attempts to elucidate the relationship between obesity and mortality from RCC have had both conflicting and paradoxical results. Both single institution retrospective series (16–18) and large meta-analyses (19) have concluded that despite the accepted association between adiposity and RCC carcinogenesis, adiposity is also an independent predictor for improved survival from RCC. This so-called “obesity paradox” was not supported by other investigations, which concluded obesity is not an independent prognostic factor after controlling for stage and grade (20, 21). Definitive conclusions have been limited by the nature of the above studies, the majority of which are single-institution series with data collected in retrospective fashion, and as such, at present the relationship between adiposity and survival from RCC remains unclear.

To our knowledge, no studies to date have evaluated the relationship between adiposity and RCC survival in prospectively collected data, the reliability of which may exceed that of retrospectively collected data (22). Our objective was to examine the relationship between adiposity, measured as BMI, and pathologic and survival outcomes of patients with ccRCC in a large prospective clinical trial of patients with ccRCC.

Study design

The ARISER Trial (NCT00087022) was a prospective, randomized, international, placebo-controlled trial comparing the efficacy and safety of adjuvant girentuximab versus placebo in patients with clinically localized, completely resected clear cell RCC (ccRCC) at high risk of recurrence. Girentuximab is a chimeric mAb specific for carbonic anhydrase IX, which is ubiquitously expressed in ccRCC and minimally expressed in normal tissues. Patients were eligible for inclusion in the ARISER trial if they had undergone partial or radical nephrectomy for ccRCC and were deemed to be at high risk for cancer recurrence by meeting one of the following high-risk pathologic criteria, based on the 6th edition UICC 2002 TNM staging: (1) pT3/T4 Nx/N0/M0, (2) pTany N+/M0, or (3) pT1b/T2 Nx/N0/M0 and high-grade. All pathologic specimens were centrally reviewed for assessment of tumor typing (i.e. confirmation of the clear cell histology, as well as grade and stage). Pathology for node-positive patients (risk group 2) was reviewed centrally to confirm lymph node metastasis. Eligible patients underwent baseline assessment no less than 3 weeks prior to randomization, at which time clinical data including age, gender, race, Eastern Cooperative Oncology Group (ECOG) performance status, height in meters, weight in kilograms, were collected and recorded in case report forms. Randomization took place not more than 12 weeks from the date of nephrectomy. Patients were randomized to drug or placebo arm and were followed for disease recurrence with CT imaging every three months for the first two years, every 6 months for the next two years, and annually thereafter until final analysis. A copy of the protocol and proposed informed consent form were reviewed and approved by the relevant Institutional Review Board or Independent Ethics Committee prior to the enrolment of the first patient at each participating site. The study sponsor and investigators were to abide by the principles of the International Conference on Harmonization Guideline for Good Clinical Practice, the latest version of the Declaration of Helsinki, as well as any local regulations.

Patient cohort

The study took place at 142 centers in 14 countries on three continents from 2004 to 2008. In total, 864 patients were accrued and randomized to two treatment arms. Of these, 19 patients were excluded from our analysis because of incomplete data regarding BMI, grade, or ECOG performance status, resulting in a final patient cohort of 845 patients.

Statistical analysis

BMI was calculated as weight in kilograms divided by height in meters squared, and stratified into the following groups: <25, 25–29.9, 30–34.5, ≥35. Descriptive statistics were calculated for the study cohort. We evaluated the association between clinicopathologic variables using bivariable analysis. Differences between groups were tested using student's t and χ2 tests for continuous and categorical variables, respectively. We then explored associations between independent predictor variables and the outcome of lymph node involvement using multivariable logistic regression analysis. Disease-free survival was calculated from the date of randomization to the date of documented relapse; patients with no documented relapse were censored at the date of their last evaluation on study. Relapse was defined as signs of metastatic disease or local recurrence as confirmed by CT, death (excluding deaths unrelated to the disease), or start of new antitumor therapy. Overall survival was calculated from the date of randomization to the date of documented death; patients with no documented death were censored at the date of their last evaluation on study (the last date the patient was known to be alive). Kaplan–Meier survival analysis was used to estimate disease-free and overall survival and tests of equality were quantified using the log-rank test. HRs for disease-free survival and overall survival were generated using multivariable Cox regression analysis. All statistical tests were two-tailed, with the probability of type 1 error set at 0.05. Statistical analysis was performed using STATA 12.1.

We identified 845 patients with complete clinical, pathologic, and follow-up data for inclusion in our analysis. The cohort was 67% male, 94% Caucasian, 86% ECOG status 0, 67% European, with a median age of 58 [interquartile range (IQR) = 51–65]. Pathologic stage T3/T4 tumors made up 84% of the cohort, and 65% of tumors were high grade. Median BMI was 27.4 (24.7–30.8) kg/m2 (Table 1). Median follow-up for the entire cohort was 54 months (IQR = 43–61), during which time 381 patients (45%) experienced disease recurrence and 178 patients (21%) died. The median time period between nephrectomy and baseline BMI measurement was 57 days (IQR = 43–75).

Table 1.

Bivariable analysis stratified by BMI category

BMI category (kg/m2)<2525–29.930–34.9≥35TotalP
Age group      0.34 
 <50 51 (22) 71 (19.5) 29 (17.7) 17 (20.2) 168 (19.9)  
 50–59 79 (34.1) 137 (37.5) 64 (39) 38 (45.2) 318 (37.6)  
 60–69 61 (26.3) 113 (31) 52 (31.7) 21 (25) 247 (29.2)  
 ≥70 41 (17.7) 44 (12.1) 19 (11.6) 8 (9.5) 112 (13.3)  
Age 58 (51–66) 58 (51–65) 58 (52–64) 58 (51–63)  0.80 
Sex      0.018 
 Female 75 (32.3) 106 (29) 66 (40.2) 36 (42.9) 283 (33.5)  
 Male 157 (67.7) 259 (71) 98 (59.8) 48 (57.1) 562 (66.5)  
Race      0.48 
 Caucasian 221 (95.3) 342 (93.7) 154 (93.9) 76 (90.5) 793 (93.8)  
 Other 11 (4.7) 23 (6.3) 10 (6.1) 8 (9.5) 52 (6.2)  
Treatment      0.71 
 Placebo 113 (48.7) 189 (51.8) 81 (49.4) 38 (45.2) 421 (49.8)  
 Girentuximab 119 (51.3) 176 (48.2) 83 (50.6) 46 (54.8) 424 (50.2)  
ECOGa      0.21 
 0 193 (83.2) 323 (88.5) 139 (84.8) 69 (82.1) 724 (85.7)  
 1 39 (16.8) 42 (11.5) 25 (15.2) 15 (17.9) 121 (14.3)  
Continent      <0.01 
 North America 41 (17.7) 76 (20.8) 51 (31.1) 53 (63.1) 221 (26.2)  
 South America 13 (5.6) 29 (7.9) 13 (7.9) 1 (1.2) 56 (6.6)  
 Europe 178 (76.7) 260 (71.2) 100 (61) 30 (35.7) 568 (67.2)  
T Stage      0.51 
 T1 13 (5.6) 22(6) 10 (6.1) 8 (9.5) 53 (6.3)  
 T2 24 (10.3) 32 (8.8) 15 (9.1) 12 (14.3) 83 (9.8)  
 T3 192 (82.8) 298 (81.6) 133 (81.1) 63 (75) 686 (81.2)  
 T4 3 (1.3) 13 (3.6) 6 (3.7) 1 (1.2) 23 (2.7)  
Nuclear grade      0.53 
 G1/G2 74 (31.9) 129 (35.3) 62 (37.8) 33 (39.3) 298 (35.3)  
 G3/G4 158 (68.1) 236 (64.7) 102 (62.2) 51 (60.7) 547 (64.7)  
N stage      0.04 
 N0/NX 207 (89.2) 337 (92.3) 156 (95.1) 82 (97.6) 782 (92.5)  
 N1/N2 25 (10.8) 28 (7.7) 8 (4.9) 2 (2.4) 63 (7.5)  
Total 232 (100) 365 (100) 164 (100) 84 (100) 845 (100)  
BMI category (kg/m2)<2525–29.930–34.9≥35TotalP
Age group      0.34 
 <50 51 (22) 71 (19.5) 29 (17.7) 17 (20.2) 168 (19.9)  
 50–59 79 (34.1) 137 (37.5) 64 (39) 38 (45.2) 318 (37.6)  
 60–69 61 (26.3) 113 (31) 52 (31.7) 21 (25) 247 (29.2)  
 ≥70 41 (17.7) 44 (12.1) 19 (11.6) 8 (9.5) 112 (13.3)  
Age 58 (51–66) 58 (51–65) 58 (52–64) 58 (51–63)  0.80 
Sex      0.018 
 Female 75 (32.3) 106 (29) 66 (40.2) 36 (42.9) 283 (33.5)  
 Male 157 (67.7) 259 (71) 98 (59.8) 48 (57.1) 562 (66.5)  
Race      0.48 
 Caucasian 221 (95.3) 342 (93.7) 154 (93.9) 76 (90.5) 793 (93.8)  
 Other 11 (4.7) 23 (6.3) 10 (6.1) 8 (9.5) 52 (6.2)  
Treatment      0.71 
 Placebo 113 (48.7) 189 (51.8) 81 (49.4) 38 (45.2) 421 (49.8)  
 Girentuximab 119 (51.3) 176 (48.2) 83 (50.6) 46 (54.8) 424 (50.2)  
ECOGa      0.21 
 0 193 (83.2) 323 (88.5) 139 (84.8) 69 (82.1) 724 (85.7)  
 1 39 (16.8) 42 (11.5) 25 (15.2) 15 (17.9) 121 (14.3)  
Continent      <0.01 
 North America 41 (17.7) 76 (20.8) 51 (31.1) 53 (63.1) 221 (26.2)  
 South America 13 (5.6) 29 (7.9) 13 (7.9) 1 (1.2) 56 (6.6)  
 Europe 178 (76.7) 260 (71.2) 100 (61) 30 (35.7) 568 (67.2)  
T Stage      0.51 
 T1 13 (5.6) 22(6) 10 (6.1) 8 (9.5) 53 (6.3)  
 T2 24 (10.3) 32 (8.8) 15 (9.1) 12 (14.3) 83 (9.8)  
 T3 192 (82.8) 298 (81.6) 133 (81.1) 63 (75) 686 (81.2)  
 T4 3 (1.3) 13 (3.6) 6 (3.7) 1 (1.2) 23 (2.7)  
Nuclear grade      0.53 
 G1/G2 74 (31.9) 129 (35.3) 62 (37.8) 33 (39.3) 298 (35.3)  
 G3/G4 158 (68.1) 236 (64.7) 102 (62.2) 51 (60.7) 547 (64.7)  
N stage      0.04 
 N0/NX 207 (89.2) 337 (92.3) 156 (95.1) 82 (97.6) 782 (92.5)  
 N1/N2 25 (10.8) 28 (7.7) 8 (4.9) 2 (2.4) 63 (7.5)  
Total 232 (100) 365 (100) 164 (100) 84 (100) 845 (100)  

NOTE: Continuous variables displayed as median (IQR).

Abbreviation: ECOG, Eastern Cooperative Oncology Group.

There was no significant difference in BMI distribution between T-stages (P = 0.51; Table 1). The proportion of patients with lymph node involvement progressively decreased with each increase in BMI category: 11% in those with BMI <25, 8% in those with BMI 25–29.9, 5% in those with BMI 30–34.5, and 2% in those with BMI ≥35. This finding was consistent when using varying cut-off points for BMI. In multivariable logistic regression analysis, as BMI increased, the rate of lymph node metastasis decreased (Table 2). When compared with men with a normal BMI, those with a BMI of ≥30 [OR 0.34; 95% confidence interval (CI), 0.15–0.76] had a significantly lower odds of lymphatic involvement.

Table 2.

Multivariable logistic regression model for prediction of lymph node involvement

OR (95% CI)P
Age group 
 <50 1.00 (referent) *** 
 50–59 0.41 (0.2–0.8) 0.01 
 60–69 0.72 (0.36–1.4) 0.33 
 ≥70 0.31 (0.11–0.87) 0.03 
Gender 
 Female 1.00 (referent) *** 
 Male 0.81 (0.46–1.42) 0.47 
Race/ethnicity 
 Other 1.00 (referent) *** 
 White 4.4 (0.57–33.88) 0.16 
BMI (kg/m2
 <25 1.00 (referent) *** 
 25.0–29.9 0.69 (0.38–1.24) 0.21 
 ≥30 0.34 (0.15–0.76) 0.01 
ECOG Performance status 
 0 1.00 (referent) *** 
 1 0.79 (0.34–1.81) 0.57 
Continent 
 North America 1.00 (referent) *** 
 South America 1.15 (0.34–3.83) 0.82 
 Europe 1.2 (0.61–2.39) 0.6 
T Stage 
 T1 1.00 (referent) *** 
 T2 0.62 (0.22–1.77) 0.38 
 T3 0.33 (0.14–0.78) 0.01 
 T4 0.5 (0.09–2.69) 0.42 
Nuclear grade 
 Low 1.00 (referent) *** 
 High 1.16 (0.63–2.14) 0.63 
OR (95% CI)P
Age group 
 <50 1.00 (referent) *** 
 50–59 0.41 (0.2–0.8) 0.01 
 60–69 0.72 (0.36–1.4) 0.33 
 ≥70 0.31 (0.11–0.87) 0.03 
Gender 
 Female 1.00 (referent) *** 
 Male 0.81 (0.46–1.42) 0.47 
Race/ethnicity 
 Other 1.00 (referent) *** 
 White 4.4 (0.57–33.88) 0.16 
BMI (kg/m2
 <25 1.00 (referent) *** 
 25.0–29.9 0.69 (0.38–1.24) 0.21 
 ≥30 0.34 (0.15–0.76) 0.01 
ECOG Performance status 
 0 1.00 (referent) *** 
 1 0.79 (0.34–1.81) 0.57 
Continent 
 North America 1.00 (referent) *** 
 South America 1.15 (0.34–3.83) 0.82 
 Europe 1.2 (0.61–2.39) 0.6 
T Stage 
 T1 1.00 (referent) *** 
 T2 0.62 (0.22–1.77) 0.38 
 T3 0.33 (0.14–0.78) 0.01 
 T4 0.5 (0.09–2.69) 0.42 
Nuclear grade 
 Low 1.00 (referent) *** 
 High 1.16 (0.63–2.14) 0.63 

Abbreviation: ECOG, Eastern Cooperative Oncology Group.

Kaplan–Meier analysis demonstrated that with each increase in BMI category there was progressively prolongation and a statistically significant difference in both disease-free and overall survival (log rank <0.01 for both, Figs. 1 and 2). Multivariable Cox proportional hazard models were used to generate HRs for disease-free survival and overall survival across categories of BMI. Covariates for both models included age, gender, race, ECOG performance status, Continent (Europe, North America, South America), T-stage, N-stage, and nuclear grade. When compared with patients with a normal BMI, there was a nonsignificant trend toward improved disease-free survival in patients with a BMI of 30–34.9 (HR 0.77; 95% CI, 0.56–1.05) and BMI ≥35 (HR 0.74; 95% CI, 0.48–1.15; Table 3). When compared with patients with a normal BMI, a BMI of 30–34.9 (HR 0.50; 95% CI, 0.31–0.81), and BMI ≥35 (HR 0.24; 95% CI, 0.09–0.60) were both associated with improved overall survival (Table 3).

Figure 1.

Kaplan–Meier estimates of disease-free survival by BMI category.

Figure 1.

Kaplan–Meier estimates of disease-free survival by BMI category.

Close modal
Figure 2.

Kaplan–Meier estimates of overall survival by BMI category.

Figure 2.

Kaplan–Meier estimates of overall survival by BMI category.

Close modal
Table 3.

Multivariable COX regression analysis of disease-free and overall survival

Disease-free survivalOverall survival
HR (95% CI)PHR (95% CI)P
Age group 
 <50 1.00 (referent) *** 1.00 (referent) *** 
 50–59 1.27 (0.93–1.74) 0.14 1.21 (0.74–1.97) 0.45 
 60–69 1.89 (1.38–2.60) <0.01 1.99 (1.24–3.21) <0.01 
 ≥70 2.23 (1.54–3.22) <0.01 2.70 (1.62–4.50) <0.01 
Gender 
 Female 1.00 (referent) *** 1.00 (referent) *** 
 Male 1.00 (0.80–1.24) 0.97 0.96 (0.69–1.32) 0.79 
Race/ethnicity 
 Other 1.00 (referent) *** 1.00 (referent) *** 
 White 0.87 (0.55–1.38) 0.57 1.01 (0.49–2.11) 0.97 
BMI (kg/m2
 <25 1.00 (referent) *** 1.00 (referent) *** 
 25.0–29.9 0.91 (0.72–1.16) 0.46 0.73 (0.53–1.01) 0.06 
 30.0–34.9 0.77 (0.56–1.05) 0.09 0.50 (0.31–0.81) <0.01 
 ≥35 0.74 (0.48–1.15) 0.18 0.24 (0.09–0.60) <0.01 
ECOG Performance status 
 0 1.00 (referent) *** 1.00 (referent) *** 
 1 1.20 (0.91–1.58) 0.19 1.18 (0.79–1.78) 0.42 
Continent 
 North America 1.00 (referent) *** 1.00 (referent) *** 
 South America 0.99 (0.62–1.59) 0.98 1.18 (0.62–2.24) 0.62 
 Europe 1.18 (0.91–1.52) 0.20 1.07 (0.73–1.55) 0.73 
T Stage 
 T1 1.00 (referent) *** 1.00 (referent) *** 
 T2 2.95 (1.53–5.69) <0.01 1.28 (0.54–3.05) 0.57 
 T3 3.23 (1.80–5.79) <0.01 1.86 (0.90–3.84) 0.09 
 T4 4.91 (2.21–10.92) <0.01 1.09 (0.29–4.14) 0.90 
N Stage 
 Nx/N0 1.00 (referent) *** 1.00 (referent) *** 
 N+ 3.23 (2.37–4.40) <0.01 3.84 (2.60–5.68) <0.01 
Nuclear grade 
 Low 1.00 (referent) *** 1.00 (referent) *** 
 High 1.98 (1.56–2.52) <0.01 2.12 (1.48–3.05) <0.01 
Disease-free survivalOverall survival
HR (95% CI)PHR (95% CI)P
Age group 
 <50 1.00 (referent) *** 1.00 (referent) *** 
 50–59 1.27 (0.93–1.74) 0.14 1.21 (0.74–1.97) 0.45 
 60–69 1.89 (1.38–2.60) <0.01 1.99 (1.24–3.21) <0.01 
 ≥70 2.23 (1.54–3.22) <0.01 2.70 (1.62–4.50) <0.01 
Gender 
 Female 1.00 (referent) *** 1.00 (referent) *** 
 Male 1.00 (0.80–1.24) 0.97 0.96 (0.69–1.32) 0.79 
Race/ethnicity 
 Other 1.00 (referent) *** 1.00 (referent) *** 
 White 0.87 (0.55–1.38) 0.57 1.01 (0.49–2.11) 0.97 
BMI (kg/m2
 <25 1.00 (referent) *** 1.00 (referent) *** 
 25.0–29.9 0.91 (0.72–1.16) 0.46 0.73 (0.53–1.01) 0.06 
 30.0–34.9 0.77 (0.56–1.05) 0.09 0.50 (0.31–0.81) <0.01 
 ≥35 0.74 (0.48–1.15) 0.18 0.24 (0.09–0.60) <0.01 
ECOG Performance status 
 0 1.00 (referent) *** 1.00 (referent) *** 
 1 1.20 (0.91–1.58) 0.19 1.18 (0.79–1.78) 0.42 
Continent 
 North America 1.00 (referent) *** 1.00 (referent) *** 
 South America 0.99 (0.62–1.59) 0.98 1.18 (0.62–2.24) 0.62 
 Europe 1.18 (0.91–1.52) 0.20 1.07 (0.73–1.55) 0.73 
T Stage 
 T1 1.00 (referent) *** 1.00 (referent) *** 
 T2 2.95 (1.53–5.69) <0.01 1.28 (0.54–3.05) 0.57 
 T3 3.23 (1.80–5.79) <0.01 1.86 (0.90–3.84) 0.09 
 T4 4.91 (2.21–10.92) <0.01 1.09 (0.29–4.14) 0.90 
N Stage 
 Nx/N0 1.00 (referent) *** 1.00 (referent) *** 
 N+ 3.23 (2.37–4.40) <0.01 3.84 (2.60–5.68) <0.01 
Nuclear grade 
 Low 1.00 (referent) *** 1.00 (referent) *** 
 High 1.98 (1.56–2.52) <0.01 2.12 (1.48–3.05) <0.01 

Abbreviation: ECOG, Eastern Cooperative Oncology Group.

We utilized data from a large prospective international phase III randomized clinical trial of patients with high-risk ccRCC to investigate the relationship between BMI and lymph node metastasis, disease-free survival, and overall survival. We observed that as BMI increased, the rate of lymph node metastasis decreased, a finding that was significant despite controlling for patient and tumor characteristics. Next, we observed an association between increased BMI and improved disease-free and overall survival. Using multivariable models, patients who were obese (30–34.9) or morbidly obese (BMI ≥35) had a statistically significant overall survival advantage and enjoyed a trend towards improved disease-free survival.

Our findings support the concept of an obesity paradox in ccRCC, however, at this time, the biological mechanisms underlying this observation are not known. One potential explanation for the paradox is that RCC developing in the obese may represent a biologically distinct and less aggressive form of the disease. There is an increasing appreciation of the genomic heterogeneity within the clear cell histologic subtype, and distinct molecular subtypes of ccRCC which demonstrate divergent survival patterns have been described (23, 24). Hakami and colleagues observed that fatty acid synthase, a gene associated with poor RCC outcomes when upregulated, appeared downregulated in tumors from obese patients. This serves as a potential mechanism to explain improved survival in this population (20, 25). Various other putative biochemical pathways linking obesity to cancer progression are the subjects of active research (26).

There is conflicting evidence as to whether improved survival in the obese can be accounted for by tumor stage and grade alone. Hakami and colleagues found that BMI was associated with lower risk of cancer-specific death and overall death; however, the association was attenuated and became nonsignificant when controlling for stage and grade. The authors concluded that the improved outcomes in obese patients were attributable to lower grade and stage tumors in this population. Other retrospective studies have similarly failed to find BMI independently prognostic of overall survival when controlling for stage (27–29). We did not find an association between BMI and T-stage in our analysis; however, our study cohort consisted predominantly of patients with pT3/pT4 disease (84%) and as such there may have been insufficient variability in stage to detect an association. We did, however, find that increases in BMI were associated with a lower risk of lymphatic involvement, supporting the concept that tumors forming in an obesogenic environment may harbor less aggressive biology, even among those with high risk of recurrence. In other words, our finding that BMI was positively associated with improved overall survival even when controlling for T-stage, N-stage, and grade, suggests that the effect of obesity is not completely accounted for in the stage and grade of tumors.

One alternative explanation for the obesity paradox may relate to the role that disease–host as well as treatment–host interactions play in modulating overall disease course. Body adiposity is known to be associated with a state of chronic systemic inflammation, with persistently elevated levels of proinflammatory cytokines including C-reactive protein (CRP), tumor necrosis factor (TNF), and IL6 and IL18, among others (30). Given the propensity of RCC to respond to immunotherapeutic treatment approaches (31, 32), the question is raised of whether these chronically elevated cytokine levels exert some form of antitumor effect. This hypothesis, however, appears unlikely given the absence of an obesity paradox among other obesity-related cancers, such as breast and colorectal carcinomas (33). An alternative explanation is that BMI may be a reflection of physiologic host reserve and overall wellness, which may confer a survival advantage, particularly in diseases associated with wasting, such as cancer. This hypothesis is supported by studies evaluating the relationship between the skeletal muscle compartment and survival. Studies of both localized and metastatic RCC have demonstrated that sarcopenia, or decreased skeletal muscle mass (34), as well as decreased skeletal muscle density (35) independently predicts worse cancer-specific and overall survival. While this may reflect greater host reserve in a disease known to cause wasting, the known propensity of the targeted agents used in the metastatic setting to exacerbate muscle wasting (36, 37), potentially leading to dose-limiting toxicities in those with pre-existing muscle wasting may also contribute to this phenomenon. Interestingly, obesity and sarcopenia are not mutually exclusive, and within each strata of BMI there is a wide range of skeletal muscle mass, including obese patients with cachectic levels of skeletal muscle mass (35, 38). Various other image-based metrics of body composition have been evaluated, including visceral adipose tissue, subcutaneous adipose tissue, and what is clear is that the relationship between body composition, both within adipose and muscular compartments, and cancer-specific outcomes is complex and somewhat poorly understood at this time (39).

Our findings should be considered in light of the increasing appreciation that many incidentally diagnosed small renal masses (<4cm) may display an indolent natural history if left untreated. The concept of the indolent small renal mass is supported by multiple epidemiologic observations, including the failure of mortality rates to decrease in the face of downward stage migration and seemingly appropriate treatment of these masses (15), along with the relative stability in incidence of more advanced stage disease (8). These observations suggest the possibility that many of these masses, if left untreated, would not be destined to cause clinically significant disease. This has raised the question of potential overtreatment of these small, incidentally detected kidney tumors (40). These considerations have resulted in the relatively infrequent but increasing use of active surveillance as a management strategy for small renal masses (41, 42). Active surveillance is particularly relevant for patients in whom there is significant perioperative risk, such as the elderly or those with severe comorbidities, or in those in whom treatment may be complex or high risk, such as in the obese patient. Given the high prevalence of obesity, coupled with the fact that the majority of newly diagnosed renal tumors are <4 cm (43) urologists are now frequently faced with the dilemma of managing an obese patient with a renal mass of unclear malignant potential. Our study suggests that in obese patients, irrespective of the underlying mechanism, renal masses may be less aggressive than those of their leaner counterparts, and that active surveillance of these masses may be oncologically sound. This, combined with the recognition of increased risks inherent in operating on the obese (44, 45), may shift the risk–benefit ratio away from intervention.

Using data from large international prospective clinical trial of surgically treated patients with high-risk ccRCC, we found that increasing BMI was associated with a lower risk of lymph node metastasis and a decreased risk of death. These findings add to the growing number of studies demonstrating a seemingly paradoxical relationship between BMI and survival in patients with ccRCC. Whether these observations relate specifically to BMI, or are rather a reflection of differences in body composition remains unknown. Molecular characterization of ccRCC and studies of tumor–host interactions in patients with different body composition will be required to understand this relationship.

P. Bevan is a director and a consultant/advisory board member at Wilex AG. A.S. Belldegrun is a consultant for Wilex AG. No potential conflicts of interest were disclosed by the other authors.

Conception and design: N.M. Donin, A. Pantuck, P. Bevan, A.S. Belldegrun, K. Chamie

Development of methodology: N.M. Donin, A. Pantuck, J. Said, K. Chamie

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): A. Pantuck, J. Said, A.S. Belldegrun

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): N.M. Donin, A. Pantuck, P. Bevan, J. Said, A.S. Belldegrun, K. Chamie

Writing, review, and/or revision of the manuscript: N.M. Donin, A. Pantuck, P. Bevan, B. Fall, A.S. Belldegrun, K. Chamie

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): N.M. Donin, K. Chamie

Study supervision: P. Klöpfer, A.S. Belldegrun, K. Chamie

Other (clinical trial manager of ARISER): P. Klöpfer

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
McLaughlin
JK
,
Lipworth
L
,
Tarone
RE
. 
Epidemiologic aspects of renal cell carcinoma
.
Semin Oncol
2006
;
33
:
527
33
.
2.
Booth
A
,
Magnuson
A
,
Fouts
J
,
Foster
M
. 
Adipose tissue, obesity and adipokines: role in cancer promotion
.
Horm Mol Biol Clin Investig
2015
;
21
:
57
74
.
3.
Moyad
MA
. 
Obesity, interrelated mechanisms, and exposures and kidney cancer
.
Semin Urol Oncol
2001
;
19
:
270
9
.
4.
Li
JJ
,
Gonzalez
A
,
Banerjee
S
,
Banerjee
SK
,
Li
SA
. 
Estrogen carcinogenesis in the hamster kidney: role of cytotoxicity and cell proliferation
.
Environ Health Perspect
1993
;
101
:
259
64
.
5.
Bergstrom
A
,
Hsieh
CC
,
Lindblad
P
,
Lu
CM
,
Cook
NR
,
Wolk
A
. 
Obesity and renal cell cancer–a quantitative review
.
Br J Cancer
2001
;
85
:
984
90
.
6.
Flegal
KM
,
Carroll
MD
,
Kit
BK
,
Ogden
CL
. 
Prevalence of obesity and trends in the distribution of body mass index among US adults, 1999–2010
.
JAMA
2012
;
307
:
491
7
.
7.
Kuczmarski
RJ
,
Flegal
KM
,
Campbell
SM
,
Johnson
CL
. 
Increasing prevalence of overweight among US adults. The national health and nutrition examination surveys, 1960 to 1991
.
JAMA
1994
;
272
:
205
11
.
8.
Gandaglia
G
,
Ravi
P
,
Abdollah
F
,
Abd-El-Barr
AE
,
Becker
A
,
Popa
I
, et al
Contemporary incidence and mortality rates of kidney cancer in the United States
.
Can Urol Assoc J
2014
;
247
52
.
9.
Yoshiike
N
,
Miyoshi
M
. 
[Epidemiological aspects of overweight and obesity in Japan–international comparisons]
.
Nihon Rinsho
2013
;
71
:
207
16
.
10.
Washio
M
,
Mori
M
,
Mikami
K
,
Miki
T
,
Watanabe
Y
,
Nakao
M
, et al
Risk factors for renal cell carcinoma in a Japanese population
.
Asian Pac J Cancer Prev
2014
;
15
:
9065
70
.
11.
Jung
KW
,
Park
S
,
Kong
HJ
,
Won
YJ
,
Boo
YK
,
Shin
HR
, et al
Cancer statistics in Korea: incidence, mortality and survival in 2006–2007
.
J Korean Med Sci
2010
;
25
:
1113
21
.
12.
Kanazawa
M
,
Yoshiike
N
,
Osaka
T
,
Numba
Y
,
Zimmet
P
,
Inoue
S
. 
Criteria and classification of obesity in Japan and Asia-Oceania
.
World Rev Nutr Diet
2005
;
94
:
1
12
.
13.
Marumo
K
,
Kanayama
H
,
Miyao
N
,
Nakazawa
H
,
Ozono
S
,
Horie
S
, et al
Prevalence of renal cell carcinoma: a nation-wide survey in Japan, 2002
.
Int J Urol
2007
;
14
:
479
82
.
14.
Khang
YH
,
Yun
SC
. 
Trends in general and abdominal obesity among Korean adults: findings from 1998, 2001, 2005, and 2007 Korea national health and nutrition examination surveys
.
J Korean Med Sci
2010
;
25
:
1582
8
.
15.
Hollingsworth
JM
,
Miller
DC
,
Daignault
S
,
Hollenbeck
BK
. 
Rising incidence of small renal masses: a need to reassess treatment effect
.
J Natl Cancer Inst
2006
;
98
:
1331
4
.
16.
Jeon
HG
,
Jeong
IG
,
Lee
JH
,
Lee
CJ
,
Kwak
C
,
Kim
HH
, et al
Prognostic value of body mass index in Korean patients with renal cell carcinoma
.
J Urol
2010
;
183
:
448
54
.
17.
Parker
AS
,
Lohse
CM
,
Cheville
JC
,
Thiel
DD
,
Leibovich
BC
,
Blute
ML
. 
Greater body mass index is associated with better pathologic features and improved outcome among patients treated surgically for clear cell renal cell carcinoma
.
Urology
2006
;
68
:
741
6
.
18.
Kamat
AM
,
Shock
RP
,
Naya
Y
,
Rosser
CJ
,
Slaton
JW
,
Pisters
LL
. 
Prognostic value of body mass index in patients undergoing nephrectomy for localized renal tumors
.
Urology
2004
;
63
:
46
50
.
19.
Choi
Y
,
Park
B
,
Jeong
BC
,
Seo
SI
,
Jeon
SS
,
Choi
HY
, et al
Body mass index and survival in patients with renal cell carcinoma: a clinical-based cohort and meta-analysis
.
Int J Cancer
2013
;
132
:
625
34
.
20.
Hakimi
AA
,
Furberg
H
,
Zabor
EC
,
Jacobsen
A
,
Schultz
N
,
Ciriello
G
, et al
An epidemiologic and genomic investigation into the obesity paradox in renal cell carcinoma
.
J Natl Cancer Inst
2013
;
105
:
1862
70
.
21.
Donat
SM
,
Salzhauer
EW
,
Mitra
N
,
Yanke
BV
,
Snyder
ME
,
Russo
P
. 
Impact of body mass index on survival of patients with surgically treated renal cell carcinoma
.
J Urol
2006
;
175
:
46
52
.
22.
Nagurney
JT
,
Brown
DF
,
Sane
S
,
Weiner
JB
,
Wang
AC
,
Chang
Y
. 
The accuracy and completeness of data collected by prospective and retrospective methods
.
Acad Emerg Med
2005
;
12
:
884
95
.
23.
Brannon
AR
,
Reddy
A
,
Seiler
M
,
Arreola
A
,
Moore
DT
,
Pruthi
RS
, et al
Molecular stratification of clear cell renal cell carcinoma by consensus clustering reveals distinct subtypes and survival patterns
.
Genes Cancer
2010
;
1
:
152
63
.
24.
Scelo
G
,
Riazalhosseini
Y
,
Greger
L
,
Letourneau
L
,
Gonzalez-Porta
M
,
Wozniak
MB
, et al
Variation in genomic landscape of clear cell renal cell carcinoma across Europe
.
Nat Commun
2014
;
5
:
5135
.
25.
Horiguchi
A
,
Asano
T
,
Ito
K
,
Sumitomo
M
,
Hayakawa
M
. 
Fatty acid synthase over expression is an indicator of tumor aggressiveness and poor prognosis in renal cell carcinoma
.
J Urol
2008
;
180
:
1137
40
.
26.
Renehan
AG
,
Zwahlen
M
,
Egger
M
. 
Adiposity and cancer risk: new mechanistic insights from epidemiology
.
Nat Rev Cancer
2015
;
15
:
484
98
.
27.
Lee
WK
,
Byun
SS
,
Kim
HH
,
Rha
KH
,
Hwang
TK
,
Sung
GT
, et al
Characteristics and prognosis of chromophobe non-metastatic renal cell carcinoma: a multicenter study
.
Int J Urol
2010
;
17
:
898
904
.
28.
Pettus
JA
,
Jang
TL
,
Thompson
RH
,
Yossepowitch
O
,
Kagiwada
M
,
Russo
P
. 
Effect of baseline glomerular filtration rate on survival in patients undergoing partial or radical nephrectomy for renal cortical tumors
.
Mayo Clin Proc
2008
;
83
:
1101
6
.
29.
Schips
L
,
Lipsky
K
,
Zigeuner
R
,
Gidaro
S
,
Salfellner
M
,
Rehak
P
, et al
Does overweight impact on the prognosis of patients with renal cell carcinoma? A single center experience of 683 patients
.
J Surg Oncol
2004
;
88
:
57
61
.
30.
Osborn
O
,
Olefsky
JM
. 
The cellular and signaling networks linking the immune system and metabolism in disease
.
Nat Med
2012
;
18
:
363
74
.
31.
Motzer
RJ
,
Escudier
B
,
McDermott
DF
,
George
S
,
Hammers
HJ
,
Srinivas
S
, et al
Nivolumab versus everolimus in advanced renal-cell carcinoma
.
N Engl J Med
2015
;
373
:
1803
13
.
32.
McDermott
DF
,
Cheng
SC
,
Signoretti
S
,
Margolin
KA
,
Clark
JI
,
Sosman
JA
, et al
The high-dose aldesleukin "select" trial: a trial to prospectively validate predictive models of response to treatment in patients with metastatic renal cell carcinoma.
Clin Cancer Res
2015
;
21
:
561
8
.
33.
Parekh
N
,
Chandran
U
,
Bandera
EV
. 
Obesity in cancer survival
.
Annu Rev Nutr
2012
;
32
:
311
42
.
34.
Psutka
SP
,
Boorjian
SA
,
Moynagh
MR
,
Schmit
GD
,
Costello
BA
,
Thompson
RH
, et al
Decreased skeletal muscle mass is associated with an increased risk of mortality after radical nephrectomy for localized renal cell cancer
.
J Urol
2016
;
195
:
270
6
.
35.
Antoun
S
,
Lanoy
E
,
Iacovelli
R
,
Albiges-Sauvin
L
,
Loriot
Y
,
Merad-Taoufik
M
, et al
Skeletal muscle density predicts prognosis in patients with metastatic renal cell carcinoma treated with targeted therapies
.
Cancer
2013
;
119
:
3377
84
.
36.
Antoun
S
,
Birdsell
L
,
Sawyer
MB
,
Venner
P
,
Escudier
B
,
Baracos
VE
. 
Association of skeletal muscle wasting with treatment with sorafenib in patients with advanced renal cell carcinoma: results from a placebo-controlled study
.
J Clin Oncol
2010
;
28
:
1054
60
.
37.
Antoun
S
,
Baracos
VE
,
Birdsell
L
,
Escudier
B
,
Sawyer
MB
. 
Low body mass index and sarcopenia associated with dose-limiting toxicity of sorafenib in patients with renal cell carcinoma
.
Ann Oncol
2010
;
21
:
1594
8
.
38.
Martin
L
,
Birdsell
L
,
Macdonald
N
,
Reiman
T
,
Clandinin
MT
,
McCargar
LJ
, et al
Cancer cachexia in the age of obesity: skeletal muscle depletion is a powerful prognostic factor, independent of body mass index
.
J Clin Oncol
2013
;
31
:
1539
47
.
39.
Antoun
S
,
Lanoy
E
,
Albiges-Sauvin
L
,
Escudier
B
. 
Clinical implications of body composition assessment by computed tomography in metastatic renal cell carcinoma
.
Expert Rev Anticancer Ther
2014
;
14
:
279
88
.
40.
Welch
HG
,
Black
WC
. 
Overdiagnosis in cancer
.
J Natl Cancer Inst
2010
;
102
:
605
13
.
41.
Pierorazio
PM
,
Johnson
MH
,
Ball
MW
,
Gorin
MA
,
Trock
BJ
,
Chang
P
, et al
Five-year analysis of a multi-institutional prospective clinical trial of delayed intervention and surveillance for small renal masses: the DISSRM registry
.
Eur Urol
2015
;
68
:
408
15
.
42.
Crispen
PL
,
Viterbo
R
,
Fox
EB
,
Greenberg
RE
,
Chen
DY
,
Uzzo
RG
. 
Delayed intervention of sporadic renal masses undergoing active surveillance
.
Cancer
2008
;
112
:
1051
7
.
43.
Kane
CJ
,
Mallin
K
,
Ritchey
J
,
Cooperberg
MR
,
Carroll
PR
. 
Renal cell cancer stage migration: analysis of the national cancer data base
.
Cancer
2008
;
113
:
78
83
.
44.
Mendoza
D
,
Newman
RC
,
Albala
D
,
Cohen
MS
,
Tewari
A
,
Lingeman
J
, et al
Laparoscopic complications in markedly obese urologic patients (a multi-institutional review)
.
Urology
1996
;
48
:
562
7
.
45.
Fazeli-Matin
S
,
Gill
IS
,
Hsu
TH
,
Sung
GT
,
Novick
AC
. 
Laparoscopic renal and adrenal surgery in obese patients: comparison to open surgery
.
J Urol
1999
;
162
:
665
9
.