Original Article

Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma

List of authors.
  • Robert J. Motzer, M.D.,
  • Bernard Escudier, M.D.,
  • David F. McDermott, M.D.,
  • Saby George, M.D.,
  • Hans J. Hammers, M.D., Ph.D.,
  • Sandhya Srinivas, M.D.,
  • Scott S. Tykodi, M.D., Ph.D.,
  • Jeffrey A. Sosman, M.D.,
  • Giuseppe Procopio, M.D.,
  • Elizabeth R. Plimack, M.D.,
  • Daniel Castellano, M.D.,
  • Toni K. Choueiri, M.D.,
  • Howard Gurney, M.D.,
  • Frede Donskov, M.D., Ph.D.,
  • Petri Bono, M.D., Ph.D.,
  • John Wagstaff, M.D.,
  • Thomas C. Gauler, M.D.,
  • Takeshi Ueda, M.D., Ph.D.,
  • Yoshihiko Tomita, M.D.,
  • Fabio A. Schutz, M.D.,
  • Christian Kollmannsberger, M.D.,
  • James Larkin, M.D., Ph.D.,
  • Alain Ravaud, M.D., Ph.D.,
  • Jason S. Simon, Ph.D.,
  • Li-An Xu, Ph.D.,
  • Ian M. Waxman, M.D.,
  • and Padmanee Sharma, M.D., Ph.D.
  • for the CheckMate 025 Investigators*

Abstract

Background

Nivolumab, a programmed death 1 (PD-1) checkpoint inhibitor, was associated with encouraging overall survival in uncontrolled studies involving previously treated patients with advanced renal-cell carcinoma. This randomized, open-label, phase 3 study compared nivolumab with everolimus in patients with renal-cell carcinoma who had received previous treatment.

Methods

A total of 821 patients with advanced clear-cell renal-cell carcinoma for which they had received previous treatment with one or two regimens of antiangiogenic therapy were randomly assigned (in a 1:1 ratio) to receive 3 mg of nivolumab per kilogram of body weight intravenously every 2 weeks or a 10-mg everolimus tablet orally once daily. The primary end point was overall survival. The secondary end points included the objective response rate and safety.

Results

The median overall survival was 25.0 months (95% confidence interval [CI], 21.8 to not estimable) with nivolumab and 19.6 months (95% CI, 17.6 to 23.1) with everolimus. The hazard ratio for death with nivolumab versus everolimus was 0.73 (98.5% CI, 0.57 to 0.93; P=0.002), which met the prespecified criterion for superiority (P≤0.0148). The objective response rate was greater with nivolumab than with everolimus (25% vs. 5%; odds ratio, 5.98 [95% CI, 3.68 to 9.72]; P<0.001). The median progression-free survival was 4.6 months (95% CI, 3.7 to 5.4) with nivolumab and 4.4 months (95% CI, 3.7 to 5.5) with everolimus (hazard ratio, 0.88; 95% CI, 0.75 to 1.03; P=0.11). Grade 3 or 4 treatment-related adverse events occurred in 19% of the patients receiving nivolumab and in 37% of the patients receiving everolimus; the most common event with nivolumab was fatigue (in 2% of the patients), and the most common event with everolimus was anemia (in 8%).

Conclusions

Among patients with previously treated advanced renal-cell carcinoma, overall survival was longer and fewer grade 3 or 4 adverse events occurred with nivolumab than with everolimus. (Funded by Bristol-Myers Squibb; CheckMate 025 ClinicalTrials.gov number, NCT01668784.)

Introduction

Each year, an estimated 338,000 new cases of renal-cell carcinoma are diagnosed worldwide,1 and approximately 30% of patients present with metastatic disease at the time of diagnosis.2 A number of targeted therapies have been approved for the treatment of advanced or metastatic renal-cell carcinoma. These agents include vascular endothelial growth factor (VEGF) pathway inhibitors and mammalian target of rapamycin (mTOR) inhibitors.3,4 Everolimus is an mTOR inhibitor that is recommended for the treatment of advanced renal-cell carcinoma after treatment with sorafenib or sunitinib has failed.3-6 Although everolimus and other agents have changed the therapeutic landscape for this disease, these treatments are associated with limited overall survival after a given agent is no longer effective.

Nivolumab is a fully human IgG4 programmed death 1 (PD-1) immune checkpoint inhibitor antibody that selectively blocks the interaction between PD-1, which is expressed on activated T cells, and PD-1 ligand 1 (PD-L1) and 2 (PD-L2), which are expressed on immune cells and tumor cells. Interaction between PD-1 and PD-L1 or PD-L2 normally results in inhibition of the cellular immune response.7-9 Previous studies have shown that PD-L1 expression is associated with a poor prognosis in renal-cell carcinoma, presumably because of its immunosuppressive function.10-12 It has been postulated that PD-L1 expression would be associated with improved overall survival in response to nivolumab therapy, because disruption of PD-1–PD-L1 signaling mediated by nivolumab leads to restored antitumor immunity.13,14

In a phase 2 dose-ranging trial involving previously treated patients with metastatic renal-cell carcinoma, nivolumab was found to produce objective responses in 20 to 22% of the patients and overall survival ranging from 18.2 to 25.5 months.15 Here, we report results from a phase 3 study comparing nivolumab with everolimus in the treatment of patients with previously treated advanced renal-cell carcinoma.

Methods

Patients

Eligible patients were 18 years of age or older, had histologic confirmation of advanced or metastatic renal-cell carcinoma with a clear-cell component and measurable disease according to the Response Evaluation Criteria in Solid Tumors (RECIST version 1.1),16 and had received one or two previous regimens of antiangiogenic therapy. Additional inclusion criteria were no more than three total previous regimens of systemic therapy, including cytokines and cytotoxic chemotherapy drugs, and disease progression during or after the last treatment regimen and within 6 months before study enrollment. All patients had a Karnofsky performance status of at least 70 at the time of study entry (Karnofsky performance status scores range from 0 to 100, with higher scores indicating better functioning).17 Key exclusion criteria were metastasis to the central nervous system, previous treatment with an mTOR inhibitor, or a condition requiring treatment with glucocorticoids (equivalent to >10 mg of prednisone daily).

Study Design

This was a randomized, open-label, phase 3 study of nivolumab in comparison with everolimus. Randomization (in a 1:1 ratio) was performed with a block size of 4, with stratification according to region (United States or Canada, Western Europe, and the rest of the world), Memorial Sloan Kettering Cancer Center (MSKCC) prognostic risk group, and the number of previous antiangiogenic therapy regimens (one or two) for advanced renal-cell carcinoma. The MSKCC prognostic risk is based on the presence of zero (favorable risk), one (intermediate risk), or two or three (poor risk) of the following prognostic factors: anemia, hypercalcemia, and poor performance status.18

Nivolumab and everolimus were provided by the sponsor, except in cases in which everolimus was procured as a local commercial product in certain countries. Nivolumab was administered at a dose of 3 mg per kilogram of body weight as a 60-minute intravenous infusion every 2 weeks. Everolimus was administered orally as a daily dose of 10 mg. Dose modifications were not permitted for nivolumab but were permitted for everolimus.

Study Oversight

This study was approved by the institutional review board or an independent ethics committee at each center and was conducted in accordance with Good Clinical Practice guidelines, as defined by the International Conference on Harmonisation. All the patients provided written informed consent that was based on the principles of the Declaration of Helsinki. A data and safety monitoring committee reviewed efficacy and safety during the study.

The study was designed by the authors in collaboration with the sponsor (Bristol-Myers Squibb). The authors vouch for the accuracy and completeness of the analyses reported and for the fidelity of the study to the protocol, which is available with the full text of this article at NEJM.org. The development of the first draft of the manuscript was led by the first author. All the authors contributed to the drafting of the manuscript and provided final approval to submit the manuscript for publication. Medical-writing support, funded by the sponsor, was provided by PPSI.

End Points and Assessments

The primary end point was overall survival, which was defined as the time from randomization to the date of death. Secondary end points included the objective response rate, progression-free survival, the association between overall survival and tumor expression of PD-L1, and the incidence of adverse events. Disease assessments were performed with the use of computed tomography or magnetic resonance imaging at baseline, every 8 weeks for the first year, and then every 12 weeks until disease progression or discontinuation of treatment. Imaging data were evaluated by the investigator to assess tumor response (according to RECIST version 1.1). Patients were allowed to continue the study therapy after initial disease progression if a clinical benefit as assessed by the investigator was noted and the study drug had an acceptable side-effect profile. Safety assessments were conducted at each clinic visit. After discontinuation of treatment, patients were followed every 3 months for assessment of survival and subsequent anticancer therapy.

The objective response rate (investigator-assessed) was defined as the number of patients with a complete response or a partial response divided by the number of patients who underwent randomization. The best overall response was defined as the investigator-assessed best response (complete response, partial response, stable disease, or progressive disease) from the time of randomization to objectively documented disease progression or subsequent therapy, whichever occurred first. Progression-free survival was defined as the time from randomization to first documented RECIST-defined tumor progression or death from any cause. Tumor PD-L1 membrane expression (≥1% vs. <1% and ≥5% vs. <5%) was assessed at a central laboratory in sections that had at least 100 tumor cells that could be evaluated and were positive for PD-L1 expression, as assessed with Dako PD-L1 immunohistochemical staining in accordance with the manufacturer’s instructions.19

Adverse events were graded according to the National Cancer Institute Common Terminology Criteria for Adverse Events, version 4.0.20 Quality of life was assessed with the use of the Functional Assessment of Cancer Therapy Kidney Symptom Index–Disease-Related Symptoms (FKSI-DRS) scoring algorithm.21 The FKSI-DRS questionnaire consists of nine symptom-specific questions that address lack of energy, pain, weight loss, bone pain, fatigue, dyspnea, cough, fevers, and hematuria. A summary score ranges from 0 to 36, with 36 as the best possible score (no symptoms) and 0 as the worst possible score (all the worst symptoms).21 Additional details are provided in the Supplementary Appendix, available at NEJM.org.

Statistical Analysis

This planned interim analysis was conducted after 398 of the 569 deaths (70%) required for the final analysis had occurred; the stopping boundary was derived on the basis of the number of deaths with the use of an O’Brien–Fleming alpha-spending function that provided 90% power to detect a hazard ratio of 0.76 with an overall type I error rate of 0.05 (two-sided).22 Interim overall survival was projected at a 0.0148 nominal significance level; if the results for overall survival were significant at that level, the study could be stopped at the recommendation of the data monitoring committee and declared to be positive for efficacy. The interim analysis would then be considered the final analysis. In July 2015, the study was stopped early because an assessment conducted by the independent data monitoring committee concluded that the study had met its end point with regard to significant results for overall survival.

All patients who underwent randomization were included in the efficacy analyses; patients who received one or more doses of study drug were included in the safety analyses. Overall survival, progression-free survival, and the duration of response were estimated with the use of Kaplan–Meier methods.16 Medians and corresponding 95% confidence intervals were determined with Brookmeyer and Crowley methods23; 95% confidence intervals were constructed by means of a log–log transformation. A stratified log-rank test was performed to compare the nivolumab group with the everolimus group with respect to overall survival and progression-free survival. We obtained a stratified hazard ratio and confidence interval for nivolumab versus everolimus by fitting a stratified Cox model with the group variable as a single covariate. The difference in response rates between the nivolumab group and the everolimus group along with the two-sided 95% confidence interval were estimated with the Cochran–Mantel–Haenszel method of weighting, with adjustment for the stratification factors.24 Survival was compared between the treatment groups with the use of the interim analysis monitoring feature of East software, version 5.4 (Cytel), which is based on the Lan–DeMets error-spending-function approach, with an O’Brien–Fleming stopping boundary used to reject the null hypothesis (i.e., that there is no treatment difference), while maintaining a two-sided overall alpha level of 0.05.22 If superiority with regard to the primary end point was demonstrated, a hierarchical statistical testing procedure was followed for the objective response rate (estimated along with the exact 95% confidence interval with the use of the Clopper–Pearson method25) and progression-free survival at an alpha level of 0.05. For quality-of-life assessments, descriptive statistics were used to assess completion rates and changes in quality of life. Wilcoxon–Mann–Whitney tests were used to evaluate the between-group differences in the median change from baseline in quality-of-life scores.

Results

Patients

Table 1. Table 1. Baseline Demographic and Clinical Characteristics of the Patients Who Underwent Randomization.

From October 2012 through March 2014, a total of 821 patients were randomly assigned to a treatment group at 146 sites in 24 countries in North America, Europe, Australia, South America, and Asia; 803 of the 821 patients who underwent randomization were treated — 406 in the nivolumab group and 397 in the everolimus group. At data cutoff (June 2015), 67 of the 406 patients (17%) in the nivolumab group and 28 of the 397 patients (7%) in the everolimus group continued to receive treatment (Fig. S1 in Supplementary Appendix). The minimum follow-up period was 14 months. The primary reason for discontinuation of treatment was disease progression (285 of 406 patients [70%] in the nivolumab group and 273 of 397 patients [69%] in the everolimus group) (Fig. S1 in Supplementary Appendix). The demographic and clinical characteristics of the patients were balanced between the treatment groups; the majority of patients (72%) had received one previous regimen of antiangiogenic therapy for advanced renal-cell carcinoma (Table 1).

Efficacy

Overall Survival

Figure 1. Figure 1. Kaplan–Meier Curve for Overall Survival.

CI denotes confidence interval, and NE not estimable.

Figure 2. Figure 2. Overall Survival in Subgroup Analyses and Kaplan–Meier Curve for Progression-free Survival.

The Memorial Sloan Kettering Cancer Center (MSKCC) prognostic risk groups are based on the presence of 0 (favorable), 1 (intermediate), or 2 or 3 (poor) of the following prognostic factors: anemia, hypercalcemia, and poor performance status. The analyses in Panel A are based on data collected with the use of an interactive voice response system.

The median overall survival was 25.0 months (95% confidence interval [CI], 21.8 to not estimable) in the nivolumab group and 19.6 months (95% CI, 17.6 to 23.1) in the everolimus group (Figure 1). Death occurred in 183 of the 410 patients (45%) randomly assigned to receive nivolumab and in 215 of the 411 patients (52%) randomly assigned to receive everolimus. The hazard ratio for death (from any cause) with nivolumab versus everolimus was 0.73 (98.5% CI, 0.57 to 0.93; P=0.002), which met the prespecified criterion for superiority. The overall survival benefit with nivolumab was observed across prespecified subgroups, including subgroups defined according to region, MSKCC prognostic score, and number of previous regimens of antiangiogenic therapy (Figure 2A). The heterogeneity of the treatment effect within each subgroup shown in Figure 2A was tested with the use of an interaction test in a Cox proportional-hazards model with treatment, subgroup, and treatment-by-subgroup interaction as covariates. None of the interaction terms were significant at the 0.05 level.

Tumor Response and Progression-free Survival

The objective response rate was higher with nivolumab than with everolimus (25% vs. 5%; odds ratio 5.98; 95% CI, 3.68 to 9.72; P<0.001) (Table S1 in the Supplementary Appendix). Partial responses were observed in 99 patients (24%) in the nivolumab group and in 20 patients (5%) in the everolimus group. Complete responses were observed in 4 patients (1%) in the nivolumab group and in 2 patients (<1%) in the everolimus group. The median time to response was 3.5 months (range, 1.4 to 24.8) among the 103 patients with a response in the nivolumab group and 3.7 months (range, 1.5 to 11.2) among the 22 patients with a response in the everolimus group; the median duration of response was 12.0 months (range, 0 to 27.6) with nivolumab and 12.0 months (range, 0 to 22.2) with everolimus (Table S1 in the Supplementary Appendix). Among the patients with a treatment response, 49 patients (48%) in the nivolumab group and 10 (45%) in the everolimus group had an ongoing response; 32 patients (31%) in the nivolumab group and 6 (27%) in the everolimus group had an ongoing response for 12 months or longer (Fig. S2 in Supplementary Appendix).

The median progression-free survival was 4.6 months (95% CI, 3.7 to 5.4) in the nivolumab group and 4.4 months (95% CI, 3.7 to 5.5) in the everolimus group (hazard ratio, 0.88; 95% CI, 0.75 to 1.03; P=0.11) (Figure 2B). To explore the apparent delayed separation of the curves, we performed an ad hoc sensitivity analysis of progression-free survival in patients who had not had disease progression or died at 6 months (145 patients [35%] in the nivolumab group and 129 patients [31%] in the everolimus group). The analysis of this subgroup of patients yielded a median progression-free survival of 15.6 months (95% CI, 11.8 to 19.6) in the nivolumab group and 11.7 months (95% CI, 10.9 to 14.7) in the everolimus group (hazard ratio, 0.64; 95% CI, 0.47 to 0.88).

PD-L1 Expression

Figure 3. Figure 3. Kaplan–Meier Curve for Overall Survival, According to Programmed Death 1 Ligand (PD-L1) Expression Level.

Of the 821 patients who underwent randomization, 756 (92%) had quantifiable tumor PD-L1 expression in pretreatment samples: 370 of the 410 patients (90%) in the nivolumab group and 386 of the 411 patients (94%) in the everolimus group (Table 1). In total, 181 of the 756 patients (24%) with quantifiable PD-L1 expression had 1% or greater PD-L1 expression, and 575 (76%) had less than 1% PD-L1 expression (Table 1). Among patients with 1% or greater PD-L1 expression, the median overall survival was 21.8 months (95% CI, 16.5 to 28.1) in the nivolumab group and 18.8 months (95% CI, 11.9 to 19.9) in the everolimus group (hazard ratio, 0.79; 95% CI, 0.53 to 1.17) (Figure 3A). Among patients with less than 1% PD-L1 expression, the median overall survival was 27.4 months (95% CI, 21.4 to not estimable) in the nivolumab group and 21.2 months (95% CI, 17.7 to 26.2) in the everolimus group (hazard ratio, 0.77; 95% CI, 0.60 to 0.97) (Figure 3B). Similar results were observed among patients with 5% or greater PD-L1 expression, as compared with patients with less than 5% PD-L1 expression, although the interpretation of these data is limited by the small numbers of patients with 5% or greater expression (Fig. S3 in Supplementary Appendix).

Treatment Administration and Safety

The median duration of treatment was 5.5 months (range, <0.1 to 29.6) with nivolumab and 3.7 months (range, 0.2 to 25.7) with everolimus. In total, 207 of the 406 patients treated with nivolumab (51%) had dose delays, and 262 of the 397 patients treated with everolimus (66%) had dose delays (including interruptions). A total of 102 of the 397 patients in the everolimus group (26%) had at least one dose reduction; dose reductions were not allowed with nivolumab.

Table 2. Table 2. Treatment-Related Adverse Events Reported in 10% or More of Treated Patients in Either Group.

Treatment-related adverse events of any grade occurred in 319 of the 406 patients (79%) treated with nivolumab and in 349 of the 397 patients (88%) treated with everolimus (Table 2). The most common treatment-related adverse events among patients who received nivolumab were fatigue (134 patients, 33%), nausea (57 patients, 14%), and pruritus (57 patients, 14%); among patients who received everolimus, the most common events were fatigue (134 patients, 34%), stomatitis (117 patients, 29%), and anemia (94 patients, 24%). Grade 3 or 4 treatment-related adverse events occurred in 76 of the 406 patients (19%) treated with nivolumab and in 145 of the 397 patients (37%) treated with everolimus; the most common grade 3 or grade 4 event was fatigue (10 patients, 2%) with nivolumab and anemia (31 patients, 8%) with everolimus.

Treatment-related adverse events leading to treatment discontinuation occurred in 31 of the 406 patients (8%) treated with nivolumab and in 52 of the 397 patients (13%) treated with everolimus. No deaths from study-drug toxic effects were reported in the nivolumab group, and two deaths were reported in the everolimus group (one from septic shock and one from acute bowel ischemia). A total of 179 of the 406 patients (44%) who received nivolumab and 183 of the 397 patients (46%) who received everolimus received treatment beyond initial RECIST version 1.1–defined progression because, as assessed by the investigator, they continued to derive clinical benefit from the treatment.

Quality of Life

The FKSI-DRS questionnaire completion rate was 80% or higher throughout the first year of the study (Table S2 in Supplementary Appendix). The median FKSI-DRS quality-of-life score was 31.0 in both treatment groups at baseline. The median changes from baseline in the FKSI-DRS score in the nivolumab group increased over time and differed significantly from the median changes in the everolimus group at each assessment point through week 104 (P<0.05) (Table S2 in Supplementary Appendix).

Subsequent Therapy

Among the 821 patients who underwent randomization, 227 of the 410 patients (55%) in the nivolumab group and 260 of the 411 patients (63%) in the everolimus group received subsequent systemic therapy. The most common therapeutic agents used after treatment with nivolumab were everolimus (105 patients, 26%), axitinib (99 patients, 24%), and pazopanib (37 patients, 9%); the most common agents used after treatment with everolimus were axitinib (149 patients, 36%), pazopanib (64 patients, 16%), and sorafenib (38 patients, 9%). Anti–PD-1 therapy was given as subsequent therapy to 7 patients in the everolimus group.

Discussion

This phase 3 randomized study showed that patients with advanced renal-cell carcinoma who had received previous antiangiogenic treatment had longer survival with nivolumab treatment than with everolimus treatment. The separation of the overall survival curves occurred early in the study, and the median overall survival was 5.4 months longer with nivolumab than with everolimus (25.0 months vs. 19.6 months), a difference that crossed the prespecified boundary for significance at the time of the interim analysis.

This study also showed a higher number of objective responses with nivolumab than with everolimus, many of which were durable. The median progression-free survival was similar in the two treatment groups and was consistent with that reported in an uncontrolled study involving patients who had previously received antiangiogenic therapy.15 Moreover, the results of a comparison of progression-free survival between the nivolumab group and the everolimus group suggest that progression-free survival was not a surrogate for overall survival in this study. The late separation of the progression-free survival curves suggested a potential delayed benefit in progression-free survival with nivolumab. This delayed benefit was subsequently quantified in a sensitivity analysis that included patients who had not had disease progression or died at 6 months; the median progression-free survival was longer with nivolumab than with everolimus in this subgroup of patients. These patients probably contributed to the overall survival benefit that was observed with nivolumab in this study.

We observed consistently prolonged survival with nivolumab, as compared with everolimus, irrespective of the MSKCC prognostic score, number of previous antiangiogenic therapies, or region. A benefit was observed with nivolumab irrespective of PD-L1 expression. Nivolumab has been reported to be associated with pharmacodynamic changes in blood and tumor markers that are consistent with PD-1 inhibition.12 Our data corroborate previous studies that have indicated that higher levels of PD-L1 expression are associated with poorer survival in renal-cell carcinoma,10,11 but they do not support PD-L1 as a marker of treatment benefit in renal-cell carcinoma. The relationship between PD-L1 expression and outcomes after treatment with nivolumab appears to depend on tumor type and histologic class. An association between PD-L1 expression and improved outcomes with nivolumab treatment has been observed for metastatic melanoma and only some types of lung cancer.26-28

Nivolumab had a safety profile consistent with that seen in other studies of this drug.13-15 Grade 3 or 4 treatment-related adverse events were less frequent with nivolumab than with everolimus, and treatment-related adverse events leading to discontinuation occurred in fewer patients in the nivolumab group than in the everolimus group. Differences between treatments in the frequency of specific adverse events were reflective of drug class. The median changes from baseline in the FKSI-DRS score suggested a significant and consistent improvement in quality of life over the 2-year study period during nivolumab treatment.

There has been considerable progress in the treatment of renal-cell carcinoma since 2005, with five VEGF-pathway inhibitors (sorafenib, sunitinib, bevacizumab, pazopanib, and axitinib) and two mTOR inhibitors (everolimus and temsirolimus) showing benefit in pivotal phase 3 trials, which led to regulatory approval. Before this era, infrequent but occasionally long-standing responses were observed with cytokines, including high doses of interleukin-2.29 With one exception,30 the benefit with approved targeted drugs has been established in phase 3 studies that showed improvements in progression-free survival but not in overall survival with those drugs as compared with standard treatment, which included interferon alfa, placebo, or an approved antiangiogenic drug.3 Among patients in the phase 3 AXIS trial who had been previously treated with sunitinib, no benefit in overall survival was detected with axitinib as compared with sorafenib (median overall survival, 15.2 months and 16.5 months, respectively).31 In addition, a phase 3 trial of cabozantinib, an investigational VEGF-pathway inhibitor, showed longer progression-free survival with cabozantinib than with standard everolimus therapy in the treatment of patients with previously treated renal-cell carcinoma.32 The median overall survival of 25.0 months with the immune checkpoint inhibitor nivolumab and the longer survival with nivolumab than with everolimus provide evidence of benefit in patients who have already undergone treatment and have advanced renal-cell carcinoma.

Funding and Disclosures

Presented in part at the European Cancer Congress, Vienna, Austria, September 25–29, 2015.

Supported by Bristol-Myers Squibb.

Disclosure forms provided by the authors are available with the full text of this article at NEJM.org.

Dr. Motzer reports receiving consulting fees from Pfizer, Novartis, and Eisai and grant support through his institution from Pfizer, Novartis, Genentech, GlaxoSmithKline, Exelixis, and Eisai; Dr. Escudier, receiving honoraria from Bayer, Pfizer, Novartis, and GlaxoSmithKline; Dr. George, receiving fees for consulting and serving on advisory boards from Bristol-Myers Squibb, Novartis, Bayer, Sanofi-Aventis, Astellas, Xcenda, and Onclive, and grant support from Bristol-Myers Squibb, Novartis, Bayer, Pfizer, Acceleron, Merck, and Agensys; Dr. Hammers, receiving grant support from Pfizer, Newlink Genetics, GlaxoSmithKline, and SFJ Pharmaceuticals; Dr. Tykodi, receiving fees for serving on advisory boards from Prometheus, consulting fees from Amgen, and grant support through his institution from Prometheus, Argos Therapeutics, Immatics Biotechnologies, Novartis, and Exelixis; Dr. Procopio, receiving fees for serving on advisory boards from Janssen and Novartis, lecture fees from Astellas and Pfizer, and grant support from Bayer; Dr. Plimack, receiving fees for serving on advisory boards from Merck, Dendreon, GlaxoSmithKline, Pfizer, Astellas, Novartis, and Genentech, and grant support from AstraZeneca, Eli Lilly, Merck, Dendreon, GlaxoSmithKline, Acceleron, and Pfizer; Dr. Choueiri, receiving fees for consulting and for serving on advisory boards from GlaxoSmithKline, Novartis, Pfizer, Merck, AstraZeneca, Bayer, and Prometheus, and grant support through his institution from Bristol-Myers Squibb, GlaxoSmithKline, Novartis, Exelixis, Pfizer, Merck, Roche, AstraZeneca, TRACON Pharmaceuticals, and Peloton; Dr. Gurney receiving fees for serving on advisory boards from Novartis, Bayer, Sanofi-Aventis, Astellas, and Pfizer; Dr. Donskov, receiving grant support from Novartis, Pfizer, and GlaxoSmithKline; Dr. Bono, receiving honoraria from GlaxoSmithKline, Pfizer, and Orion; Dr. Wagstaff, receiving fees for serving on advisory boards, paid to his institution, from Bristol-Myers Squibb, Novartis, GlaxoSmithKline, Roche, and Amgen; Dr. Tomita, receiving fees for serving on advisory boards from ONO Pharmaceuticals and Pfizer, and honoraria and grant support from ONO Pharmaceuticals, Novartis, and Pfizer; Dr. Gauler, receiving fees for consulting and serving on advisory boards from Boehringer Ingelheim, Merck Serono, Novartis, Pfizer, GlaxoSmithKline, Merck Sharp & Dohme, Bayer HealthCare, Roche, Bristol-Myers Squibb, Eli Lilly, and Janssen-Cilag, honoraria from Boehringer Ingelheim, Merck Serono, Novartis, Pfizer, GlaxoSmithKline, Bayer, Roche, Eli Lilly, Janssen-Cilag, and Sanofi-Aventis, travel support from Boehringer Ingelheim, Merck Serono, Pfizer, Roche, and Eli Lilly, and owning stock in Bayer; Dr. Schutz, receiving fees for serving on advisory boards from Pfizer, GlaxoSmithKline, and Novartis, and lecture fees from GlaxoSmithKline; Dr. Kollmannsberger, receiving fees for serving on advisory boards from Pfizer, Novartis, Sanofi-Aventis, Bayer, and Seattle Genetics, and lecture fees from Pfizer and Novartis; Dr. Ravaud, receiving lecture fees from Merck Sharp & Dohme; Dr. Simon, Dr. Xu, and Dr. Waxman are employees of and hold stock in Bristol-Myers Squibb; and Dr. Sharma reports receiving consulting fees from Jounce Therapeutics, Amgen, Bristol-Myers Squibb, GlaxoSmithKline, and AstraZeneca/MedImmune; he is also founder of and holds stock in Jounce Therapeutics. No other potential conflict of interest relevant to this article was reported.

This article was published on September 25, 2015, and updated on January 14, 2016, at NEJM.org.

We thank the patients and their families, as well as the research colleagues and clinical teams for making the study possible; Jennifer McCarthy for serving as a protocol manager for the CheckMate 025 study; Justin Doan for involvement with quality-of-life analyses and interpretation; and the staff at Dako, for collaborative development of the automated PD-L1 immunohistochemical assay. Earlier versions of the manuscript were prepared with medical-writing and editorial assistance provided by Jennifer Granit and Maria Soushko of PPSI (a PAREXEL company), with funding from Bristol-Myers Squibb.

Author Affiliations

From Memorial Sloan Kettering Cancer Center, New York (R.J.M.), and Roswell Park Cancer Institute, Buffalo (S.G.) — both in New York; Institut Gustave Roussy, Villejuif (B.E.), and Bordeaux University Hospital, Hôpital Saint André, Bordeaux (A.R.) — both in France; Beth Israel Deaconess Medical Center (D.F.M.) and Dana–Farber Cancer Institute, Brigham and Women’s Hospital, and Harvard Medical School (T.K.C.) — all in Boston; Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, Baltimore (H.J.H.); Stanford Cancer Institute, Stanford, CA (S.S.); University of Washington and Fred Hutchinson Cancer Research Center, Seattle (S.S.T.); Vanderbilt University Medical Center, Nashville (J.A.S.); Fondazione Istituto Nazionale Tumori, Milan (G.P.); Fox Chase Cancer Center, Philadelphia (E.R.P.); Hospital Universitario 12 De Octubre, Madrid (D.C.); Westmead Hospital and Macquarie University, Sydney (H.G.); Aarhus University Hospital, Aarhus, Denmark (F.D.); Helsinki University Central Hospital and University of Helsinki, Helsinki (P.B.); South West Wales Cancer Institute and Swansea University College of Medicine, Swansea (J.W.), and Royal Marsden Hospital, London (J.L.) — both in the United Kingdom; University Hospital Essen of University of Duisburg–Essen, Germany (T.C.G.); Chiba Cancer Center, Chiba (T.U.), and Niigata University, Niigata (Y.T.) — both in Japan; Hospital Sao Jose, Beneficencia Portuguesa de São Paulo, São Paulo (F.A.S.); British Columbia Cancer Agency, Vancouver, BC, Canada (C.K.); Bristol-Myers Squibb, Lawrenceville (J.S.S., I.M.W.) and Hopewell (L.-A.X.) — both in New Jersey; and M.D. Anderson Cancer Center, University of Texas, Houston (P.S.).

Address reprint requests to Dr. Motzer at Memorial Sloan Kettering Cancer Center, Memorial Hospital, 1275 York Ave., New York, NY 10021, or at ; or to Dr. Sharma at M.D. Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, or at .

A complete list of investigators in the CheckMate 025 study is provided in the Supplementary Appendix, available at NEJM.org

Supplementary Material

References (32)

  1. 1. Ferlay J, Soerjomataram I, Dikshit R, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer 2015;136:E359-E386

  2. 2. Fisher R, Gore M, Larkin J. Current and future systemic treatments for renal cell carcinoma. Semin Cancer Biol 2013;23:38-45

  3. 3. National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology: kidney cancer. Fort Washington, PA: National Comprehensive Cancer Network, 2015. Version 3.2015 (www.nccn.org/professionals/physician_gls/f_guidelines.asp).

  4. 4. Escudier B, Porta C, Schmidinger M, et al. Renal cell carcinoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2014;25:Suppl 3:iii49-iii56

  5. 5. Afinitor (everolimus) prescribing information. East Hanover, NJ: Novartis Pharmaceuticals, 2015 (www.pharma.us.novartis.com/product/pi/pdf/afinitor.pdf).

  6. 6. Dabney R, Devine R, Sein N, George B. New agents in renal cell carcinoma. Target Oncol 2014;9:183-193

  7. 7. Hamanishi J, Mandai M, Iwasaki M, et al. Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer. Proc Natl Acad Sci U S A 2007;104:3360-3365

  8. 8. Hamid O, Carvajal RD. Anti-programmed death-1 and anti-programmed death-ligand 1 antibodies in cancer therapy. Expert Opin Biol Ther 2013;13:847-861

  9. 9. Nurieva RI, Liu X, Dong C. Molecular mechanisms of T-cell tolerance. Immunol Rev 2011;241:133-144

  10. 10. Thompson RH, Kuntz SM, Leibovich BC, et al. Tumor B7-H1 is associated with poor prognosis in renal cell carcinoma patients with long-term follow-up. Cancer Res 2006;66:3381-3385

  11. 11. Thompson RH, Gillett MD, Cheville JC, et al. Costimulatory B7-H1 in renal cell carcinoma patients: indicator of tumor aggressiveness and potential therapeutic target. Proc Natl Acad Sci U S A 2004;101:17174-17179

  12. 12. Choueiri T, Fishman MN, Escudier B. Immunomodulatory activity of nivolumab in metastatic renal cell carcinoma (mRCC): association of biomarkers with clinical outcomes. J Clin Oncol 2015;33:Suppl:4500. abstract.

  13. 13. Brahmer JR, Drake CG, Wollner I, et al. Phase I study of single-agent anti-programmed death-1 (MDX-1106) in refractory solid tumors: safety, clinical activity, pharmacodynamics, and immunologic correlates. J Clin Oncol 2010;28:3167-3175

  14. 14. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012;366:2443-2454

  15. 15. Motzer RJ, Rini BI, McDermott DF, et al. Nivolumab for metastatic renal cell carcinoma: results of a randomized phase II trial. J Clin Oncol 2015;33:1430-1437

  16. 16. Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 2009;45:228-247

  17. 17. Schag CC, Heinrich RL, Ganz PA. Karnofsky performance status revisited: reliability, validity, and guidelines. J Clin Oncol 1984;2:187-193

  18. 18. Motzer RJ, Bacik J, Schwartz LH, et al. Prognostic factors for survival in previously treated patients with metastatic renal cell carcinoma. J Clin Oncol 2004;22:454-463

  19. 19. Cogswell JP, Goldberg SM, Gupta AK, Jure-Kunkel M, Wang XT, Wigginton JM. Cancer immunotherapy by disrupting pd-1/pd-l1 signaling. FreePatentsOnline.com (http://www.freepatentsonline.com/20130309250.pdf).

  20. 20. National Cancer Institute. Common Terminology Criteria for Adverse Events (CTCAE). Version 4.0. 2010 (http://evs.nci.nih.gov/ftp1/CTCAE/CTCAE_4.03_2010-06-14_QuickReference_5x7.pdf).

  21. 21. Cella D, Yount S, Brucker PS, et al. Development and validation of a scale to measure disease-related symptoms of kidney cancer. Value Health 2007;10:285-293

  22. 22. O’Brien PC, Fleming TR. A multiple testing procedure for clinical trials. Biometrics 1979;35:549-556

  23. 23. Brookmeyer R, Crowley J. A confidence interval for the median survival time. Biometrics 1982;38:29-41

  24. 24. Categorical data analysis. In: Berry DA, ed. Statistical methodology in the pharmaceutical sciences. New York: Marcel Dekker, 1990:391-475.

  25. 25. Clopper CJ, Pearson ES. The use of confidence or fiducial limits illustrated in the case of the binomial. Biometrika 1934;26:404-414

  26. 26. Robert C, Long GV, Brady B, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med 2015;372:320-330

  27. 27. Paz-Ares L, Horn L, Borghaei H, et al. Phase III, randomized trial (CheckMate 057) of nivolumab (NIVO) versus docetaxel (DOC) in advanced non-squamous cell (non-SQ) non-small cell lung cancer (NSCLC). J Clin Oncol 2015;33:Suppl:LBA109. abstract.

  28. 28. Brahmer J, Reckamp KL, Baas P, et al. Nivolumab versus docetaxel in advanced squamous-cell non–small-cell lung cancer. N Engl J Med 2015;373:123-135

  29. 29. Lee S, Margolin K. Cytokines in cancer immunotherapy. Cancers (Basel) 2011;3:3856-3893

  30. 30. Hudes G, Carducci M, Tomczak P, et al. Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med 2007;356:2271-2281

  31. 31. Motzer RJ, Escudier B, Tomczak P, et al. Axitinib versus sorafenib as second-line treatment for advanced renal cell carcinoma: overall survival analysis and updated results from a randomised phase 3 trial. Lancet Oncol 2013;14:552-562

  32. 32. Choueiri TK, Escudier B, Powles T, et al. Cabozantinib versus everolimus in advanced renal-cell carcinoma. N Engl J Med 2015;373:1814-1823

Citing Articles (4030)

Only the 1000 most recent citing articles are listed here.

    Letters

    Comments (3)

    Figures/Media

    1. Table 1. Baseline Demographic and Clinical Characteristics of the Patients Who Underwent Randomization.
      Table 1. Baseline Demographic and Clinical Characteristics of the Patients Who Underwent Randomization.
    2. Figure 1. Kaplan–Meier Curve for Overall Survival.
      Figure 1. Kaplan–Meier Curve for Overall Survival.

      CI denotes confidence interval, and NE not estimable.

    3. Figure 2. Overall Survival in Subgroup Analyses and Kaplan–Meier Curve for Progression-free Survival.
      Figure 2. Overall Survival in Subgroup Analyses and Kaplan–Meier Curve for Progression-free Survival.

      The Memorial Sloan Kettering Cancer Center (MSKCC) prognostic risk groups are based on the presence of 0 (favorable), 1 (intermediate), or 2 or 3 (poor) of the following prognostic factors: anemia, hypercalcemia, and poor performance status. The analyses in Panel A are based on data collected with the use of an interactive voice response system.

    4. Figure 3. Kaplan–Meier Curve for Overall Survival, According to Programmed Death 1 Ligand (PD-L1) Expression Level.
      Figure 3. Kaplan–Meier Curve for Overall Survival, According to Programmed Death 1 Ligand (PD-L1) Expression Level.
    5. Table 2. Treatment-Related Adverse Events Reported in 10% or More of Treated Patients in Either Group.
      Table 2. Treatment-Related Adverse Events Reported in 10% or More of Treated Patients in Either Group.