3,306
Views
5
CrossRef citations to date
0
Altmetric
Immunotherapy - Cancer

The emerging landscape of neo/adjuvant immunotherapy in renal cell carcinoma

, & ORCID Icon
Article: 2178217 | Received 02 Nov 2022, Accepted 05 Feb 2023, Published online: 12 Feb 2023

ABSTRACT

Adjuvant and neoadjuvant therapies that reduce the risk of renal cell carcinoma (RCC) recurrence remain an area of unmet need. Advances have been made in metastatic RCC recently by leveraging PD-1/PD-L1 immune checkpoint inhibitors (ICIs). These agents are currently being investigated in the adjuvant and neoadjuvant settings to determine if intervention early in the disease trajectory offers a clinically meaningful benefit. While a disease-free survival benefit has been demonstrated with pembrolizumab, results from other ICI studies have not been positive to date. More mature data from these studies are needed to determine whether there is a survival benefit to ICIs in the curative-intent setting. The success of ICIs has also ushered a new wave of studies combining ICIs with other agents such as targeted therapies and vaccines, which are in early stages of investigation. We review the current state of adjuvant/neoadjuvant therapy in RCC and highlight opportunities for ongoing study.

This article is part of the following collections:
Immunotherapy of Renal Cell Carcinoma

Introduction

Renal cell carcinoma (RCC) is the 14th most common cancer globally, with over 400,000 new diagnoses each year.Citation1 For patients who are fit for surgery, the standard of care for localized RCC is radical nephrectomy (RN) or partial nephrectomy (PN). Other options include active surveillance or ablative techniques for selected patients.Citation2 Despite the definitive surgical treatment, the recurrence rate of RCC at 5-years is significant, varying from 2.2% to 58.1% depending on risk factors such as tumor size, histology, and other clinical features.Citation3 Therefore, adjuvant and/or neoadjuvant treatment to reduce the risk of recurrence and improve survival remains an area of unmet clinical need.

RCCs have long been recognized as tumors that are sensitive to immune-based therapies. In part, this may be owing to significant infiltration of CD8+ T-lymphocytes and the high cytolytic activity.Citation4 Historically, immune-based therapies such as interleukin-2Citation5 and interferon-αCitation6 were the standard of care in metastatic RCC. However, these therapies had low response rates of only 10–20%, were poorly tolerated, and demonstrated limited benefit in the adjuvant setting.Citation7–10 In recent years, immune checkpoint inhibitors (ICIs) targeting programmed death 1 (PD-1), programmed death ligand 1 (PD-L1), and cytotoxic T-lymphocyte associated protein 4 (CTLA-4) have made significant advances in the management of metastatic RCC.Citation11–15 Currently, the standard of care in metastatic RCC involves a combination of two ICIs,Citation12 or one ICI combined with a vascular endothelial growth factor tyrosine kinase inhibitor (VEGF-TKI).Citation11,Citation13,Citation14 The success of ICIs in the metastatic setting has generated immense interest in their perioperative application to reduce disease recurrence.

In this paper we aim to summarize the various prognostic models for recurrence in patients who have undergone nephrectomy, which may guide decision-making in patient selection for adjuvant and/or neoadjuvant therapy. We will review a brief historical background on prior adjuvant and neoadjuvant therapies, and then examine the current literature around ICIs and other immune-based therapies. Finally, challenges in the application of immunotherapy including patient selection, intensity of treatment, and incorporating adjuvant ICIs in our current treatment algorithm will be discussed.

Prognostic models

The ability to accurately predict the individualized risk of cancer recurrence after definitive therapy is a key aspect of patient care. In resected RCC, prognostic models highlighting recurrence risk scores are essential tools to guide surveillance approaches for localized disease and to help patients make informed decisions about ongoing care. As the treatment landscape of RCC evolves toward using systemic therapies in the curative setting, prognostic models will become crucial in selecting patients who would benefit from perioperative treatments.

The TNM system has been a longstanding staging tool, however its ability to predict mortality from RCC after RN is limited.Citation16 Consequently, several validated prognostic models have been developed for localized RCC. The Stage, Size, Grade and Necrosis (SSIGN) Score was reported in 2002. A total of 1801 patients with unilateral clear cell RCC treated with RN between 1970 and 1998 were identified. In addition to the 1997 TNM stage, other pathologic factors (nuclear grade, necrosis and tumor size) were found to be significantly associated with cancer-specific survival.Citation16 The SSIGN score has been validated as a predictive tool for death among patients treated with contemporary RN and PN.Citation17

The UCLA Integrated Staging System (UISS) was developed in 2001, among a cohort of 661 patients undergoing PN or RN for RCC between 1989 and 1999. All histologic subtypes were included. The TNM stage, nuclear grade and Eastern Cooperative Oncology Group (ECOG) performance status were found to be predictive factors for survival and are used to stratify patients into five prognostic groups, ranging from favorable disease (UISS I) to unfavorable disease (UISS V).Citation18

The Memorial Sloan Kettering Cancer Centre (MSKCC) developed a prognostic nomogram in 2001, that estimates the probability of disease recurrence at 5 years. The nomogram was validated among 601 patients who underwent RN or PN for unilateral, localized RCC of any histologic subtype. Patient symptoms, histology, tumor size and pathological stage were found to have a predictive value for disease recurrence.Citation19

The Leibovich score was subsequently developed in 2003, with the main goal of predicting progression to metastatic disease after radical nephrectomy. The purpose was to create a tool for patient stratification for potential participation in adjuvant clinical trials. The model was validated against 1671 patients with clinically localized, unilateral clear cell RCC, who underwent RN between 1970 and 2000. Multivariate analysis showed that tumor stage, regional lymph node status, tumor size, nuclear grade and necrosis were associated with progression to metastatic disease.Citation20 The Leibovich scoring system was updated in 2018 to incorporate updates from the AJCC system and to provide specific scoring systems for different RCC histologic subtypes, predicting both progression-free and cancer-specific survival.Citation21

Several predictive clinical variables have been integrated into the Karakiewicz postoperative nomogram, which was developed in 2007 among a cohort of 2530 patients who underwent RN or PN for renal cancer. A second cohort of 1377 patients was used for external validation. The nomogram combines TNM stage, tumor size, nuclear grade and symptoms at presentation to provide an estimation of RCC-specific survival at 10 years.Citation22 The same group developed a preoperative nomogram that also predicts cancer-specific survival among patients with both localized and metastatic disease, based on a number of clinicopathologic features, including age, gender, symptoms, tumor size, T-stage and presence of metastasis.Citation23

Despite the availability of several prognostic models (), there is a lack of consensus on the optimal stratification approach for localized RCC. Current practice guidelines provide different recommendations on the clinical implementation of prognostic models. The European Association of Urology states that prognostic models are more accurate than TNM stage or grade alone for predicting oncological outcomes (level of evidence 3). The guideline panel provides a strong recommendation for the use of models, both in localized and metastatic disease, but does not provide directions on which specific model should be used.Citation24 A recent prospective validation study evaluating eight different RCC recurrence models showed a considerable decrease in the predictive ability of all models compared to previous retrospective validations.Citation25 Based on these results, the American Urological Association guideline panel created a system that stratifies patients into low, intermediate, high and very high-risk disease based on T and N staging, grading and pathologic features. The presence of a surgical positive margin, which is associated with an increased risk of local recurrence,Citation26 is also incorporated into this system, unlike other prognostic models.Citation27 Similarly, the Canadian Urological Association uses pathological staging to stratify patients into four groups, from low- to very high-risk disease, and provides a surveillance strategy for each group. Although other recognized prognostic factors, such as nuclear grade, ECOG performance status and presence of symptoms are not included in their stratification strategy, the Canadian panel states that some patients may benefit from more intensive surveillance based on the presence of these risk factors.Citation28

Table 1. Comparison of six prognostic models for localized RCC.

The substantial variability among the performance of these prognostic models was recently shown in a head-to-head comparison of five prognostic models (UISS, Leibovich score 2003, Leibovich score 2018, VENUSS score and GRANT score). Additionally, the accuracy of all models differed according to histologic subgroup, highlighting histology as a key prognostic factor in RCC.Citation29

Whether historical prognostic models can be applicable to contemporary patients remains an ongoing question. A recent prospective observational evaluation of the performance of the Leibovich score among a contemporary patient cohort in the UK found an improved rate of metastases-free survival (MFS) in the contemporary cohort among intermediate- and high-risk patients (5-year MFS 85% and 50% respectively) compared to the original cohort (74% and 31%), generating hypotheses regarding evolution of clinical practice and tumor biology.Citation30

As data regarding the benefit of perioperative targeted and immune-based systemic therapies emerges, accurate and applicable prognostic models will become crucial tools in selecting patients who would benefit from neoadjuvant and adjuvant strategies. Several molecular and genomic factors have been identified as having prognostic value in localized RCCCitation31 and might be incorporated to clinicopathologic factors in novel prognostic models.

Adjuvant therapies – a historical perspective

Most recurrences of RCC tend to occur within the first 2 years post-nephrectomy,Citation32 although late recurrences beyond 10 years can occur in about 6% of patients.Citation33 The aim of adjuvant therapy is to target residual microscopic disease after curative surgery and reduce recurrence risk. In contrast to advanced disease, a clinically meaningful benefit is more challenging to demonstrate in an adjuvant trial, as clinical events are fewer (i.e., a proportion of patients may be cured with surgery alone and never develop a recurrence), which would require trials with larger sample sizes and/or longer follow-up. As such, the gold standard outcome of overall survival (OS) can be difficult to demonstrate in adjuvant trials, and instead disease-free survival (DFS) is often used as a primary endpoint. In RCC, DFS has been demonstrated to moderately correlate with OS post-nephrectomy,Citation34,Citation35 and has been used as the primary endpoint in past adjuvant trials. Whether this correlation holds in the domain of immunotherapy is part of ongoing study.

Cytokines such as interferon-α and interleukin-2 were some of the first successful systemic treatments in RCC. However, they have not been demonstrated to have a significant benefit in the adjuvant setting.Citation7,Citation8,Citation10 Combinations of interferon-α, interleukin-2, and 5-fluorouracil have also been studied with no DFS benefit.Citation36,Citation37 Vaccines are another approach to immunotherapy, which have the potential to induce a tumor-specific, durable immune response.Citation38 Historically, irradiated autologous tumor cells and tumor-derived antigens have been studied in the adjuvant setting.Citation39,Citation40 One phase III study using autologous renal tumor cell vaccine did demonstrate a statistically significant progression-free survival and OS benefit in the experimental group;Citation39 however, the two arms of this study were not balanced with respect to stage and prognostic scoring, and the positive results were not replicated in subsequent vaccine studies. To date, there is no effective adjuvant vaccine therapy in RCC.

Regarding VEGF-TKIs, the only agent to have demonstrated benefit in the adjuvant setting was sunitinib, which is approved by the US Food & Drug Administration in this setting.Citation41 The S-TRAC trial enrolled high-risk clear cell RCC patients based on the modified UISS criteria, and randomized patients to sunitinib or placebo post-nephrectomy for one year. The study demonstrated a median DFS of 6.8 years in the sunitinib group, and 5.6 years in the control group (HR = 0.76; 95% CI, 0.59 to 0.98), although no OS benefit was demonstrated in a subsequent updated analysis.Citation42,Citation43 The success of sunitinib has not been replicated in subsequent studies of other VEGF TKIs in the adjuvant setting.Citation44–46 Given the absence of survival benefit and side effect profile of TKIs, their use in the adjuvant setting has not been widely adopted.

Emerging data for adjuvant ICIs

Pembrolizumab (anti-PD-1 antibody) is the first ICI to have demonstrated a clinically meaningful DFS benefit in the adjuvant setting in KEYNOTE-564.Citation47,Citation48 This study randomized 994 patients with histologically confirmed clear cell RCC post-nephrectomy to either 1 year of pembrolizumab every 3 weeks started within 12 weeks of surgery, or placebo. Patients included in this study were of intermediate-to-high risk, high-risk, or had resected synchronous or metachronous metastases within 12 months of the initial nephrectomy with no evidence of disease (M1 NED, ). In the most recent 30-month update, the risk of recurrence or death was 37% lower in the pembrolizumab arm (HR 0.63, 95% CI 0.50–0.80). The estimated 24-month DFS rates were 78.3% vs 67.3% in the pembrolizumab and placebo arms, respectively.Citation49 In subgroup analyses, the DFS benefit was the greatest in the M1 NED patients (HR 0.28, 95% CI 0.12–0.66). OS data remains immature, but favors pembrolizumab with an estimated 24-month OS of 96.2% vs. 93.8%. The hazard ratio for death was 0.52 (23 vs 43 deaths in the pembrolizumab and placebo arms, respectively), although statistical significance has not yet been reached for OS and longer follow-up is required. With regards to adverse events (AEs), grade 3 or higher events were observed in 18.9% in the pembrolizumab arm, compared to 1.2% in placebo. The most common events were fatigue, diarrhea, rash/pruritus, and thyroid dysfunction. Treatment-related AEs led to therapy discontinuation in 18.2% of patients receiving pembrolizumab vs. 0.8% in the placebo arm. Based on the demonstrated DFS benefit, pembrolizumab was approved for use in the adjuvant setting by the US Food and Drug Administration in Nov 2021,Citation48 and the Canadian Agency for Drugs and Technologies in Health in Sept 2022.Citation53

Table 2. Summary of adjuvant and neoadjuvant immune checkpoint inhibitors in renal cell carcinoma.

IMmotion 010 is a phase III study of adjuvant atezolizumab (anti-PD-L1 antibody) that included resected RCC with either clear cell and/or sarcomatoid component at increased risk of recurrence ().Citation50 Notably in this study, the definition of M1 NED allowed resection of metachronous metastases beyond 12 months after the initial nephrectomy. Patients were randomized to either atezolizumab (n = 390) or placebo (n = 388) for 1 year. With a median follow up of 44.7 months, the study found no significant difference in the DFS rate between the atezolizumab and control arms (median DFS 57.2 vs. 49.5 months, HR 0.93, 95% CI 0.75–1.15). DFS at two years was 67.3% in the experimental arm and 65.0% control arm. The subgroup of M1 NED patients in this study did not demonstrate improved outcomes. The rate of AEs leading to treatment discontinuation was 11.5% vs. 2.6%.

The role of adjuvant dual ICIs with nivolumab (anti-PD-1) and ipilimumab (anti-CTLA-4) was evaluated by Part A of the CheckMate 914 trial.Citation51 This phase III trial randomized patients with completely resected RCC with predominantly clear cell histology and at increased risk of recurrence based on TNM stage and histologic grade () to 6 months of combined ipilimumab/nivolumab versus placebo. Importantly, this study excluded patients with any metastatic disease. In the primary analysis with a median follow-up of 37 months, no statistical difference was observed in median DFS (not reached vs. 50.7 months, HR 0.92, 95% CI 0.71–1.19). The estimated 24-month DFS rates were 76.4% in the experimental and 74% in the control arms. Subgroup analysis from this study demonstrated that those with sarcomatoid features had a significant benefit from ipilimumab/nivolumab, although this observation is limited by small subgroup size. The rate of AEs leading to treatment discontinuation was 29% in the experimental arm vs 1% in the control arm. Of note, 23% of patients in the experimental arm required treatment with corticosteroids to manage immune-related AEs. Checkmate-914 also has an ongoing Part B, which will examine the role of nivolumab monotherapy as adjuvant treatment. This will add to the body of evidence around single PD-1 inhibition in the adjuvant setting (NCT03138512).

RAMPART (NCT03288532) is an ongoing multi-arm study that randomizes patients at high risk of recurrence post-nephrectomy to durvalumab (anti-PD-L1) monotherapy, combined durvalumab and tremelimumab (anti-CTLA-4), or active monitoring.Citation54 The Leibovich Score (scores 3–11) was used to select for patients with elevated risk of recurrence, which could include patients with small tumors and aggressive features (e.g. T1 with higher histologic grade or presence of necrosis). The study also allowed for patients with resected ipsilateral adrenal metastases, but other metastatic disease was excluded. Primary completion of the study is expected in 2024, and it will contribute to our understanding of combined PD-L1/CTLA-4 inhibition in the adjuvant setting.

It is postulated that neoadjuvant use of ICIs can prime the immune system prior to surgery,Citation55 and has been demonstrated in phase II studies of several tumor sites to have significant partial or complete pathological response rates, including urothelial carcinoma,Citation56,Citation57 non-small cell lung cancer,Citation58 melanoma,Citation59 and colon cancer.Citation60 In RCC, at present there is no established role for neoadjuvant immunotherapy.Citation27 PROSPER is a phase III trial of nivolumab in high-risk RCC that randomized patients to perioperative nivolumab (one neoadjuvant cycle and nine adjuvant cycles) or surgery alone followed by active surveillance.Citation52 High-risk patients were selected based on clinical T stage, lymph node involvement, and/or M1 NED status (). Radiological staging was used to enroll participants, possibly resulting in a lower risk population compared to adjuvant studies that incorporated histologic information in their risk stratification. An interim analysis for futility revealed no difference in recurrence-free survival and the trial was stopped early for inefficacy (HR 0.97, 95% CI 0.74–1.28). Importantly, approximately 80% of patients had clear cell RCC, while the remaining 20% represented other RCC histologies that may be less sensitive to immune-checkpoint blockade. With only 16 months of median follow-up, significant censoring was observed. As the initial analysis of this study is still ongoing, several questions remain regarding surgery and (neo)adjuvant therapy completion rates, treatment delays, and AEs.

Combination approaches

Since the benefit of pembrolizumab was demonstrated in KEYNOTE-564, several agents are being studied in combination with pembrolizumab in the adjuvant/neoadjuvant settings.

Lenvatinib is a tyrosine kinase inhibitor that targets the VEGF pathway, which has been demonstrated in the CLEAR trial to have an OS and PFS benefit when used in combination with pembrolizumab in metastatic RCC.Citation13 In the non-metastatic setting, the rapid onset of action of TKIs (compared to ICIs) presents a case for neoadjuvant therapy to downstage tumors and to improve the resectability of locally advanced RCC. Currently, a phase II trial (NCT04393350) is underway to examine the combination of lenvatinib and pembrolizumab in the neoadjuvant setting in locally advanced, non-metastatic disease that is at high-risk of recurrence (T3+, any N+, or unresectable disease).Citation61 Another instance where neoadjuvant treatment has a theoretical benefit is in locally advanced RCC with inferior vena cava (IVC) thrombosis. These tumors can be surgically challenging to resect and present a risk of tumor emboli to the pulmonary circulation. A phase II trial (NCT05319015) will examine neoadjuvant lenvatinib and pembrolizumab in patients with IVC thrombosis, with primary outcomes of tumor progression and post-operative complication rates.Citation62

Belzutifan is a novel oral agent that inhibits the hypoxia-inducible factor (HIF) 2α transcription factor. HIF-2α acts upstream to the expression of several genes implicated in oncogenesis, including VEGF, erythropoietin, cyclin D1, and glucose transporter 1.Citation63 Inhibition of this pathway with belzutifan has been demonstrated in a phase II trial to have activity against von Hippel-Lindau associated RCC.Citation64 Litespark-022 (NCT05239728) is a phase III trial that aims to examine the role of adjuvant belzultifan and pembrolizumab in patients with RCC at intermediate-high or high-risk of recurrence according to the KEYNOTE-564 criteria.Citation65 This study also allows patients with M1 NED, defined as synchronous or metachronous metastases resected within 2 years from nephrectomy. The experimental arm will receive belzutifan and pembrolizumab, while the placebo arm will receive pembrolizumab and an oral placebo for up to 54 weeks. The primary end point is DFS, with OS as secondary endpoint. Primary results are expected in 2027.

Vaccines

While ICIs have demonstrated clinical efficacy in the adjuvant setting, they represent only one therapeutic approach in the cancer-immunity cycle through the enhancement of antigen recognition and cytotoxic effect of T-cells.Citation66 Vaccines are another approach to immune therapy that introduces tumor antigens that can activate the immune system. While vaccines derived from autologous tumor cells have been studied in the past, none have demonstrated clinical benefit in a phase III setting.Citation39,Citation40 However, there is now immense interest in combining ICIs with vaccines to achieve a multi-pronged approach to immunotherapy. NeoVax is a personalized vaccine that uses synthetic neoantigens based on tumor DNA sequencing, combined with a toll-like receptor agonist (poly-ICLC) to stimulate the immune system.Citation67 It has been demonstrated in a Phase I study of high-risk melanoma patients to have a durable response post-resection.Citation68 NeoVax is currently being investigated in RCC combined with ipilimumab in a phase I trial (NCT02950766).Citation69

Discussion

Efficacious therapies that significantly decrease the risk of RCC recurrence after nephrectomy and improve survival remains an area of unmet need. The DFS benefit observed in KEYNOTE-564 establishes the use of ICIs in the adjuvant setting and has garnered it regulatory approvals. While future studies appear to build upon the pembrolizumab backbone, at present other similar ICI trials have not shown similar efficacious top line data. Therefore, this peri-operative space in RCC remains an area worthy of ongoing study.

Examination of the differences in trial design, patient selection, and backbone of treatment may provide explanations for differences in outcomes across currently available trials, and possibly help optimize future immunotherapy trial designs. The intensity (i.e., single vs dual) and duration of treatment with ICIs are important variables among the studies. In the adjuvant setting for RCC, the results from Checkmate 914 showed that a 6-month treatment with combined PD-1/CTLA-4 inhibition is associated with 29% treatment discontinuation in the experimental arm due to treatment-related AEs without any DFS benefit. On the other hand, a 12-month treatment with single PD-1/PD-L1 inhibition was associated with lower rates of grade 3 or higher AEs in KEYNOTE-564 and IMmotion 010 (18.9% and 11.5%, respectively). The results from the ongoing RAMPART study will help to understand whether the higher toxicity associated with addition of CTLA-4 inhibitors could have a significant clinical benefit to justify higher intensity of treatment.

The mechanism of action of ICIs may also play a role in the efficacy of adjuvant ICIs in RCC. One possible explanation for the difference in the primary outcome of DFS between KEYNOTE-564 and IMmotion 010 may be the molecular targets of pembrolizumab and atezolizumab (PD-1 vs PD-L1), which could elicit distinct immune responses and interrupt different cancer-to-immune cell interfaces (i.e., PD-1/PD-L2 interaction). Although atezolizumab has shown some activity in metastatic RCC,Citation50 its efficacy appears less remarkable than its ICI counterparts and has not been proven to improve OS in the metastatic setting.

Patient selection is another area that requires further investigation. Despite several prognostic tools becoming available over the years (), concerns remain regarding their applicability in the contemporary immunotherapy era. The adjuvant ICI studies outlined here have been selected for high recurrence using tools that are primarily based on TNM staging, tumor grade and histology. However, as the control arms of these trials demonstrate, 65–75% of patients are disease free at the 24-month timepoint without adjuvant treatment.Citation49–51 The evidence from KEYNOTE-564 suggests M1 NED patients as one potential risk group that may derive the greatest benefit from adjuvant ICI therapy, with an HR of 0.28. It should be noted that in KEYNOTE-564, the M1 NED subgroup only included those who had metastatic disease at the time of screening and underwent a metastatectomy either at the time of, or within 12 months of initial nephrectomy. In contrast, IMmotion 010 employed a different criterion for M1 NED, which included patients who presented with resectable metastases either at the time of screening, or a recurrence beyond 12 months after the initial nephrectomy. The M1 NED group represented 13.9% of the total study population (compared to 5.8% in KEYNOTE-564), with the majority having metachronous metastases. A DFS benefit was not observed in the IMmotion 010 subgroup, which may reflect differences in tumor biology and a more indolent course in those with late metastases. This observation is limited by the small subgroup size in both studies.

Tumor histology could also play a role in patient selection. Sarcomatoid differentiation is known to be a poor prognostic feature in RCC,Citation70 and historically has had poor response to chemotherapy and TKIs.Citation71 However, subset analyses from metastatic RCC trials suggest that sarcomatoid RCC are responsive to ICIs.Citation72 Interpretation of neoadjuvant/adjuvant ICI in sarcomatoid RCC is limited by the small subgroup sizes, however in KEYNOTE-564 and IMmotion 010, there were trends toward improved DFS, and in Checkmate 914, the sarcomatoid subgroup had a substantial benefit from ICIs (HR = 0.29, 95% CI: 0.09–0.91).Citation47,Citation50,Citation51 Given the poor natural history and the positive response to ICIs in multiple trials, sarcomatoid histology would be one-factor clinicians should consider when offering adjuvant ICI. With respect to other non-clear cell histologies, RAMPART and PROSPER trials were designed with broader inclusion criteria for various RCC subtypes. Trials with inclusion of non-clear cell histologies may help inform the applicability of adjuvant strategies largely studied to date in clear cell RCC.Citation52,Citation54 Improvements in patient selection using biomarkers that can identify patients at high-risk of recurrence and predict response to ICIs are needed.

The use of genomic expression profiles (GEPs) in predicting recurrence after nephrectomy is an active area of research. ClearCode34 is a validated prognostic tool for clear cell RCC, which stratifies patients into low-risk (ccA) and high-risk (ccB) categories based on the expression of 34 genes.Citation73,Citation74 It has been demonstrated in retrospective datasets to be superior to the UISS and SSIGN clinical models at assessing risk of death from RCC. Rini et al. developed a “recurrence score” based on a 16-gene GEP to independently predict risk of recurrence after nephrectomy.Citation75 This study found the combination of the recurrence score and Leibovich score improved risk differentiation. Other peripheral blood biomarkers such as circulating tumor DNA, microRNA, and long non-coding RNA are also in early stages of investigation.Citation76,Citation77 Further research into incorporating GEPs and biomarkers to improve patient selection using larger, prospective studies are needed to better tailor adjuvant therapy.

Predicting the benefit from ICIs may also improve patient selection. In many immunogenic tumors, PD-L1 and tumor mutational burden are predictive of response to ICIs. In RCC, however, the evidence is inconsistent. Analyses of metastatic trials demonstrate that RCCs respond to ICIs irrespective of PD-L1 expression levels, and that higher expression of PD-L1 may not correlate with improved outcomes uniformly.Citation78–81 Similarly, tumor mutational burden has not been demonstrated to be a predictive marker in RCC.Citation78,Citation79,Citation81 Retrospective analyses of metastatic ICI studies have identified GEPs that are predictive of response to TKIs and/or ICIs.Citation75–78Citation81–84 These have yet to be incorporated in the adjuvant setting.

At present, many jurisdictions have approved pembrolizumab for use in the adjuvant setting.Citation48,Citation53,Citation85 Although an OS benefit has not yet been established, adjuvant treatment has been shown to reduce the risk of recurrence. In a real-world setting, eligible patients should be made aware of the option for adjuvant pembrolizumab including the benefits, toxicities, and the need for longer term OS data. These discussions may also highlight ongoing trials to further study this space. Ultimately, care plans for adjuvant therapy should remain patient-centered, taking into consideration patients’ values, goals, and comorbidities.

As adjuvant ICIs become part of routine clinical practice in RCC, one question that remains unanswered is whether patients with disease recurrence should be rechallenged with ICIs in the metastatic setting. Evidence in this area is mostly retrospective and has been mostly examined in melanoma and non-small cell lung cancer where ICIs have been standards of care for over a decade.Citation86,Citation87 Ideally, prospective data would inform whether there is utility for ICI rechallenge, heretofore lacking in RCC. In this void, individualized decisions may take into consideration several factors. First, the timing of progression is important: whether it was during or after completion of adjuvant treatment, and if so time since last treatment with ICI. Secondly, the expected activity with combination approaches after adjuvant monotherapy should be considered. In the mRCC setting, salvage addition of CTLA-4 agents after progression on PD-1 therapy has shown significant activity.Citation88 Finally, the patient’s performance status and prior immune-related toxicities need to be taken into consideration. We suggest that standard first-line ICI-based combinations could be offered to those who have a recurrence 6 months beyond completion of adjuvant immunotherapy. In contrast, in patients who recur during adjuvant ICI or within 6 months of completion, next line standard therapy with TKIs should be offered. Similar approaches have been described in the literature for RCC,Citation89 however will require contextualizing within jurisdictional drug approval bodies globally.

In summary, the current literature provides support for the use of pembrolizumab in the adjuvant setting post-nephrectomy for those at high risk of RCC recurrence based on tumor stage, grade, and histology. ICIs hold great promise in the adjuvant treatment of RCC both as monotherapy and in combination with other agents such as TKIs, HIF 2α inhibitors, and personalized vaccines. More mature OS data from completed and ongoing trials may help reassure routine clinical utility in most patients with resected RCC. Further research into risk assessment, GEPs as well as pathomics and radiomics may help better refine patient selection for peri-operative therapy. At present, an individualized approach that takes into consideration an assessment of disease recurrence risk, other comorbidities, and patient preferences would be important in determining candidacy for adjuvant ICIs.

Disclosure statement

PD and LC report no competing interests to declare. AAL: Grants/Research Support: BMS (Inst), BioCanRx (Inst), Novartis (Inst), Roche (Inst), Ipsen (Inst), EMD Serrono (Inst); Honoraria/Consultancy: AbbVie, Astellas, AstraZeneca, Bayer, BMS, Eisai, EMD Serono, Ipsen, Janssen, McKesson, Merck, Novartis, Pfizer, Roche, TerSera.

Additional information

Funding

The author(s) reported there is no funding associated with the work featured in this article.

References

  • Em FJ, Lam F, Colombet M, Mery L, Piñeros M, Znaor A, Soerjomataram I, Bray F. Cancer today (powered by GLOBOCAN 2020). In: Observatory TGC, editor. International Agency for Research on Cancer; 2020. publications.iarc.fr.
  • NCCN. Clinical practice Guidelines in Oncology - Kidney Cancer (Version 2.2023); 2022 [updated 2022 Aug 2; accessed 2022 Aug 3]. https://www.nccn.org/professionals/physician_gls/pdf/kidney.pdf
  • Speed JM, Trinh Q-D, Choueiri TK, Sun M. Recurrence in localized renal cell carcinoma: a systematic review of contemporary data. Curr Urol Rep. 2017 Feb 17;18(2):15. doi:10.1007/s11934-017-0661-3.
  • Şenbabaoğlu Y, Gejman RS, Winer AG, Liu M, Van Allen EM, de Velasco G, Miao D, Ostrovnaya I, Drill E, Luna A, et al. Tumor immune microenvironment characterization in clear cell renal cell carcinoma identifies prognostic and immunotherapeutically relevant messenger RNA signatures. Genome Biol. 2016 Nov 17;17(1):231. doi:10.1186/s13059-016-1092-z.
  • Klapper JA, Downey SG, Smith FO, Yang JC, Hughes MS, Kammula US, Sherry RM, Royal RE, Steinberg SM, Rosenberg S. High-dose interleukin-2 for the treatment of metastatic renal cell carcinoma: a retrospective analysis of response and survival in patients treated in the surgery branch at the National Cancer Institute between 1986 and 2006. Cancer. 2008 Jul 15;113(2):293–10. doi:10.1002/cncr.23552.
  • Canil C, Hotte S, Mayhew LA, Waldron TS, Winquist E. Interferon-alfa in the treatment of patients with inoperable locally advanced or metastatic renal cell carcinoma: a systematic review. Can Urol Assoc J. 2010 Jun;4(3):201–08. doi:10.5489/cuaj.853.
  • Pizzocaro G, Piva L, Colavita M, Ferri S, Artusi R, Boracchi P, Parmiani G, Marubini E. Interferon adjuvant to radical nephrectomy in Robson stages II and III renal cell carcinoma: a multicentric randomized study. J Clin Oncol. 2001 Jan 15;19(2):425–31. doi:10.1200/JCO.2001.19.2.425.
  • Messing EM, Manola J, Wilding G, Propert K, Fleischmann J, Crawford ED, Pontes JE, Hahn R, Trump D. Phase III study of interferon alfa-NL as adjuvant treatment for resectable renal cell carcinoma: an Eastern Cooperative Oncology Group/Intergroup trial. J Clin Oncol. 2003 Apr 1;21(7):1214–22. doi:10.1200/JCO.2003.02.005.
  • Atzpodien J, Kirchner H, Illiger HJ, Metzner B, Ukena D, Schott H, Funke PJ, Gramatzki M, Jürgenson SV, Wandert T, et al. IL-2 in combination with IFN-αand 5-FU versus tamoxifen in metastatic renal cell carcinoma: long-term results of a controlled randomized clinical trial. Br J Cancer. 2001 Oct 19;85(8):1130–36. doi:10.1054/bjoc.2001.2076.
  • Clark JI, Atkins MB, Urba WJ, Creech S, Figlin RA, Dutcher JP, Flaherty L, Sosman JA, Logan TF, White R, 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 Aug 15;21(16):3133–40. doi:10.1200/JCO.2003.02.014.
  • Rini BI, Plimack ER, Stus V, Gafanov R, Hawkins R, Nosov D, Pouliot F, Alekseev B, Soulières D, Melichar B, et al. Pembrolizumab plus Axitinib versus Sunitinib for advanced renal-cell carcinoma. N Engl J Med. 2019 Mar 21;380(12):1116–27. doi:10.1056/NEJMoa1816714.
  • Motzer RJ, Tannir NM, McDermott DF, Arén Frontera O, Melichar B, Choueiri TK, Plimack ER, Barthélémy P, Porta C, George S, et al. Nivolumab plus Ipilimumab versus Sunitinib in advanced renal-cell carcinoma. N Engl J Med. 2018;378(14):1277–90. doi:10.1056/NEJMoa1712126.
  • Motzer R, Alekseev B, Rha SY, Porta C, Eto M, Powles T, Grünwald V, Hutson TE, Kopyltsov E, Méndez-Vidal MJ, et al. Lenvatinib plus Pembrolizumab or Everolimus for advanced renal cell carcinoma. N Engl J Med. 2021 Apr 8;384(14):1289–300. doi:10.1056/NEJMoa2035716.
  • Choueiri TK, Powles T, Burotto M, Escudier B, Bourlon MT, Zurawski B, Oyervides Juárez VM, Hsieh JJ, Basso U, Shah AY, et al. Nivolumab plus Cabozantinib versus Sunitinib for advanced renal-cell carcinoma. N Engl J Med. 2021 Mar 4;384(9):829–41. doi:10.1056/NEJMoa2026982.
  • Motzer RJ, Penkov K, Haanen J, Rini B, Albiges L, Campbell MT, Venugopal B, Kollmannsberger C, Negrier S, Uemura M, et al. Avelumab plus Axitinib versus Sunitinib for advanced renal-cell carcinoma. N Engl J Med. 2019 Mar 21;380(12):1103–15. doi:10.1056/NEJMoa1816047.
  • Frank I, Blute ML, Cheville JC, Lohse CM, Weaver AL, Zincke H. An outcome prediction model for patients with clear cell renal cell carcinoma treated with radical nephrectomy based on tumor stage, size, grade and necrosis: the SSIGN score. J Urol. 2002 Dec;168(6):2395–400. doi:10.1016/S0022-5347(05)64153-5.
  • Parker WP, Cheville JC, Frank I, Zaid HB, Lohse CM, Boorjian SA, Leibovich BC, Thompson RH. Application of the stage, size, grade, and necrosis (SSIGN) score for clear cell renal cell carcinoma in contemporary patients. Eur Urol. 2017 Apr;71(4):665–73. doi:10.1016/j.eururo.2016.05.034.
  • Zisman A, Pantuck AJ, Dorey F, Said JW, Shvarts O, Quintana D, Gitlitz BJ, deKernion JB, Figlin RA, Belldegrun AS. Improved prognostication of renal cell carcinoma using an integrated staging system. J Clin Oncol. 2001;19(6):1649–57. doi:10.1200/JCO.2001.19.6.1649.
  • Kattan MW, Reuter V, Motzer RJ, Katz J, Russo P. A postoperative prognostic nomogram for renal cell carcinoma. J Urol. 2001 Jul;166(1):63–67. doi:10.1016/S0022-5347(05)66077-6.
  • Leibovich BC, Blute ML, Cheville JC, Lohse CM, Frank I, Kwon ED, Weaver AL, Parker AS, Zincke H. Prediction of progression after radical nephrectomy for patients with clear cell renal cell carcinoma: a stratification tool for prospective clinical trials. Cancer. 2003 Apr 1;97(7):1663–71. doi:10.1002/cncr.11234.
  • Leibovich BC, Lohse CM, Cheville JC, Zaid HB, Boorjian SA, Frank I, Thompson RH, Parker WP. Predicting oncologic outcomes in renal cell carcinoma after surgery. Eur Urol. 2018 May;73(5):772–80. doi:10.1016/j.eururo.2018.01.005.
  • Karakiewicz PI, Briganti A, Chun FK, Trinh Q-D, Perrotte P, Ficarra V, Cindolo L, De La Taille A, Tostain J, Mulders PFA, et al. Multi-institutional validation of a new renal cancer–specific survival nomogram. J Clin Oncol. 2007 Apr 10;25(11):1316–22. doi:10.1200/JCO.2006.06.1218.
  • Karakiewicz PI, Suardi N, Capitanio U, Jeldres C, Ficarra V, Cindolo L, de la Taille A, Tostain J, Mulders PFA, Bensalah K, et al. A preoperative prognostic model for patients treated with nephrectomy for renal cell carcinoma. Eur Urol. 2009 Feb;55(2):287–95. doi:10.1016/j.eururo.2008.07.037.
  • EAU Guidelines. Edn. presented at the EAU Annual Congress Amsterdam 2022. Arnhem (The Netherlands): EAU Guidelines Office [accessed 2022 Aug 17]. http://uroweb.org/guidelines/compilations-of-all-guidelines/
  • Correa AF, Jegede O, Haas NB, Flaherty KT, Pins MR, Messing EM, Manola J, Wood CG, Kane CJ, Jewett MAS, et al. Predicting renal cancer recurrence: defining limitations of existing prognostic models with prospective trial-based validation. J Clin Oncol. 2019 Aug 10;37(23):2062–71. doi:10.1200/JCO.19.00107.
  • Wood EL, Adibi M, Qiao W, Brandt J, Zhang M, Tamboli P, Matin SF, Wood CG, Karam JA. Local tumor bed recurrence following partial nephrectomy in patients with small renal masses. J Urol. 2018 Feb;199(2):393–400. doi:10.1016/j.juro.2017.09.072.
  • Campbell SC, Uzzo RG, Karam JA, Chang SS, Clark PE, Souter L. Renal mass and localized renal cancer: evaluation, management, and follow-up: AUA guideline: part II. J Urol. 2021 Aug;206(2):209–18. doi:10.1097/JU.0000000000001912.
  • Kassouf W, Monteiro LL, Drachenberg DE, Fairey AS, Finelli A, Kapoor A, Lattouf J-B, Leveridge MJ, Power NE, Pouliot F, et al. Canadian urological association guideline for follow up of patients after treatment of non-metastatic renal cell carcinoma. Can Urol Assoc J. 2018 Aug;12(8):231–38. doi:10.5489/cuaj.5462.
  • Rosiello G, Larcher A, Fallara G, Giancristofaro C, Martini A, Re C, Cei F, Musso G, Tian Z, Karakiewicz PI, et al. Head-to-head comparison of all the prognostic models recommended by the European Association of Urology guidelines to predict oncologic outcomes in patients with renal cell carcinoma. Urol Oncol. 2022 Jun;40(6):e271 19–27. doi:10.1016/j.urolonc.2021.12.010.
  • Vasudev NS, Hutchinson M, Trainor S, Ferguson R, Bhattarai S, Adeyoju A, Cartledge J, Kimuli M, Datta S, Hanbury D, et al. UK multicenter prospective evaluation of the leibovich score in localized renal cell carcinoma: performance has altered over time. Urology. 2020 Feb;136:162–68. doi:10.1016/j.urology.2019.09.044.
  • Graham J, Dudani S, Heng DYC. Prognostication in Kidney Cancer: recent advances and future directions. J Clin Oncol. 2018 Oct 29;36:3567–73. doi:10.1200/JCO.2018.79.0147.
  • Saidi JA, Newhouse JH, Sawczuk IS. Radiologic follow-up of patients with T1–3a,b,c or T4N+M0 renal cell carcinoma after radical nephrectomy. Urology. 1998 Dec 1;52(6):1000–03. doi:10.1016/S0090-4295(98)00423-3.
  • Miyao N, Naito S, Ozono S, Shinohara N, Masumori N, Igarashi T, Nakao M, Tsushima T, Senga Y, Horie S, et al. Late recurrence of renal cell carcinoma: retrospective and collaborative study of the Japanese Society of Renal Cancer. Urology. 2011 Feb;77(2):379–84. doi:10.1016/j.urology.2010.07.462.
  • Haas NB, Song Y, Rogerio JW, Zhang S, Adejoro O, Carley C, Zhu J, Bhattacharya R, Signorovitch J, Sundaram M. Disease-free survival as a predictor of overall survival in localized renal cell carcinoma (RCC) following first nephrectomy. J Clin Oncol. 2021;39(15_suppl):4581. doi:10.1200/JCO.2021.39.15_suppl.4581.
  • George DJ, Pantuck AJ, Figlin R, Escudier B, Halabi S, Casey M, Lin X, Serfass L, Lechuga Frean MJ, Ravaud A. Correlations between disease-free survival (DFS) and overall survival (OS) in patients (pts) with renal cell carcinoma (RCC) at high risk for recurrence: results from S-TRAC trial. Ann Oncol. 2018;29:viii312. doi:10.1093/annonc/mdy283.090.
  • Atzpodien J, Schmitt E, Gertenbach U, Fornara P, Heynemann H, Maskow A, Ecke M, Wöltjen HH, Jentsch H, Wieland W, 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 Mar 14;92(5):843–46. doi:10.1038/sj.bjc.6602443.
  • Passalacqua R, Caminiti C, Buti S, Porta C, Camisa R, Braglia L, Tomasello G, Vaglio A, Labianca R, Rondini E, et al. Adjuvant low-dose interleukin-2 (IL-2) plus interferon-α (IFN-α) in operable renal cell carcinoma (RCC). J Immunother (1991). 2014 Nov–Dec;37(9):440–47. doi:10.1097/CJI.0000000000000055.
  • Braun DA, Bakouny Z, Hirsch L, Flippot R, Van Allen EM, Wu CJ, Choueiri TK. Beyond conventional immune-checkpoint inhibition—novel immunotherapies for renal cell carcinoma. Nat Rev Clin Oncol. 2021 Apr 1;18(4):199–214. doi:10.1038/s41571-020-00455-z.
  • Jocham D, Richter A, Hoffmann L, Iwig K, Fahlenkamp D, Zakrzewski G, Schmitt E, Dannenberg T, Lehmacher W, von Wietersheim J, 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 Feb 21;363(9409):594–99. doi:10.1016/S0140-6736(04)15590-6.
  • Wood C, Srivastava P, Bukowski R, Lacombe L, Gorelov AI, Gorelov S, Mulders P, Zielinski H, Hoos A, Teofilovici F, 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 Jul 12;372(9633):145–54. doi:10.1016/S0140-6736(08)60697-2.
  • Administration UFD FDA approves sunitinib malate for adjuvant treatment of renal cell carcinoma fda.Gov2017 [accessed 2022 Aug 30]. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-sunitinib-malate-adjuvant-treatment-renal-cell-carcinoma
  • Ravaud A, Motzer RJ, Pandha HS, George DJ, Pantuck AJ, Patel A, Chang Y-H, Escudier B, Donskov F, Magheli A, et al. Adjuvant Sunitinib in high-risk renal-cell carcinoma after nephrectomy. N Engl J Med. 2016 Dec 8;375(23):2246–54. doi:10.1056/NEJMoa1611406.
  • Motzer RJ, Ravaud A, Patard JJ, Pandha HS, George DJ, Patel A, Chang Y-H, Escudier B, Donskov F, Magheli A, et al. Adjuvant Sunitinib for high-risk renal cell carcinoma after nephrectomy: subgroup analyses and updated overall survival results. Eur Urol. 2018 Jan;73(1):62–68. doi:10.1016/j.eururo.2017.09.008.
  • Haas NB, Manola J, Dutcher JP, Flaherty KT, Uzzo RG, Atkins MB, DiPaola RS, Choueiri TK. Adjuvant treatment for high-risk clear cell renal cancer: updated results of a high-risk subset of the ASSURE randomized trial. JAMA Oncol. 2017 Sep 1;3(9):1249–52. doi:10.1001/jamaoncol.2017.0076.
  • Motzer RJ, Russo P, Haas N, Doehn C, Donskov F, Gross-Goupil M, Varlamov S, Kopyltsov E, Lee JL, Lim HY, 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 Mar;79(3):334–38. doi:10.1016/j.eururo.2020.12.029.
  • Gross-Goupil M, Kwon TG, Eto M, Ye D, Miyake H, Seo SI, Byun S-S, Lee JL, Master V, Jin J, et al. Axitinib versus placebo as an adjuvant treatment of renal cell carcinoma: results from the phase III, randomized ATLAS trial. Ann Oncol. 2018 Dec 1;29(12):2371–78. doi:10.1093/annonc/mdy454.
  • Choueiri TK, Tomczak P, Park SH, Venugopal B, Ferguson T, Chang Y-H, Hajek J, Symeonides SN, Lee JL, Sarwar N, et al. Adjuvant Pembrolizumab after nephrectomy in renal-cell carcinoma. N Engl J Med. 2021 Aug 19;385(8):683–94. doi:10.1056/NEJMoa2106391.
  • Administration UFa D FDA approves pembrolizumab for adjuvant treatment of renal cell carcinoma fda.Gov2021 [accessed 2022 Aug 30]. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-pembrolizumab-adjuvant-treatment-renal-cell-carcinoma
  • Choueiri TK, Tomczak P, Park SH, Venugopal B, Ferguson T, Symeonides SN, Hajek J, Chang Y-H, Lee J-L, Sarwar N, et al. Pembrolizumab as post nephrectomy adjuvant therapy for patients with renal cell carcinoma: results from 30-month follow-up of KEYNOTE-564. J Clin Oncol. 2022;40(6_suppl):290. doi:10.1200/JCO.2022.40.6_suppl.290.
  • Pal SK, Uzzo R, Karam JA, Master VA, Donskov F, Suarez C, Albiges L, Rini B, Tomita Y, Kann AG, 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(10358):1103–16. doi:10.1016/S0140-6736(22)01658-0.
  • Motzer RJ, Russo P, Gruenwald V, Tomita Y, Zurawski B, Parikh OA, Buti S, Barthelemy P, Goh JCH, Ye D, et al. Adjuvant nivolumab plus ipilimumab (NIVO+IPI) vs placebo (PBO) for localized renal cell carcinoma (RCC) at high risk of relapse after nephrectomy: results from the randomized, phase III CheckMate 914 trial. Presidential Symposium II European Society of Medical Oncology Paris, France 2022 Sep 11; 33 (suppl_7): S808-S869.
  • Allaf Sek M, Harshman LC, McDermott DF, Master VA, Signoretti S, Cole S, Moon H, Adra N, Singer EA, Gills J, et al. 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. Proffered Paper session 1: GU tumours, non-prostate. Paris, France: European Society of Medical Oncology; 2022 Sept 10.
  • CADTH. Reimbursement Recommendation - Pembrolizumab [Internet]; 2022 [accessed 2022 Oct 17]. https://www.cadth.ca/sites/default/files/DRR/2022/PC0273%20Keytruda%20RCC%20-%20Confidential%20Draft%20CADTH%20Recommendation%20with%20Corrections%20Made_for%20posting%20September%201%2C%202022.pdf.
  • Oza B, Frangou E, Smith B, Bryant H, Kaplan R, Choodari-Oskooei B, Powles T, Stewart GD, Albiges L, Bex A, et al. RAMPART: a phase III multi-arm multi-stage trial of adjuvant checkpoint inhibitors in patients with resected primary renal cell carcinoma (RCC) at high or intermediate risk of relapse. Contemp Clin Trials. 2021 Sep;108:106482. doi:10.1016/j.cct.2021.106482.
  • Keung EZ, Ukponmwan EU, Cogdill AP, Wargo JA. The Rationale and emerging use of neoadjuvant immune checkpoint blockade for solid malignancies. Ann Surg Oncol. 2018 Jul;25(7):1814–27. doi:10.1245/s10434-018-6379-8.
  • van Dijk N, Gil-Jimenez A, Silina K, van Dijk N, Hendricksen K, Smit LA, de Feijter JM, van Montfoort ML, van Rooijen C, Peters D, et al. Preoperative ipilimumab plus nivolumab in locoregionally advanced urothelial cancer: the NABUCCO trial. Nat Med. 2020 Dec;26(12):1839–44. doi:10.1038/s41591-020-1085-z.
  • Necchi A, Anichini A, Raggi D, Briganti A, Massa S, Lucianò R, Colecchia M, Giannatempo P, Mortarini R, Bianchi M, et al. Pembrolizumab as neoadjuvant therapy before radical cystectomy in patients with muscle-invasive urothelial bladder carcinoma (PURE-01): an open-label, single-arm, phase II study. J Clin Oncol. 2018 Dec 1;36(34):3353–60. doi:10.1200/JCO.18.01148.
  • Forde PM, Chaft JE, Smith KN, Anagnostou V, Cottrell TR, Hellmann MD, Zahurak M, Yang SC, Jones DR, Broderick S, et al. Neoadjuvant PD-1 blockade in resectable lung cancer. N Engl J Med. 2018;378(21):1976–86. doi:10.1056/NEJMoa1716078.
  • Rozeman EA, Menzies AM, van Akkooi AC, Adhikari C, Bierman C, van de Wiel BA, Scolyer RA, Krijgsman O, Sikorska K, Eriksson H, et al. Identification of the optimal combination dosing schedule of neoadjuvant ipilimumab plus nivolumab in macroscopic stage III melanoma (OpACIN-neo): a multicentre, phase 2, randomised, controlled trial. Lancet Oncol. 2019;20(7):948–60. doi:10.1016/S1470-2045(19)30151-2.
  • Verschoor YL, Jvd B, Beets G, Sikorska K, Aalbers A, van Lent A, Grootscholten C, Huibregtse I, Marsman H, Oosterling S, et al. Neoadjuvant nivolumab, ipilimumab, and celecoxib in MMR-proficient and MMR-deficient colon cancers: final clinical analysis of the NICHE study. J Clin Oncol. 2022;40(16_suppl):3511. doi:10.1200/JCO.2022.40.16_suppl.3511.
  • Clinicaltrials.gov. Lenvatinib and Pembrolizumab before surgery for the treatment of locally advanced non-metastatic kidney cancer. US National Library of Medicine; 2022 Jan 12.
  • Clinicaltrials.gov. Neoadjuvant Lenvatinib and Pembrolizumab for IVC tumor thrombus. US National Library of Medicine; 2022 Jun 30.
  • Choueiri TK, Kaelin WG. Targeting the HIF2–VEGF axis in renal cell carcinoma. Nat Med. 2020 Oct 1;26(10):1519–30. doi:10.1038/s41591-020-1093-z.
  • Jonasch E, Donskov F, Iliopoulos O, Rathmell WK, Narayan VK, Maughan B, Oudard S, Else T, Maranchie JK, Welsh SJ, et al. Belzutifan for renal cell carcinoma in von Hippel–Lindau disease. N Engl J Med. 2021;385(22):2036–46. doi:10.1056/NEJMoa2103425.
  • Choueiri TK, Bedke J, Karam JA, McKay RR, Motzer RJ, Pal SK, Suárez C, Uzzo R, Liu H, Burgents JE, et al. LITESPARK-022: a phase 3 study of pembrolizumab + belzutifan as adjuvant treatment of clear cell renal cell carcinoma (ccRCC). J Clin Oncol. 2022;40(16_suppl):TPS4602. doi:10.1200/JCO.2022.40.16_suppl.TPS4602.
  • Chen DS, Mellman I. Oncology meets immunology: the cancer-immunity cycle. Immunity. 2013 Jul 25;39(1):1–10. doi:10.1016/j.immuni.2013.07.012.
  • Ott PA, Hu Z, Keskin DB, Shukla SA, Sun J, Bozym DJ, Zhang W, Luoma A, Giobbie-Hurder A, Peter L, et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature. 2017 Jul 13;547(7662):217–21. doi:10.1038/nature22991.
  • Hu Z, Leet DE, Allesøe RL, Oliveira G, Li S, Luoma AM, Liu J, Forman J, Huang T, Iorgulescu JB, et al. Personal neoantigen vaccines induce persistent memory T cell responses and epitope spreading in patients with melanoma. Nat Med. 2021 Mar 1;27(3):515–25. doi:10.1038/s41591-020-01206-4.
  • Clinicaltrials G NeoVax Plus Ipilimumab in renal cell carcinoma. US National Library of Medicine; 2022 Mar 24.
  • Alevizakos M, Gaitanidis A, Nasioudis D, Msaouel P, Appleman LJ. Sarcomatoid renal cell carcinoma: population-based study of 879 patients. Clin Genitourin Cancer. 2019 Jun;17(3):e447–53. doi:10.1016/j.clgc.2019.01.005.
  • Blum KA, Gupta S, Tickoo SK, Chan TA, Russo P, Motzer RJ, Karam JA, Hakimi AA. Sarcomatoid renal cell carcinoma: biology, natural history and management. Nat Rev Urol. 2020 Dec 1;17(12):659–78. doi:10.1038/s41585-020-00382-9.
  • Lombardi P, Filetti M, Falcone R, Di Bidino R, Iacovelli R, Ciccarese C, Bria E, Tortora G, Scambia G, Daniele G. New first-line immunotherapy-based combinations for metastatic renal cell carcinoma: a systematic review and network meta-analysis. Cancer Treat Rev. 2022 May;106:102377. doi:10.1016/j.ctrv.2022.102377.
  • Brooks SA, Brannon AR, Parker JS, Fisher JC, Sen O, Kattan MW, Hakimi AA, Hsieh JJ, Choueiri TK, Tamboli P, et al. ClearCode34: a prognostic risk predictor for localized clear cell renal cell carcinoma. Eur Urol. 2014 Jul;66(1):77–84. doi:10.1016/j.eururo.2014.02.035.
  • Ghatalia P, Rathmell WK. Systematic review: ClearCode 34 - a validated prognostic signature in clear cell renal cell carcinoma (ccRCC). Kidney Cancer. 2018;2(1):23–29. doi:10.3233/KCA-170021.
  • Rini B, Goddard A, Knezevic D, Maddala T, Zhou M, Aydin H, Campbell S, Elson P, Koscielny S, Lopatin M, et al. A 16-gene assay to predict recurrence after surgery in localised renal cell carcinoma: development and validation studies. Lancet Oncol. 2015 Jun;16(6):676–85. doi:10.1016/S1470-2045(15)70167-1.
  • Wan J, Zhu L, Jiang Z, et al. Monitoring of plasma cell-free DNA in predicting postoperative recurrence of clear cell renal cell carcinoma. Urol Int. 2013;91(3):273–78. doi:10.1159/000351409.
  • Kubiliute R, Jarmalaite S. Epigenetic biomarkers of renal cell carcinoma for liquid biopsy tests. Int J Mol Sci. 2021 Aug 17;22(16):8846. doi:10.3390/ijms22168846.
  • Motzer RJ, Robbins PB, Powles T, Albiges L, Haanen JB, Larkin J, Mu XJ, Ching KA, Uemura M, Pal SK, et al. Avelumab plus axitinib versus sunitinib in advanced renal cell carcinoma: biomarker analysis of the phase 3 JAVELIN renal 101 trial. Nat Med. 2020 Nov;26(11):1733–41. doi:10.1038/s41591-020-1044-8.
  • Motzer RJ, Choueiri TK, McDermott DF, Powles T, Vano Y-A, Gupta S, Yao J, Han C, Ammar R, Papillon-Cavanagh S, et al. Biomarker analysis from CheckMate 214: nivolumab plus ipilimumab versus sunitinib in renal cell carcinoma. J Immunother Cancer. 2022 Mar;10(3):e004316. doi:10.1136/jitc-2021-004316.
  • Powles T, Plimack ER, Soulieres D, Waddell T, Stus V, Gafanov R, Nosov D, Pouliot F, Melichar B, Vynnychenko I, et al. Pembrolizumab plus axitinib versus sunitinib monotherapy as first-line treatment of advanced renal cell carcinoma (KEYNOTE-426): extended follow-up from a randomised, open-label, phase 3 trial. Lancet Oncol. 2020 Dec;21(12):1563–73. doi:10.1016/S1470-2045(20)30436-8.
  • McDermott DF, Huseni MA, Atkins MB, Motzer RJ, Rini BI, Escudier B, Fong L, Joseph RW, Pal SK, Reeves JA, et al. Clinical activity and molecular correlates of response to atezolizumab alone or in combination with bevacizumab versus sunitinib in renal cell carcinoma. Nat Med. 2018 Jun;24(6):749–57. doi:10.1038/s41591-018-0053-3.
  • Motzer RJ, Martini JF, Mu XJ, Staehler M, George DJ, Valota O, Lin X, Pandha HS, Ching KA, Ravaud A. Molecular characterization of renal cell carcinoma tumors from a phase III anti-angiogenic adjuvant therapy trial. Nat Commun. 2022 Oct 10;13(1):5959. doi:10.1038/s41467-022-33555-8.
  • Roussel E, Verbiest A, Kinget L, Boeckx B, Zucman-Rossi J, Couchy G, Caruso S, Job S, de Reyniès A, De Wever L, et al. Molecular subtypes and gene expression signatures as prognostic features in fully resected clear cell renal cell carcinoma: a tailored approach to adjuvant trials. Clin Genitourin Cancer. 2021 Dec;19(6):e382–94. doi:10.1016/j.clgc.2021.07.005.
  • McDermott DF, Lee J-L, Donskov F, Tykodi SS, Bjarnason GA, Larkin JMG, Gafanov R, Kochenderfer MD, Malik J, Poprach A, et al. Association of gene expression with clinical outcomes in patients with renal cell carcinoma treated with pembrolizumab in KEYNOTE-427. J Clin Oncol. 2020;38(15_suppl):5024. doi:10.1200/JCO.2020.38.15_suppl.5024.
  • Agency EM. Keytruda - pembrolizumab; 2022.
  • Gobbini E, Charles J, Toffart AC, Leccia MT, Moro-Sibilot D, Giaj Levra M. Current opinions in immune checkpoint inhibitors rechallenge in solid cancers. Crit Rev Oncol Hematol. 2019 Dec;144:102816. doi:10.1016/j.critrevonc.2019.102816.
  • Keilholz U, Ascierto PA, Dummer R, Robert C, Lorigan P, van Akkooi A, Arance A, Blank CU, Chiarion Sileni V, Donia M, et al. ESMO consensus conference recommendations on the management of metastatic melanoma: under the auspices of the ESMO Guidelines Committee. Ann Oncol. 2020 Nov;31(11):1435–48. doi:10.1016/j.annonc.2020.07.004.
  • Gul A, Stewart TF, Mantia CM, Shah NJ, Gatof ES, Long Y, Allman KD, Ornstein MC, Hammers HJ, McDermott DF, et al. Salvage Ipilimumab and Nivolumab in patients with metastatic renal cell carcinoma after prior immune checkpoint inhibitors. J Clin Oncol. 2020 Sep 20;38(27):3088–94. doi:10.1200/JCO.19.03315.
  • Bimbatti D, Maruzzo M, Pierantoni F, Diminutto A, Dionese M, Deppieri FM, Lai E, Zagonel V, Basso U. Immune checkpoint inhibitors rechallenge in urological tumors: an extensive review of the literature. Crit Rev Oncol Hematol. 2022 Feb;170:103579. doi:10.1016/j.critrevonc.2022.103579.