Adjuvant therapy in renal cell carcinoma (RCC): progress, at last
Introduction
In the United States, renal cell carcinoma (RCC) is the sixth most common cancer in men and the ninth most common in women, with an estimated 14,890 people dying from kidney cancer in 2023 (1). The incidence has grown over time, doubling since 1975 and affecting males disproportionately in a 1.5–2:1 ratio (1,2). Treatment options have also dramatically improved in recent decades, bringing the 5-year survival rate in the U.S. from 50% in 1970s to 77% as of 2018 (1). RCC has traditionally been divided into histological subtypes with differing prevalence, with the most common being clear cell (75%), papillary (10%), and chromophobe (5%) (3). Of these three major subtypes, clear cell is the most aggressive and is by far the most highly represented subtype in most therapeutic clinical trials of RCC (3).
Advances in the field of RCC treatment include improved surgical technique for removal of primary tumors and metastases, improved radiation treatment modalities, as well as new systemic therapies for advanced disease. Researchers have worked in recent decades to expand these therapies to the localized setting in order to prolong survival and improve cure rates in RCC. This review will address the role and rationale of adjuvant therapy in patients at high risk of recurrence, provide a historical overview of adjuvant therapies in RCC, and summarize current guidelines and clinical practice, highlighting trials to anticipate in the years to come.
Treatment approaches
RCC, like other cancers, is treated by a variety of modalities depending on clinical criteria (grading, staging, histology, etc.) as well as a patient’s prognosis and personal decision-making process. While the mainstay of treatment in metastatic disease is systemic therapy (4,5), the first-line and often curative treatment for localized RCC [i.e., tumor node metastasis (TNM) stages I–III] is removal of the primary tumor (6). Patients with tumor spread to the ipsilateral adrenal gland are also considered to have localized disease for treatment purposes (7). Depending on factors including tumor size, tumor location, and involvement of veins or lymph nodes, patients may be eligible to receive a partial nephrectomy rather than a radical nephrectomy (8). Multiple meta-analyses showing comparable survival and better post-operative outcomes in partial nephrectomies have spurred a movement towards this approach in recent decades (9-11). It is worth noting that surgery with curative intent is not only an option in stages I–III but is increasingly being used in patients with advanced disease, including those with 1 to 3 resectable metastases. Although no randomized clinical trials have been performed of surgical metastasectomy (SM) in RCC, a systematic review showed SM to be associated with lower all-cause mortality while its morbidity is comparable to primary tumor surgery (12).
Adjuvant therapy in localized RCC: rationale and risk stratification
For many patients with localized RCC, surgery is definitive and curative; however, a significant portion will develop recurrence. The percentage cited in the literature depends on the source, although historically it is thought that approximately 25–50% of patients who undergo surgery for localized RCC will eventually develop metastatic RCC (13). A more recent analysis of European patients with localized clear cell RCC (ccRCC) in the RECUR database showed that recurrence rates can differ dramatically depending on risk factors, with those deemed intermediate-risk having a 5-year recurrence rate of 23.2% compared to 61.6% in the high-risk group (14). The question of how best to determine a patient’s risk for recurrence is an evolving area of research, however the models used clinically today rely on a variety of factors, including TNM staging, histology, and/or a patient’s symptoms and functional status.
The main such systems used in the clinic today are the UCLA Integrated Staging System (UISS), the Leibovich systems, the Stage, Size, Grade, and Necrosis (SSIGN) system, and the Karakiewicz nomogram (15-20). Since each model has unique variables, many of which are subjective, there is some heterogeneity in risk calculations depending on the system used. However, all have been validated to provide valuable information to patients and clinicians in predicting overall survival (OS) or progression to metastatic disease (see Table 1). These systems are routinely used as part of the inclusion criteria for clinical trials in the field of adjuvant therapy for RCC.
Table 1
Prognostic models | Clinical characteristics | Recurrence risks |
---|---|---|
UISS | TNM stage, Furman Grade, ECOG | 2-year survival, 5-year survival |
Leibovich, localized unilateral ccRCC patients radical nephrectomy [2003] | Tumor stage, regional lymph node status, tumor size, nuclear grade, histologic tumor necrosis | Disease progression after radical nephrectomy |
SSIGN [2002], ccRCC patients s/p radical nephrectomy | TNM stage, Furman Grade, tumor size, presence of necrosis, metastasis | Cancer-specific survival |
Karakiewicz Nomogram [2007], patients of all histological types with node-positive RCC in the absence of distant metastases who underwent partial or radical nephrectomy | TNM stage, grade, size, symptoms | RCC-specific survival |
Leibovich [2018], patients with ccRCC, papRCC, and chrRCC who underwent partial or radical nephrectomy | ccRCC: age, ECOG, constitutional symptoms, adrenalectomy, surgical margins, grade, coagulative necrosis, sarcomatoid differentiation, tumor size, perinephric or renal sinus fat invasion, tumor thrombus, extension beyond kidney, nodal involvement | PFS and cancer-specific survival at 5-, 10-, and 15-year |
papRCC: grade, perinephric or renal sinus fat invasion, tumor thrombus | ||
chrRCC: sarcomatoid differentiation, perinephric or renal sinus fat invasion, nodal involvement |
RCC, renal cell carcinoma; UISS, UCLA Integrated Staging System; TNM, tumor node metastasis; ECOG, Eastern Cooperative Oncology Group; ccRCC, clear cell RCC; SSIGN, Stage, Size, Grade, and Necrosis; papRCC, papillary RCC; chrRCC, chromophobe RCC; PFS, progression-free survival.
Although traditional risk-stratification strategies have proven useful to the field of localized RCC treatment, some newer models now incorporate molecular characteristics and genetic data from patients. Armed with this data, clinicians hope to more accurately risk-stratify patients and, in some cases, find actionable features of the disease which can be targeted with precision therapies. Studies have looked at chromosomal copy number variants or the presence of certain immune cell populations in tumor tissue as predictors of disease course (21,22). Other research has found numerous genetic biomarkers whose mutations, differential transcriptions, or differential protein expressions are associated with clinical outcomes (23). For patients with localized ccRCC, for example, one study found that tumor mutations in SETD2, BAP1, and PBRM1 were significantly associated with a higher risk of metastatic disease after nephrectomy (24). Other research groups have successfully developed multigene signatures of tumor transcription or translation that can predict recurrence, cancer-specific mortality, or overall mortality (23). Most published studies of these signatures, and indeed most other exploratory research on genomic and molecular biomarkers, have included only patients with ccRCC. Such signatures include ClearCode34 (25), the 16-gene recurrence score (26), and the epithelial-mesenchymal transition (EMT) score (27,28); the cell-cycle progression (CCP) score been validated in ccRCC and non-ccRCC patients as well (29). While molecular and genetic approaches to risk-stratification remain an exciting prospect, these have not yet been consistently applied to clinical practice or incorporated in official guidelines (30).
Adjuvant therapy after metastasectomy
Surgical intervention to remove metastatic lesions has been used with favorable long-term outcomes in a subset of patients, particularly in those with fully resectable oligometastatic disease with good performance status (31,32). After surgery, these patients are known in the literature as M1 with no evidence of disease (NED), and some have been included in trials of adjuvant therapy for localized RCC (33). In discussing these trials, this review will note when M1 NED patients have been included.
Historical overview of adjuvant therapy for localized RCC
Historically, RCC (localized or metastatic) was not responsive to traditional cytotoxic or hormonal treatments (34). Since the approval of interleukin 2 (IL-2) in 1992 by the Food and Drug Administration (FDA) as the first drug for metastatic RCC, we have made significant strides in therapeutics for patients with RCC (35). Despite these advances, relatively few options exist to prevent recurrence for localized RCC patients post-operatively. In the majority of adjuvant therapy trials time, drugs shown to work in the metastatic setting were trialed as adjuvant therapy and did not show an OS benefit for patients (see Figure 1) (34).
Trials of cytokine therapies in the adjuvant setting
The first systemic agents shown to improve outcomes in advanced RCC, albeit with low response rates and numerous side effects, were the cytokines IL-2 and interferon alpha (34). Subsequent randomized clinical trials published in the early- to mid-2000s put these agents to the test in the adjuvant setting; however, they uniformly demonstrated no benefit for patients.
A study conducted by Pizzocaro et al. [2001] of 247 patients in Italy with Stage II and Stage III RCC (any histology) compared adjuvant recombinant interferon alfa-2b to observation. The two groups did not differ in 5-year OS probability (0.665 for observation and 0.660 for treated, P=0.861) and 5-year event free survival (0.671 for observation and 0.567 for treated, P=0.107). Of note, sub-analysis showed a protective effect for pN2 and pN3 patients and a harmful effect for pN0 patients. Several years later in 2003, Messing et al. published a trial of interferon alfa-NL as adjuvant therapy for 283 patients with recent radical nephrectomy and/or lymphadenectomy for pT3–4a and/or node-positive disease. There was also no significant difference in OS and recurrence-free survival (RFS) between those who received interferon alfa-NL (median OS 5.1 years and median RFS 2.2 years) or observation (median OS 7.4 years and median RFS 3.0 years). Two-sided P values were 0.09 and 0.33 for OS and RFS, respectively.
In a trial published by Clark et al. in 2003 (36), 69 patients with completely resected RCC (locally advanced with pT3b-4 or N1–3; or M1) were randomized to receive either one course of high dose IL-2 or observation. The trial was terminated early due to lack of signal on interim analysis of the locally advanced subset of patients, for which the study was powered to detect a 30% absolute difference in disease-free survival (DFS). The authors concluded that the study was underpowered in the setting of an overly ambitious expected treatment benefit.
Other randomized trials tested combinations of cytokine drugs rather than individual agents. Atzpodien et al. [2005] used a two-arm design to compare groups who received either (A) 8 weeks of IL-2, interferon alpha-2a, and 5-fluorouracil or (B) observation (37). The trial included 203 RCC patients at high risk of relapse after surgery (pT3b-pT4 and/or nodal involvement; or with resected tumor relapse or solitary metastasis). The treatment group actually had worse OS compared to the observation group, with 2-, 5-, and 8-year survival rates of 81%, 58%, and 58% while the observation group had 2-, 5-, and 8-year survival rates of 91%, 76%, and 66% (log rank P=0.0278). The two groups did not differ significantly with regards to relapse-free survival. Another study looking at combination cytokines was performed by the Italian Oncology Group for Clinical Research (2014). This trial tested low doses of IL-2 and interferon alpha given over a 5-year period after partial or radical nephrectomy in 303 patients with RCC (pT2–3b pN0–3 M0, any histology) (38). Patients were randomized to receive cytokine treatment or observation, however, there was no significant difference in relapse-free survival [hazard ratio (HR) 0.84, 95% confidence interval (CI): 0.54–1.31, P=0.44].
Trials of autologous tumor cell vaccines in the adjuvant setting
Spurred by initial success in phase III trials of adjuvant therapy for colon cancer (39), researchers in 1990s started investigating autologous tumor cell vaccines as adjuvant therapy for RCC in attempts to increase patients’ immune responses to their own tumor cells. The first study was published in 1996 by Galligioni et al. in a cohort of 120 patients who were randomized to receive either injections of irradiated autologous tumor cells with Bacillus Calmette-Guèrin (BCG) or observation (40). Patients were stages I–III with fully resected tumor and Eastern Cooperative Oncology Group (ECOG) 0–1. Although the treatment group showed a significant delayed cutaneous hypersensitivity reaction to tumor cells compared to normal renal cells (indicating immunization to tumor antigen), the study failed to show a significant difference in DFS or OS between groups. The rate of 5-year DFS was 63% in the immunized group and 72% for the control group (P=0.21). The probability of 5-year OS in the immunized group and control group were 69% and 78%, respectively (P=0.28).
One study published by Jocham et al. in 2004 did show a DFS benefit from an adjuvant autologous renal tumor cell vaccine (41). Researchers randomly assigned 379 RCC patients after radical nephrectomy (pT2–3b pN0–N3 M0, any histology) to receive either an autologous renal tumor cell vaccine or observation. After 5 years of follow-up, analysis showed a hazard ratio of 1.58 (95% CI: 1.05–2.37, P=0.0204) in favor of the vaccine group. The authors concluded that autologous tumor cell therapy could be considered as adjuvant therapy for high-risk patients after radical nephrectomy. The excitement caused by this study was tempered by fundamental critiques of the trial, including unblinded design and concerns over differences in attrition rates between groups (42).
Finally in 2008, Wood et al. compared an autologous tumor-derived heat-shock protein complex to observation alone for 818 patients at high-risk of recurrence after nephrectomy (pT1b–T4 N0 M0 or pTany N1–2 M0) (43). Results showed no significant difference in RFS between groups (HR 0.923, 95% CI: 0.729–1.169, P=0.506). A sub-analysis showed some signal of improvement for patients with stage I or II cancer, however this did not reach statistical significance (HR 0.576, 95% CI: 0.324–1.023, P=0.056).
Trial of a carbonic anhydrase inhibitor in the adjuvant setting
Carbonic anhydrase IX (CAIX) is a cell surface glycoprotein that is expressed in ccRCC but not normal renal tissue, and as such showed promise as a drug target (44). Girentuximab, a monoclonal antibody that binds CAIX, had been routinely used in PET CT imaging of RCC (45) and certain phase II studies in metastatic RCC suggested that it could slow disease progression (some in combination with cytokine therapy interferon alpha) (46-48). In this context, the ARISER trial published in 2017 investigated girentuximab as an adjuvant therapy versus placebo in 864 patients with high risk ccRCC (defined as pT3/pT4Nx/N0M0 or pTanyN+M0 or pT1b/pT2Nx/N0M0 with nuclear grade 3 or greater) (49). The trial did not show any significant difference between groups in either of the primary endpoints of DFS (HR 0.97, 95% CI: 0.79–1.18) or OS (HR 0.99, 95% CI: 0.74–1.32).
Trials of VEGF receptor tyrosine kinase inhibitors (TKIs) in the adjuvant setting
After cytokine therapies, the next innovation in the field of RCC treatment came with the advent of drugs targeting angiogenesis, which is implicated in the pathogenesis of ccRCC via the von Hippel-Lindau (VHL) gene (50). Motzer et al. first published a trial in 2007 showing sunitinib, a receptor tyrosine kinase (RTK) inhibitor of the vascular endothelial growth factor (VEGF) pathway, to be superior to interferon alpha in metastatic RCC with a clear cell component (51). Sunitinib as well as other vascular endothelial growth factor receptor (VEGFR) TKIs pazopanib, axitinib, and sorafenib were all assayed in the adjuvant setting in trials published from 2016–2020. Of these trials, the only one to show a significant difference in a primary endpoint was the S-TRAC trial of sunitinib. Negative placebo-controlled trials of VEGFR TKIs in the adjuvant setting were ASSURE (2016) with sunitinib and sorafenib (52), PROTECT (2017) with pazopanib (53), ATLAS (2018) with axitinib (54), and SORCE (2020) with sorafenib (55). Each is explored in more detail in this section.
The S-TRAC trial enrolled 615 patients with recently resected ccRCC (≥pT3 and/or regional nodal involvement, ECOG ≤2, M=0) (56). Patients were randomly assigned 1:1 to receive either 50 mg of sunitinib or placebo for 1 year in a 4 weeks on, 2 weeks off schedule. Results published in 2016 showed that sunitinib was associated with a median DFS (determined by central review) of 6.8 years compared to 5.6 years in the placebo group (HR 0.76, 95% CI: 0.59–0.98). This improvement in the treatment group came at a cost of greater toxic events, with 63.4% and 21.7% of patients experiencing an adverse event of grade 3 or higher in the sunitinib and placebo groups, respectively. The rate of adverse events labeled as serious, however, was similar between the two groups (21.9% for sunitinib versus 17.1% for placebo). The most common adverse events in the sunitinib group included diarrhea, palmar-plantar erythrodysesthesia, and hypertension. Patients in the treatment group also reported significantly lower health-related quality of life on many items via questionnaire, with diarrhea and loss of appetite considered clinically meaningful by the investigators. The follow-up analysis continued to show increased DFS in the sunitinib group compared to placebo, 6.8 vs. 5.6 years respectively (HR 0.76, 95% CI: 0.76–0.98) (57). The HR for OS comparing sunitinib to placebo was 0.92 (95% CI: 0.66–1.28, P=0.6).
ASSURE (adjuvant sunitinib or sorafenib vs. placebo in resected unfavorable renal cell carcinoma), was a multicenter North American study that enrolled 1,943 patients in a 1:1:1 three-arm design to compare sunitinib, sorafenib, and placebo (52). Inclusion criteria were different from the S-TRAC trial with participants having T1b or greater high grade RCC and inclusion of patients with non-ccRCC (21% of total). Patients in the active drug groups suffered high toxicities, with rates of discontinuation in the sunitinib and sorafenib groups 44% and 45%, respectively. In response to this, the dosing scheme was revised after the trial began, however toxicity remained a serious issue. The most commonly reported adverse events were hypertension, hand-foot syndrome, rash, and fatigue. The results published in 2016 showed no difference between groups in the primary end point of DFS (by investigator review), with 5.8 years for sunitinib (HR 1.02, 97.5% CI: 0.85–1.23, P=0.8038), 6.1 years for sorafenib (HR 0.97, 97.5% CI: 0.80–1.17, P=0.7184), and 6.6 years for placebo. Secondary analysis of a high-risk subset of patients (node-positive or pT3 or greater), showed no effect on 5-year DFS and no effect of dosing on outcome (58).
PROTECT (Pazopanib As Adjuvant Therapy in Localized/Locally Advanced RCC After Nephrectomy) was a trial that enrolled 1,538 patients with ccRCC and M=0 after partial or radical nephrectomy (pT2G3–4N0, pT3–T4 GanyN0, or pTanyGanyN1) to compare pazopanib versus placebo (59). In response to a higher than anticipated discontinuation rate due to toxicity, the starting dose was amended from 800 to 600 mg and the primary endpoint became DFS in the 600 mg group (n=571) versus placebo (n=564). Elevations in AST and ALT were the most common adverse events for patients. Initial results published in 2017 showed improved but not significant DFS in the 600 mg pazopanib group (HR 0.86, 95% CI: 0.70–1.06, P=0.165). Final follow-up analysis in 2021 showed no significant difference in OS between groups (HR 1.0, 95% CI: 0.80–1.26, P>0.9) (53). Subgroup analysis of the PROTECT trial showed that treatment with sunitinib was actually associated with significantly lower OS in older women (60).
The ATLAS (Adjuvant Axitinib Therapy of Renal Cell Cancer in High Risk Patients) trial enrolled 724 patients in centers across Asia, Europe, and the U.S. with recently diagnosed RCC (>50% clear cell component) and ≥pT2 and/or nodal involvement with any grade and ECOG 0–1 (54). Patients were assigned 1:1 to receive either 5 mg axitinib twice per day or placebo for between 1 and 3 years. About 56% of the study sample was considered “highest risk” with pT3 and grade ≥3, pT4, or nodal involvement. The study was stopped early due to futility, failing to show a difference in the primary end point of DFS (determined by central committee) with a hazard ratio of 0.870 (95% CI: 0.660–1.147, P=0.3211). Axitinib was associated with greater grade 3 or 4 adverse events (61% versus 30% in placebo).
Lastly, the SORCE trial assigned patients into three groups (in a 2:3:3 ratio): the first group received 3 years of placebo, the second group received 1 year of sorafenib followed by 2 years of placebo, and the third group received 3 years of sorafenib (55). 1,711 patients were enrolled with completely resected RCC of any histology, M=0, and an intermediate or high risk of relapse according to the 2003 Leibovich risk model. Similar to other trials of VEGFR inhibitors, the protocol was amended in response to high discontinuation rates, with the dose of sorafenib reduced from 400 mg twice per day to 400 mg once per day. Despite this change, 58.6% of patients receiving sorafenib for 1 year and 63.9% of patients receiving sorafenib for 3 years reported an adverse reaction of grade 3 or higher, compared to 29.2% in the placebo group. The primary outcome analysis published in 2020 showed no difference in survival between the group who received sorafenib for 3 years versus placebo for three years (HR 1.01, 95% CI: 0.83–1.23, P=0.95).
Based on the positive signal in the S-TRAC trial, the FDA approved sunitinib in 2017 for treatment of RCC after surgery for resection, the first therapy approved for this indication (61). This move was widely criticized by providers who were not convinced that the treatment toxicities were outweighed by the potential benefits, which had not been replicated in other trials of VEGFR TKIs (62). Indeed, meta-analysis by Riaz et al. [2021] showed with high certainty that there was no benefit in OS (HR 1.01, 95% CI: 0.91–1.12) or DFS (HR 0.92, 95% CI: 0.86–1.00), including across subgroups and in the highest-risk patients (63). Because of this conflicting data and the drug’s significant toxicity, sunitinib was not widely adopted by clinicians despite being the first RCC drug formally approved in the adjuvant setting.
Trial of a mammalian target of rapamycin (mTOR)R pathway inhibitor in the adjuvant setting
The mTOR pathway has been another target of investigation given that this pathway is often inappropriately upregulated in RCC (64). mTOR inhibitor temsirolimus was approved by the FDA as first line therapy for metastatic RCC in 2007 after a phase III trial showed superiority in OS for patients taking temsirolimus plus interferon alpha compared to those taking interferon-alpha alone (65). Another mTOR inhibitor, everolimus, was approved by the FDA in 2009 for patients with metastatic RCC who had failed therapy with a VEGFR TKI (66). Despite mTOR inhibitors’ success in treating metastatic disease, the only published phase III trial to date of an mTOR inhibitor as adjuvant therapy has not shown improvement in outcomes for patients with resected RCC. The EVEREST trial [2023] looked at 1,545 RCC patients (any histology) who were deemed to have intermediate-high or very high risk of recurrence per a modified UISS, comparing daily everolimus for one year to placebo (67). The everolimus group did have longer RFS than the placebo group, with 5-year RFS of 67% and 63%, respectively (log rank P=0.05). However, this did not meet the study’s pre-specified threshold of significance of P=0.044. Sub-analysis showed that only the very-high risk group had longer RFS with everolimus compared with placebo (HR 0.79, 95% CI: 0.65–0.97, P=0.022). The everolimus group also had significantly more grade 3 or higher adverse events compared to placebo (46% vs. 11%). The authors concluded that the results do not support the use of everolimus for RCC in the adjuvant setting.
Trials of immune checkpoint inhibitors (ICIs) in the adjuvant setting
Therapies harnessing the human immune system to fight cancer, in particular the ICIs of programmed death-1 (PD-1), programmed death-ligand 1 (PD-L1), and cytotoxic T-lymphocyte antigen-4 (CTLA-4), have shown great promise in treating a wide variety of cancers over the last decade. Nivolumab was the first such therapy approved in 2015 for patients with metastatic RCC previously treated with anti-angiogenic therapy, based on data from a phase III trial showing an OS benefit when compared to everolimus (68,69). Since then, several phase III trials of ICIs have been performed in the adjuvant setting (see Table 2), with only one (KEYNOTE-564) meeting its primary endpoints and demonstrating a survival benefit.
Table 2
KEYNOTE-564 [2021] | PROSPER [2022] | IMmotion-010 [2022] | Checkmate-914 [2023] | |
---|---|---|---|---|
Agent (target) | Pembrolizumab (PD-1) | Nivolumab (PD-1) | Atezolizumab (PD-L1) | Nivolumab (PD-1) + ipilimumab (CTLA-4) |
M1 NED? | Yes | Yes | Yes | No |
Histology | ccRCC | Any | ccRCC or sarcomatoid | – |
Number of patients | 994 | 819 | 778 | 816 |
Inclusion criteria | Stage II with nuclear grade 4 or sarcomatoid differentiation, stage III or higher, regional lymph node-metastasis, and/or M1 with NED | ≥pT2, pTanyN+, or M1 with NED | pT2 with grade 4, pT3a with grade 3–4, pT3b through pT4 with any grade, nodal involvement, and/or M1 with NED | pT2a (grade III–IV) N0M0, pT2b-4N0M0 (any grade), or pTany (any grade) N1M0 |
Treatment duration | 200 mg of pembrolizumab every 3 weeks for up to a year | 1 pre-op dose of nivolumab followed by 9 post-op doses, 450 mg every 4 weeks | 1,200 mg atezolizumab every 3 weeks for 1 year | 12 doses of 240 mg nivolumab every 2 weeks; 4 doses of 1 mg/kg ipilimumab every 6 weeks |
Rates of discontinuation due to toxicity | 21.3% in treatment, 2% in placebo | 12% overall discontinuation in treatment and control (reason not specified) | 11.5% in treatment, 2.6% in placebo | 29.0% in treatment, 1.0% in placebo |
Primary end point & results | DFS: HR for recurrence or death 0.68, 95% CI: 0.53–0.87, P=0.002 | RFS: HR 0.97, 95% CI: 0.74–1.28, P1-sided=0.43 | DFS: HR 0.93, 95% CI: 0.75–1.15, P=0.50 | DFS: HR 0.92, 95% CI: 0.71–1.19, P=0.53 |
OS: HR for death 0.54, 95% CI: 0.30–0.96 (data immature) | OS: HR 1.48, 95% CI: 0.89–2.48, P1-sided =0.93 (data not mature) | |||
Further data expected? | Yes, per most recent follow-up | Ended early due to futility | No | No |
PD-1, programmed death-1; PD-L1, programmed death-ligand 1; CTLA-4, cytotoxic T-lymphocyte antigen-4; NED, no evidence of disease; ccRCC, clear cell renal cell carcinoma; DFS, disease-free survival; HR, hazard ratio; CI, confidence interval; OS, overall survival.
The KEYNOTE-564 trial randomly assigned 994 patients to either receive placebo or 200 mg of PD-1 inhibitor pembrolizumab every 3 weeks for up to a year (70). Patients exclusively had ccRCC and included those at stage II with nuclear grade 4 or sarcomatoid differentiation, stage III or higher, regional lymph node-metastasis, and/or M1 with NED. Initial analysis showed significantly longer DFS in the pembrolizumab group compared to the placebo group, with 24-month DFS rates of 77.3% and 68.1%, respectively (HR for recurrence or death 0.68, 95% CI: 0.53–0.87, P=0.002). OS was higher in the treatment group (HR for death 0.54, 95% CI: 0.30–0.96), however the data were immature, preventing definitive interpretation. Toxicities due to treatment were as expected based on prior experience with pembrolizumab, with 32.4% of patients experiencing a grade 3 or higher adverse reaction compared to 17.7% in placebo, the most common including hypertension and increased alanine aminotransferase levels. In response to this trial, the FDA approved pembrolizumab in 2021 as adjuvant therapy for patients at intermediate-high or high risk of recurrence following nephrectomy with or without metastasectomy (71). Follow-up analysis published in 2022 showed that the pembrolizumab group continued to demonstrate superior DFS (HR 0.63, 95% CI: 0.5–0.8) (72) and recent data from the third prespecified interim analysis showed a clinically meaningful and statistically significant increase in the secondary endpoint of OS in the pembrolizumab group compared to placebo, with no new safety signals observed (73).
The PROSPER study was an open-label trial comparing PD-1 inhibitor nivolumab versus observation in the perioperative setting for 819 patients with ≥pT2, pTanyN+, or M1 with NED after partial or total nephrectomy for RCC of any histology (74). This study was unique compared to other adjuvant ICI trials in that it included a pre-operative dose of nivolumab in addition to 9 post-operative doses (480 mg every 4 weeks). The study was stopped early due to futility after interim analysis showed similar RFS between arms (HR 0.97, 95% CI: 0.74–1.28, P1-sided=0.43). OS was not mature at the time the study was stopped but this was also similar between groups (HR 1.48, 95% CI: 0.89–2.48, P1-sided=0.93).
Published in 2022, IMmotion010 randomly assigned patients to receive either PD-L1 inhibitor atezolizumab or placebo (75). Its sample included 778 patients with RCC (with a clear cell or sarcomatoid histological component) and pT2 with grade 4, pT3a with grade 3–4, pT3b through pT4 with any grade, nodal involvement, and/or M1 with NED after nephrectomy and/or metastasectomy. DFS as assessed by investigator did not differ significantly between groups (HR 0.93, 95% CI: 0.75–1.15, P=0.50). 18% of patients on nivolumab and 12% of patients on placebo had a serious adverse event.
The most recent trial to be published of an ICI in the adjuvant setting for RCC, CheckMate-914, was a 2-part trial comparing the combination of nivolumab plus ipilimumab and nivolumab monotherapy to placebo (76). The Part A (ipi+nivo vs. placebo) treatment group received 240 mg of nivolumab every 2 weeks for 12 doses with 1 mg/kg of ipilimumab every 6 weeks for four doses. The study’s patient sample included 816 patients at high risk of recurrence [staging pT2a(grade III–IV)N0M0, pT2b(any grade)N0M0, pT3(any grade)N0M0, pT4(any grade)N0M0, or pTany(any grade) N1M0]. Notably, those who were at stage M1 with NED were excluded. Primary analysis showed no significant difference between the treatment and placebo group with regards to DFS (HR 0.92, 95% CI: 0.71–1.19, P=0.53). OS data were not mature at the time of data cutoff. Those receiving nivolumab and ipilimumab experienced considerably more toxicity than the placebo group, with rates of adverse events leading to discontinuation of 32% and 2% for treatment and placebo, respectively. Part B (ipi+nivo vs. nivo vs. placebo) presented at the 2024 ASCO Genitourinary Cancers Symposium showed no DFS benefit from adjuvant nivolumab monotherapy.
A meta-analysis of the four trials with data available showed considerable heterogeneity across studies due to inclusion criteria and statistical methods (I2=64%) as well as no significant improvement in DFS in the pooled dataset (HR 0.85, 95% CI: 0.69–1.04, P=0.11) (77). Differential DFS was seen, however, when patients were stratified by PD-L1 expression (positive versus negative, HR 0.72, 95% CI: 0.55–0.94, P=0.02) and presence of sarcomatoid features (HR 0.59, 95% CI: 0.38–0.91, P=0.02). Stratification by nephrectomy type (partial vs. radical) or risk category (intermediate-high, high, M1 NED) did not show a differential effect of ICIs.
Differences in patient inclusion criteria, dosing, treatment discontinuation and treatment duration across studies have been posited as explanations for disparate results (76). Patients who were M1 with NED were able to enroll in IMmotion010, PROSPER, and KEYNOTE-564 but not CheckMate-914. PROSPER and IMmotion010 allowed patients with non-clear cell histology to enroll but KEYNOTE-564 and CheckMate-914 did not. Whereas patients in KEYNOTE-564 and IMmotion010 were protocoled to receive treatment for 1 year, those in PROSPER and CheckMate-914 were planned for 9 months and 6 months, respectively. Given these factors, it is tough to put each individual trial’s results in the context of the others when determining whether these agents have a potential role for RCC patients in the adjuvant setting.
Current standard of care: guidelines and clinical practice
Recent developments in the field of ICIs, and the resulting FDA approval of pembrolizumab as adjuvant therapy in 2021, have given patients and clinicians more options for post-surgical treatment of RCC. However, many clinical societies have yet to incorporate trials from the last several years into official guidelines, including the National Comprehensive Cancer Network (NCCN) and American Urological Association (AUA) (8,78). The European Association of Urology (EAU) issued a guideline update in 2022 with a “weak” recommendation for pembrolizumab but advised caution given the negative trials published of other ICI drugs (6).
The only other drug approved for adjuvant therapy in RCC, sunitinib, has not been routinely used in clinical practice in the United States despite its FDA approval, with many citing concerns about negative trials and toxicities for patients (62). NCCN and AUA guidelines recommend that patients with stage III Category 3 ccRCC at high risk of recurrence receive counseling on systemic treatment with sunitinib and consider enrolling in a clinical trial of adjuvant therapy (preferred) or active surveillance (8,78). Sunitinib never achieved regulatory approval in Europe, and up until its 2022 update incorporating the KEYNOTE-564 trial, EAU’s guidelines recommended systemic adjuvant therapy only in the context of a clinical trial (79,80).
Within the existing NCCN framework for counseling patients on options after nephrectomy, many clinicians are offering adjuvant pembrolizumab to those patients at high risk of recurrence. Patients who received benefit per the KEYNOTE-564 included those with “intermediate-high” risk (pT2N0M0 with grade 4 or sarcomatoid features or pT3N0M0 with any grade), “high risk” (pT4N0M0 any grade or any pT, any grade, N+, M0), and M1 NED (81). Those who were M1 NED received the highest DFS benefit from pembrolizumab in subgroup analysis of the trial (72). Patients had good functional status, with ECOG 0 or 1, and received the therapy within 12 weeks of surgery (81). For patients at high risk of recurrence as defined by these criteria or by other commonly used risk stratifiers (UISS, Leibovich, etc.), enrolling in a clinical trial or active surveillance remain valid treatment strategies, especially for those concerned about efficacy of ICIs and side effects of systemic therapy. Ultimately, clinicians must engage in conversation with patients so that they can make their own informed decision of how to proceed with care after a nephrectomy and/or metastasectomy, as summarized in Figure 2.
Future directions
Adjuvant therapy in patients with high risk localized ccRCC is a rapidly evolving field. The most exciting and consequential recent development is the data from KEYNOTE-564 showing that adjuvant pembrolizumab increases OS in a high-risk population, representing a new standard of care for ccRCC patients after surgery. Trials that are ongoing are summarized in Table 3 below. In particular, two phase III trials are in the pipeline with primary results expected in coming years. The first, RAMPART, is a study based in the United Kingdom that uses a three-arm design to compare PD-1 inhibitor durvalumab +/− CTLA-4 inhibitor tremelimumab versus placebo. It has a goal recruitment of 1750 RCC patients of any histology with Leibovich scores from 3–11. It is estimated to reach its primary completion date in July 2024. The other phase III study, LITESPARK-022, aims to test the efficacy of an PD-1 inhibitor pembrolizumab in combination with hypoxia-inducible factor 2 alpha inhibitor belzutifan as adjuvant RCC therapy. It is a two-arm, placebo-controlled study organized by Merck Sharp & Dohme LLC. Patients will have ccRCC that is intermediate-high risk (pT2, grade 4 or sarcomatoid, N0M0; pT2, any grade, N0M0), high risk (pT4, any grade, N0M0; pTany stage, any grade, N+, M0), or M1 with NED. The recruitment goal is 1600 patients with an expected primary completion date in October 2027.
Table 3
Trial name (NCT number) | Sponsor | Phase | Sample size | Primary end point(s) | Summary of design | Est. primary completion date |
---|---|---|---|---|---|---|
KEYNOTE-564 (NCT 03142334) | Merck Sharp & Dohme LLC | 3 | 994 (actual) | DFS as assessed by investigator | Initial data analysis showed pembrolizumab superior to placebo for PFS in intermediate-high risk, high risk, or M1 NED RCC with clear cell component | Completed 12/14/2020 |
Recently met secondary endpoint of OS | ||||||
CheckMate 914 (NCT03138512) | Bristol-Myers Squibb | 3 | 1,653 (actual) | DFS as assessed by BICR | Three-arm study in patients with predominantly clear cell histology who received nephrectomy: nivolumab vs. nivolumab + ipilimumab vs. placebo | 9/12/2023 |
RAMPART (NCT03288532) | University College, London, UK | 3 | 1,750 | DFS, OS (high-risk only) | Three-arm design assessing no intervention (active monitoring) vs. durvalumab vs. durvalumab + tremelimumab | 7/1/2024 |
PE-PE (NCT05578664) | Consorzio Oncotech | 2 | 81 | RFS | Study enrolling patients with oligometastatic disease (max 3 mets) who will have locally directed therapy (surgery or radiation). The experimental arm will receive pembrolizumab in addition to locally directed therapy (with first dose given three weeks before) and the control arm will receive locally directed therapy alone | 10/2024 |
MRD GATE RCC (NCT06005818) | University of Alabama at Birmingham, USA | 2 | 100 | PFS | Patients who have undergone surgery for localized ccRCC will be assessed for MRD with pembrolizumab given only to patients in whom MRD is detected | 12/31/2024 |
IUNU-RC (NCT05768464) | Nanjing University Medical School, China | 2 | 30 | DFS | Single arm study looking at efficacy/safety of toripalimab + axitinib in patients with non-clear cell RCC with high risk for recurrence | 12/30/2026 |
LITESPARK-022 (NCT05239728) | Merck Sharp & Dohme LLC | 3 | 1,600 | DFS | Two-arm study to compare pembrolizumab plus belzutifan vs. pembrolizumab plus placebo in patients with ccRCC after nephrectomy | 10/25/2027 |
RCC, renal cell carcinoma; NCT, national clinical trial; DFS, disease-free survival; PFS, progression-free survival; NED, no evidence of disease; OS, overall survival; BICR, Blinded Independent Central Review; RFS, recurrence-free survival; ccRCC, clear cell RCC; MRD, molecular residual disease.
Besides these phase III trials, there are several phase II trials to follow in the pipeline as well. IUNU-RC is a phase II study developed by Nanjing University in China looking at the efficacy and safety of toripalimab (PD-1 inhibitor) plus VEGF inhibitor axitinib, specifically in patients with non-ccRCC at high risk for recurrence. This represents one of the few adjuvant studies performed specifically for patients with non-ccRCC. MRD GATE RCC is a trial at the University of Alabama at Birmingham evaluating pembrolizumab after surgery for localized RCC only for patients with molecular residual disease (MRD) as evidenced by presence of circulating tumor DNA (ctDNA). Patients without MRD in this study will receive surveillance only. Lastly, PE-PE is an Italian phase II study looking at pembrolizumab as adjuvant treatment after local therapy with surgery or radiation for up to 3 metastases. The treatment group will receive pembrolizumab in addition to local therapy while the control group will receive local therapy alone.
Conclusions
In conclusion, we have reached a pivotal moment for adjuvant treatment of patients with high risk localized ccRCC after surgery. The data from KEYNOTE-564 trial data showing not only an improved progression-free survival but also an OS benefit is transformative and this represents a new standard of care. We are optimistic about new trials such as RAMPART and LITESPARK-022 which may provide additional treatment options in the adjuvant setting. Further, genomic and clinical biomarkers may help to objectively risk stratify patients so that we may identify those who would derive the most benefit from adjuvant therapy. Overall, the future looks far brighter now than it did even several years ago: after a long streak of unsuccessful trials, progress is here at last.
Acknowledgments
Funding: None.
Footnote
Peer Review File: Available at https://tcr.amegroups.com/article/view/10.21037/tcr-23-2247/prf
Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tcr.amegroups.com/article/view/10.21037/tcr-23-2247/coif). M.N.S. reports receiving grants or contracts from Janssen Oncology, Advaxis, Bristol-Myers Squibb, Lilly, Seattle Genetics, Xencor, Tmunity, Exelixis, Bellicum Pharmaceuticals, Regeneron, Bicycle Therapeutics and AstraZeneca, all of which have provided payment to his institution; and also receiving consulting fees from Exelixis, Xencor, Janssen, Vaccitech, Merck, and Bristol-Myers Squibb/Medarex. K.R. reports receiving funding from the 2023 Robert A. Winn Diversity in Clinical Trials Career Development Award, and a payment for serving as a steering committee for a phase III trial for Janseen. The other author has no conflicts of interest to declare.
Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.
Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.
References
- Siegel RL, Miller KD, Wagle NS, et al. Cancer statistics, 2023. CA Cancer J Clin 2023;73:17-48. [Crossref] [PubMed]
- Capitanio U, Bensalah K, Bex A, et al. Epidemiology of Renal Cell Carcinoma. Eur Urol 2019;75:74-84. [Crossref] [PubMed]
- Padala SA, Barsouk A, Thandra KC, et al. Epidemiology of Renal Cell Carcinoma. World J Oncol 2020;11:79-87. [Crossref] [PubMed]
- Hanna N, Sun M, Meyer CP, et al. Survival Analyses of Patients With Metastatic Renal Cancer Treated With Targeted Therapy With or Without Cytoreductive Nephrectomy: A National Cancer Data Base Study. J Clin Oncol 2016;34:3267-75. [Crossref] [PubMed]
- Albiges L, Powles T, Staehler M, et al. Updated European Association of Urology Guidelines on Renal Cell Carcinoma: Immune Checkpoint Inhibition Is the New Backbone in First-line Treatment of Metastatic Clear-cell Renal Cell Carcinoma. Eur Urol 2019;76:151-6. [Crossref] [PubMed]
- Ljungberg B, Albiges L, Abu-Ghanem Y, et al. European Association of Urology Guidelines on Renal Cell Carcinoma: The 2022 Update. Eur Urol 2022;82:399-410. [Crossref] [PubMed]
- Weight CJ, Mulders PF, Pantuck AJ, et al. The Role of Adrenalectomy in Renal Cancer. Eur Urol Focus 2016;1:251-7. [Crossref] [PubMed]
- Campbell SC, Clark PE, Chang SS, et al. Renal Mass and Localized Renal Cancer: Evaluation, Management, and Follow-Up: AUA Guideline: Part I. J Urol 2021;206:199-208. [Crossref] [PubMed]
- Kim SP, Thompson RH, Boorjian SA, et al. Comparative effectiveness for survival and renal function of partial and radical nephrectomy for localized renal tumors: a systematic review and meta-analysis. J Urol 2012;188:51-7. [Crossref] [PubMed]
- Mir MC, Derweesh I, Porpiglia F, et al. Partial Nephrectomy Versus Radical Nephrectomy for Clinical T1b and T2 Renal Tumors: A Systematic Review and Meta-analysis of Comparative Studies. Eur Urol 2017;71:606-17. [Crossref] [PubMed]
- Pierorazio PM, Johnson MH, Patel HD, et al. Management of Renal Masses and Localized Renal Cancer: Systematic Review and Meta-Analysis. J Urol 2016;196:989-99. [Crossref] [PubMed]
- Ouzaid I, Capitanio U, Staehler M, et al. Surgical Metastasectomy in Renal Cell Carcinoma: A Systematic Review. Eur Urol Oncol 2019;2:141-9. [Crossref] [PubMed]
- Janzen NK, Kim HL, Figlin RA, et al. Surveillance after radical or partial nephrectomy for localized renal cell carcinoma and management of recurrent disease. Urol Clin North Am 2003;30:843-52. [Crossref] [PubMed]
- Dabestani S, Beisland C, Stewart GD, et al. Long-term Outcomes of Follow-up for Initially Localised Clear Cell Renal Cell Carcinoma: RECUR Database Analysis. Eur Urol Focus 2019;5:857-66. [Crossref] [PubMed]
- Larroquette M, Peyraud F, Domblides C, et al. Adjuvant therapy in renal cell carcinoma: Current knowledges and future perspectives. Cancer Treat Rev 2021;97:102207. [Crossref] [PubMed]
- Ciccarese C, Strusi A, Arduini D, et al. Post nephrectomy management of localized renal cell carcinoma. From risk stratification to therapeutic evidence in an evolving clinical scenario. Cancer Treat Rev 2023;115:102528. [Crossref] [PubMed]
- Zisman A, Pantuck AJ, Dorey F, et al. Improved prognostication of renal cell carcinoma using an integrated staging system. J Clin Oncol 2001;19:1649-57. [Crossref] [PubMed]
- Leibovich BC, Blute ML, Cheville JC, et al. Prediction of progression after radical nephrectomy for patients with clear cell renal cell carcinoma: a stratification tool for prospective clinical trials. Cancer 2003;97:1663-71. [Crossref] [PubMed]
- Frank I, Blute ML, Cheville JC, et al. 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. [Crossref] [PubMed]
- Karakiewicz PI, Trinh QD, Bhojani N, et al. Renal cell carcinoma with nodal metastases in the absence of distant metastatic disease: prognostic indicators of disease-specific survival. Eur Urol 2007;51:1616-24. [Crossref] [PubMed]
- Gupta S, Sukov WR, Vanderbilt CM, et al. A contemporary guide to chromosomal copy number profiling in the diagnosis of renal cell carcinoma. Urol Oncol 2022;40:512-24. [Crossref] [PubMed]
- Şenbabaoğlu Y, Gejman RS, Winer AG, et al. Tumor immune microenvironment characterization in clear cell renal cell carcinoma identifies prognostic and immunotherapeutically relevant messenger RNA signatures. Genome Biol 2016;17:231. [Crossref] [PubMed]
- Cotta BH, Choueiri TK, Cieslik M, et al. Current Landscape of Genomic Biomarkers in Clear Cell Renal Cell Carcinoma. Eur Urol 2023;84:166-75. [Crossref] [PubMed]
- Mano R, Duzgol C, Ganat M, et al. Somatic mutations as preoperative predictors of metastases in patients with localized clear cell renal cell carcinoma - An exploratory analysis. Urol Oncol 2021;39:791.e17-24. [Crossref] [PubMed]
- Brooks SA, Brannon AR, Parker JS, et al. ClearCode34: A prognostic risk predictor for localized clear cell renal cell carcinoma. Eur Urol 2014;66:77-84. [Crossref] [PubMed]
- Rini B, Goddard A, Knezevic D, et al. A 16-gene assay to predict recurrence after surgery in localised renal cell carcinoma: development and validation studies. Lancet Oncol 2015;16:676-85. [Crossref] [PubMed]
- Harada K, Miyake H, Kusuda Y, et al. Expression of epithelial-mesenchymal transition markers in renal cell carcinoma: impact on prognostic outcomes in patients undergoing radical nephrectomy. BJU Int 2012;110:E1131-7. [Crossref] [PubMed]
- Nallandhighal S, Vince R, Karim R, et al. Molecular Characterization of Clear Cell Renal Cell Carcinoma Reveals Prognostic Significance of Epithelial-mesenchymal Transition Gene Expression Signature. Eur Urol Oncol 2022;5:92-9. [Crossref] [PubMed]
- Morgan TM, Mehra R, Tiemeny P, et al. A Multigene Signature Based on Cell Cycle Proliferation Improves Prediction of Mortality Within 5 Yr of Radical Nephrectomy for Renal Cell Carcinoma. Eur Urol 2018;73:763-9. [Crossref] [PubMed]
- Dizman N, Philip EJ, Pal SK. Genomic profiling in renal cell carcinoma. Nat Rev Nephrol 2020;16:435-51. [Crossref] [PubMed]
- Kato S, Demura S, Murakami H, et al. Surgical metastasectomy for renal cell carcinoma: which patients are the real candidates for surgery? Ann Transl Med 2019;7:S273. [Crossref] [PubMed]
- Loh J, Davis ID, Martin JM, et al. Extracranial oligometastatic renal cell carcinoma: current management and future directions. Future Oncol 2014;10:761-74. [Crossref] [PubMed]
- Bedke J, Albiges L, Capitanio U, et al. 2021 Updated European Association of Urology Guidelines on the Use of Adjuvant Pembrolizumab for Renal Cell Carcinoma. Eur Urol 2022;81:134-7. [Crossref] [PubMed]
- Geynisman DM, Maranchie JK, Ball MW, et al. A 25 year perspective on the evolution and advances in an understanding of the biology, evaluation and treatment of kidney cancer. Urol Oncol 2021;39:548-60. [Crossref] [PubMed]
- Clement JM, McDermott DF. The high-dose aldesleukin (IL-2) "select" trial: a trial designed to prospectively validate predictive models of response to high-dose IL-2 treatment in patients with metastatic renal cell carcinoma. Clin Genitourin Cancer 2009;7:E7-E9. [Crossref] [PubMed]
- Clark JI, Atkins MB, Urba WJ, et al. Adjuvant high-dose bolus interleukin-2 for patients with high-risk renal cell carcinoma: a cytokine working group randomized trial. J Clin Oncol 2003;21:3133-40. [Crossref] [PubMed]
- Atzpodien J, Schmitt E, Gertenbach U, et al. Adjuvant treatment with interleukin-2- and interferon-alpha2a-based chemoimmunotherapy in renal cell carcinoma post tumour nephrectomy: results of a prospectively randomised trial of the German Cooperative Renal Carcinoma Chemoimmunotherapy Group (DGCIN). Br J Cancer 2005;92:843-6. [Crossref] [PubMed]
- Passalacqua R, Caminiti C, Buti S, et al. Adjuvant low-dose interleukin-2 (IL-2) plus interferon-α (IFN-α) in operable renal cell carcinoma (RCC): a phase III, randomized, multicentre trial of the Italian Oncology Group for Clinical Research (GOIRC). J Immunother 2014;37:440-7. [Crossref] [PubMed]
- Hanna MG Jr, Hoover HC Jr, Vermorken JB, et al. Adjuvant active specific immunotherapy of stage II and stage III colon cancer with an autologous tumor cell vaccine: first randomized phase III trials show promise. Vaccine 2001;19:2576-82. [Crossref] [PubMed]
- Galligioni E, Quaia M, Merlo A, et al. Adjuvant immunotherapy treatment of renal carcinoma patients with autologous tumor cells and bacillus Calmette-Guèrin: five-year results of a prospective randomized study. Cancer 1996;77:2560-6. [Crossref] [PubMed]
- Jocham D, Richter A, Hoffmann L, et al. Adjuvant autologous renal tumour cell vaccine and risk of tumour progression in patients with renal-cell carcinoma after radical nephrectomy: phase III, randomised controlled trial. Lancet 2004;363:594-9. [Crossref] [PubMed]
- Fishman M, Antonia S. Specific antitumour vaccine for renal cancer. Lancet 2004;363:583-4. [Crossref] [PubMed]
- Wood C, Srivastava P, Bukowski R, et al. An adjuvant autologous therapeutic vaccine (HSPPC-96; vitespen) versus observation alone for patients at high risk of recurrence after nephrectomy for renal cell carcinoma: a multicentre, open-label, randomised phase III trial. Lancet 2008;372:145-54. [Crossref] [PubMed]
- Stillebroer AB, Mulders PF, Boerman OC, et al. Carbonic anhydrase IX in renal cell carcinoma: implications for prognosis, diagnosis, and therapy. Eur Urol 2010;58:75-83. [Crossref] [PubMed]
- Divgi CR, Uzzo RG, Gatsonis C, et al. Positron emission tomography/computed tomography identification of clear cell renal cell carcinoma: results from the REDECT trial. J Clin Oncol 2013;31:187-94. [Crossref] [PubMed]
- Siebels M, Rohrmann K, Oberneder R, et al. A clinical phase I/II trial with the monoclonal antibody cG250 (RENCAREX®) and interferon-alpha-2a in metastatic renal cell carcinoma patients. World J Urol 2011;29:121-6. [Crossref] [PubMed]
- Bleumer I, Knuth A, Oosterwijk E, et al. A phase II trial of chimeric monoclonal antibody G250 for advanced renal cell carcinoma patients. Br J Cancer 2004;90:985-90. [Crossref] [PubMed]
- Bleumer I, Oosterwijk E, Oosterwijk-Wakka JC, et al. A clinical trial with chimeric monoclonal antibody WX-G250 and low dose interleukin-2 pulsing scheme for advanced renal cell carcinoma. J Urol 2006;175:57-62. [Crossref] [PubMed]
- Chamie K, Donin NM, Klöpfer P, et al. Adjuvant Weekly Girentuximab Following Nephrectomy for High-Risk Renal Cell Carcinoma: The ARISER Randomized Clinical Trial. JAMA Oncol 2017;3:913-20. [Crossref] [PubMed]
- Posadas EM, Limvorasak S, Sharma S, et al. Targeting angiogenesis in renal cell carcinoma. Expert Opin Pharmacother 2013;14:2221-36. [Crossref] [PubMed]
- Motzer RJ, Hutson TE, Tomczak P, et al. Sunitinib versus interferon alfa in metastatic renal-cell carcinoma. N Engl J Med 2007;356:115-24. [Crossref] [PubMed]
- Haas NB, Manola J, Uzzo RG, 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. [Crossref] [PubMed]
- Motzer RJ, Russo P, Haas N, et al. Adjuvant Pazopanib Versus Placebo After Nephrectomy in Patients With Localized or Locally Advanced Renal Cell Carcinoma: Final Overall Survival Analysis of the Phase 3 PROTECT Trial. Eur Urol 2021;79:334-8. [Crossref] [PubMed]
- Gross-Goupil M, Kwon TG, Eto M, 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. [Crossref] [PubMed]
- Eisen T, Frangou E, Oza B, et al. Adjuvant Sorafenib for Renal Cell Carcinoma at Intermediate or High Risk of Relapse: Results From the SORCE Randomized Phase III Intergroup Trial. J Clin Oncol 2020;38:4064-75. [Crossref] [PubMed]
- Ravaud A, Motzer RJ, Pandha HS, et al. Adjuvant Sunitinib in High-Risk Renal-Cell Carcinoma after Nephrectomy. N Engl J Med 2016;375:2246-54. [Crossref] [PubMed]
- Motzer RJ, Ravaud A, Patard JJ, et al. Adjuvant Sunitinib for High-risk Renal Cell Carcinoma After Nephrectomy: Subgroup Analyses and Updated Overall Survival Results. Eur Urol 2018;73:62-8. [Crossref] [PubMed]
- Haas NB, Manola J, Dutcher JP, et al. Adjuvant Treatment for High-Risk Clear Cell Renal Cancer: Updated Results of a High-Risk Subset of the ASSURE Randomized Trial. JAMA Oncol 2017;3:1249-52. [Crossref] [PubMed]
- Motzer RJ, Haas NB, Donskov F, 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. [Crossref] [PubMed]
- Mamtani R, Wang XV, Gyawali B, et al. Association between age and sex and mortality after adjuvant therapy for renal cancer. Cancer 2019;125:1637-44. [Crossref] [PubMed]
- Martinez Chanza N, Tripathi A, Harshman LC. Adjuvant Therapy Options in Renal Cell Carcinoma: Where Do We Stand? Curr Treat Options Oncol 2019;20:44. [Crossref] [PubMed]
- Gyawali B, Goldstein DA. The US Food and Drug Administration's Approval of Adjuvant Sunitinib for Renal Cell Cancer: A Case of Regulatory Capture?. JAMA Oncol 2018;4:623-4. [Crossref] [PubMed]
- Riaz IB, Siddiqi R, Islam M, et al. Adjuvant Tyrosine Kinase Inhibitors in Renal Cell Carcinoma: A Concluded Living Systematic Review and Meta-Analysis. JCO Clin Cancer Inform 2021;5:588-99. [Crossref] [PubMed]
- Battelli C, Cho DC. mTOR inhibitors in renal cell carcinoma. Therapy 2011;8:359-67. [Crossref] [PubMed]
- Kwitkowski VE, Prowell TM, Ibrahim A, et al. FDA approval summary: temsirolimus as treatment for advanced renal cell carcinoma. Oncologist 2010;15:428-35. [Crossref] [PubMed]
- Yuan R, Kay A, Berg WJ, et al. Targeting tumorigenesis: development and use of mTOR inhibitors in cancer therapy. J Hematol Oncol 2009;2:45. [Crossref] [PubMed]
- Ryan CW, Tangen CM, Heath EI, et al. Adjuvant everolimus after surgery for renal cell carcinoma (EVEREST): a double-blind, placebo-controlled, randomised, phase 3 trial. Lancet 2023;402:1043-51. [Crossref] [PubMed]
- Xu JX, Maher VE, Zhang L, et al. FDA Approval Summary: Nivolumab in Advanced Renal Cell Carcinoma After Anti-Angiogenic Therapy and Exploratory Predictive Biomarker Analysis. Oncologist 2017;22:311-7. [Crossref] [PubMed]
- Motzer RJ, Escudier B, McDermott DF, et al. Nivolumab versus Everolimus in Advanced Renal-Cell Carcinoma. N Engl J Med 2015;373:1803-13. [Crossref] [PubMed]
- Choueiri TK, Tomczak P, Park SH, et al. Adjuvant Pembrolizumab after Nephrectomy in Renal-Cell Carcinoma. N Engl J Med 2021;385:683-94. [Crossref] [PubMed]
- FDA approves pembrolizumab for adjuvant treatment of renal cell carcinoma. 2021 (cited 2023 10/18). Available online: https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-pembrolizumab-adjuvant-treatment-renal-cell-carcinoma
- Powles T, Tomczak P, Park SH, et al. Pembrolizumab versus placebo as post-nephrectomy adjuvant therapy for clear cell renal cell carcinoma (KEYNOTE-564): 30-month follow-up analysis of a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2022;23:1133-44. [Crossref] [PubMed]
- Choueiri TK, Tomczak P, Park SH, et al. Overall Survival with Adjuvant Pembrolizumab in Renal-Cell Carcinoma. N Engl J Med 2024;390:1359-71. [Crossref] [PubMed]
- Allaf M, Kim SE, Harshman LC, et al. LBA67 Phase III randomized study comparing perioperative nivolumab (nivo) versus observation in patients (Pts) with renal cell carcinoma (RCC) undergoing nephrectomy (PROSPER, ECOG-ACRIN EA8143), a National Clinical Trials Network trial. Ann Oncol 2022;33:S1432-3. [Crossref]
- Pal SK, Uzzo R, Karam JA, et al. Adjuvant atezolizumab versus placebo for patients with renal cell carcinoma at increased risk of recurrence following resection (IMmotion010): a multicentre, randomised, double-blind, phase 3 trial. Lancet 2022;400:1103-16. [Crossref] [PubMed]
- Motzer RJ, Russo P, Grünwald V, et al. Adjuvant nivolumab plus ipilimumab versus placebo for localised renal cell carcinoma after nephrectomy (CheckMate 914): a double-blind, randomised, phase 3 trial. Lancet 2023;401:821-32. [Crossref] [PubMed]
- Riveros C, Huang E, Ranganathan S, et al. Adjuvant immunotherapy in renal cell carcinoma: a systematic review and meta-analysis. BJU Int 2023;131:553-61. [Crossref] [PubMed]
- Motzer RJ, Jonasch E, Agarwal N, et al. Kidney Cancer, Version 3.2022, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw 2022;20:71-90. [Crossref] [PubMed]
- Bex A, Albiges L, Ljungberg B, et al. Updated European Association of Urology Guidelines Regarding Adjuvant Therapy for Renal Cell Carcinoma. Eur Urol 2017;71:719-22. [Crossref] [PubMed]
- Ljungberg B, Albiges L, Abu-Ghanem Y, et al. European Association of Urology Guidelines on Renal Cell Carcinoma: The 2019 Update. Eur Urol 2019;75:799-810. [Crossref] [PubMed]
- Choueiri TK, Tomczak P, Park SH, et al. Pembrolizumab versus placebo as post-nephrectomy adjuvant therapy for patients with renal cell carcinoma: Randomized, double-blind, phase III KEYNOTE-564 study. J Clin Oncol 2021;39:LBA5. [Crossref]