Purpose:

No circulating biomarkers are currently available to identify patients at highest risk of recurrence after nephrectomy for renal cell carcinoma (RCC). Kidney injury molecule-1 (KIM-1) is overexpressed in RCC and its ectodomain circulates in plasma. We investigated whether plasma KIM-1 is a prognostic biomarker in patients with localized RCC after nephrectomy.

Experimental Design:

The ECOG-ACRIN E2805 (ASSURE) trial evaluated adjuvant sunitinib, sorafenib, or placebo in resected high-risk RCC. KIM-1 levels were measured from banked plasma at trial enrollment 4–12 weeks after nephrectomy. Lognormal accelerated failure time models were used to test for association between KIM-1 and disease-free survival (DFS) as well as overall survival (OS).

Results:

Plasma from 418 patients was analyzed. Higher post-nephrectomy KIM-1 was associated with worse DFS across all study arms after adjustment for Fuhrman grade, T stage, N stage, and tumor histology [survival time ratio 0.56 for 75th vs. 25th percentile of KIM-1; 95% confidence interval (CI), 0.42–0.73; P < 0.001]. The association between KIM-1 and DFS was stronger among patients with pathologic nodal involvement (Pinteraction = 0.0086). The addition of post-nephrectomy KIM-1 improved the concordance of clinical prognostic models [Stage, Size, Grade, and Necrosis (SSIGN) concordance 0.57 vs. 0.43, P = 0.05; UCLA International Staging System (UISS) concordance 0.60 vs. 0.40, P = 0.0005]. Higher post-nephrectomy KIM-1 was also associated with worse OS after multivariable adjustment (survival time ratio 0.71 for 75th vs. 25th percentile of KIM-1; 95% CI, 0.56–0.91; P < 0.001).

Conclusions:

Post-nephrectomy plasma KIM-1 is associated with DFS and OS in RCC, and may be a biomarker for microscopic residual disease.

Translational Relevance

Disease recurrence after nephrectomy for localized renal cell carcinoma (RCC) is common, and the identification of a tumor marker could help risk stratify patients after nephrectomy, identify patients who may benefit from adjuvant treatment, and enhance surveillance. Kidney injury molecule-1 (KIM-1) is a transmembrane protein overexpressed in RCC and detectable in circulating plasma. We report that elevated post-nephrectomy KIM-1 is associated with worse progression-free survival and overall survival in banked plasma from a large randomized trial, particularly among patients with pathologic node involvement at time of nephrectomy. We also show that the inclusion of plasma KIM-1 into existing post-nephrectomy risk assessment models improves their concordance. If validated in further studies, KIM-1 is a potential circulating biomarker in RCC to identify patients who are at high risk of recurrence after nephrectomy.

In 2018, there were more than 400,000 new cases and 175,000 deaths attributed to renal cell carcinoma (RCC; ref. 1). Although some patients with localized disease are cured by surgical excision, 30% or more experience subsequent disease recurrence leading to poor long-term outcomes (2–6). Despite many recent advances in the treatment of metastatic RCC, the standard of care for high-risk localized disease after nephrectomy remains imaging surveillance alone. Although sunitinib has FDA regulatory approval status for adjuvant therapy, it is rarely used because of toxicity, lack of known survival benefit, and the inability to determine a priori which patients may benefit (2, 4, 7). These problems are compounded by the lack of a circulating biomarker for RCC. Such a biomarker, if available, could help to identify patients at high risk for recurrence after surgical excision, identify patients for adjuvant therapy, and assist in the early detection of recurrent disease.

Kidney injury molecule-1 (KIM-1) is a transmembrane glycoprotein expressed by injured renal proximal tubular cells and RCC cells (8, 9). The ectodomain of KIM-1 undergoes cleavage and can be detected in both urine and blood (10, 11). Urinary KIM-1 levels have been shown to decrease after nephrectomy in people with RCC that expresses KIM-1 (10). In addition, elevated KIM-1 levels have been identified in patients up to 5 years prior to a subsequent diagnosis of RCC, and are associated with higher risk of RCC mortality (12). These findings suggest that KIM-1 may be a marker for overall RCC tumor burden. In patients with resected RCC and no radiographic evidence of disease, KIM-1 could indicate the presence of occult tumor and therefore high risk for future recurrence. However, it is currently not known whether post-nephrectomy KIM-1 levels are associated with clinical outcomes.

We therefore performed a post hoc analysis of the ECOG-ACRIN E2805 (ASSURE) trial to determine whether post-nephrectomy plasma KIM-1 levels are associated with worse disease-free survival (DFS) among patients with high-risk localized RCC.

Patient population

ECOG-ACRIN E2805 (ASSURE) was a double-blinded, placebo-controlled phase III trial of 1,943 participants with completely resected, high-grade, pathologic stage T1b or higher RCC (2). Participants were randomized in a 1:1:1 manner to 54 weeks of treatment with adjuvant sunitinib, sorafenib, or placebo. The primary outcome was DFS, defined as time from randomization to either recurrence, development of a second primary cancer, or death from any cause. Because of an interim calculation of low conditional power for the primary endpoint, the Data Safety Monitoring Committee determined in 2014 that blinded follow-up should cease; analysis for the primary endpoint subsequently showed no difference in DFS between the study arms. Further data regarding the conduct and outcomes of the ECOG-ACRIN E2805 study have been previously reported in detail (2).

Banked plasma from ECOG-ACRIN E2805 was randomly selected and allocated by study statisticians for preplanned biomarker analyses. Plasma samples from 418 patients who had undergone total nephrectomy across all treatment arms were allocated for this KIM-1 study. Patient demographics including age, sex, race/ethnicity, pathologic information, and clinical outcomes were collected prospectively as per trial protocol. Patients who underwent partial nephrectomy (5.4% of the E2805 study cohort) were excluded from the analysis to minimize possible confounding due to KIM-1 release from injured postsurgical kidney epithelium.

All participants provided written informed consent prior to participation, and consent for exploratory biomarker analysis was embedded within the ECOG-ACRIN E2805 trial consent. Approval for the study was granted by the institutional review board at each participating study center. This study was conducted in accordance with the Declaration of Helsinki.

Specimen characteristics

As part of the ECOG-ACRIN E2805 study protocol, plasma was collected in sodium citrate at the pretreatment baseline (4–12 weeks after nephrectomy) and at cycle 2 day 1 (either 4 or 6 weeks after initiation of therapy). All plasma samples were processed centrally and stored at −80°C until the time of analysis.

KIM-1 measurement

Plasma KIM-1 levels were measured in duplicate using a microbead-based assay as described previously (11). This analysis was performed at the laboratory of Venkata Sabbisetti, and investigators were blinded to the identity of samples at the time of analysis. Each sample was diluted 10-fold in sample diluent buffer (0.1 mol/L HEPES, 0.1 mol/L NaCl, 0.1% Tween-20, and 1% BS; pH 7.4; filter sterilized). Thirty microliters (30 μL) of diluted sample, recombinant standards, and internal control urine samples were incubated with approximately 6,000 microbeads coupled with KIM-1 capture antibody for 1 hour (R&D Systems, catalog no. AF1750). Beads were then washed 3× with PBST and incubated with detection antibody for 45 minutes (R&D Systems, catalog no. BAF1750). Beads were washed 3× with PBS-Tween and incubated with Streptavidin-PE (Invitrogen) for 15 minutes. The signal from the fluorochrome was captured using Bio-Plex 200 system (Bio-Rad). Data were generated and interpreted using a five parametric logistic regression analysis. The lower limit of detection for KIM-1 was set at 9.1 pg/mL, and all values lower than this were considered undetectable.

Statistical analysis

Univariable analyses were performed to assess for associations between KIM-1 level and baseline participant characteristics. Univariable and multivariable lognormal accelerated failure time (AFT) models were used to assess the association between post-nephrectomy plasma KIM-1 and DFS (13). The AFT model was used to avoid making a proportional hazards assumption. The multivariable model was adjusted for sex, Eastern Cooperative Oncology Group performance status (ECOG PS), Fuhrman grade, tumor stage, nodal stage, histology, and the presence of sarcomatoid features. KIM-1 was fit as a restricted cubic spline with three knots and was stratified by nodal stage. An additional AFT multivariable model was used to assess the association between post-nephrectomy plasma KIM-1 level and overall survival (OS).

We analyzed post-nephrectomy, baseline plasma KIM-1 levels from 418 patients randomly selected from among the 1,837 participants in the ASSURE trial who underwent total nephrectomy. Baseline characteristics of these 418 participants were similar to those of the overall ASSURE trial population and are shown in Table 1. The majority of patients had clear cell histology (79%). Eleven percent (11%) of the participants had sarcomatoid features; 19% had Fuhrman grade 4 disease. Forty-seven percent (47%) of patients were very-high risk as classified by the modified UCLA International Staging System (UISS) Criteria and pathologic grading (14, 15). Thirty-six percent (36%) were randomized to sunitinib, 31% were randomized to sorafenib, and 33% were randomized to placebo. There were no significant differences in baseline characteristics between our cohort and the overall ECOG-ACRIN E2805 study population who underwent total nephrectomy (2).

Table 1.

Baseline characteristics.

Number of patients (%) or median (IQR)
Total 418 (100) 
Male sex 292 (70) 
Race 
 African American 21 (5) 
 Asian 10 (2) 
 Other/unknown 11 (3) 
 White 376 (90) 
Ethnicity 
 Hispanic 25 (6) 
 Non-Hispanic 367 (88) 
 Missing 26 (6) 
Age 56 (49–64) 
Histology 
 Clear cell 329 (79) 
 Papillary 37 (9) 
 Chromophobe 22 (5) 
 Other 30 (7) 
ECOG PS 
 0 317 (76) 
 1 89 (21) 
 2 0 (0) 
 3 0 (0) 
 Missing 12 (3) 
Surgical approach 
 Open 236 (56) 
 Laparoscopic 182 (44) 
Sarcomatoid features 
 Absent 370 (89) 
 Present 47 (11) 
 Missing 1 (0) 
Fuhrman grade 
 1 7 (2) 
 2 123 (29) 
 3 204 (49) 
 4 79 (19) 
 Missing 5 (1) 
T stage 
 1 46 (11) 
 2 115 (28) 
 3 250 (60) 
 4 7 (2) 
N stage 
 0/X 386 (92) 
 1/2 32 (8) 
UISS risk group 
 Intermediate high 222 (53) 
 Very high 196 (47) 
Treatment arm 
 Sunitinib 150 (36) 
 Sorafenib 128 (31) 
 Placebo 140 (33) 
Number of patients (%) or median (IQR)
Total 418 (100) 
Male sex 292 (70) 
Race 
 African American 21 (5) 
 Asian 10 (2) 
 Other/unknown 11 (3) 
 White 376 (90) 
Ethnicity 
 Hispanic 25 (6) 
 Non-Hispanic 367 (88) 
 Missing 26 (6) 
Age 56 (49–64) 
Histology 
 Clear cell 329 (79) 
 Papillary 37 (9) 
 Chromophobe 22 (5) 
 Other 30 (7) 
ECOG PS 
 0 317 (76) 
 1 89 (21) 
 2 0 (0) 
 3 0 (0) 
 Missing 12 (3) 
Surgical approach 
 Open 236 (56) 
 Laparoscopic 182 (44) 
Sarcomatoid features 
 Absent 370 (89) 
 Present 47 (11) 
 Missing 1 (0) 
Fuhrman grade 
 1 7 (2) 
 2 123 (29) 
 3 204 (49) 
 4 79 (19) 
 Missing 5 (1) 
T stage 
 1 46 (11) 
 2 115 (28) 
 3 250 (60) 
 4 7 (2) 
N stage 
 0/X 386 (92) 
 1/2 32 (8) 
UISS risk group 
 Intermediate high 222 (53) 
 Very high 196 (47) 
Treatment arm 
 Sunitinib 150 (36) 
 Sorafenib 128 (31) 
 Placebo 140 (33) 

Abbreviations: ECOG PS, Eastern Cooperative Oncology Group performance status; UISS, UCLA Integrated Staging System.

The distribution of post-nephrectomy KIM-1 values observed is shown in Supplementary Fig. S1. The median level of KIM-1 after nephrectomy and prior to the start of study treatment was 76.00 pg/mL [interquartile range (IQR), 40.55–219.75 pg/mL]. Because we observed a right skewed KIM-1 distribution with very high values seen in a minority of patients, the KIM-1 values were log transformed for univariable and multivariable analyses. We observed no significant associations between post-nephrectomy KIM-1 and sex, age, or ECOG PS. There were also no significant associations between KIM-1 and histologic subtype, sarcomatoid features, Fuhrman grade, T stage, or N stage, suggesting that post-nephrectomy KIM-1 is independent of known clinicopathologic predictors of recurrence (Supplementary Fig. S2).

To evaluate whether plasma KIM-1 levels correlate over time within a given patient, we compared post-nephrectomy KIM-1 levels with those at C2D1 of follow-up (Supplementary Fig. S3). This analysis showed an association between post-nephrectomy KIM-1 and C2D1 KIM-1 levels (Spearman ρ = 0.633).

Among 418 studied patients, we observed 199 DFS events and 128 deaths. Kaplan–Meier curves representing the rate of DFS and OS events per KIM-1 quartile are presented in Fig. 1A and B, respectively. A nonlinear relationship was seen where higher KIM-1 was associated with worse DFS (logrank P = 0.004), and this effect appeared to be driven by DFS events within the first 2 years after nephrectomy. Similar associations were noted among subgroups of patients with and without lymph node involvement at time of nephrectomy (Supplementary Fig. S4). A univariable AFT model showed a survival time ratio of 0.65 for the 75th versus 25th percentile of baseline KIM-1 [95% confidence interval (CI), 0.39–1.06, Wald P = 0.0004].

Figure 1.

Kaplan–Meier curves of clinical outcomes by baseline KIM-1. Patients are stratified by quartile of plasma KIM-1 measured at post-nephrectomy baseline. A, DFS stratified by baseline KIM-1 quartile. B, OS stratified by baseline KIM-1 quartile.

Figure 1.

Kaplan–Meier curves of clinical outcomes by baseline KIM-1. Patients are stratified by quartile of plasma KIM-1 measured at post-nephrectomy baseline. A, DFS stratified by baseline KIM-1 quartile. B, OS stratified by baseline KIM-1 quartile.

Close modal

To further understand the relationship between KIM-1 and disease recurrence in relation to known clinicopathologic variables, a multivariable AFT model was used to evaluate the association between baseline KIM-1 and DFS while adjusting for sex, PS, Fuhrman grade, nodal stage, tumor stage, presence of sarcomatoid features, and tumor histology (Table 2A). Higher post-nephrectomy baseline KIM-1 was associated with worse DFS in this multivariable model (survival time ratio 0.56 for 75th vs. 25th percentile of KIM-1; 95% CI, 0.42–0.73; P < 0.001). Of note, no significant interaction was seen between KIM-1 and assigned treatment arm on the risk of recurrence (P = 0.53), suggesting that KIM-1 is not a predictive biomarker for response to adjuvant therapy with sunitinib or sorafenib.

Table 2A.

Multivariable model of KIM-1 and DFS.

Survival time ratio95% CI
KIM-1 75th percentile vs. 25th percentilea 0.56 (0.42–0.73) 
N stage N1/N2 vs. N0/NX 0.41 (0.21–0.80) 
Fuhrman grade 4 vs. <4 0.34 (0.21–0.54) 
Histology Papillary vs. CC 1.24 (0.67–2.28) 
 Chromophobe vs. CC 2.88 (1.09–7.58) 
 Other vs. CC 0.58 (0.3–1.09) 
ECOG performance status 1 vs. 0 0.61 (0.41–0.91) 
T stage 1 vs. 3 2.01 (1.14–3.55) 
 2 vs. 3 1.51 (1.01–2.26) 
 4 vs. 3 1.79 (0.46–6.98) 
Sarcomatoid features Present vs. absent 0.76 (0.43–1.34) 
Sex Female vs. male 1.22 (0.84–1.78) 
Survival time ratio95% CI
KIM-1 75th percentile vs. 25th percentilea 0.56 (0.42–0.73) 
N stage N1/N2 vs. N0/NX 0.41 (0.21–0.80) 
Fuhrman grade 4 vs. <4 0.34 (0.21–0.54) 
Histology Papillary vs. CC 1.24 (0.67–2.28) 
 Chromophobe vs. CC 2.88 (1.09–7.58) 
 Other vs. CC 0.58 (0.3–1.09) 
ECOG performance status 1 vs. 0 0.61 (0.41–0.91) 
T stage 1 vs. 3 2.01 (1.14–3.55) 
 2 vs. 3 1.51 (1.01–2.26) 
 4 vs. 3 1.79 (0.46–6.98) 
Sarcomatoid features Present vs. absent 0.76 (0.43–1.34) 
Sex Female vs. male 1.22 (0.84–1.78) 

Note: A multivariable lognormal AFT model was used to assess the association between post-nephrectomy plasma KIM-1 and DFS or OS. In these models, lower survival time ratio represents shorter DFS relative to the covariate of interest. The reported KIM-1 survival time ratio compares the 75th percentile with the 25th percentile of KIM-1.

aThe 25th percentile of KIM-1 was 40.55 pg/mL. The 75th percentile of KIM-1 was 219.8 pg/mL.

We assessed for interaction terms on the effect of baseline KIM-1 on DFS. The presence of nodal disease at time of nephrectomy was noted to be an effect modifier, with greater effect size of KIM-1 on DFS seen among patients with at least N1 nodal disease (Pinteraction 0.0086). A restricted cubic spline was used to illustrate the effect of nodal status on the relationship between baseline KIM-1 and recurrence risk (Fig. 2).

Figure 2.

Log DFS time in relation to KIM-1. A restricted cubic spline was used to model the relationship between DFS and baseline KIM-1, in patients with and without pathologic nodal involvement at time of nephrectomy.

Figure 2.

Log DFS time in relation to KIM-1. A restricted cubic spline was used to model the relationship between DFS and baseline KIM-1, in patients with and without pathologic nodal involvement at time of nephrectomy.

Close modal

We evaluated whether subsequent changes in KIM-1 during follow-up provide additional prognostic information. We found that C2D1 KIM-1 was not independently predictive of recurrence risk after adjustment for baseline KIM-1 (P = 0.67). In addition, no difference was seen in the pattern of KIM-1 change between the placebo and treatment arms (Supplementary Fig. S3).

To provide clinically interpretable estimates for KIM-1 and recurrence risk, we calculated model-derived survival rates for patients both with and without nodal disease at time of nephrectomy, based on various percentiles of baseline KIM-1. Model-predicted survival curves for representative post-nephrectomy plasma KIM-1 levels are presented in Supplementary Fig. S5. For a representative male with PS 0, clear-cell histology, T3 tumor, no sarcomatoid features, Fuhrman grade 3 or less, and N1 or N2 disease at nephrectomy, the predicted DFS is 4.93 (2.74–8.85) times longer when post-nephrectomy KIM-1 is at the 10th percentile compared with the 90th percentile of KIM-1.

We evaluated whether KIM-1 adds to the prognostic value of previously published clinical models (14, 16, 17). When added as a spline term to either the Stage, Size, Grade, and Necrosis (SSIGN) score or the UISS score for disease recurrence after nephrectomy, baseline KIM-1 improved the predictive value of both models (likelihood ratio test P = 0.078 and P = 0.0022, respectively). In addition, when applied to the E2805 cohort, both the SSIGN and UISS models produced higher concordance when KIM-1 was included in the model (SSIGN concordance 0.57 vs. 0.43, P = 0.052; UISS concordance 0.60 vs. 0.40, P = 0.0005).

We evaluated the association between post-nephrectomy plasma KIM-1 and OS. Kaplan–Meier analysis showed a trend toward worse OS for patients with higher baseline KIM-1 (logrank P = 0.056; Fig. 1B). Higher post-nephrectomy baseline KIM-1 was associated with worse OS in a multivariable AFT model, after adjustment for age, sex, PS, Fuhrman grade, nodal stage, tumor stage, presence of sarcomatoid features, and tumor histology (survival time ratio 0.71 for 75th vs. 25th percentile of KIM-1; 95% CI, 0.56–0.91; P < 0.001; Table 2B; Supplementary Fig. S6).

Table 2B.

Multivariable model of KIM-1 and OS.

Survival time ratio95% CI
KIM-1 75th percentile vs. 25th percentile* 0.71 (0.56–0.91) 
N-stage N1/N2 vs. N0/NX 0.65 (0.37–1.15) 
Fuhrman grade 4 vs. <4 0.50 (0.33–0.75) 
Histology Papillary vs. CC 0.84 (0.50–1.41) 
 Chromophobe vs. CC 4.15 (1.28–13.47) 
 Other vs. CC 0.51 (0.30–0.87) 
ECOG performance status 1 vs. 0 0.73 (0.51–1.03) 
T stage 1 vs. 3 1.91 (1.13–3.24) 
 2 vs. 3 1.14 (0.80–1.63) 
 4 vs. 3 2.12 (0.56–8.08) 
Sarcomatoid features Present vs. absent 0.87 (0.54–1.41) 
Age at enrollment 75th percentile vs. 25th percentilea 0.75 (0.61–0.93) 
Sex Female vs. male 1.21 (0.87–1.70) 
Survival time ratio95% CI
KIM-1 75th percentile vs. 25th percentile* 0.71 (0.56–0.91) 
N-stage N1/N2 vs. N0/NX 0.65 (0.37–1.15) 
Fuhrman grade 4 vs. <4 0.50 (0.33–0.75) 
Histology Papillary vs. CC 0.84 (0.50–1.41) 
 Chromophobe vs. CC 4.15 (1.28–13.47) 
 Other vs. CC 0.51 (0.30–0.87) 
ECOG performance status 1 vs. 0 0.73 (0.51–1.03) 
T stage 1 vs. 3 1.91 (1.13–3.24) 
 2 vs. 3 1.14 (0.80–1.63) 
 4 vs. 3 2.12 (0.56–8.08) 
Sarcomatoid features Present vs. absent 0.87 (0.54–1.41) 
Age at enrollment 75th percentile vs. 25th percentilea 0.75 (0.61–0.93) 
Sex Female vs. male 1.21 (0.87–1.70) 

aThe 75th percentile of age was 64 years. The 25th percentile of age was 49.5 years.

A sensitivity analysis was performed with statistical imputation of KIM-1 levels that fell below the lower limit of detection. Similar associations between KIM-1 and DFS were seen when using the imputed low KIM-1 values (Supplementary Table S1).

In this study, we demonstrate that higher post-nephrectomy plasma KIM-1 is associated with worse DFS among patients with high-risk localized RCC. This association was strongest among patients with pathologic nodal disease at the time of nephrectomy.

To our knowledge, this is the largest study to date to evaluate the prognostic performance of a circulating tumor-derived biomarker in assessing post-nephrectomy risk of recurrence in RCC. Plasma KIM-1 is a minimally invasive biomarker that we show improves the risk stratification of patients with RCC after nephrectomy. Indeed, we found that the addition of post-nephrectomy plasma KIM-1 to both the UISS (14) and SSIGN (18) risk stratification scores significantly improved both models' discriminatory performance. Proposed circulating biomarkers for RCC have included circulating cytokines, tumor proteins including CA-IX, tumor cells, cell-free tumor DNA and RNA, and DNA methylation patterns (19–24). In a previous analysis of circulating cytokines in ECOG-ACRIN E2805, we demonstrated that cytokine levels are sensitive to host–drug interactions and that post-nephrectomy baseline CXCL10/IP-10 is associated with worse DFS (25). However, methodologic limitations have thus far limited the widespread clinical adoption of known circulating biomarkers. Some potential barriers have included low sensitivity (for instance with circulating tumor cells), fluctuations in response to host factors and antitumor therapy (seen with cytokine biomarkers), and high interpatient variability and computational complexity (circulating tumor DNA, transcriptomic and methylation signatures). Compared with other circulating biomarkers, KIM-1 is a single protein overexpressed in the majority of patients with RCC, directly correlated with tumor burden, and easily measurable in banked plasma with a relatively straightforward antibody-based microbead assay with the potential for implementation at scale. The management of high-risk localized RCC continues to pose a clinical dilemma. Despite a multitude of completed phase III trials, no systemic adjuvant therapy for RCC has ever been shown to improve OS (2–5, 26). Sunitinib received FDA approval for adjuvant therapy after nephrectomy for high-risk RCC based on improved DFS seen in the S-TRAC randomized trial (4). However, there was no OS benefit and patients receiving sunitinib experienced substantial toxicity and reduction in health-related quality of life. All other adjuvant trials of VEGF inhibitors, including the ECOG-ACRIN E2805 ASSURE trial, failed to show improvement in DFS or OS compared with placebo in resected RCC (2, 3, 5, 26).

In this study, KIM-1 was a prognostic but not predictive biomarker in setting of therapy with VEGFR tyrosine kinase inhibitors (TKI). There was no trend seen in the pattern of plasma KIM-1 change over time when comparing the placebo and treatment arms (Supplementary Fig. S3). This is consistent with prior preclinical studies suggesting that macroscopic tumors are limited by vascular supply whereas microscopic tumors are not, and therefore VEGFR TKIs may have limited activity in treating micrometastatic disease (27, 28).

It is therefore possible that KIM-1 may yet prove to be a predictive biomarker for adjuvant immune checkpoint inhibition. Currently, there are several ongoing phase III trials evaluating adjuvant anti-PD-1/PD-L1 blockade alone or in combination with CTLA-4 blockade. These include PROSPER (NCT03055013), IMmotion 010 (NCT03024996), KEYNOTE 564 (NCT03142334), RAMPART (NCT03288532), and CheckMate 914 (NCT03138512; refs 29–33). Several of these adjuvant trials have completed enrollment and results are expected in the coming years. Although the vast majority of circulating KIM-1 in RCC is thought to derive from tumor cells, KIM-1 is also expressed in regulatory B cells and some T cells and appears to play a role in immune tolerance signaling (34, 35). It is not known whether circulating KIM-1 is immunologically active in the adjuvant setting, and whether plasma KIM-1 could help to select patients for adjuvant immune checkpoint blockade. We are planning preclinical and clinical studies using specimens from these ongoing adjuvant immune checkpoint trials to better elucidate these questions.

Strengths of this study include the use of the ECOG-ACRIN E2805 (ASSURE) cohort, which consists of highly annotated, prospectively collected clinical data and outcomes, in conjunction with patient plasma that was collected at prespecified time points across multiple institutions (2). These strengths have made the ECOG-ACRIN E2805 cohort uniquely valuable for the development of new clinical and laboratory prognostic biomarkers (25, 36, 37).

Our results are consistent with the prior literature describing KIM-1 in RCC. Urinary KIM-1 has been shown to decrease in patients with RCC following nephrectomy, consistent with the hypothesis that it is released directly from tumors (10). Similarly, KIM-1 has been previously shown to be higher in patients with RCC pre-diagnosis compared with healthy controls, and also associated with subsequent risk of death (12). While one prior study showed that urine and tissue KIM-1 levels were associated with T stage and tumor histology (38), we did not see such an association, likely because all patients in this study underwent complete macroscopic resection of disease and the primary tumor was no longer the main source of KIM-1. It is not clear why higher post-nephrectomy KIM-1 levels had a stronger effect on DFS among node-positive (N1 or N2) participants compared with node-negative (N0 or NX) participants. One possibility is that this simply reflects higher recurrence risk among node-positive patients, but it is also possible that patients who do experience disease recurrence despite no nodal disease at time of surgery have less well-differentiated RCC that is less likely to express KIM-1.

Our study has several limitations. Because of the schedule of plasma collection, we were unable to reassess KIM-1 levels at time of relapse among those participants whose cancer recurred. Such information could help to further characterize the utility of KIM-1 in surveillance after nephrectomy. We excluded patients who underwent partial nephrectomy because surgically injured nephrons are expected to release KIM-1 during healing, but patients with partial nephrectomy represented only a small proportion (5.4%) of the overall ASSURE study cohort (2, 11). In addition, in this study, we investigated a single biomarker and it is possible that comeasurement of additional investigational biomarkers could further improve prognostic performance.

Because KIM-1 is an established biomarker for kidney injury, impaired renal function may confound the relationship between KIM-1 and clinical outcomes. Eligibility criteria for ECOG-ACRIN E2805 required serum creatinine ≤ 2 times the normal limit or CrCl ≥ 30 mL/minute and our study therefore did not include patients with severe renal impairment (2). However, in a prior cohort of prediagnostic plasma samples from patients with RCC, we saw no correlation between plasma KIM-1 and plasma creatinine, and the addition of creatinine did not change the multivariable risk estimate for KIM-1 and new RCC (12). In patients with kidney cancer, the amount of KIM-1 shed by RCC into plasma likely predominates compared with the smaller amount shed via ongoing injury of normal kidney epithelium.

In this study, we did not have nephrectomy tumor tissue available to assess transcriptomic expression of KIM-1. However, previous investigators have assessed tissue expression of KIM-1 using 133 nephrectomy samples from patients who participated in the S-TRAC randomized trial of adjuvant sunitinib versus placebo (39). In data presented at the 2019 ESMO Congress, a nonsignificant trend was seen between KIM-1 expression above the median and favorable DFS (HR in sunitinib arm 0.50; 95% CI, 0.22–1.15; HR in placebo arm 0.48; 95% CI, 0.22–1.02; unadjusted P = 0.06). These data are consistent with our hypothesis that circulating KIM-1 likely reflects residual tumor burden rather than inherently unfavorable tumor biology.

Because our study was carried out using patients from a single randomized trial, we have planned further studies to validate these findings in additional cohorts, including independent adjuvant therapy trials. In addition, studies are warranted to investigate the role of KIM-1 in patients who underwent partial nephrectomy or ablative therapies, and in patients with chronic kidney disease. If validated, these studies could help to define appropriate cut-off levels for plasma KIM-1 for use in risk stratification and/or surveillance after nephrectomy.

Conclusions

Among patients at high risk for relapse for localized RCC, elevated plasma KIM-1 post-nephrectomy is associated with worse DFS and OS. Plasma KIM-1 may be a minimally invasive circulating biomarker for the detection of microscopic residual disease after nephrectomy.

K.T. Flaherty reports personal fees from Loxo Oncology, Clovis Oncology, Strata Oncology, Checkmate Pharmaceuticals, Vivid Biosciences, Kinnate Biopharma, Scorpion Therapeutics, X4 Pharmaceuticals, PIC Therapeutics, Shattuck Labs, Apricity, Oncoceutics, Fog Pharma, Tvardi, xCures, Monopteros, Vibliome, Sanofi, Amgen, Asana, Neon Therapeutics, Tolero, Lilly, Genentech, Bristol Myers Squibb, Merck, Takeda, Verastem, Boston Biomedical, Pierre Fabre, Debiopharm, and ALX Oncology, as well as grants and personal fees from Novartis and Sanofi during the conduct of the study. R.G. Uzzo reports other from Urogen, Genentech, Pfizer, Janssen, and Amgen outside the submitted work. V. Sabbisetti reports a patent 10712349 issued. No disclosures were reported by the other authors.

The Editor-in-Chief of Clinical Cancer Research is an author on this article. In keeping with AACR editorial policy, a senior member of the Clinical Cancer Research editorial team managed the consideration process for this submission and independently rendered the final decision concerning acceptability.

W. Xu: Conceptualization, formal analysis, funding acquisition, investigation, methodology, writing–original draft, writing–review and editing. M. Puligandla: Conceptualization, data curation, formal analysis, investigation, methodology, writing–original draft, writing–review and editing. B. Halbert: Formal analysis, writing–original draft, writing–review and editing. N.B. Haas: Conceptualization, resources, supervision, methodology, project administration, writing–review and editing. K.T. Flaherty: Conceptualization, resources, supervision, project administration, writing–review and editing. R.G. Uzzo: Conceptualization, resources, supervision, project administration, writing–review and editing. J.P. Dutcher: Conceptualization, resources, supervision, project administration, writing–review and editing. R.S. DiPaola: Conceptualization, resources, supervision, project administration, writing–review and editing. V. Sabbisetti: Conceptualization, resources, formal analysis, supervision, investigation, methodology, writing–review and editing. R.S. Bhatt: Conceptualization, resources, supervision, funding acquisition, investigation, methodology, project administration, writing–review and editing.

This study was conducted by the ECOG-ACRIN Cancer Research Group (Peter J. O'Dwyer and Mitchell D. Schnall) and supported by the NCI of the NIH under the following award numbers: U10CA180820, U10CA180794, and UG1CA233180. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH. Mention of trade names, commercial products, or organizations does not imply endorsement by the U.S. government. Funding for biomarker analysis was also provided in part by a Conquer Cancer Foundation of the American Society of Clinical Oncology Young Investigator Award (to W. Xu). This work was also supported by NIH R01 CA196996 (to R.S. Bhatt), and NIH P50 CA10194212 (to R.S. Bhatt).

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.
Bray
F
,
Ferlay
J
,
Soerjomataram
I
,
Siegel
RL
,
Torre
LA
,
Jemal
A
. 
Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries
.
CA Cancer J Clin
2018
;
68
:
394
424
.
2.
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
:
2008
16
.
3.
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
:
3916
23
.
4.
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
:
2246
54
.
5.
Gross-Goupil
M
,
Kwon
TG
,
Eto
M
,
Ye
D
,
Miyake
H
,
Seo
SI
, et al
Axitinib versus placebo as an adjuvant treatment of renal cell carcinoma: results from the phase III, randomized ATLAS trial
.
Ann Oncol
2018
;
29
:
2371
8
.
6.
Mclaughlin
J
,
Lipworth
L
,
Tarone
R
. 
Epidemiologic aspects of renal cell carcinoma
.
Semin Oncol
2006
;
33
:
527
33
.
7.
Porta
C
,
Cosmai
L
,
Leibovich
BC
,
Powles
T
,
Gallieni
M
,
Bex
A
. 
The adjuvant treatment of kidney cancer: a multidisciplinary outlook
.
Nat Rev Nephrol
2019
;
15
:
423
33
.
8.
Ichimura
T
,
Bonventre
J V
,
Bailly
V
,
Wei
H
,
Hession
CA
,
Cate
RL
, et al
Kidney injury molecule-1 (KIM-1), a putative epithelial cell adhesion molecule containing a novel immunoglobulin domain, is up-regulated in renal cells after injury
.
J Biol Chem
1998
;
273
:
4135
42
.
9.
Lin
F
,
Zhang
PL
,
Yang
XJ
,
Shi
J
,
Blasick
T
,
Han
WK
, et al
Human kidney injury molecule-1 (hKIM-1): a useful immunohistochemical marker for diagnosing renal cell carcinoma and ovarian clear cell carcinoma
.
Am J Surg Pathol
2007
;
31
:
371
81
.
10.
Zhang
PL
,
Mashni
JW
,
Sabbisetti
VS
,
Schworer
CM
,
Wilson
GD
,
Wolforth
SC
, et al
Urine kidney injury molecule-1: a potential non-invasive biomarker for patients with renal cell carcinoma
.
Int Urol Nephrol
2014
;
46
:
379
88
.
11.
Sabbisetti
VS
,
Waikar
SS
,
Antoine
DJ
,
Smiles
A
,
Wang
C
,
Ravisankar
A
, et al
Blood kidney injury molecule-1 is a biomarker of acute and chronic kidney injury and predicts progression to ESRD in type I diabetes
.
J Am Soc Nephrol
2014
;
25
:
2177
86
.
12.
Scelo
G
,
Muller
DC
,
Riboli
E
,
Johansson
M
,
Cross
AJ
,
Vineis
P
, et al
KIM-1 as a blood-based marker for early detection of kidney cancer: a prospective nested case–control study
.
Clin Cancer Res
2018
;
24
:
5594
601
.
13.
Bradburn
MJ
,
Clark
TG
,
Love
SB
,
Altman
DG
. 
Survival analysis part II: multivariate data analysis–an introduction to concepts and methods
.
Br J Cancer
2003
;
89
:
431
6
.
14.
Zisman
A
,
Pantuck
AJ
,
Wieder
J
,
Chao
DH
,
Dorey
F
,
Said
JW
, et al
Risk group assessment and clinical outcome algorithm to predict the natural history of patients with surgically resected renal cell carcinoma
.
J Clin Oncol
2002
;
20
:
4559
66
.
15.
Lam
JS
,
Shvarts
O
,
Leppert
JT
,
Pantuck
AJ
,
Figlin
RA
,
Belldegrun
AS
. 
Postoperative surveillance protocol for patients with localized and locally advanced renal cell carcinoma based on a validated prognostic nomogram and risk group stratification system
.
J Urol
2005
;
174
:
466
72
.
16.
Pencina
MJ
,
D' Agostino
RB
,
D' Agostino
RB
,
Vasan
RS
. 
Evaluating the added predictive ability of a new marker: from area under the ROC curve to reclassification and beyond
.
Stat Med
2008
;
27
:
157
72
.
17.
Zigeuner
R
,
Hutterer
G
,
Chromecki
T
,
Imamovic
A
,
Kampel-Kettner
K
,
Rehak
P
, et al
External validation of the Mayo Clinic stage, size, grade, and necrosis (SSIGN) score for clear-cell renal cell carcinoma in a single European centre applying routine pathology
.
Eur Urol
2010
;
57
:
102
9
.
18.
Frank
I
,
Blute
ML
,
Cheville
JC
,
Lohse
CM
,
Weaver
AL
,
Zincke
H
. 
An outcome prediction model for patients with clear cell renal cell carcinoma treated with radical nephrectomy based on tumor stage, size, grade and necrosis: the SSIGN score
.
J Urol
2002
;
168
:
2395
400
.
19.
Zurita
AJ
,
Jonasch
E
,
Wang
X
,
Khajavi
M
,
Yan
S
,
Du
DZ
, et al
A cytokine and angiogenic factor (CAF) analysis in plasma for selection of sorafenib therapy in patients with metastatic renal cell carcinoma
.
Ann Oncol
2012
;
23
:
46
52
.
20.
Rini
B
. 
Dissecting responsive phenotypes through cytokine and angiogenic factor analysis
.
Ann Oncol
2012
;
23
:
6
7
.
21.
Kim
TH
,
Kang
Y-T
,
Cho
Y-H
,
Kim
JH
,
Jeong
BC
,
Seo
SI
, et al
Detection of circulating tumour cells and their potential use as a biomarker for advanced renal cell carcinoma
.
Can Urol Assoc J
2019
;
13
:
E285
91
.
22.
Nuzzo
PV
,
Berchuck
JE
,
Korthauer
K
,
Spisak
S
,
Nassar
AH
,
Abou Alaiwi
S
, et al
Detection of renal cell carcinoma using plasma and urine cell-free DNA methylomes
.
Nat Med
2020
;
26
:
1041
3
.
23.
Farber
NJ
,
Kim
CJ
,
Modi
PK
,
Hon
JD
,
Sadimin
ET
,
Singer
EA
. 
Renal cell carcinoma: the search for a reliable biomarker
.
Transl Cancer Res
2017
;
6
:
620
32
.
24.
Ball
MW
,
Gorin
MA
,
Guner
G
,
Pierorazio
PM
,
Netto
G
,
Paller
CJ
, et al
Circulating tumor DNA as a marker of therapeutic response in patients with renal cell carcinoma: a pilot study
.
Clin Genitourin Cancer
2016
;
14
:
e515
20
.
25.
Xu
W
,
Puligandla
M
,
Manola
J
,
Bullock
AJ
,
Tamasauskas
D
,
McDermott
DF
, et al
Angiogenic factor and cytokine analysis among patients treated with adjuvant VEGFR TKIs in resected renal cell carcinoma
.
Clin Cancer Res
2019
;
25
:
6098
106
.
26.
Eisen
TQG
,
Frangou
E
,
Smith
B
,
Ritchie
A
,
Kaplan
RS
,
Oza
B
, et al
Primary efficacy analysis from the SORCE trial (RE05): adjuvant sorafenib for renal cell carcinoma at intermediate or high risk of relapse: an international, randomized double-blind phase III trial led by the MRC CTU at UCL
.
Ann Oncol
2019
;
30
:
v891
2
.
27.
Tonini
T
,
Rossi
F
,
Claudio
PP
. 
Molecular basis of angiogenesis and cancer
.
Oncogene
2003
;
22
:
6549
56
.
28.
Holash
J
,
Maisonpierre
PC
,
Compton
D
,
Boland
P
,
Alexander
CR
,
Zagzag
D
, et al
Vessel cooption, regression, and growth in tumors mediated by angiopoietins and VEGF
.
Science
1999
;
284
:
1994
8
.
29.
US National Library of Medicine
. 
A study comparing the combination of nivolumab and ipilimumab versus placebo in participants with localized renal cell carcinoma - ClinicalTrials.gov
; 
2019
.
Available from
: https://clinicaltrials.gov/ct2/show/NCT03138512.
30.
US National Library of Medicine
. 
Safety and efficacy study of pembrolizumab (MK-3475) as monotherapy in the adjuvant treatment of renal cell carcinoma post nephrectomy (MK-3475–564/KEYNOTE-564) - ClinicalTrials.gov
; 
2019
.
Available from
: https://clinicaltrials.gov/ct2/show/NCT03142334.
31.
US National Library of Medicine
. 
A study of atezolizumab as adjuvant therapy in participants with renal cell carcinoma (RCC) at high risk of developing metastasis following nephrectomy - ClinicalTrials.gov
; 
2019
.
Available from
: https://clinicaltrials.gov/ct2/show/NCT03024996.
32.
US National Library of Medicine
. 
Nivolumab in treating patients with localized kidney cancer undergoing nephrectomy - ClinicalTrials.gov
; 
2019
.
Available from
: https://clinicaltrials.gov/ct2/show/NCT03055013.
33.
US National Library of Medicine
. 
Renal Adjuvant MultiPle Arm Randomised Trial (RAMPART) - ClinicalTrials.gov
; 
2019
.
Available from
: https://clinicaltrials.gov/ct2/show/NCT03288532.
34.
Ding
Q
,
Yeung
M
,
Camirand
G
,
Zeng
Q
,
Akiba
H
,
Yagita
H
, et al
Regulatory B cells are identified by expression of TIM-1 and can be induced through TIM-1 ligation to promote tolerance in mice
.
J Clin Invest
2011
;
121
:
3645
56
.
35.
Freeman
GJ
,
Casasnovas
JM
,
Umetsu
DT
,
DeKruyff
RH
. 
TIM genes: a family of cell surface phosphatidylserine receptors that regulate innate and adaptive immunity
.
Immunol Rev
2010
;
235
:
172
89
.
36.
Correa
AF
,
Jegede
O
,
Haas
NB
,
Flaherty
KT
,
Pins
MR
,
Messing
EM
, et al
Predicting renal cancer recurrence: defining limitations of existing prognostic models with prospective trial-based validation
.
J Clin Oncol
2019
;
37
:
2062
71
.
37.
Jilaveanu
LB
,
Puligandla
M
,
Weiss
SA
,
Wang
XV
,
Zito
C
,
Flaherty
KT
, et al
Tumor microvessel density as a prognostic marker in high-risk renal cell carcinoma patients treated on ECOG-ACRIN E2805
.
Clin Cancer Res
2018
;
24
:
217
23
.
38.
Mijuskovic
M
,
Stanojevic
I
,
Milovic
N
,
Cerovic
S
,
Petrovic
D
,
Maksic
D
, et al
Tissue and urinary KIM-1 relate to tumor characteristics in patients with clear renal cell carcinoma
.
Int Urol Nephrol
2018
;
50
:
63
70
.
39.
Ravaud
A
,
Martini
J-F
,
Ching
K
,
Staehler
M
,
Magheli
A
,
Escudier
B
, et al
Phase III trial of adjuvant sunitinib in patients with high-risk renal cell carcinoma: comprehensive tumour genomic and transcriptomic analyses
.
Ann Oncol
2019
;
30
:
v358
9
.