Abstract

Much attention has been paid to immune checkpoint inhibitors to various cancer treatments. In urothelial cancer, pembrolizumab was initially approved for patients who either recurred or progressed following platinum-based chemotherapy. For the platinum-fit population, although the standard first-line treatment is still platinum-based systemic chemotherapy, avelumab has been recently approved as a maintenance therapy for patients who have not had disease progression with four to six cycles of first-line chemotherapy. In addition, adjuvant nivolumab has just prolonged disease-free survival (DFS) by ~10 months, compared with placebo in patients with muscle-invasive bladder urothelial cancer or upper tract urothelial cancer at high-risk of recurrence after radical surgical resection.

On the other hand, in kidney cancer, nivolumab was initially approved for advanced renal cell carcinoma patients after one or two prior anti-angiogenic therapies. Next, combinations of two immune checkpoint inhibitors (nivolumab + ipilimumab) and immune checkpoint inhibitor + tyrosine kinase inhibitors (pembrolizumab + axitinib and avelumab + axitinib) were approved for the first-line treatment for patients with advanced renal cell carcinoma. Recently, new generation tyrosine kinase inhibitors, such as cabozantinib and lenvatinib have been combined with immune checkpoint inhibitors. Both nivolumab + cabozantinib and pembrolizumab + lenvatinib have demonstrated superior progression-free survival and objective response rate, compared with sunitinib. So far, no prospective trials have demonstrated the duration of immune checkpoint inhibitor treatments. We are now doing the Japan Clinical Oncology Group 1905 trial, where patients with advanced renal cell carcinoma who have received an immune checkpoint inhibitor for 24 weeks are divided into two groups: those who continue immune checkpoint inhibitor treatment and those who discontinue immune checkpoint inhibitor treatment.

Introduction

Urothelial and kidney cancer have been historically considered to be sensitive to immunotherapy because of their standard treatments of intravesical instillation of bacillus Calmette-Guerin (BCG) (1) and administration of interferon-α (2). In addition, after the introduction of immune checkpoint inhibitors to urothelial (3) and kidney cancer (4), recent clinical trials have been drastically changing their standard treatments. Here, we summarize current status and future perspectives of immunotherapy (including various combination therapies) against urothelial and kidney cancer.

Urothelial cancer

Characteristics of urothelial cancer as a target of immune checkpoint inhibitor therapy

Urothelial cancer is known to be one of the malignancies with a high frequency of gene mutation (5). Molecular mechanisms for urothelial carcinogenesis are complicated (6,7), and it is often not caused by a single driver gene mutation (8). Recent accumulation of genomic analyses has revealed that UC can be categorized into several molecular subtypes, suggesting high intertumoral heterogeneity (9). Therefore, there has never been a highly effective molecular-targeting agent for UC with rare exceptions (10).

UC is also characterized by its immunogenicity as evident by the fact that intravesical instillation of BCG has been used for non-muscle-invasive bladder cancer (NMIBC) for decades (1) in addition to its high mutation burden (5,8). Thus, UC was one of the malignancies that had been expected to respond to immunotherapy. Indeed, the efficacy of anti-PD-1/PD-L1 treatment was reported ahead of many other cancers (11). Since then, the effectiveness of immune checkpoint inhibitors (ICIs) has been tested in UC patients in various treatment settings. Some of them have already been shown to improve treatment outcomes. Furthermore, it is expected that the effectiveness of ICI will be proved in other settings, leading to expanded indication in the future. This part of the article reviews current status and discusses future perspective of ICIs in the treatment of UC.

Current standard treatment for stage IV disease

Methotrexate, vinblastine, adriamycin plus cisplatin (MVAC) was shown to prolong survival compared with older chemotherapy combinations in 1990’s (12), followed by gemcitabine plus cisplatin (GC) shown to be less toxic than MVAC without compromising survival outcomes in 2000 (13). Those two regimens are approved in Japan as the current standard for patients with stage IV UC (14). Gemcitabine plus carboplatin (G-CBDCA) is often used for cisplatin-ineligible patients as an approved regimen, although it was reported to yield lower complete response rate and shorter overall survival (OS) (15). Better efficacy by modifications of dose density has been reported (16), although those modified regimens have not been approved in Japan. Additionally, those regimens are also frequently used as perioperative chemotherapy either at the neoadjuvant or adjuvant setting (14). Current standard chemotherapy is active in the majority of the patients; objective response (OR; partial response [PR] or complete remission [CR]) in 40 to 50% and disease control (stable disease, PR or CR) in 75 to 80%. However, most patients have disease progression within ~9 months, and the median OS was 14–15 months.

Immune checkpoint inhibitors for unresectable, chemoresistant UC

There had been no effective treatment for patients with UC that progressed against first-line systemic chemotherapy. This long-standing problem was overcome by KEYNOTE-045 study (ClinicalTrials.gov number, NCT02256436) in 2017 (3). In this phase 3 trial, 542 patients who either recurred or progressed following platinum-based chemotherapy were randomized to receive either pembrolizumab or chemotherapy (paclitaxel, docetaxel or vinflunine). OS of those who received pembrolizumab was significantly superior to that of those who received chemotherapy (10.3 vs 7.4 months; 95% confidence interval (CI) 8.0–11.8 vs 6.1–8.3; hazard ratio [HR] 0.73; 95% CI 0.59–0.91; P = 0.002). The OR rate was 21.1% for pembrolizumab group compared with 11.4% for chemotherapy group. Similar trends for the better oncological outcomes were observed in the Japanese sub-population (17) and after >2-year follow-up (18).

In terms of tolerability, treatment-related adverse event (AE) was reported in 60.9% of patients in the pembrolizumab group compared with 90.2% of those in the chemotherapy group. Another report of exploratory results demonstrated that the quality of life in patients administered pembrolizumab was significantly better than that in those administered chemotherapy (19) despite several criticisms were raised (20).

These results prompted US Food and Drug Administration (FDA) and Japan Pharmaceuticals and Medical Devices Agency (PMDA) to give pembrolizumab a full approval for treatment of post-platinum UC patients in both countries by the end of 2017. Since then, as the only life-prolonging treatment in UC refractory to the standard first-line chemotherapy, the use of second-line pembrolizumab has increased rapidly without clear guidance for validated prognostication. Several investigators reported relatively small-scale outcome studies from their initial experiences with prognostic markers related to performance status (PS), number or site of metastasis, neutrophil-to-lymphocyte ratio (NLR), C-reactive protein (CRP) and time from previous chemotherapy (Table 1) (21–26).

Table 1

Retrospective patient series of second-line pembrolizumab for urothelial cancer

AuthorNumber of patientsPFS (months)OS (months)Risk factors
Tamura et al. (21)412.511.9OS: ECOG PS (>1), Number of metastatic site (>1), NLR kinetics (+6.12% at 6 M)
Yasuoka et al. (22)404.110.0OS: ECOG PS (>1), liver metastasis (yes), CRP (>0.5 mg/dL)
Ogihara et al. (24)78N.S.N.S.CSM: NLR (≥3.35)
Disease progression: ECOG PS (>0), liver metastasis (yes), NLR (≥3.35)
Furubayashi et al. (23)343.311.7OS: liver metastasis (yes), time from previous chemotherapy (≥3 M)
Yamamoto et al. (25)121N.S.N.S.OS: ECOG PS (>1), LN metastasis only (no), CRP (>0.56 mg/dL), NLR (>3.0)
Kijima et al. (26)977.513.1OS: ECOG PS (>1), liver meta (yes), CRP (>0.5 mg/dL, kinetics)
Kobayashi et al. (27)Discovery: 463
Validation: 292
N.S.10.2
12.5
OS: ECOG PS (>1), Metastasis site (Liver/Other organs/LN only), Hb (<11), NLR (>3.0)
AuthorNumber of patientsPFS (months)OS (months)Risk factors
Tamura et al. (21)412.511.9OS: ECOG PS (>1), Number of metastatic site (>1), NLR kinetics (+6.12% at 6 M)
Yasuoka et al. (22)404.110.0OS: ECOG PS (>1), liver metastasis (yes), CRP (>0.5 mg/dL)
Ogihara et al. (24)78N.S.N.S.CSM: NLR (≥3.35)
Disease progression: ECOG PS (>0), liver metastasis (yes), NLR (≥3.35)
Furubayashi et al. (23)343.311.7OS: liver metastasis (yes), time from previous chemotherapy (≥3 M)
Yamamoto et al. (25)121N.S.N.S.OS: ECOG PS (>1), LN metastasis only (no), CRP (>0.56 mg/dL), NLR (>3.0)
Kijima et al. (26)977.513.1OS: ECOG PS (>1), liver meta (yes), CRP (>0.5 mg/dL, kinetics)
Kobayashi et al. (27)Discovery: 463
Validation: 292
N.S.10.2
12.5
OS: ECOG PS (>1), Metastasis site (Liver/Other organs/LN only), Hb (<11), NLR (>3.0)

PFS; progression-free survival, OS; overall survival, NLR; neutrophil-to-lymphocyte ratio, CRP; C-reactive protein, CSM; cancer-specific mortality, Hb; hemoglobin.

Table 1

Retrospective patient series of second-line pembrolizumab for urothelial cancer

AuthorNumber of patientsPFS (months)OS (months)Risk factors
Tamura et al. (21)412.511.9OS: ECOG PS (>1), Number of metastatic site (>1), NLR kinetics (+6.12% at 6 M)
Yasuoka et al. (22)404.110.0OS: ECOG PS (>1), liver metastasis (yes), CRP (>0.5 mg/dL)
Ogihara et al. (24)78N.S.N.S.CSM: NLR (≥3.35)
Disease progression: ECOG PS (>0), liver metastasis (yes), NLR (≥3.35)
Furubayashi et al. (23)343.311.7OS: liver metastasis (yes), time from previous chemotherapy (≥3 M)
Yamamoto et al. (25)121N.S.N.S.OS: ECOG PS (>1), LN metastasis only (no), CRP (>0.56 mg/dL), NLR (>3.0)
Kijima et al. (26)977.513.1OS: ECOG PS (>1), liver meta (yes), CRP (>0.5 mg/dL, kinetics)
Kobayashi et al. (27)Discovery: 463
Validation: 292
N.S.10.2
12.5
OS: ECOG PS (>1), Metastasis site (Liver/Other organs/LN only), Hb (<11), NLR (>3.0)
AuthorNumber of patientsPFS (months)OS (months)Risk factors
Tamura et al. (21)412.511.9OS: ECOG PS (>1), Number of metastatic site (>1), NLR kinetics (+6.12% at 6 M)
Yasuoka et al. (22)404.110.0OS: ECOG PS (>1), liver metastasis (yes), CRP (>0.5 mg/dL)
Ogihara et al. (24)78N.S.N.S.CSM: NLR (≥3.35)
Disease progression: ECOG PS (>0), liver metastasis (yes), NLR (≥3.35)
Furubayashi et al. (23)343.311.7OS: liver metastasis (yes), time from previous chemotherapy (≥3 M)
Yamamoto et al. (25)121N.S.N.S.OS: ECOG PS (>1), LN metastasis only (no), CRP (>0.56 mg/dL), NLR (>3.0)
Kijima et al. (26)977.513.1OS: ECOG PS (>1), liver meta (yes), CRP (>0.5 mg/dL, kinetics)
Kobayashi et al. (27)Discovery: 463
Validation: 292
N.S.10.2
12.5
OS: ECOG PS (>1), Metastasis site (Liver/Other organs/LN only), Hb (<11), NLR (>3.0)

PFS; progression-free survival, OS; overall survival, NLR; neutrophil-to-lymphocyte ratio, CRP; C-reactive protein, CSM; cancer-specific mortality, Hb; hemoglobin.

More recently, a multicenter study by Japan Urological Oncology Group (JUOG) analyzed a total of 755 UC patients who received pembrolizumab for chemoresistant disease (27). The study proposed risk stratification based on four factors namely Eastern Cooperative Oncology Group (ECOG) PS (>1), metastasis sites (liver/other organs/lymph node only), hemoglobin (<11 g/dL) and NLR (>3.0), which was established by multivariate Cox proportional hazard analysis on 463 patients and externally validated using data on distinct 292 patients. At median follow up of 17.7 months, median (95% CI) OS after the initiation of pembrolizumab treatment was 2.3 (1.2–2.6) months for high-risk, 6.8 (5.8–8.9) months for intermediate and not reached (NR) (NR–19.1) for low-risk patients. C-index of the model for the validation cohort was 0.747. Importantly, the risk stratification was significantly associated with OR rate (ORR) as well. Because there have been no prognostic or predictive biomarkers for second-line pembrolizumab treatment, the model using conventional clinical parameters is clinically useful and also informative for the design of future clinical trials. These prognostic markers will be growingly important as novel agents including enfortumab vedotin and erdafitinib are introduced as an alternative or subsequent treatment.

Similarly, Sternberg et al. (28) reported results from SAUL (NCT02928406), a single-arm multicenter international open-label phase 3B safety study of atezolizumab in a real-world population, which specified a ‘difficult-to-treat’ group defined as having ECOG PS 2 or more or central nervous system metastasis. In another report, Sonpavde et al. (29) analyzed data from phase 1/2 trials for chemoresistant UC patients receiving PD-L1 inhibitors (avelumab or durvalumab). They reported that a five-factor prognostic model incorporating ECOG-PS, liver metastasis, platelet count, NLR ratio and lactate dehydrogenase yielded robust discrimination of survival of between low, intermediate and high-risk groups.

In terms of PS, Khaki et al. (30) reported real-world data of UC patients receiving ICI and suggested that those with an ECOG PS 3 were unlikely to benefit from ICIs although a meaningful number of patients with an ECOG PS 2 appeared to benefit, at least on the basis of the ORR. In this regard, another JUOG study showed similar OS outcome between those ECOG PS 2 and ≥ 3, which was better discriminated by NLR and liver metastasis (31), suggesting that even those with ECOG PS 3 may be benefitted from pembrolizumab if they do not have high NLR nor liver metastasis.

NLR is also robustly associated with the survival of UC patients treated with ICIs described above. However, it is not a specific prognosticator for this patient population but for UC patients in other treatment or disease stages, those with other malignant or non-malignant diseases, and even in a general population (27). It is noteworthy that several investigators reported better prognosis of patients achieving the reduction of NLR after pembrolizumab treatment (21,24,25). Additionally, a JUOG study showed better prognosis after pembrolizumab treatment of patients achieving the reduction of NLR after first-line chemotherapy (32). Biological and clinical significance of this unique biomarker should be further studied in the future.

First-line treatment for unresectable locally advanced or metastatic UC patients

Use of ICI as the first-line treatment was initially approved for CDDP-unfit population (33). In a phase 2 IMvigor 210 trial (NCT02108652), atezolizumab showed 23.5% of ORR including radiological CR in 6.7% of the patients (34). Another phase 2 KEYNOTE-052 trial (NCT02335424) reported 28.6% of ORR including radiological CR in 8.9% (35). Based on these findings both atezolizumab and pembrolizumab have been approved for this patient population in the USA but not in Japan as neither of the two studies enrolled Japanese patients.

Several other phase 3 trials explored ICIs as first-line treatment using different approaches (DANUBE [NCT02516241] (36), KEYNOTE-361 [NCT02853305] (37) and IMvigor 130 [NCT02807636] (38) (Table 2). Although atezolizumab in combination with chemotherapy showed superior progression-free survival (PFS) compared with chemotherapy alone without increasing AEs rate, difference in OS did NR the predefined threshold of statistical significance at least at the time of interim analysis (38), which means we have to wait until the final OS analysis to have definitive conclusion for OS benefit by additional atezolizumab to standard chemotherapy at the first-line treatment setting. The other two failed to show OS advantage over standard chemotherapy in intention-to-treat (ITT) population (36,37). There are several more trials that are still ongoing (CheckMate-901 [NCT03036098], NILE [NCT03682068], LEAP-011 [NCT03898180], and EV-302 [NCT04223856]). (Table 2 and ref. (39)) Until any of these trials shows survival advantage over the standard of care, first-line chemotherapy and subsequent, whatever salvage or maintenance, ICI will stay at the center of treatment strategy in this setting. Then the timing of switching from chemotherapy to ICI, particularly in those who have their disease controlled with chemotherapy, will become a very important clinical question. Usually accumulation of chemotherapy usually substantially worsens patient quality of life, and probably systemic immune status as well, which may affect the effectiveness of subsequent ICI (40). On the other hand, it takes courage to change an effective treatment to another as someone said ‘never mess with the winning team’.

Table 2

Key phase-3 trials on first-line treatment regimen using immune checkpoint inhibitor for unresectable or metastatic urothelial cancer

NameTrial IDTotal sample sizeTreatment regimenControl regimenPrimary endpointsKey secondary oncological endpointsKey results
DANUBEa (36)NCT 025162411032Durva.
Durva. + Tremeli.
G-C or G-CBDCAOScPFS, ORR, DOR
  • OS in high PD-L1 population: 14.4 M for Durva. vs 12.1 M for CT (HR 0.89, 95%CI 0.71–1.11, P = 0.30, not reached predefined threshold)

  • OS in the ITT population: 15.1 M for Durva. + Tremeli. vs 12.1 M for CT (HR 0.85, 95%CI 0.72–1.02, P = 0.0075, not reached predefined threshold)

  • OS in high PD-L1 population: 17.9 M for Durva. + Tremeli. vs 12.1 M for CT (HR 0.74, 95%CI 0.59–0.93)

IMvigor130a (38)NCT 028076361213Atezo.
Atezo. + G-C or G-CBDCA
G-C or G-CBDCAPFSd, OSe, safetyORR, DOR
  • PFS in the ITT population: 8.2 M for Atezo. + CT. vs 6.3 M for CT (HR 0.82, 95%CI 0.70–0.96, 1-sided P = 0.007)

  • OS in the ITT population: 16.0 M for Atezo. + CT. vs 13.4 M for CT (HR 0.83, 95%CI 0.69–1.00, 1-sided P = 0.027, not reached predefined threshold)

  • OS in high PD-L1 population: Not reached for Atezo. vs 17.8 M for CT (HR 0.68, 95%CI 0.43–1.08)

KEYNOTE-361a (37)NCT 028533051010Pembro.
Pembro. + G-C or G-CBDCA
G-C or G-CBDCAPFS, OSORR, DOR, DCR
  • PFS: 8.3 M for Pembro. + CT vs 7.1 M for CT (HR 0.78, 95%CI 0.65–0.93, P = 0.0033, not reached predefined threshold)

  • OS: 17.0 M for Pembro. + CT vs 14.3 M for CT (HR 0.86, 95%CI 0.72–1.02, P = 0.0407, not reached predefined threshold)

CheckMate-901aNCT 030360981290bNivo. + Ipi.
Nivo. + G-C
G-C or G-CBDCAPFS, OSNot yet reported
NILEaNCT 036820681434bDurva. + G-C or G-CBDCA
Durva. + Tremeli. + G-C or G-CBDCA
G-C or G-CBDCAOSPFS, ORR, DOR, DCR, PFS2Not yet reported
EV-302aNCT 04223856760bPembro. + EVG-C or G-CBDCAPFS, OSORR, DORNot yet reported
NameTrial IDTotal sample sizeTreatment regimenControl regimenPrimary endpointsKey secondary oncological endpointsKey results
DANUBEa (36)NCT 025162411032Durva.
Durva. + Tremeli.
G-C or G-CBDCAOScPFS, ORR, DOR
  • OS in high PD-L1 population: 14.4 M for Durva. vs 12.1 M for CT (HR 0.89, 95%CI 0.71–1.11, P = 0.30, not reached predefined threshold)

  • OS in the ITT population: 15.1 M for Durva. + Tremeli. vs 12.1 M for CT (HR 0.85, 95%CI 0.72–1.02, P = 0.0075, not reached predefined threshold)

  • OS in high PD-L1 population: 17.9 M for Durva. + Tremeli. vs 12.1 M for CT (HR 0.74, 95%CI 0.59–0.93)

IMvigor130a (38)NCT 028076361213Atezo.
Atezo. + G-C or G-CBDCA
G-C or G-CBDCAPFSd, OSe, safetyORR, DOR
  • PFS in the ITT population: 8.2 M for Atezo. + CT. vs 6.3 M for CT (HR 0.82, 95%CI 0.70–0.96, 1-sided P = 0.007)

  • OS in the ITT population: 16.0 M for Atezo. + CT. vs 13.4 M for CT (HR 0.83, 95%CI 0.69–1.00, 1-sided P = 0.027, not reached predefined threshold)

  • OS in high PD-L1 population: Not reached for Atezo. vs 17.8 M for CT (HR 0.68, 95%CI 0.43–1.08)

KEYNOTE-361a (37)NCT 028533051010Pembro.
Pembro. + G-C or G-CBDCA
G-C or G-CBDCAPFS, OSORR, DOR, DCR
  • PFS: 8.3 M for Pembro. + CT vs 7.1 M for CT (HR 0.78, 95%CI 0.65–0.93, P = 0.0033, not reached predefined threshold)

  • OS: 17.0 M for Pembro. + CT vs 14.3 M for CT (HR 0.86, 95%CI 0.72–1.02, P = 0.0407, not reached predefined threshold)

CheckMate-901aNCT 030360981290bNivo. + Ipi.
Nivo. + G-C
G-C or G-CBDCAPFS, OSNot yet reported
NILEaNCT 036820681434bDurva. + G-C or G-CBDCA
Durva. + Tremeli. + G-C or G-CBDCA
G-C or G-CBDCAOSPFS, ORR, DOR, DCR, PFS2Not yet reported
EV-302aNCT 04223856760bPembro. + EVG-C or G-CBDCAPFS, OSORR, DORNot yet reported

aJapanese patients enrolled.

bEstimated enrollment.

cTwo OS comparisons; (1) between the Durva. vs CT groups in the population of patients with high PD-L1 expression and (2) between the Durva. + Treme. vs CT groups in the ITT population.

dPFS between the Atezo. + CT vs CT groups in the ITT population.

eOS between the Atezo. vs CT groups in the ITT population, which was to be formally tested only if OS was positive for the Atezo. + CT vs CT groups. Only the interim result is reported as of the end of 2020.

G-C, Gemcitabine + cisplatin; G-CBDCA, Gemcitabine + carboplatin; Durva., Durvalumab; Tremeli., Tremelimumab; Atezo., Atezolizumab; Pembro., Pembrolizumab; Nivo., Nivolumab; Ipi., Ipilimumab; EV, Enfortumab vedotin; PFS, Progression-free survival; OS, Overall survival; ORR, Objective response rate; DOR, Duration of response; DCR, Disease control rate; PFS2, Second profression-free survival; CT, Chemotherapy; ITT, Intention to treat.

Table 2

Key phase-3 trials on first-line treatment regimen using immune checkpoint inhibitor for unresectable or metastatic urothelial cancer

NameTrial IDTotal sample sizeTreatment regimenControl regimenPrimary endpointsKey secondary oncological endpointsKey results
DANUBEa (36)NCT 025162411032Durva.
Durva. + Tremeli.
G-C or G-CBDCAOScPFS, ORR, DOR
  • OS in high PD-L1 population: 14.4 M for Durva. vs 12.1 M for CT (HR 0.89, 95%CI 0.71–1.11, P = 0.30, not reached predefined threshold)

  • OS in the ITT population: 15.1 M for Durva. + Tremeli. vs 12.1 M for CT (HR 0.85, 95%CI 0.72–1.02, P = 0.0075, not reached predefined threshold)

  • OS in high PD-L1 population: 17.9 M for Durva. + Tremeli. vs 12.1 M for CT (HR 0.74, 95%CI 0.59–0.93)

IMvigor130a (38)NCT 028076361213Atezo.
Atezo. + G-C or G-CBDCA
G-C or G-CBDCAPFSd, OSe, safetyORR, DOR
  • PFS in the ITT population: 8.2 M for Atezo. + CT. vs 6.3 M for CT (HR 0.82, 95%CI 0.70–0.96, 1-sided P = 0.007)

  • OS in the ITT population: 16.0 M for Atezo. + CT. vs 13.4 M for CT (HR 0.83, 95%CI 0.69–1.00, 1-sided P = 0.027, not reached predefined threshold)

  • OS in high PD-L1 population: Not reached for Atezo. vs 17.8 M for CT (HR 0.68, 95%CI 0.43–1.08)

KEYNOTE-361a (37)NCT 028533051010Pembro.
Pembro. + G-C or G-CBDCA
G-C or G-CBDCAPFS, OSORR, DOR, DCR
  • PFS: 8.3 M for Pembro. + CT vs 7.1 M for CT (HR 0.78, 95%CI 0.65–0.93, P = 0.0033, not reached predefined threshold)

  • OS: 17.0 M for Pembro. + CT vs 14.3 M for CT (HR 0.86, 95%CI 0.72–1.02, P = 0.0407, not reached predefined threshold)

CheckMate-901aNCT 030360981290bNivo. + Ipi.
Nivo. + G-C
G-C or G-CBDCAPFS, OSNot yet reported
NILEaNCT 036820681434bDurva. + G-C or G-CBDCA
Durva. + Tremeli. + G-C or G-CBDCA
G-C or G-CBDCAOSPFS, ORR, DOR, DCR, PFS2Not yet reported
EV-302aNCT 04223856760bPembro. + EVG-C or G-CBDCAPFS, OSORR, DORNot yet reported
NameTrial IDTotal sample sizeTreatment regimenControl regimenPrimary endpointsKey secondary oncological endpointsKey results
DANUBEa (36)NCT 025162411032Durva.
Durva. + Tremeli.
G-C or G-CBDCAOScPFS, ORR, DOR
  • OS in high PD-L1 population: 14.4 M for Durva. vs 12.1 M for CT (HR 0.89, 95%CI 0.71–1.11, P = 0.30, not reached predefined threshold)

  • OS in the ITT population: 15.1 M for Durva. + Tremeli. vs 12.1 M for CT (HR 0.85, 95%CI 0.72–1.02, P = 0.0075, not reached predefined threshold)

  • OS in high PD-L1 population: 17.9 M for Durva. + Tremeli. vs 12.1 M for CT (HR 0.74, 95%CI 0.59–0.93)

IMvigor130a (38)NCT 028076361213Atezo.
Atezo. + G-C or G-CBDCA
G-C or G-CBDCAPFSd, OSe, safetyORR, DOR
  • PFS in the ITT population: 8.2 M for Atezo. + CT. vs 6.3 M for CT (HR 0.82, 95%CI 0.70–0.96, 1-sided P = 0.007)

  • OS in the ITT population: 16.0 M for Atezo. + CT. vs 13.4 M for CT (HR 0.83, 95%CI 0.69–1.00, 1-sided P = 0.027, not reached predefined threshold)

  • OS in high PD-L1 population: Not reached for Atezo. vs 17.8 M for CT (HR 0.68, 95%CI 0.43–1.08)

KEYNOTE-361a (37)NCT 028533051010Pembro.
Pembro. + G-C or G-CBDCA
G-C or G-CBDCAPFS, OSORR, DOR, DCR
  • PFS: 8.3 M for Pembro. + CT vs 7.1 M for CT (HR 0.78, 95%CI 0.65–0.93, P = 0.0033, not reached predefined threshold)

  • OS: 17.0 M for Pembro. + CT vs 14.3 M for CT (HR 0.86, 95%CI 0.72–1.02, P = 0.0407, not reached predefined threshold)

CheckMate-901aNCT 030360981290bNivo. + Ipi.
Nivo. + G-C
G-C or G-CBDCAPFS, OSNot yet reported
NILEaNCT 036820681434bDurva. + G-C or G-CBDCA
Durva. + Tremeli. + G-C or G-CBDCA
G-C or G-CBDCAOSPFS, ORR, DOR, DCR, PFS2Not yet reported
EV-302aNCT 04223856760bPembro. + EVG-C or G-CBDCAPFS, OSORR, DORNot yet reported

aJapanese patients enrolled.

bEstimated enrollment.

cTwo OS comparisons; (1) between the Durva. vs CT groups in the population of patients with high PD-L1 expression and (2) between the Durva. + Treme. vs CT groups in the ITT population.

dPFS between the Atezo. + CT vs CT groups in the ITT population.

eOS between the Atezo. vs CT groups in the ITT population, which was to be formally tested only if OS was positive for the Atezo. + CT vs CT groups. Only the interim result is reported as of the end of 2020.

G-C, Gemcitabine + cisplatin; G-CBDCA, Gemcitabine + carboplatin; Durva., Durvalumab; Tremeli., Tremelimumab; Atezo., Atezolizumab; Pembro., Pembrolizumab; Nivo., Nivolumab; Ipi., Ipilimumab; EV, Enfortumab vedotin; PFS, Progression-free survival; OS, Overall survival; ORR, Objective response rate; DOR, Duration of response; DCR, Disease control rate; PFS2, Second profression-free survival; CT, Chemotherapy; ITT, Intention to treat.

Despite various approaches using ICIs are exploring as first-line treatment, it is unclear whether ICI can outperform chemotherapy in the ITT population. Subgroup analyses from several trials suggest that use of a predictive biomarker is critically important. According to the interim analysis of IMvigor 130 trial, atezolizumab plus chemotherapy did not achieve OS advantage over chemotherapy alone based on the predefined statistical significance in the ITT population. Subgroup analysis showed benefit from atezolizumab plus chemotherapy in those with high PD-L1 expression (38). In DANUBE (36), anti-PD-L1 durvalumab alone or durvalumab plus anti-CTLA-4 tremelimumab failed to show OS advantage over chemotherapy in the ITT population. Subgroup analysis showed OS benefit from durvalumab plus tremelimumab in the high PD-L1 group. Based on these results, the target of the primary endpoints may be changed from the ITT population to the PD-L1 high expression group in some ongoing trials. In that case, the introduction of companion diagnostics will be inevitable. However, standardization of the evaluation method for PD-L1 expression remains a critical issue (41).

As shown above, for the platinum-fit population, the standard first-line treatment is still platinum-based systemic chemotherapy. Even if OR or disease control is achieved initially, early chemotherapy resistance is often occurred, which limits PFS and OS after chemotherapy. In a phase 3 JAVELIN bladder 100 trial (NCT02603432), patients who did not have disease progression with first-line chemotherapy (4–6 cycles of GC or G-CBDCA) were randomized to receive best supportive care with or without maintenance avelumab (42). OS was significantly better for the avelumab group (21.4 vs 14.3 months; HR for death, 0.69 [0.56 to 0.86]; P = 0.001). As for maintenance ICI after first-line chemotherapy yielding disease control, pembrolizumab is currently being tested in a phase 2, randomized placebo-controlled trial (NCT02500121). In this setting, Abe et al. (43) reported that maintenance chemotherapy at two- to three-month interval yielded median OS of 37 months (95%CI: 24–60 months) from the initiation of induction chemotherapy, whereas median OS for propensity score-matched patients who did not receive maintenance chemotherapy was 19 months (95%CI: 11–30 months) (P = 0.0573). Because the patient in the control group of JAVELIN bladder 100 trial did not receive maintenance chemotherapy, it is still inclusive which of chemotherapy or immunotherapy is superior as the maintenance treatment. Novel predictive markers to discriminate patients who are benefited from chemotherapy or immunotherapy are strongly warranted in near future.

Perioperative therapy for stage II-III disease

Current consensus is that cisplatin-based neoadjuvant chemotherapy (NAC) improves OS after radical cystectomy (RC) in muscle-invasive bladder cancer (MIBC) (44). Advantage of NAC in pathological complete response (pCR) rate over immediate RC was also demonstrated in Japan; neoadjuvant M-VAC (methotrexate, doxorubicin, vinblastine and cisplatin) yielded higher pCR rate and favorable OS (45). As for adjuvant chemotherapy (AC), recent phase 3 trial (46) and large scale observational study with propensity score matching analysis (47) showed survival advantage of AC over differed treatment strategy. There has not been definitive data to support perioperative therapy in cisplatin-ineligible patients. Because it is considered that patients who achieved pCR are most benefited from NAC, pCR is widely accepted as an intermediate surrogate for survival. Recent reports showed that high-dose or dose-dense modification of M-VAC (48) or GC (49) yielded high pCR rates compared with conventional regimens. As for upper urinary tract cancer (UTUC), the POUT trial has shown advantage in DFS for AC (50) after long time lack of definitive evidence compared with bladder cancer. Although only relatively low-level evidence exists in NAC for UTUC (51), an increasing trend for the use of NAC was reported (52).

Use of ICI for perioperative treatment in MIBC has received an increasing attention in recent years. As extensively reviewed in recent literature (53–55), many phase 1 or 2 trials have been reported or are ongoing; they are testing mono-immunotherapy, combinatorial immunotherapy (e.g. anti-PD-L1 plus anti-CTLA-4) and chemo-immunotherapy combination (e.g. GC plus anti-PD-1). PURE-1 trial (NCT02736266) is an open-label single-arm phase 2 study testing the effect of three cycles of pembrolizumab (200 mg/body q3w) before RC on pCR rate (56). It was reported that pCR rate of 42% and pathological downstage (<pT2) rate of 54%. Similarly, ABACUS trial (NCT02662309) showed that two cycles (1200 mg/body, three times weekly) of preoperative atezolizumab achieved pCR rate of 31% and pathological downstage rate of 39% (57). In a phase 1b NABUCCO trial (NCT03387761), patients with MIBC received 3 mg/kg of ipilimumab (day 1), 3 mg/kg of ipilimumab plus 1 mg/kg nivolumab (day 22) and 3 mg/kg of nivolumab (day 43) followed by RC (58), resulting in pCR rate of 46% and pathological downstage rate of 58%. These proximity outcomes are comparable with those from dd-M-VAC (48) or dd-GC (49). Importantly, biomarker analyses suggested that higher response rate can be expected for those with higher PD-L1 expression. Currently, an international phase 3 study is ongoing; testing the efficacy of neoadjuvant GC plus perioperative nivolumab or neoadjuvant GC plus perioperative nivolumab and BMS-986205 (an oral IDO1 inhibitor) compared with neoadjuvant GC alone (NCT03661320). It will need several years to have primary outcomes including pCR rate and event-free survival. As for UTUC, several phase 2 trials on neoadjuvant or adjuvant immunotherapy are currently ongoing (51).

As for adjuvant therapy, CheckMate-274 (NCT02632409), a phase 3, randomized, double-blind, multicenter trial has revealed that adjuvant nivolumab prolonged DFS by ~10 months, which corresponds to a 30% reduction in the risk of disease recurrence or death, compared with placebo (median DFS 21.0 vs 10.9; HR, 0.70; 98.31% CI, 0.54–0.89; P < 0.001) in patients with muscle-invasive bladder UC or UTUC at high risk of recurrence after radical surgical resection (59). High risk of recurrence was determined as having adverse pathological findings; either ypT2–4a or ypN+ (ypT2–4 or ypN+ for UTUC) in patients treated with previous NAC or pT3–4a or pN+ (pT3–4 or pN+ for UTUC) in patients without previous NAC. On the other hand, IMVigor010 (NCT02450331), a phase 3, randomized, open-label, multicenter trial did not meet its primary endpoint of improved DFS in the atezolizumab group over observation (median DFS 19.4 vs 16.6 months; HR, 0.89; 95% CI, 0.74–1.08; P = 0.24) (60). Despite the almost identical eligibility criteria in terms of high risk for post-surgical recurrence, DFS for the patients in the control arms showed an impressive difference, whereas DFS for the patients in the treatment arms did not differ much. Although we cannot compare the outcomes between independently conducted clinical trials, careful scrutiny is needed for the conflicting results of two closely designed clinical trials.

Bladder-sparing approach for stage I-III disease

RC is the standard therapy for patients with BCG-unresponsive NMIBC or MIBC. However, quite a few patients are considered intolerable of highly invasive surgery or refuse due to postoperative impairment in quality of life. KEYNOTE-057, an open-label single-arm phase 2 trial (NCT02625961) demonstrated the efficacy of pembrolizumab (200 mg/body q3w) with CR rate of 38.8%, 80.2% of which had a CR duration of ≥6 months, an acceptable AE rate (grade 3/4, 12.6%), leading to FDA approval in January 2020 (61). Furthermore, an international phase 3 trial is currently ongoing; testing durvalumab plus BCG versus BCG alone in high-risk, BCG-naïve NMIBC (POTMAC, NCT03528694).

For MIBC, combinatorial radio-immunotherapy has attracted as a promising therapeutic approach (62). Combination of radiotherapy with immunotherapy has been reported to induce immunogenic cell death and an increase in immune markers thus leading to improved tumor control. Because there are some safety concerns for concomitant use of radiotherapy with immunotherapy, dose and timing of each modality including concomitant or sequential administration are still under optimization. As extensively reviewed in a recent literature, many early, phase 1/2 studies are currently ongoing. In Japan, an open-label, single arm phase 2 study (TSUKUBA-002, jRCT2031180060) (https://rctportal.niph.go.jp/en/detail?trial_id=jRCT2031180060) is testing atezolizumab (1200 mg/body q3w) plus radiotherapy (41.4 Gy/23 Fr to the lesser pelvis followed by additional 16.2 Gy/9 Fr to whole bladder) in patients who are not tolerable or refuse RC, with pCR as the primary endpoint.

A phase 3 trial testing pembrolizumab in combination with chemoradiotherapy (CRT) versus CRT alone in cT2-T4a MIBC (KEYNOTE-992, NCT04241185) is also ongoing. Cisplatin monotherapy (35 mg/m2 weekly), 5-fluorouracil (500 mg/m2 on days 1–5 and days 22–26) plus mitomycin C (12 mg/m2 on day 1) or gemcitabine monotherapy (27 mg/m2 twice weekly) are administered intravenously (IV) as radio-sensitizing agents during the CRT (63).

Molecular biomarkers as a predictor of ICI treatment

Treatment with ICI is characterized by the remarkable disparity between responders and non-responders. In KEYNOTE-045 trial, survival rate for pembrolizumab-treated group during the first 3 months was lower than that for the control group, suggesting the impact of initial non-responders (3,18). However, survival curves were then reversed and being separated over time thereafter, suggesting the presence of long-term responders (18). Long-term survivors in the control group are considered to be attributed to crossover use of pembrolizumab after the initial report (3). Therefore, it is particularly important to identify predictive marker for the response to ICI treatment.

Exploratory biomarker analysis from IMvigor010 trial (NCT02450331) has demonstrated that patients with muscle-invasive UC who had detectable circulating tumor DNA (ctDNA) were more likely to benefit from treatment with adjuvant atezolizumab (64). These findings suggest that liquid-base biomarker could help determine who may benefit most from ICI treatment particularly at adjuvant or maintenance settings. Indeed, another clinical trial IMvigor011 has been initiated to evaluate the efficacy and safety of adjuvant treatment with atezolizumab compared with placebo in patients with MIBC who are ctDNA positive and are at high risk for recurrence following cystectomy (NCT04660344).

There have been several clinical biomarkers associated with prognosis of the patients receiving ICI treatment including NLR, CRP, Hb, PS, metastasis site, etc. as described above. Several molecular biomarkers have been reported to act as predictive biomarkers (65). High PD-L1 expression assessed by immunohistochemistry was reportedly associated with favorable response to or survival after ICI treatment in most of clinical trials for the first-line (36,38) or perioperative (36,56) treatment settings. Because high PD-L1 expression is associated with unfavorable response to conventional chemotherapy, it may act as a predictive marker for the benefit of ICI treatment. However, there is no consensus in terms of the definition of ‘PD-L1 high’ cases including which antibody to use and how to count positive cells (41).

Another promising approach to predict clinical response to anti-PD-1/PD-L1 therapy is positron-emission tomography (PET) imaging using radioisotope-labeled antibody. Bensch et al. (66) reported the initial results from MPDL3280A-imaging-IST-UMCG study (NCT02453984) to assess the feasibility of PET imaging with zirconium-89-labeled atezolizumab (89Zr-atezolizumab) in patients with metastatic bladder cancer, non-small-cell lung cancer or triple-negative breast cancer. Uptake was generally high in tumors, particularly in bladder cancer. High heterogeneity within and among lesions, patients, and tumor types was observed in PET uptake, which was correlated with clinical responses to atezolizumab treatment. This approach is reminiscent of 177Lu-PSMA therapy in castration-resistant prostate cancer and seems highly promising. Further development is awaited.

High tumor mutation burden (TMB) (67,68) and microsatellite instability (MSI) (68,69) have been reported to be associated with better response to ICI treatment in UC. However, other studies demonstrated that TMB or MSI alone did not clearly discriminate responders from non-responders in UC and other cancers (70,71). High TMB or MSI is thought to enhance the effects of ICI through the induction of neoantigens. Recent studies suggested that true neo-antigen burden or the number of mutations actually targeted by T cells have a stronger relationship with the ICI response than TMB (65). Indeed, it was reported that the computationally predicted neoantigen burden based on single nucleotide variation data was associated with response to atezolizumab (68). Gene expression profiles provide molecular subtypes (8,9) and extent of infiltration of CD8+ (57) or M1 macrophage (72), which were also reported to be associated with ICI response (57,68,72). A recent report showed that association of these factors with TGFβ signaling pathway (68). The finding of the report is clinically important since TGFβ signaling can be a more direct, stronger predictor ICI response; moreover, it can be a therapeutic target to overcome resistance to ICI treatment. Further studies are warranted in the future.

Renal cell carcinoma

Characteristics of Renal cell carcinoma as a target of ICI therapy

ICIs have changed the approach to the treatment of advanced renal cell carcinoma (aRCC). For example, nivolumab improves the OS in patients with aRCC after anti-angiogenic therapy. In addition, nivolumab plus ipilimumab (73), pembrolizumab plus axitinib (74) and avelumab plus axitinib (75) combination therapies improve the OS and ORR when implemented as the first-line treatment for patients with aRCC. In contrast, questions have been raised regarding the efficacy of sunitinib, which served as the control agent in the randomized phase III trials of these first-line treatments. In addition, the search for biomarkers is important to more effectively select patients who may achieve maximum benefits. Unfortunately, little progress has been made in this area, but a few reports have been published. We believe that continued improvement in the emergence of anti-tumor immunity will lead to changes in the management of aRCC in the near future.

Nivolumab

Nivolumab, a monoclonal antibody that blocks PD-1, has been approved by the US FDA as a second-line treatment for patients with aRCC (4). The approval was based on the results of the Phase III CheckMate025 trial, which showed that in patients with aRCC, the OS was higher in the nivolumab group (n = 406) than in the everolimus group (n = 397) (25 months vs 19.6 months, respectively). The ORR was also higher in patients who received nivolumab for aRCC (4).

Combination of nivolumab and ipilimumab

The combination of ipilimumab, a monoclonal antibody that inhibits CTLA-4 and nivolumab has been approved by the US FDA as the first-line treatment for patients with intermediate- and poor-risk aRCC. The approval was based on the results of the Phase III CheckMate214 trial, which included 1096 patients divided into two groups, with 546 patients receiving ipilimumab and nivolumab (73). The survival rate at a median follow-up of 25.2 months was 75% in the ipilimumab plus nivolumab group and 60% in the sunitinib group. In addition, although the median OS for the ipilimumab plus nivolumab group was NR, the median OS for the sunitinib group was 26.0 months. The ORR was 42% in the ipilimumab plus nivolumab group and 27% in the sunitinib group. Median progression-free survival was 11.6 months and 8.4 months, respectively (73).

Combination of ICIs with molecular target agents

Some trials have been carried out to evaluate the different combinations of anti-angiogenic drugs (tyrosine kinase inhibitor; TKI) with ICIs in aRCC. For example, KEYNOTE-426 (74) was a randomized phase III trial in which 861 patients with previously untreated aRCC were randomly divided into two groups, one receiving pembrolizumab (200 mg) IV every 3 weeks plus axitinib (5 mg) orally twice daily (432 patients) and the other receiving sunitinib (50 mg) orally once daily (4 weeks on and 2 weeks off) (429 patients) (Table 3). The primary endpoint was OS and PFS in the ITT population, and the secondary endpoint was ORR. The median PFS was significantly longer in patients treated with the combination than in patients treated with sunitinib (15.1 months vs 11.1 months, HR 0.69, 95% CI = 0.57–0.84, P < 0.001). The most commonly reported G3 or G4 treatment-related AEs were diarrhea and hypertension in both groups. The incidence of hepatotoxicity was higher in the pembrolizumab-axitinib combination therapy group. However, there were no deaths related to hepatotoxicity. There were four treatment-related deaths in the combination group and seven deaths in the sunitinib group (74).

Table 3

Phase III studies of immune checkpoint inhibitors in combination with vascular endothelial growth factor (VEGF)-targeted therapy for patients with untreated advanced RCC

Trial nameRef NoTherapyPopulationN
KEYNOTE-42674Pembrolizumab + axitinibClear cell component840
JAVELIN Renal 10175Avelumab + axitinibClear cell component583
CLEAR/KEYNOTE-581(307)76Penbrolizumab + lenvatinibClear cell component1069
CheckMate 9 ER77Nivolumab + cabozantinibClear cell component651
Trial nameRef NoTherapyPopulationN
KEYNOTE-42674Pembrolizumab + axitinibClear cell component840
JAVELIN Renal 10175Avelumab + axitinibClear cell component583
CLEAR/KEYNOTE-581(307)76Penbrolizumab + lenvatinibClear cell component1069
CheckMate 9 ER77Nivolumab + cabozantinibClear cell component651
Table 3

Phase III studies of immune checkpoint inhibitors in combination with vascular endothelial growth factor (VEGF)-targeted therapy for patients with untreated advanced RCC

Trial nameRef NoTherapyPopulationN
KEYNOTE-42674Pembrolizumab + axitinibClear cell component840
JAVELIN Renal 10175Avelumab + axitinibClear cell component583
CLEAR/KEYNOTE-581(307)76Penbrolizumab + lenvatinibClear cell component1069
CheckMate 9 ER77Nivolumab + cabozantinibClear cell component651
Trial nameRef NoTherapyPopulationN
KEYNOTE-42674Pembrolizumab + axitinibClear cell component840
JAVELIN Renal 10175Avelumab + axitinibClear cell component583
CLEAR/KEYNOTE-581(307)76Penbrolizumab + lenvatinibClear cell component1069
CheckMate 9 ER77Nivolumab + cabozantinibClear cell component651

JAVELIN Renal 101 (75) was a randomized phase III trial evaluating the efficacy of avelumab (10 mg/kg, IV every 2 weeks) plus axitinib (5 mg orally twice daily) or sunitinib (50 mg orally once daily for 4 weeks on and 2 weeks off) in 886 patients with previously untreated aRCC (Table 3). OS and PFS were the two primary endpoints in patients with PD-L1 positive tumors. PFS, ORR, and safety in the overall population were secondary endpoints. A total of 560 patients (63.2%) had PD-L1-positive tumors. Among these patients, median PFS in the overall population was 13.8 months in the combination group and 7.2 months in the sunitinib group (HR 0.61, 95% CI = 0.47–0.79, P < 0.001). Median PFS was 13.8 months and 8.4 months in the overall population, respectively (HR 0.69, 95% CI = 0). In patients with PD-L1 positive tumors, ORR was 55.2% in the avelumab plus axitinib arm and 25.5% in the control arm. At the time of analysis, the OS data were immature. The most commonly reported G3 or G4 treatment-related AEs were hypertension, diarrhea, elevated alanine aminotransferase levels and palmar plantar erythrocytosis in the combination group, and hypertension, palmar plantar erythrocytosis and hematologic toxicity in the sunitinib group, with three and one treatment-related deaths, respectively (75).

Results from a Phase III clinical trial of CLEAR/KEYNOTE-581(307) were presented (Table 3) (76). The study reported that the combination of lenvatinib (20 mg orally once daily) plus pembrolizumab (200 mg every 3 weeks IV) was superior to sunitinib in terms of PFS in patients with untreated aRCC (23.9 months vs 9.2 months, HR = 0.39, 95% CI = 0.32–0.49, P < 0.001) (76). Moreover, results from a Phase III clinical trial of CheckMate9ER were presented (Table 3) (77). The study reported that the combination of cabozantinib and nivolumab was superior to sunitinib in terms of PFS in patients with untreated aRCC (16.6 months vs 8.3 months, HR = 0.51, 95% CI = 0.41–0.64, P = 0.0001). Long-term data on OS are still in its infancy and awaits further reports. These positive results support the possibility that TKIs enhances the response to ICIs (77).

How long should we use ICIs?

Treatment with ICIs differs from treatment with cytotoxic anticancer agents in several ways. First, in patients who obtain the treatment benefit, the tumors shrink significantly and the effect is sustained over time. Second, the types and timing of side effects are different from those of cytotoxic anticancer drugs. In particular, autoimmune disease-like side effects, which occur when immune cells react to normal tissues, are known to appear in multiple organs. Third, the effects persist after administration is discontinued; PD-1 pathway inhibitors have been proposed to induce immune memory, similar to vaccine therapy for infectious diseases, to activate and sustain immune responses against tumors (78,79). A patient with malignant melanoma was the first patient in whom the effect was sustained after discontinuation of the drug. In the same study, patients who discontinued ICIs had a sustained response to treatment, leading to the hypothesis that ICIs re-educate the immune system against the tumor, resulting in the acquisition of immune memory (80). A phase II trial is ongoing to prove this hypothesis (81). In this single-arm phase II trial, patients who achieved a reduction in tumor volume were withdrawn from treatment and given intermittent nivolumab therapy monitored by CT imaging (81). Four of five patients (80%) who discontinued treatment showed sustained anti-tumor responses and discontinued therapy. It is conceivable that such intermittent therapy could improve toxicity and tolerability without diminishing the therapeutic effect. If the withdrawal period could be extended, the cumulative toxicity and the cost of treatment could be reduced. Despite the limitations of this trial, this prospective study provides important possibility that an intermittent approach to immunotherapy is feasible in patients with aRCC. Regarding the possibility of a pause in ICI, we are now doing the Japan Clinical Oncology Group 1905 trial, where patients with renal cancer who have received an ICI for 24 weeks are randomized into two groups: those who continued ICI treatments and those who discontinued ICI treatments (Fig. 1). In the CheckMate 025 study, patients on nivolumab monotherapy after one or two prior TKI therapies had a relatively early response (6–8 weeks); however, some patients demonstrated early progression in spite of initial responses. Therefore, we have decided suspending ICI treatments only in patients who have continuous responses to the treatments for a certain period of time, rather than suspending treatments immediately after initial responses are obtained. Although there is no clear data to support when it is appropriate to stop ICI treatments, it seems more logical to stop them when tumors stopped shrinking and have reached a plateau rather than when tumors are continuously shrinking. The spider plot in the CheckMate 025 trial shows that the rate of tumor size reduction often reaches a plateau around 24 to 36 weeks after the start of nivolumab treatment (4). Tumors treated with pembrolizumab plus axitinib or avelumab plus axitinib also seem to reach a plateau around 4–6 months (74,75). Therefore, we have decided to enroll patients who have no exacerbations at 24 weeks after starting ICI therapies.

Trial design of JCOG1905.
Figure 1.

Trial design of JCOG1905.

Biomarkers to predict prognosis and response to ICIs

Unfortunately, to date, no reliable biomarkers that can predict the efficacy of immunotherapy and define the specific subgroup of patients who respond best to anti-PD-1 agents have been revealed (82,83). Although PD-L1 expression is the most studied predictive biomarker for ICI outcomes, stratification of patient outcomes based on PD-L1 expression is currently met with skepticism due to limitations in sensitivity, specificity, reproducibility, and the unreliability of staining. The reasons for this are, first, that PD-L1 expression is highly heterogeneous both within tissue samples and between primary tumors and metastatic sites. Second, PD-L1 expression is dynamic and has been reported to be profoundly affected by treatments.

In a cohort of renal cell carcinoma (RCC) patients treated with nivolumab, whole exome sequencing was performed on tumor and normal tissue pairs. This study showed that there was an association between PBRM1 loss and survival (84). In other words, PBRM1 loss was identified as a biomarker of response to ICI treatment: patients with tumors with PBMR1 loss had higher ORR, longer PFS, and longer OS (85). Conversely, there was no difference in PFS, OS or ORR by PBRM1 status in patients treated with TKIs (85). Furthermore, TMB has recently been proposed as a predictive biomarker of response to ICI in patients with various cancer types. However, the percentage of TMB in RCC is 10–400 times lower than that in melanoma and NSCLC, even though the low TMB in RCC is characterized by a high absolute number of frameshift indel mutations and a high percentage of indels in total mutations (86). Frameshift indel mutation is known to be a major mechanism involved in the production of neoantigens through which T cell responses are induced (85). The association of frameshift indel mutations with response rates has been confirmed in patients with RCC treated with anti-PD-1 agents (87). Despite the limited sample size, the number of frameshift indel mutations was significantly associated with OS (87). In contrast, in patients treated with TKIs, there was no statistical difference in OS (88). Recently reported data on the clinical efficacy of pembrolizumab (an anti-PD-1 drug) in MMR-deficient patients support the hypothesis that MMR-deficient tumors are more likely to benefit from anti-PD-1 drugs than MMR-proficient tumors (88).

Conclusions and future vision

UC is a cancer for which immunotherapy is expected to be effective. ICI has been approved in chemoresistant UC as the novel standard treatment that prolongs prognosis. Approval of ICI is being expanded to other treatment settings for which no standard treatment currently exists, including maintenance therapy after effective first-line chemotherapy for metastatic urothelial carcinoma (mUC), first-line therapy for platinum-unfit mUC and salvage therapy for BCG-unresponsive NMIBC. Furthermore, there are a number of trials that explore the possibility for ICI to replace conventional standard of care, including cisplatin-based first-line chemotherapy for mUC and perioperative chemotherapy for MIBC prior to RC. There is an urgent need for predictive biomarkers for personalized clinical decision-making in the future. On the other hand, several problems need to be solved in order to optimize ICI treatment for patients with aRCC. It is yet unclear as to in which cases should we switch to different therapeutic agents, how long should ICI be used in the case of adjuvant therapy, and whether surgical resection of the primary tumor or metastases should be performed prior to ICI therapy. Furthermore, if ICI therapy is successful, then when should ICI therapy be discontinued? Further prospective trials are needed to answer these questions. It is expected that a number of prospective clinical trials will lead to the appropriate use of ICI in patients with recurrent renal cancer.

Funding

This work was supported in part by AMED under Grant Number JP20ck0106585.

Conflict of interest statement

Takashi Kobayashi received honoraria for lectures from Janssen Pharma, AstraZneca, Chugai, Bayer, MSD, Sanofi, Takeda, Astellas, Nippon Shinyaku, Nihon Kayaku, Merck, Pfizer and research funding from AstraZneca, Chugai. Ario Takeuchi has no conflicts of interest. Hiroyuki Nishiyama received honoraria for lectures from MSD, AstraZneca, and research funding from Chugai, Bayer, Ono, Takeda Astellas. Masatoshi Eto received honoraria for lectures from ONO, BMS, Pfizer, Novartis, Bayer, Takeda and research funding from ONO, Pfizer, Astellas, Takeda and Kissei.

References

1.

Kamat
 
AM
,
Hahn
 
NM
,
Efstathiou
 
JA
, et al.  
Bladder cancer
.
Lancet
 
2016
;
388
:
2796
810
.

2.

Negrier
 
S
,
Escudier
 
B
,
Lasset
 
C
, et al.  
Recombinant human interleukin-2, recombinant human interferon alfa-2a, or both in metastatic renal-cell carcinoma. Groupe Francais d'Immunotherapie
.
N Engl J Med
 
1998
;
338
:
1272
8
.

3.

Bellmunt
 
J
,
de
 
Wit
 
R
,
Vaughn
 
DJ
, et al.  
Pembrolizumab as second-line therapy for advanced urothelial carcinoma
.
N Engl J Med
 
2017
;
376
:
1015
26
.

4.

Motzer
 
RJ
,
Escudier
 
B
,
McDermott
 
DF
, et al.  
Nivolumab versus everolimus in advanced renal-cell carcinoma
.
N Engl J Med
 
2015
;
373
:
1803
13
.

5.

Kandoth
 
C
,
McLellan
 
MD
,
Vandin
 
F
, et al.  
Mutational landscape and significance across 12 major cancer types
.
Nature
 
2013
;
502
:
333
9
.

6.

Knowles
 
MA
,
Hurst
 
CD
.
Molecular biology of bladder cancer: new insights into pathogenesis and clinical diversity
.
Nat Rev Cancer
 
2015
;
15
:
25
41
.

7.

Kobayashi
 
T
,
Owczarek
 
TB
,
McKiernan
 
JM
,
Abate-Shen
 
C
.
Modelling bladder cancer in mice: opportunities and challenges
.
Nat Rev Cancer
 
2015
;
15
:
42
54
.

8.

Robertson
 
AG
,
Kim
 
J
,
Al-Ahmadie
 
H
, et al.  
Comprehensive molecular characterization of muscle-invasive bladder cancer
.
Cell
 
2017
;
171
:
540
56
 
e25
.

9.

Kamoun
 
A
,
de
 
Reynies
 
A
,
Allory
 
Y
, et al.  
A consensus molecular classification of muscle-invasive bladder cancer
.
Eur Urol
 
2020
;
77
:
420
33
.

10.

Iyer
 
G
,
Hanrahan
 
AJ
,
Milowsky
 
MI
, et al.  
Genome sequencing identifies a basis for everolimus sensitivity
.
Science
 
2012
;
338
:
221
.

11.

Powles
 
T
,
Eder
 
JP
,
Fine
 
GD
, et al.  
MPDL3280A (anti-PD-L1) treatment leads to clinical activity in metastatic bladder cancer
.
Nature
 
2014
;
515
:
558
62
.

12.

Logothetis
 
CJ
,
Dexeus
 
FH
,
Finn
 
L
, et al.  
A prospective randomized trial comparing MVAC and CISCA chemotherapy for patients with metastatic urothelial tumors
.
J Clin Oncol
 
1990
;
8
:
1050
5
.

13.

von der
 
Maase
 
H
,
Hansen
 
SW
,
Roberts
 
JT
, et al.  
Gemcitabine and cisplatin versus methotrexate, vinblastine, doxorubicin, and cisplatin in advanced or metastatic bladder cancer: results of a large, randomized, multinational, multicenter, phase III study
.
J Clin Oncol
 
2000
;
18
:
3068
77
.

14.

Matsumoto
 
H
,
Shiraishi
 
K
,
Azuma
 
H
, et al.  
Clinical practice guidelines for bladder cancer 2019 edition by the Japanese Urological Association: revision working position paper
.
Int J Urol
 
2020
;
27
:
362
8
.

15.

Galsky
 
MD
,
Chen
 
GJ
,
Oh
 
WK
, et al.  
Comparative effectiveness of cisplatin-based and carboplatin-based chemotherapy for treatment of advanced urothelial carcinoma
.
Ann Oncol
 
2012
;
23
:
406
10
.

16.

Hanna
 
KS
.
Updates and novel treatments in urothelial carcinoma
.
J Oncol Pharm Pract
 
2019
;
25
:
648
56
.

17.

Nishiyama
 
H
,
Yamamoto
 
Y
,
Sassa
 
N
, et al.  
Pembrolizumab versus chemotherapy in recurrent, advanced urothelial cancer in Japanese patients: a subgroup analysis of the phase 3 KEYNOTE-045 trial
.
Int J Clin Oncol
 
2020
;
25
:
165
74
.

18.

Fradet
 
Y
,
Bellmunt
 
J
,
Vaughn
 
DJ
, et al.  
Randomized phase III KEYNOTE-045 trial of pembrolizumab versus paclitaxel, docetaxel, or vinflunine in recurrent advanced urothelial cancer: results of >2 years of follow-up
.
Ann Oncol
 
2019
;
30
:
970
6
.

19.

Vaughn
 
DJ
,
Bellmunt
 
J
,
Fradet
 
Y
, et al.  
Health-related quality-of-life analysis from KEYNOTE-045: a phase III study of pembrolizumab versus chemotherapy for previously treated advanced urothelial cancer
.
J Clin Oncol
 
2018
;
36
:
1579
87
.

20.

Slongo
 
J
,
Jain
 
RK
,
Spiess
 
PE
.
Important caveats of KEYNOTE-045: relevance of these findings in the current and future therapeutic paradigm
.
Ann Transl Med
 
2019
;
7
:
S23
.

21.

Tamura
 
D
,
Jinnouchi
 
N
,
Abe
 
M
, et al.  
Prognostic outcomes and safety in patients treated with pembrolizumab for advanced urothelial carcinoma: experience in real-world clinical practice
.
Int J Clin Oncol
 
2020
;
25
:
899
905
.

22.

Yasuoka
 
S
,
Yuasa
 
T
,
Nishimura
 
N
, et al.  
Initial experience of pembrolizumab therapy in Japanese patients with metastatic urothelial cancer
.
Anticancer Res
 
2019
;
39
:
3887
92
.

23.

Furubayashi
 
N
,
Kuroiwa
 
K
,
Tokuda
 
N
, et al.  
Treating Japanese patients with pembrolizumab for platinum-refractory advanced urothelial carcinoma in real-world clinical practice
.
J Clin Med Res
 
2020
;
12
:
300
6
.

24.

Ogihara
 
K
,
Kikuchi
 
E
,
Shigeta
 
K
, et al.  
The pretreatment neutrophil-to-lymphocyte ratio is a novel biomarker for predicting clinical responses to pembrolizumab in platinum-resistant metastatic urothelial carcinoma patients
.
Urol Oncol
 
2020
;
38
:
602 e1
e10
.

25.

Yamamoto
 
Y
,
Yatsuda
 
J
,
Shimokawa
 
M
, et al.  
Prognostic value of pre-treatment risk stratification and post-treatment neutrophil/lymphocyte ratio change for pembrolizumab in patients with advanced urothelial carcinoma
.
Int J Clin Oncol
 
2021
;
26
:
169
177
.

26.

Kijima
 
T
,
Yamamoto
 
H
,
Saito
 
K
, et al.  
Early C-reactive protein kinetics predict survival of patients with advanced urothelial cancer treated with pembrolizumab
.
Cancer Immunol Immunother
 
2021
;
70
:
657
665
.

27.

Kobayashi
 
T
,
Ito
 
K
,
Kojima
 
T
, et al.  
Risk stratification for the prognosis of patients with chemoresistant urothelial cancer treated with pembrolizumab
.
Cancer Sci
 
2021
;
112
:
760
73
.

28.

Sternberg
 
CN
,
Loriot
 
Y
,
James
 
N
, et al.  
Primary results from SAUL, a multinational single-arm safety study of atezolizumab therapy for locally advanced or metastatic urothelial or nonurothelial carcinoma of the urinary tract
.
Eur Urol
 
2019
;
76
:
73
81
.

29.

Sonpavde
 
G
,
Manitz
 
J
,
Gao
 
C
, et al.  
Five-factor prognostic model for survival of post-platinum patients with metastatic urothelial carcinoma receiving PD-L1 inhibitors
.
J Urol
 
2020
;
204
:
1173
79
.

30.

Khaki
 
AR
,
Li
 
A
,
Diamantopoulos
 
LN
, et al.  
Impact of performance status on treatment outcomes: a real-world study of advanced urothelial cancer treated with immune checkpoint inhibitors
.
Cancer
 
2020
;
126
:
1208
16
.

31.

Ito
 
K
,
Kobayashi
 
T
,
Kojima
 
T
, et al.  
Pembrolizumab for treating advanced urothelial carcinoma in patients with impaired performance status: analysis of a Japanese nationwide cohort
.
Cancer Med
 
2021
;
10
:
3188
96
.

32.

Kobayashi
 
T
,
Ito
 
K
,
Kojima
 
T
, et al.  
Effect of change in NLR by 1st-line chemotherapy on the efficacy of 2nd-line pembrolizumab therapy in urothelial cancer
.
2020
;
submitted
.

33.

Suzman
 
DL
,
Agrawal
 
S
,
Ning
 
YM
, et al.  
FDA approval summary: atezolizumab or pembrolizumab for the treatment of patients with advanced urothelial carcinoma ineligible for cisplatin-containing chemotherapy
.
Oncologist
 
2019
;
24
:
563
9
.

34.

Necchi
 
A
,
Joseph
 
RW
,
Loriot
 
Y
, et al.  
Atezolizumab in platinum-treated locally advanced or metastatic urothelial carcinoma: post-progression outcomes from the phase II IMvigor210 study
.
Ann Oncol
 
2017
;
28
:
3044
50
.

35.

Vuky
 
J
,
Balar
 
AV
,
Castellano
 
D
, et al.  
Long-term outcomes in KEYNOTE-052: phase II study investigating first-line pembrolizumab in cisplatin-ineligible patients with locally advanced or metastatic urothelial cancer
.
J Clin Oncol
 
2020
;
38
:
2658
66
.

36.

Powles
 
T
,
van der
 
Heijden
 
MS
,
Castellano
 
D
, et al.  
Durvalumab alone and durvalumab plus tremelimumab versus chemotherapy in previously untreated patients with unresectable, locally advanced or metastatic urothelial carcinoma (DANUBE): a randomised, open-label, multicentre, phase 3 trial
.
Lancet Oncol
 
2020
;
21
:
1574
88
.

37.

Alva
 
A
,
Csőszi
 
T
,
Ozguroglu
 
M
, et al.  
Pembrolizumab (P) combined with chemotherapy (C) vs C alone as first-line (1L) therapy for advanced urothelial carcinoma (UC): KEYNOTE-361
.
Ann Oncol
 
2020
;
31
:
S1142
S215
.

38.

Galsky
 
MD
,
Arija
 
JAA
,
Bamias
 
A
, et al.  
Atezolizumab with or without chemotherapy in metastatic urothelial cancer (IMvigor130): a multicentre, randomised, placebo-controlled phase 3 trial
.
Lancet
 
2020
;
395
:
1547
57
.

39.

Nadal
 
R
,
Bellmunt
 
J
.
Management of metastatic bladder cancer
.
Cancer Treat Rev
 
2019
;
76
:
10
21
.

40.

van
 
Dijk
 
N
,
Funt
 
SA
,
Blank
 
CU
,
Powles
 
T
,
Rosenberg
 
JE
,
van der
 
Heijden
 
MS
.
The cancer immunogram as a framework for personalized immunotherapy in urothelial cancer
.
Eur Urol
 
2019
;
75
:
435
44
.

41.

Rouanne
 
M
,
Radulescu
 
C
,
Adam
 
J
,
Allory
 
Y
.
PD-L1 testing in urothelial bladder cancer: essentials of clinical practice
.
World J Urol
 
2021
;
39
:
1335
55
.

42.

Powles
 
T
,
Park
 
SH
,
Voog
 
E
, et al.  
Avelumab maintenance therapy for advanced or metastatic urothelial carcinoma
.
N Engl J Med
 
2020
;
383
:
1218
30
.

43.

Abe
 
T
,
Minami
 
K
,
Harabayashi
 
T
, et al.  
Outcome of maintenance systemic chemotherapy with drug-free interval for metastatic urothelial carcinoma
.
Jpn J Clin Oncol
 
2019
;
49
:
965
71
.

44.

Meeks
 
JJ
,
Bellmunt
 
J
,
Bochner
 
BH
, et al.  
A systematic review of neoadjuvant and adjuvant chemotherapy for muscle-invasive bladder cancer
.
Eur Urol
 
2012
;
62
:
523
33
.

45.

Kitamura
 
H
,
Tsukamoto
 
T
,
Shibata
 
T
, et al.  
Randomised phase III study of neoadjuvant chemotherapy with methotrexate, doxorubicin, vinblastine and cisplatin followed by radical cystectomy compared with radical cystectomy alone for muscle-invasive bladder cancer: Japan Clinical Oncology Group Study JCOG0209
.
Ann Oncol
 
2014
;
25
:
1192
8
.

46.

Sternberg
 
CN
,
Skoneczna
 
I
,
Kerst
 
JM
, et al.  
Immediate versus deferred chemotherapy after radical cystectomy in patients with pT3-pT4 or N+ M0 urothelial carcinoma of the bladder (EORTC 30994): an intergroup, open-label, randomised phase 3 trial
.
Lancet Oncol
 
2015
;
16
:
76
86
.

47.

Galsky
 
MD
,
Stensland
 
KD
,
Moshier
 
E
, et al.  
Effectiveness of adjuvant chemotherapy for locally advanced bladder cancer
.
J Clin Oncol
 
2016
;
34
:
825
32
.

48.

Zargar
 
H
,
Shah
 
JB
,
van de
 
Putte
 
EEF
, et al.  
Dose dense MVAC prior to radical cystectomy: a real-world experience
.
World J Urol
 
2017
;
35
:
1729
36
.

49.

Iyer
 
G
,
Balar
 
AV
,
Milowsky
 
MI
, et al.  
Multicenter prospective phase II trial of neoadjuvant dose-dense gemcitabine plus cisplatin in patients with muscle-invasive bladder cancer
.
J Clin Oncol
 
2018
;
36
:
1949
56
.

50.

Birtle
 
A
,
Johnson
 
M
,
Chester
 
J
, et al.  
Adjuvant chemotherapy in upper tract urothelial carcinoma (the POUT trial): a phase 3, open-label, randomised controlled trial
.
Lancet
 
2020
;
395
:
1268
77
.

51.

Leow
 
JJ
,
Chong
 
YL
,
Chang
 
SL
,
Valderrama
 
BP
,
Powles
 
T
,
Bellmunt
 
J
.
Neoadjuvant and adjuvant chemotherapy for upper tract urothelial carcinoma: a 2020 systematic review and meta-analysis, and future perspectives on systemic therapy
.
Eur Urol
 
2021
;
79
:
635
654
.

52.

Hamaya
 
T
,
Hatakeyama
 
S
,
Tanaka
 
T
, et al.  
Trends in the use of neoadjuvant chemotherapy and oncological outcomes for high-risk upper tract urothelial carcinoma: a multicentre retrospective study
.
BJU Int
 
2021
. .
Online ahead of print.

53.

Lee
 
HH
,
Ham
 
WS
.
Perioperative immunotherapy in muscle-invasive bladder cancer
.
Transl Cancer Res
 
2020
;
9
:
6546
53
.

54.

Zucali
 
PA
,
Cordua
 
N
,
D'Antonio
 
F
, et al.  
Current perspectives on immunotherapy in the peri-operative setting of muscle-infiltrating bladder cancer
.
Front Oncol
 
2020
;
10
:
568279
.

55.

Rouanne
 
M
,
Bajorin
 
DF
,
Hannan
 
R
, et al.  
Rationale and outcomes for neoadjuvant immunotherapy in urothelial carcinoma of the bladder
.
Eur Urol Oncol
 
2020
;
3
:
728
38
.

56.

Necchi
 
A
,
Anichini
 
A
,
Raggi
 
D
, 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
;
36
:
3353
60
.

57.

Powles
 
T
,
Kockx
 
M
,
Rodriguez-Vida
 
A
, et al.  
Clinical efficacy and biomarker analysis of neoadjuvant atezolizumab in operable urothelial carcinoma in the ABACUS trial
.
Nat Med
 
2019
;
25
:
1706
14
.

58.

van
 
Dijk
 
N
,
Gil-Jimenez
 
A
,
Silina
 
K
, et al.  
Preoperative ipilimumab plus nivolumab in locoregionally advanced urothelial cancer: the NABUCCO trial
.
Nat Med
 
2020
;
26
:
1839
44
.

59.

Bajorin
 
DF
,
Witjes
 
JA
,
Gschwend
 
JE
, et al.  
First results from the phase 3 Check Mate 274 trial of adjuvant nivolumab vs placebo in patients who underwent radical surgery for high-risk muscle-invasive urothelial carcinoma (MIUC)
.
J Clin Oncol
 
2021
;
39
:391. doi: .

60.

Bellmunt
 
J
,
Hussain
 
M
,
Gschwend
 
JE
, et al.  
Adjuvant atezolizumab versus observation in muscle-invasive urothelial carcinoma (IMvigor010): a multicentre, open-label, randomised, phase 3 trial
.
Lancet Oncol
 
2021
;
22
:
525
37
.

61.

Balar
 
AV
,
Kulkarni
 
GS
,
Uchio
 
EM
, et al.  
Keynote 057: phase II trial of pembrolizumab (pembro) for patients (pts) with high-risk (HR) nonmuscle invasive bladder cancer (NMIBC) unresponsive to bacillus calmette-guérin (BCG)
.
J Clin Oncol
 
2019
;
37
:
350
.

62.

Daro-Faye
 
M
,
Kassouf
 
W
,
Souhami
 
L
, et al.  
Combined radiotherapy and immunotherapy in urothelial bladder cancer: harnessing the full potential of the anti-tumor immune response
.
World J Urol
 
2021
;
39
:
1131
43
.

63.

Balar
 
AV
,
James
 
ND
,
Shariat
 
SF
,
Shore
 
ND
,
van der
 
Heijden
 
MS
,
Weickhardt
 
AJ
.
Phase III study of pembrolizumab (pembro) plus chemoradiotherapy (CRT) versus CRT alone for patients (pts) with muscle-invasive bladder cancer (MIBC): KEYNOTE-992
.
J Clin Oncol
 
2020
;
38
:
5093
.

64.

Powles
 
TB
,
Assaf
 
ZJ
,
Davarpanah
 
N
, et al.  
Clinical outcomes in post-operative ctDNA-positive muscle-invasive urothelial carcinoma (MIUC) patients after atezolizumab adjuvant therapy
.
Ann Oncol
 
2020
;
31
:S1417.

65.

Roviello
 
G
,
Catalano
 
M
,
Nobili
 
S
,
Santi
 
R
,
Mini
 
E
,
Nesi
 
G
.
Focus on biochemical and clinical predictors of response to immune checkpoint inhibitors in metastatic urothelial carcinoma: Where do we stand?
 
Int J Mol Sci
 
2020
;
21
:
7935
.

66.

Bensch
 
F
,
van der
 
Veen
 
EL
,
Lub-de Hooge
 
MN
, et al.  
(89)Zr-atezolizumab imaging as a non-invasive approach to assess clinical response to PD-L1 blockade in cancer
.
Nat Med
 
2018
;
24
:
1852
8
.

67.

Balar
 
AV
,
Galsky
 
MD
,
Rosenberg
 
JE
, et al.  
Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, phase 2 trial
.
Lancet
 
2017
;
389
:
67
76
.

68.

Mariathasan
 
S
,
Turley
 
SJ
,
Nickles
 
D
, et al.  
TGFbeta attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells
.
Nature
 
2018
;
554
:
544
8
.

69.

Castro
 
MP
,
Goldstein
 
N
.
Mismatch repair deficiency associated with complete remission to combination programmed cell death ligand immune therapy in a patient with sporadic urothelial carcinoma: immunotheranostic considerations
.
J Immunother Cancer
 
2015
;
3
:
58
.

70.

Leiserson
 
MD
,
Vandin
 
F
,
Wu
 
HT
, et al.  
Pan-cancer network analysis identifies combinations of rare somatic mutations across pathways and protein complexes
.
Nat Genet
 
2015
;
47
:
106
14
.

71.

Roh
 
W
,
Chen
 
PL
,
Reuben
 
A
, et al.  
Integrated molecular analysis of tumor biopsies on sequential CTLA-4 and PD-1 blockade reveals markers of response and resistance
.
Sci Transl Med
 
2017
;
9
:
eaah3560
.

72.

Zeng
 
D
,
Ye
 
Z
,
Wu
 
J
, et al.  
Macrophage correlates with immunophenotype and predicts anti-PD-L1 response of urothelial cancer
.
Theranostics
 
2020
;
10
:
7002
14
.

73.

Motzer
 
RJ
,
Tannir
 
NM
,
McDermott
 
DF
, et al.  
Nivolumab plus ipilimumab versus sunitinib in advanced renal-cell carcinoma
.
N Engl J Med
 
2018
;
378
:
1277
90
.

74.

Rini
 
BI
,
Plimack
 
ER
,
Stus
 
V
, et al.  
Pembrolizumab plus axitinib versus sunitinib for advanced renal-cell carcinoma
.
N Engl J Med
 
2019
;
380
:
1116
27
.

75.

Motzer
 
RJ
,
Penkov
 
K
,
Haanen
 
J
, et al.  
Avelumab plus axitinib versus sunitinib for advanced renal-cell carcinoma
.
N Engl J Med
 
2019
;
380
:
1103
15
.

76.

Motzer
 
RJ
,
Alekseev
 
B
,
Rha
 
SY
, et al.  
Lenvatinib plus pembrolizumab or everolimus for advanced renal cell carcinoma
.
N Engl J Med
 
2021 Apr 8
;
384
:
1289
300
.

77.

Choueiri
 
TK
,
Powles
 
T
,
Burotto
 
M
, et al.  
Nivolumab + cabozantinib vs sunitinib in first-line treatment for advanced renal cell carcinoma: first results from the randomized phase 3 Check Mate 9ER trial
.
Ann Oncol
 
2020
;
31
:
S1142
215
.

78.

Weber
 
JS
,
Kahler
 
KC
,
Hauschild
 
A
.
Management of immune-related adverse events and kinetics of response with ipilimumab
.
J Clin Oncol
 
2012
;
30
:
2691
7
.

79.

Schreiber
 
RD
,
Old
 
LJ
,
Smyth
 
MJ
.
Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion
.
Science
 
2011
;
331
:
1565
70
.

80.

Weber
 
JS
,
Hodi
 
FS
,
Wolchok
 
JD
, et al.  
Safety profile of nivolumab monotherapy: a pooled analysis of patients with advanced melanoma
.
J Clin Oncol
 
2017
;
35
:
785
92
.

81.

Ornstein
 
MC
,
Wood
 
LS
,
Hobbs
 
BP
, et al.  
A phase II trial of intermittent nivolumab in patients with metastatic renal cell carcinoma (mRCC) who have received prior anti-angiogenic therapy
.
J Immunother Cancer
 
2019
;
7
:
127
.

82.

Madore
 
J
,
Vilain
 
RE
,
Menzies
 
AM
, et al.  
PD-L1 expression in melanoma shows marked heterogeneity within and between patients: implications for anti-PD-1/PD-L1 clinical trials
.
Pigment Cell Melanoma Res
 
2015
;
28
:
245
53
.

83.

McLaughlin
 
J
,
Han
 
G
,
Schalper
 
KA
, et al.  
Quantitative assessment of the heterogeneity of PD-L1 expression in non-small-cell lung cancer
.
JAMA Oncol
 
2016
;
2
:
46
54
.

84.

Miao
 
D
,
Margolis
 
CA
,
Gao
 
W
, et al.  
Genomic correlates of response to immune checkpoint therapies in clear cell renal cell carcinoma
.
Science
 
2018
;
59
:
801
6
.

85.

Braun
 
DA
,
Ishii
 
Y
,
Walsh
 
AM
, et al.  
Clinical validation of PBRM1 alterations as a marker of immune checkpoint inhibitor response in renal cell carcinoma
.
JAMA Oncol
 
2019
;
5
:
1631
3
.

86.

Lavacchi
 
D
,
Pellegrini
 
E
,
Palmieri
 
V
, et al.  
Immune checkpoint inhibitors in the treatment of renal cancer: current state and future perspective
.
Int J Mol Sci
 
2020
;
21
:
4691
.

87.

Voss
 
MH
,
Novik
 
JB
,
Hellmann
 
MD
, et al.  
Correlation of degree of tumor immune infiltration and insertion-and-deletion (indel) burden with outcome on programmed death 1 (PD1) therapy in advanced renal cell cancer (RCC)
.
J Clin Oncol
 
2018
;
36
:
4518
.

88.

Patel
 
S
,
Longacre
 
T
,
Ladabaum
 
U
, et al.  
Tumor molecular testing guides anti-pd-1 therapy and provides evidence for pathogenicity of mismatch repair variants
.
Oncologist
 
2018 Dec
;
23
:
1395
400
.

Author notes

Takashi Kobayashi, Ario Takeuchi and Masatoshi Eto Contributed equally

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/journals/pages/open_access/funder_policies/chorus/standard_publication_model)