Next Article in Journal
Genetic Heterogeneity, Therapeutic Hurdle Confronting Sorafenib and Immune Checkpoint Inhibitors in Hepatocellular Carcinoma
Next Article in Special Issue
Immune Checkpoint Inhibitors in Urothelial Bladder Cancer: State of the Art and Future Perspectives
Previous Article in Journal
Host–Microbiota Interactions in Liver Inflammation and Cancer
Previous Article in Special Issue
Immune Checkpoint Inhibition in Advanced Non-Clear Cell Renal Cell Carcinoma: Leveraging Success from Clear Cell Histology into New Opportunities
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Efficacy of Immune Checkpoint Inhibitors in Upper Tract Urothelial Carcinomas: Current Knowledge and Future Directions

by
Jonathan Thouvenin
1,2,*,
Nieves Martínez Chanzá
2,
Omar Alhalabi
3,
Hervé Lang
4,
Nizar M. Tannir
3,
Philippe Barthélémy
1 and
Gabriel G. Malouf
1,*
1
Oncology Department, Institut de Cancérologie Strasbourg (ICANS), 67200 Strasbourg, France
2
Oncology Department, Institut Jules Bordet, 1000 Bruxelles, Belgium
3
Genitourinary Medical Oncology Department, The University of Texas MD Anderson Cancer Center (MDACC), Houston, TX 77030, USA
4
Urology Department, Strasbourg University Hospital, Nouvel Hôpital Civil, 67000 Strasbourg, France
*
Authors to whom correspondence should be addressed.
Cancers 2021, 13(17), 4341; https://doi.org/10.3390/cancers13174341
Submission received: 28 July 2021 / Revised: 21 August 2021 / Accepted: 24 August 2021 / Published: 27 August 2021
(This article belongs to the Special Issue Immune Checkpoint Inhibitors for Genitourinary Cancers)

Abstract

:

Simple Summary

Upper tract urothelial carcinoma (UTUC) represents 5 to 10% of urothelial carcinoma. Their mutational profile is different as compared to bladder urothelial carcinoma (UC). While immune checkpoint inhibitors are now part of the therapeutic landscape of urothelial carcinoma, data concerning their use in UTUC patient’s treatment remain scarce. The aim of this review is to summarize the available evidence and the biological rationale of using immune checkpoint inhibitors in high-grade UTUC. We reviewed the latest molecular characterization data and proposed an insight for future therapeutic strategies based on molecular alteration profiles.

Abstract

Upper tract urothelial carcinoma (UTUC) represents a rare and aggressive malignancy arising from the renal pelvis or ureter. It can develop sporadically or have a hereditary origin, such as Lynch syndrome, caused by DNA mismatch repair deficiency, leading to microsatellite instability phenotype. According to molecular characterization studies, UTUC presents different mutational profiles as compared to urinary bladder urothelial carcinomas. In particular, it has been reported that UTUC harbored a higher level of FGFR3 alterations associated with a T-cell depleted immune microenvironment. The therapeutic landscape in urothelial carcinoma is rapidly evolving, with immune checkpoint inhibitors forming part of the standard of care. A greater understanding of the molecular alterations and immune microenvironment leads to the development of new treatment combinations and targeted therapy. This review summarizes the available evidence concerning the use of immune checkpoint inhibitors and the biological rationale underlying their use in high-grade UTUC.

1. Introduction

Urothelial carcinoma (UC) represents the fourth most common malignancy worldwide, with an urgent need for tailored approaches in the management of the metastatic disease [1]. Depending on the level of muscle invasion seen on the pathological exam, UC is divided into muscle-invasive (MI) and non-muscle invasive (NMI) disease. MIUC of the bladder represents 25% of tumors [2] as compared to 60% in upper tract urothelial carcinoma (UTUC), explaining their increased aggressiveness [3,4,5,6]. The 5-year extravesical recurrence and overall survival rates are 28% and 23% for UTUC and bladder UC, respectively [7]. While bladder origin represents 90–95% of UCs, UTUC is less common. It represents 5–10% of UCs and can arise within the renal pelvis or ureter, which are derived from a different embryologic origin as compared to the bladder [1,8].
There is a strong relationship between UC of the bladder and UTUC since approximately 50% of patients with UTUC will have urinary bladder urothelial carcinomas either at presentation or subsequently, justifying the need to perform annual cystoscopy in the follow-up of these patients [1].
For high-risk localized disease, nephroureterectomy along with peri-operative chemotherapy is the standard of care management approach [1]. In the metastatic setting, platinum-based chemotherapy regimen remains the first-line recommended treatment [1]. However, there is a growing body of evidence concerning the use of immune checkpoint inhibitors (ICI) in the treatment of urothelial carcinoma [1,9] with the approval of several compounds in the first and second-line settings of advanced UCs. However, given their rarity, patients with UTUC represent a minority of patients included in clinical trials, and there is a paucity of data concerning ICI use in this setting.
UTUC has a different behavior as compared to bladder UC [10], and while molecular alterations of urothelial bladder carcinoma have been widely studied by The Cancer Genome Atlas, data about such alterations in UTUC remain scarce [4,11,12]. However, the novel molecular insights provided by these studies led to a better understanding of this aggressive disease and provided a rationale for new therapeutic approaches.
This review summarizes the available literature regarding the use of ICIs and the biological rationale underlying their use in high-grade urothelial upper tract carcinoma management.

2. The Molecular Landscape of UTUC

Despite histological similarities, UTUC displays different molecular and genetic features as compared to bladder UC [11,13,14,15]. Bladder UC was classified into 5 molecular subtypes, according to the TCGA: luminal-papillary, luminal-infiltrated, luminal, basal/squamous, and neuronal [14,16]. However, UTUC was not included in this analysis.
In the past few years, several studies have focused on the molecular characterization of UTUC using next-generation sequencing [4,5,6,11,12,13,17,18,19,20,21,22,23,24]. The most frequently mutated genes were FGFR3 (40–80%), KMT2D (35–56%), KMT2A (32–47%), and TP53 (18–26%) [4,5,11,13,22]. Other common mutations involving oncogenes or tumor suppressor genes, such as HRAS, NRAS, KRAS, ARID1A, PIK3CA and CDKN2A were also reported in these studies. Alteration of TP53/MDM2 was associated with more aggressive disease and worse outcomes, whereas FGFR3 alterations were linked to a better prognosis [13,20]. Comparable mutation rates were seen in UTUC and bladder UC, with a strong APOBEC signature, but FGFR3, chromatin remodeling gene such as KMT2D, and CDKN2A were mutated at a higher frequency in UTUC compared in bladder UC [4]. Conversely, the frequency of TP53 mutations was lower. However, the associations between these somatic alterations and response to immunotherapy in UTUC are unclear. This is, for instance, the case for CDKN2A loss, which has been shown to be associated with decreased response to atezolizumab in bladder UC [25].
UTUC is represented in the spectrum of Lynch syndrome, an autosomal-dominant familial cancer syndrome caused by germline mutations in the DNA mismatch repair (MMR) genes MLH1, MSH2, MSH6, or PMS2 [26,27,28]. It is particularly associated with MSH2 mutations, with such mutations found in approximately 70% of UC integrated in Lynch syndrome [26,27,29,30]. Loss of function in the MMR system, either caused by an inherited mutation or a sporadic event, results in microsatellite instability (MSI) throughout the genome [31]. MSI have been found between 3.9% to 20% in UTUC as compared to <1% in UBC [32,33]. High MSI is correlated with a better prognosis, particularly in patients younger than 71 years old with T2-T3N0M0 tumors [34]. Furthermore, high MSI is associated with a higher tumor mutational burden (TMB) and correlated to a higher response rate with ICI treatment [35,36]. Recent retrospective data in advanced high MSI UTUC patients treated with immune checkpoint inhibitors have demonstrated excellent clinical activity [28].
Using a combination of whole-exome sequencing and RNA sequencing, some studies described biological differences between UTUC and bladder UC [11]. Four molecular subgroups of UTUC were defined through RNA-sequencing by Moss et al. [4]. Comparison to the TCGA dataset has revealed that cluster 1 was similar to luminal-subtype, cluster 2 was close to basal subtype, and cluster 4 demonstrated a high frequency of upregulated immune checkpoint related genes [4]. Moreover, UTUC is found to be predominantly luminal-like tumors, and, more precisely, luminal-papillary and T-cell depleted as compared to bladder UC [11,14].
Recently, we reported the UTUC methylation profiles and identified 2 epigenetic subtypes, namely EpiC-low and EpiC-high. While the earlier one was hypomethylated, immune depleted, and enriched for FGFR3 mutations, the latter was hypermethylated, immune infiltrated, and associated with SWI/SNF genes somatic mutations [12] (Figure 1). Moreover, we identified for the first time a high rate of mutations in the ZFP36 family in almost one-quarter of UTUC. Further mechanistic studies showed that ZFP36L1 loss of function experiments in urothelial cell lines led to increased cell migration and epithelial-mesenchymal transition [12]. More recently, Fujii et al. conducted a comprehensive molecular study of 198 UTUC patients through whole-exome sequencing (WES), single nucleotide polymorphism (SNP) array, RNA sequencing, and methylation analysis [37]. They reported 5 molecular subtypes (hypermutated (5.5%), TP53/MDM2 (37.7%), RAS (15.1%), FGFR3 (35.2%) and triple-negative (6.5%)) that correlates with clinicopathological features [37]. Indeed, patients classified in TP53/MDM2 and triple-negative subgroups had the worse disease-specific survival. Moreover, similar to previous studies, they found a majority of luminal-like tumors (71.5%) [37]. They also confirmed a different mutational profile between tumors arising in ureters versus renal pelvis, suggesting a distinct carcinogenesis mechanism depending on location in the urothelium [37]. They also sequenced urinary sediment-derived DNA and found 82.2% (95% CI, 71.5−90.2%) of sensitivity and 100% (95% CI, 81.5−100%) of specificity of this approach for UTUC diagnosis [37].

3. The Immune Microenvironment of UTUC

The immune landscape of tumors is an important determinant of the host’s antitumoral response and clinical outcomes [38]. According to the results of molecular profiling studies, it is well established that sporadic UTUC displays a high rate of FGFR3 mutations (40–80%) [4,5,11,13,22]. Moreover, it has also been shown that upper tract tumors with FGFR3-alterations express lower CD8 T-cell gene signatures [11,39]. In addition, FGFR3 upregulation has been shown to be important in shaping the observed T-cell depleted phenotype [11]. Indeed, genes related to IFNG pathway, such as BTS2 and IRF9, were found downregulated in T-cell depleted cluster and the use of FGFR3 inhibitors correlated with upregulation of these genes [11]. However, how FGFR3 signaling mediates IFNG related genes and a T-cell depleted microenvironment has to be determined. Recently, Rose et al. found that upregulated FGFR3 signaling significantly correlates with upregulated PPARG gene signatures [40]. They suggested it as a potential mechanism given that the upregulation of PPARG signaling appears to suppress pro-inflammatory cytokine signaling and correlates with a non-T cell-inflamed phenotype [40].
Therefore, sporadic UTUC with FGFR3 mutations might be considered immune cold tumors. Consistent with previous studies, we recently observed similar findings. In addition, we found that FGFR3 mutated UTUC were hypomethylated compared to FGFR3 wild-type tumors, suggesting crosstalk between genetic and epigenetic phenotypes of these tumors [12]. Notably, FGFR3 wild-type tumors harbored a high rate of SWI/SNF genetic tumor alterations and were associated with a higher level of tumor infiltrated lymphocytes (TILs) [12]. Further studies are needed to fully understand these observations at the mechanistic level allowing us to elucidate better how the genetic alterations of the cancer cells might shape the immune contexture. Importantly, UTUC developed in a Lynch syndrome context because of an MMR loss of function could be considered immune hot tumors [21].
Finally, correlations were observed between PD-L1 positivity on tumoral cells and the worse outcomes in UTUC patients, although PD-L1 expression rate varied between 20−25% depending on the study and the cutoff used [41,42].

4. Current Systemic Management of UTUC

4.1. Localized Disease

The gold standard treatment for high-risk disease, defined by at least one of the following criteria: hydronephrosis, tumor size ≥ 2 cm, high-grade cytology, high-grade on biopsy, multifocal disease, previous radical cystectomy for bladder cancer, and variant histology [1], the standard is radical nephro-ureterectomy (RNU) with bladder cuff excision associated with lymph node dissection, according to the European Association of Urology (EAU) guidelines [1]. Kidney-sparing surgery could be proposed for patients with low-risk disease or even in patients with high-risk tumors located in the distal ureter to reduce morbidity. This strategy could allow optimal adjuvant chemotherapy administration by preserving kidney function [1]. For locally advanced UTUC, adjuvant chemotherapy with platinum-gemcitabine combination chemotherapy is recommended.
Indeed, the POUT trial, a multi-center randomized controlled trial including 261 patients, has reported a significant improvement in disease-free survival (DFS) (hazard ratio (HR): 0.45, 95% CI 0.30–0.68; p = 0.0001) at a median follow-up of 30.3 months (IQR 18.0–47.5) with adjuvant platinum-gemcitabine combination chemotherapy beginning within 90 days following nephroureterectomy [43]. The recently updated results of this trial, with a median follow-up of 48.1 months (IQR: 36.0–60.1), confirmed the chemotherapy benefit in DFS (HR: 0.50, 95% CI: 0.34–0.75; p = 0.001) [44]. There was no detrimental long-term toxicity observed. Furthermore, no statistically significant improvement in OS was observed [44]. Unfortunately, the optimal administration of cisplatin was not feasible for some patients due to the surgical impact on renal function [45,46]. Therefore, neoadjuvant chemotherapy was proposed, with interesting results in terms of pathological downstaging and survival [47]. Moreover, there is a benefit in terms of pathological response, disease recurrence, and mortality rate compared to RNU alone, even if no randomized controlled trials have been published yet [48]. The use of ICIs in the peri-operative setting is not yet a standard of care in UTUC management [1]. Recently published data about the use of ICI in this setting are discussed below.

4.2. Metastatic Disease

The management of the metastatic disease should be multimodal, with the use of local therapy in the case of oligometastatic disease, even in the absence of a randomized controlled trial [1,49]. RNU could also be performed in the oligometastatic situation, although providing only a small benefit [50,51,52,53], or in the palliative setting to manage symptomatic disease [1,54].
In the metastatic setting, systemic chemotherapy is extrapolated from data collected in metastatic bladder UC, since few data are available for advanced UTUC [1]. Indeed, cisplatin-based regimens, such as MVAC (methotrexate, vinblastine, doxorubicin, and cisplatin), or GC (gemcitabine and cisplatin) remained standard of care in first-line. Furthermore, a recent retrospective study found that the location of the primary tumor (upper vs. lower tract) did not change the progression-free survival (PFS) and overall survival (OS) in patients treated with platinum-based chemotherapy for metastatic UC [52]. Immune checkpoint inhibitors are also part of the therapeutic armamentarium.
Recently, avelumab as first-line maintenance therapy for platinum-based chemotherapy responder or stable disease became the standard of care [55]. The JAVELIN-100 trial is a randomized controlled phase 3 trial comparing avelumab versus placebo as maintenance therapy after platinum-based chemotherapy in first-line for patients with metastatic UC. A total of 700 patients were randomized, 30.3% (n = 106/350) versus 23.1% (n = 81/350) of UTUC patients were, respectively, in the avelumab and placebo arm. Median overall survival was significantly longer in the avelumab arm, 21.4 (18.9–26.1) versus 14.3 (12.9–17.9) hazard ratio for death 0.69 (0.56–0.86) p = 0.001 [55]. Based on subgroup analysis, the second-line systemic standard of care includes ICI, such as pembrolizumab. Indeed, in the phase 3 trial KEYNOTE-045, 542 UC patients were randomly assigned between pembrolizumab or chemotherapy chosen by the investigator. There was 14.1% (n = 38/270) UTUC patients in the pembrolizumab arm versus 13.6% (n = 37/272) in the chemotherapy arm. In the total population, median overall survival was 10.3 months (95% CI, 8.0 to 11.8) versus 7.4 months (95% CI, 6.1 to 8.3) in the chemotherapy group (HR, 0.73; 95% CI, 0.59 to 0.91; p = 0.002) [56,57]. Nevertheless, based on subgroup analysis, a systemic chemotherapy regimen with a single agent such as paclitaxel, docetaxel, or vinflunine could still be proposed [58].

5. Immune Checkpoint Inhibition in UTUC

Given their relative rarity, there are no studies specifically focusing on UTUC. Therefore, data related to ICI efficacy are extracted from a larger cohort of patients with UC that included a small subgroup of UTUC.

5.1. Immune Checkpoint Inhibitors in the Perioperative Setting

In the adjuvant setting, ICI-based therapy has been widely tested since the role of adjuvant treatment in high-risk muscle-invasive urothelial carcinoma after radical surgery was not clear.
Since the first promising results of adjuvant pembrolizumab in the management of UC [59,60], several studies have now included UTUC patients in ICI-based adjuvant treatment (Table 1).
The IMvigor 010 study enrolled 809 high-risk UC patients to be randomized between adjuvant atezolizumab versus placebo. There were only 7% of UTUC patients in the atezolizumab arm as compared to 6% in the placebo arm (Table 1). There was no statistical difference in terms of median disease-free survival, 19.4 months (95% CI 15.9–24.8) with atezolizumab and 16.6 months (11.2–24.8) with observation (stratified hazard ratio 0.89 [95% CI 0.74–1.08]; p = 0.24) [61]. More recently, the data of the Checkmate 274 trial were reported. It was a phase 3 trial, including 709 patients randomized between adjuvant nivolumab versus placebo. A total of 21% of enrolled patients were patients with UTUC (Table 1). However, based on the results of the POUT trial, the inclusion of UTUC patients was prematurely interrupted. In the intention-to-treat population, median disease-free survival was 20.8 months (95% CI, 16.5 to 27.6) with nivolumab and 10.8 months (95% CI, 8.3 to 13.9) with placebo; (HR, 0.70; 98.22% CI, 0.55 to 0.90; p < 0.001). For the UTUC subgroup, the HR for disease recurrence or death were 1.23 (CI 95% 0.67–2.23) and 1.56 (CI 95% 0.7–3.48) for UTUC arising in renal pelvis and ureter, respectively. The percentage of patients was 74.5% and 55.7%, respectively (hazard ratio, 0.55; 98.72% CI, 0.35 to 0.85; p < 0.001), for those expressing PD-L1 more than 1% [62]. Moreover, several neoadjuvant trials combining chemotherapy with ICI are actively recruiting, but available data are currently limited [63,64,65].

5.2. Immune Checkpoint Inhibitors in the Metastatic Setting

In the metastatic setting, ICI are widely used in the management of UC (Table 2). Indeed, avelumab as maintenance therapy after platinum-based chemotherapy is currently the standard of care according to the results of the JAVELIN-100 trial [55]. For cisplatin-ineligible UC patients, based on phase 2 trials IMvigor 210 and KEYNOTE 052 provided interesting results for the use of ICI in this frail population (Table 2) [66,67]. The overall response rate for UTUC patients was 39% with atezolizumab [66] and 22% with pembrolizumab in monotherapy in this setting [67].
The IMvigor 130 trial, a randomized phase 3 trial, showed significant PFS improvement of the addition of atezolizumab to platinum-based chemotherapy [68] (Table 2). Specific outcomes of UTUC patients were not assessed. However, unfortunately, atezolizumab in monotherapy failed to improve overall survival compared to chemotherapy in pretreated metastatic UC [70]. In the same manner, the addition of pembrolizumab to first-line platinum-based chemotherapy was not associated with a survival benefit compared to chemotherapy alone (Table 2) [69].
In the second or later line, atezolizumab, durvalumab, avelumab, nivolumab, and pembrolizumab have been demonstrated safe and efficient in platinum pretreated UC population. However, UTUC patients’ data remained scarce (Table 2).

6. Perspectives in UTUC Management

According to the recent advances in the molecular characterization of UTUC, there is a rationale to develop new treatment combinations. Indeed, given the high prevalence of FGFR3 mutations and their association with a T-cell depleted phenotype in UTUC, there is a rationale for combining ICI with FGFR3 inhibitors (Figure 1). Erdafitinib, a pan-FGFR inhibitor, is now approved based on the results of the phase 2 trial in metastatic bladder cancer, with a 40% of response rate in patients with FGFR actionable alterations [71]. Moreover, Ding et al. reported the case of a 67 years old metastatic, chemo-refractory UTUC’s patient having a dramatic response to pembrolizumab in association with erdafitinib [72]. However, reliable response biomarkers are still needed to improve precision medicine in urothelial carcinoma. The ongoing trials assessing immune checkpoint inhibitors-based combinations therapies in UTUC metastatic setting are reported in Table 3. They often include backbone ICI in combination with chemotherapy, antibody-drug conjugates, and tyrosine kinase inhibitors.

7. Conclusions

ICI inhibitors are now widely used in daily practice to treat urothelial carcinoma patients. Based on the recent advancement in the comprehension of the molecular biology of UTUC and the differences between bladder UC and UTUC, further studies focused on UTUC patients are needed to personalize the therapeutic approach and find new treatment combinations.

Author Contributions

Conceptualization, J.T., O.A., N.M.C., P.B., N.M.T. and G.G.M.; methodology, J.T. and G.G.M.; validation, P.B., G.G.M. and N.M.C.; writing—original draft preparation, J.T.; writing—review and editing, J.T., O.A., N.M.C., H.L., P.B., N.M.T. and G.G.M.; visualization, J.T., P.B. and G.G.M.; supervision, N.M.T. and G.G.M.; project administration, G.G.M.; All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

J.T. declares no conflict of interest related to this manuscript, N.M.C. reports support for research travel from Pfizer, Janssen and Ipsen, and consulting fees for BMS, Pfizer, Sanofi and Bayer, O.A. declares no conflict of interest related to this manuscript, H.L. declares no conflict of interest related to this manuscript, N.M.T. reports grants from Bristol Myers Squibb during the conduct of the study; grants and personal fees from Exelixisa and Nektar Therapeutics; grants from Calithera Bioscience, Takeda, Arrowhead Pharmaceuticals, and Epizyme, Inc.; personal fees from Bristol Myers Squibb, Pfizer, Lilly Oncology, Neolukin Therapeutics, Eisai, Novartis, Oncorena, Ono Pharmaceutical, Surface Oncology, and Ipsen outside the submitted work, P.B. reports other from BMS (consulting, travel); other from MSD (consulting, travel), Pfizer (consulting, travel), Ipsen (consulting, travel), Janssen Cilag (consulting, travel), AstraZeneca (consulting), Amgen (consulting, travel), Novartis (consulting), Eusapharma (consulting), Roche (consulting, travel), Esai (consulting, travel), Merck (consulting, travel) and Astellas (consulting, travel) outside the submitted work, G.G.M. reports consulting fees from Pfizer, Ipsen, BMS, Astellas; received travel support from Pfizer, Ipsen, BMS, MSD and Astellas outside of the submitted work.

References

  1. Rouprêt, M.; Babjuk, M.; Burger, M.; Capoun, O.; Cohen, D.; Compérat, E.M.; Cowan, N.C.; Dominguez-Escrig, J.L.; Gontero, P.; Mostafid, A.H.; et al. European association of urology guidelines on upper urinary tract urothelial carcinoma: 2020 update. Eur. Urol. 2020, 79, 62–79. [Google Scholar] [CrossRef] [PubMed]
  2. Sylvester, R.J.; Rodríguez, O.; Hernández, V.; Turturica, D.; Bauerová, L.; Bruins, H.M.; Bründl, J.; van der Kwast, T.H.; Brisuda, A.; Rubio-Briones, J.; et al. European association of urology (EAU) prognostic factor risk groups for non–muscle-invasive bladder cancer (NMIBC) incorporating the WHO 2004/2016 and WHO 1973 classification systems for grade: An update from the EAU NMIBC guidelines panel. Eur. Urol. 2021, 79, 480–488. [Google Scholar] [CrossRef] [PubMed]
  3. Bersanelli, M.; Buti, S.; Giannatempo, P.; Raggi, D.; Necchi, A.; Leonetti, A.; Banna, G.L.; Petrelli, F. Outcome of patients with advanced upper tract urothelial carcinoma treated with immune checkpoint inhibitors: A systematic review and meta-analysis. Crit. Rev. Oncol. 2021, 159, 103241. [Google Scholar] [CrossRef] [PubMed]
  4. Moss, T.J.; Qi, Y.; Xi, L.; Peng, B.; Kim, T.-B.; Ezzedine, N.E.; Mosqueda, M.E.; Guo, C.C.; Czerniak, B.A.; Ittmann, M.; et al. Comprehensive genomic characterization of upper tract urothelial carcinoma. Eur. Urol. 2017, 72, 641–649. [Google Scholar] [CrossRef]
  5. Sfakianos, J.P.; Cha, E.K.; Iyer, G.; Scott, S.N.; Zabor, E.C.; Shah, R.; Ren, Q.; Bagrodia, A.; Kim, P.H.; Hakimi, A.A.; et al. Genomic characterization of upper tract urothelial carcinoma. Eur. Urol. 2015, 68, 970–977. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Lee, J.Y.; Kim, K.; Sung, H.H.; Jeon, H.G.; Jeong, B.C.; Seo, S.I.; Jeon, S.S.; Lee, H.M.; Choi, H.-Y.; Kwon, G.-Y.; et al. Molecular characterization of urothelial carcinoma of the bladder and upper urinary tract. Transl. Oncol. 2017, 11, 37–42. [Google Scholar] [CrossRef] [PubMed]
  7. Margulis, V.; Shariat, S.F.; Matin, S.F.; Kamat, A.M.; Zigeuner, R.; Kikuchi, E.; Lotan, Y.; Weizer, A.; Raman, J.; Wood, C.G.; et al. Outcomes of radical nephroureterectomy: A series from the upper tract urothelial carcinoma collaboration. Cancer 2009, 115, 1224–1233. [Google Scholar] [CrossRef] [PubMed]
  8. Leow, J.J.; Chong, K.T.; Chang, S.L.; Bellmunt, J. Upper tract urothelial carcinoma: A different disease entity in terms of management. ESMO Open 2016, 1, e000126. [Google Scholar] [CrossRef] [PubMed]
  9. Witjes, J.A.; Bruins, H.M.; Cathomas, R.; Compérat, E.M.; Cowan, N.C.; Gakis, G.; Hernández, V.; Espinós, E.L.; Lorch, A.; Neuzillet, Y.; et al. European association of urology guidelines on muscle-invasive and metastatic bladder cancer: Summary of the 2020 guidelines. Eur. Urol. 2020, 79, 82–104. [Google Scholar] [CrossRef] [PubMed]
  10. Green, D.A.; Rink, M.; Xylinas, E.; Matin, S.F.; Stenzl, A.; Roupret, M.; Karakiewicz, P.I.; Scherr, D.; Shariat, S.F. Urothelial carcinoma of the bladder and the upper tract: Disparate twins. J. Urol. 2012, 189, 1214–1221. [Google Scholar] [CrossRef] [PubMed]
  11. Robinson, B.D.; Vlachostergios, P.; Bhinder, B.; Liu, W.; Li, K.; Moss, T.J.; Bareja, R.; Park, K.; Tavassoli, P.; Cyrta, J.; et al. Upper tract urothelial carcinoma has a luminal-papillary T-cell depleted contexture and activated FGFR3 signaling. Nat. Commun. 2019, 10, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Su, X.; Lu, X.; Bazai, S.K.; Compérat, E.; Mouawad, R.; Yao, H.; Rouprêt, M.; Spano, J.-P.; Khayat, D.; Davidson, I.; et al. Comprehensive integrative profiling of upper tract urothelial carcinomas. Genome Biol. 2021, 22, 1–25. [Google Scholar] [CrossRef] [PubMed]
  13. Audenet, F.; Isharwal, S.; Cha, E.K.; Donoghue, M.T.A.; Drill, E.N.; Ostrovnaya, I.; Pietzak, E.J.; Sfakianos, J.P.; Bagrodia, A.; Murugan, P.; et al. Clonal relatedness and mutational differences between upper tract and bladder urothelial carcinoma. Clin. Cancer Res. 2018, 25, 967–976. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Robertson, A.G.; Kim, J.; Al-Ahmadie, H.; Bellmunt, J.; Guo, G.; Cherniack, A.D.; Hinoue, T.; Laird, P.W.; Hoadley, K.; Akbani, R.; et al. Comprehensive molecular characterization of muscle-invasive bladder cancer. Cell 2017, 171, 540–556.e25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Atlas, N.C.G. The cancer genome atlas research network comprehensive molecular characterization of urothelial bladder carcinoma. Nature 2014, 507, 315–322. [Google Scholar] [CrossRef] [Green Version]
  16. Kamoun, A.; de Reyniès, A.; Allory, Y.; Sjödahl, G.; Robertson, A.G.; Seiler, R.; Hoadley, K.A.; Groeneveld, C.S.; Al-Ahmadie, H.; Choi, W.; et al. A consensus molecular classification of muscle-invasive bladder cancer. Eur. Urol. 2019, 77, 420–433. [Google Scholar] [CrossRef] [PubMed]
  17. Hassler, M.R.; Bray, F.; Catto, J.W.; Grollman, A.P.; Hartmann, A.; Margulis, V.; Matin, S.F.; Roupret, M.; Sfakianos, J.P.; Shariat, S.F.; et al. Molecular characterization of upper tract urothelial carcinoma in the era of next-generation sequencing: A systematic review of the current literature. Eur. Urol. 2020, 78, 209–220. [Google Scholar] [CrossRef] [PubMed]
  18. Necchi, A.; Madison, R.; Pal, S.K.; Ross, J.S.; Agarwal, N.; Sonpavde, G.; Joshi, M.; Yin, M.; Miller, V.A.; Grivas, P.; et al. Comprehensive genomic profiling of upper-tract and bladder urothelial carcinoma. Eur. Urol. Focus 2020, in press. [Google Scholar] [CrossRef]
  19. Bagrodia, A.; Cha, E.K.; Sfakianos, J.P.; Zabor, E.C.; Bochner, B.; Al-Ahmadie, H.A.; Solit, D.B.; Coleman, J.; Iyer, G.; Scott, S.N.; et al. Genomic biomarkers for the prediction of stage and prognosis of upper tract urothelial carcinoma. J. Urol. 2016, 195, 1684–1689. [Google Scholar] [CrossRef] [Green Version]
  20. Bagrodia, A.; Audenet, F.; Pietzak, E.J.; Kim, K.; Murray, K.S.; Cha, E.K.; Sfakianos, J.P.; Iyer, G.; Singla, N.; Arcila, M.; et al. Genomic profile of urothelial carcinoma of the upper tract from ureteroscopic biopsy: Feasibility and validation using matched radical nephroureterectomy specimens. Eur. Urol. Focus 2018, 5, 365–368. [Google Scholar] [CrossRef] [PubMed]
  21. Donahue, T.F.; Bagrodia, A.; Audenet, F.; Donoghue, M.T.; Cha, E.K.; Sfakianos, J.P.; Sperling, D.; Al-Ahmadie, H.; Clendenning, M.; Rosty, C.; et al. Genomic characterization of upper-tract urothelial carcinoma in patients with lynch syndrome. JCO Precis. Oncol. 2018, 2018, 1–13. [Google Scholar] [CrossRef] [PubMed]
  22. Nassar, A.H.; Umeton, R.; Kim, J.; Lundgren, K.; Harshman, L.; Van Allen, E.M.; Preston, M.; Dong, F.; Bellmunt, J.; Mouw, K.W.; et al. Mutational analysis of 472 urothelial carcinoma across grades and anatomic sites. Clin. Cancer Res. 2018, 25, 2458–2470. [Google Scholar] [CrossRef] [PubMed]
  23. Hoang, M.L.; Chen, C.-H.; Sidorenko, V.S.; He, J.; Dickman, K.G.; Yun, B.H.; Moriya, M.; Niknafs, N.; Douville, C.; Karchin, R.; et al. Mutational signature of aristolochic acid exposure as revealed by whole-exome sequencing. Sci. Transl. Med. 2013, 5, 197ra102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Castells, X.; Karanović, S.; Ardin, M.; Tomić, K.; Xylinas, E.; Durand, G.; Villar, S.; Forey, N.; Le Calvez-Kelm, F.; Voegele, C.; et al. Low-coverage exome sequencing screen in formalin-fixed paraffin-embedded tumors reveals evidence of exposure to carcinogenic aristolochic acid. Cancer Epidemiol. Biomark. Prev. 2015, 24, 1873–1881. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Banchereau, R.; Leng, N.; Zill, O.; Sokol, E.; Liu, G.; Pavlick, D.; Maund, S.; Liu, L.-F.; Kadel, E.; Baldwin, N.; et al. Molecular determinants of response to PD-L1 blockade across tumor types. Nat. Commun. 2021, 12, 1–11. [Google Scholar] [CrossRef] [PubMed]
  26. Engel, C.; Loeffler, M.; Steinke, V.; Rahner, N.; Holinski-Feder, E.; Dietmaier, W.; Schackert, H.K.; Goergens, H.; Doeberitz, M.V.K.; Goecke, T.O.; et al. Risks of less common cancers in proven mutation carriers with lynch syndrome. J. Clin. Oncol. 2012, 30, 4409–4415. [Google Scholar] [CrossRef]
  27. Van Der Post, R.S.; Kiemeney, L.; Ligtenberg, M.J.L.; Witjes, J.A.; De Kaa, C.A.H.-V.; Bodmer, D.; Schaap, L.; Kets, C.M.; Van Krieken, J.H.J.M.; Hoogerbrugge, N. Risk of urothelial bladder cancer in Lynch syndrome is increased, in particular among MSH2 mutation carriers. J. Med. Genet. 2010, 47, 464–470. [Google Scholar] [CrossRef] [PubMed]
  28. Watson, P.; Vasen, H.; Mecklin, J.-P.; Bernstein, I.; Aarnio, M.; Järvinen, H.J.; Myrhøj, T.; Sunde, L.; Wijnen, J.T.; Lynch, H.T. The risk of extra-colonic, extra-endometrial cancer in the lynch syndrome. Int. J. Cancer 2008, 123, 444–449. [Google Scholar] [CrossRef] [Green Version]
  29. Joost, P.; Therkildsen, C.; Dominguez-Valentin, M.; Jönsson, M.; Nilbert, M. Urinary tract cancer in lynch syndrome; increased risk in carriers of MSH2 mutations. Urology 2015, 86, 1212–1217. [Google Scholar] [CrossRef]
  30. Andreev-Drakhlin, A.; Shah, A.Y.; Adriazola, A.C.; Shaw, L.; Lopez, L.; James, M.; Matin, S.F.; Alhalabi, O.; Gao, J.; Siefker-Radtke, A.O.; et al. Efficacy of immune checkpoint blockade in patients with advanced upper tract urothelial cancer and mismatch repair deficiency or microsatellite instability (MSI). J. Clin. Oncol. 2021, 39, 487. [Google Scholar] [CrossRef]
  31. Umar, A.; Boland, C.R.; Terdiman, J.P.; Syngal, S.; de la Chapelle, A.; Rüschoff, J.; Fishel, R.; Lindor, N.M.; Burgart, L.J.; Hamelin, R.; et al. Revised Bethesda guidelines for hereditary nonpolyposis colorectal cancer (lynch syndrome) and microsatellite instability. J. Natl. Cancer Inst. 2004, 96, 261–268. [Google Scholar] [CrossRef] [PubMed]
  32. Rao, A.; McGrath, J.E.; Xiu, J.; de Souza, A.L.; Gulati, S.; Abuali, I.; Sagaram, S.; Nabhan, C.; Korn, W.M.; Ryan, C.J.; et al. Characterization of microsatellite instability (dMMR/MSI-H) and mutational landscape in a large contemporary cohort of upper tract urothelial cancer (UTUC) patients. J. Clin. Oncol. 2021, 39, 465. [Google Scholar] [CrossRef]
  33. Bonneville, R.; Krook, M.A.; Kautto, E.; Miya, J.; Wing, M.R.; Chen, H.-Z.; Reeser, J.W.; Yu, L.; Roychowdhury, S. Landscape of microsatellite instability across 39 cancer types. JCO Precis. Oncol. 2017, 1, 1–15. [Google Scholar] [CrossRef] [PubMed]
  34. Rouprêt, M.; Fromont, G.; Azzouzi, A.-R.; Catto, J.W.; Vallancien, G.; Hamdy, F.C.; Cussenot, O. Microsatellite instability as predictor of survival in patients with invasive upper urinary tract transitional cell carcinoma. Urology 2005, 65, 1233–1237. [Google Scholar] [CrossRef] [PubMed]
  35. Luchini, C.; Bibeau, F.; Ligtenberg, M.; Singh, N.; Nottegar, A.; Bosse, T.; Miller, R.; Riaz, N.; Douillard, J.-Y.; Andre, F.; et al. ESMO recommendations on microsatellite instability testing for immunotherapy in cancer, and its relationship with PD-1/PD-L1 expression and tumour mutational burden: A systematic review-based approach. Ann. Oncol. 2019, 30, 1232–1243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Le, D.T.; Uram, J.N.; Wang, H.; Bartlett, B.; Kemberling, H.; Eyring, A.D.; Skora, A.D.; Luber, B.S.; Azad, N.S.; Laheru, D.; et al. PD-1 blockade in tumors with mismatch-repair deficiency. N. Engl. J. Med. 2015, 372, 2509–2520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Fujii, Y.; Sato, Y.; Suzuki, H.; Kakiuchi, N.; Yoshizato, T.; Lenis, A.T.; Maekawa, S.; Yokoyama, A.; Takeuchi, Y.; Inoue, Y.; et al. Molecular classification and diagnostics of upper urinary tract urothelial carcinoma. Cancer Cell 2021, 39, 793–809.e8. [Google Scholar] [CrossRef]
  38. Wellenstein, M.D.; De Visser, K.E. Cancer-cell-intrinsic mechanisms shaping the tumor immune landscape. Immunity 2018, 48, 399–416. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Wang, L.; Gong, Y.; Saci, A.; Szabo, P.M.; Martini, A.; Necchi, A.; Siefker-Radtke, A.; Pal, S.; Plimack, E.R.; Sfakianos, J.P.; et al. Fibroblast growth factor receptor 3 alterations and response to PD-1/PD-L1 blockade in patients with metastatic urothelial cancer. Eur. Urol. 2019, 76, 599–603. [Google Scholar] [CrossRef] [PubMed]
  40. Rose, T.L.; Weir, W.H.; Mayhew, G.M.; Shibata, Y.; Eulitt, P.; Uronis, J.M.; Zhou, M.; Nielsen, M.; Smith, A.B.; Woods, M.; et al. Fibroblast growth factor receptor 3 alterations and response to immune checkpoint inhibition in metastatic urothelial cancer: A real world experience. Br. J. Cancer 2021, 125, 1–10. [Google Scholar] [CrossRef]
  41. Chen, C.-H.; Tsai, M.-Y.; Chiang, P.-C.; Sung, M.-T.; Luo, H.-L.; Suen, J.-L.; Tsai, E.-M.; Chiang, P.-H. Prognostic value of PD-L1 combined positive score in patients with upper tract urothelial carcinoma. Cancer Immunol. Immunother. 2021, 70, 1–10. [Google Scholar] [CrossRef]
  42. Lu, Y.; Kang, J.; Luo, Z.; Song, Y.; Tian, J.; Li, Z.; Wang, X.; Liu, L.; Yang, Y.; Liu, X. The prevalence and prognostic role of PD-L1 in upper tract urothelial carcinoma patients underwent radical nephroureterectomy: A systematic review and meta-analysis. Front. Oncol. 2020, 10, 1400. [Google Scholar] [CrossRef] [PubMed]
  43. Birtle, A.; Johnson, M.; Chester, J.; Jones, R.; Dolling, D.; Bryan, R.; Harris, C.; Winterbottom, A.; Blacker, A.; Catto, J.W.F.; et al. Adjuvant chemotherapy in upper tract urothelial carcinoma (the POUT trial): A phase 3, open-label, randomised controlled trial. Lancet 2020, 395, 1268–1277. [Google Scholar] [CrossRef]
  44. Birtle, A.J.; Chester, J.D.; Jones, R.J.; Jenkins, B.; Johnson, M.; Catto, J.W.; Powles, T.; Bryan, R.T.; Blacker, A.; Chakraborti, P.R.; et al. Updated outcomes of POUT: A phase III randomized trial of peri-operative chemotherapy versus surveillance in upper tract urothelial cancer (UTUC). J. Clin. Oncol. 2021, 39, 455. [Google Scholar] [CrossRef]
  45. Lane, B.R.; Smith, A.K.; Larson, B.T.; Gong, M.C.; Campbell, S.C.; Raghavan, D.; Dreicer, R.; Hansel, D.E.; Stephenson, A.J. Chronic kidney disease after nephroureterectomy for upper tract urothelial carcinoma and implications for the administration of perioperative chemotherapy. Cancer 2010, 116, 2967–2973. [Google Scholar] [CrossRef]
  46. Kaag, M.G.; O’Malley, R.L.; O’Malley, P.; Godoy, G.; Chen, M.; Smaldone, M.C.; Hrebinko, R.L.; Raman, J.D.; Bochner, B.; Dalbagni, G.; et al. Changes in renal function following nephroureterectomy may affect the use of perioperative chemotherapy. Eur. Urol. 2010, 58, 581–587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Zennami, K.; Sumitomo, M.; Takahara, K.; Nukaya, T.; Takenaka, M.; Fukaya, K.; Ichino, M.; Fukami, N.; Sasaki, H.; Kusaka, M.; et al. Two cycles of neoadjuvant chemotherapy improves survival in patients with high-risk upper tract urothelial carcinoma. BJU Int. 2020, 127, 332–339. [Google Scholar] [CrossRef] [PubMed]
  48. Leow, J.J.; Chong, Y.L.; Chang, S.L.; Valderrama, B.P.; 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. [Google Scholar] [CrossRef] [PubMed]
  49. Paciotti, M.; Nguyen, D.-D.; Modonutti, D.; Haeuser, L.; Lipsitz, S.; Mossanen, M.; Kibel, A.S.; Lughezzani, G.; Trinh, Q.-D.; Cole, A.P. Impact of high-intensity local treatment on overall survival in stage IV upper tract urothelial carcinoma. Urol. Oncol. Semin. Orig. Investig. 2021, 39, 436.e1–436.e10. [Google Scholar] [CrossRef]
  50. Dong, F.; Fu, H.; Shi, X.; Shen, Y.; Xu, T.; Gao, F.; Wang, X.; Zhong, S.; Ding, Q.; Shen, Z.; et al. How do organ-specific metastases affect prognosis and surgical treatment for patients with metastatic upper tract urothelial carcinoma: First evidence from population based data. Clin. Exp. Metastasis 2017, 34, 467–477. [Google Scholar] [CrossRef] [PubMed]
  51. Seisen, T.; Jindal, T.; Karabon, P.; Sood, A.; Bellmunt, J.; Rouprêt, M.; Leow, J.J.; Vetterlein, M.; Sun, M.; Alanee, S.; et al. Efficacy of Systemic Chemotherapy Plus Radical Nephroureterectomy for Metastatic Upper Tract Urothelial Carcinoma. Eur. Urol. 2016, 71, 714–718. [Google Scholar] [CrossRef]
  52. Moschini, M.; Xylinas, E.; Zamboni, S.; Mattei, A.; Niegisch, G.; Yu, E.Y.; Bamias, A.; Agarwal, N.; Sridhar, S.S.; Sternberg, C.N.; et al. Efficacy of surgery in the primary tumor site for metastatic urothelial cancer: Analysis of an international, multicenter, multidisciplinary database. Eur. Urol. Oncol. 2019, 3, 94–101. [Google Scholar] [CrossRef] [PubMed]
  53. Nazzani, S.; Preisser, F.; Mazzone, E.; Marchioni, M.; Bandini, M.; Tian, Z.; Mistretta, F.; Shariat, S.; Soulières, D.; Saad, F.; et al. Survival effect of nephroureterectomy in metastatic upper urinary tract urothelial carcinoma. Clin. Genitourin. Cancer 2019, 17, e602–e611. [Google Scholar] [CrossRef] [PubMed]
  54. Leow, J.; Liu, Z.; Tan, T.W.; Lee, Y.M.; Yeo, E.K.; Chong, Y.-L. Optimal management of upper tract urothelial carcinoma: Current perspectives. OncoTargets Ther. 2020, 13, 1–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Powles, T.; Park, S.H.; Voog, E.; Caserta, C.; Valderrama, B.P.; Gurney, H.; Kalofonos, H.; Radulović, S.; Demey, W.; Ullén, A.; et al. Avelumab Maintenance Therapy for Advanced or Metastatic Urothelial Carcinoma. N. Engl. J. Med. 2020, 383, 1218–1230. [Google Scholar] [CrossRef] [PubMed]
  56. Bellmunt, J.; De Wit, R.; Vaughn, D.J.; Fradet, Y.; Lee, J.-L.; Fong, L.; Vogelzang, N.J.; Climent, M.A.; Petrylak, D.P.; Choueiri, T.K.; et al. Pembrolizumab as second-line therapy for advanced urothelial carcinoma. N. Engl. J. Med. 2017, 376, 1015–1026. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Fradet, Y.; Bellmunt, J.; Vaughn, D.; Lee, J.; Fong, L.; Vogelzang, N.; Climent, M.; Petrylak, D.; Choueiri, T.; Necchi, A.; 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, 970–976. [Google Scholar] [CrossRef] [PubMed]
  58. Raggi, D.; Miceli, R.; Sonpavde, G.; Giannatempo, P.; Mariani, L.; Galsky, M.D.; Bellmunt, J.; Necchi, A. Second-line single-agent versus doublet chemotherapy as salvage therapy for metastatic urothelial cancer: A systematic review and meta-analysis. Ann. Oncol. 2016, 27, 49–61. [Google Scholar] [CrossRef]
  59. 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, 36, 3353–3360. [Google Scholar] [CrossRef] [Green Version]
  60. Necchi, A.; Raggi, D.; Gallina, A.; Madison, R.; Colecchia, M.; Lucianò, R.; Montironi, R.; Giannatempo, P.; Farè, E.; Pederzoli, F.; et al. Updated results of PURE-01 with preliminary activity of neoadjuvant pembrolizumab in patients with muscle-invasive bladder carcinoma with variant histologies. Eur. Urol. 2020, 77, 439–446. [Google Scholar] [CrossRef]
  61. Bellmunt, J.; Hussain, M.; Gschwend, J.E.; Albers, P.; Oudard, S.; Castellano, D.; Daneshmand, S.; Nishiyama, H.; Majchrowicz, M.; Degaonkar, V.; et al. Adjuvant atezolizumab versus observation in muscle-invasive urothelial carcinoma (IMvigor010): A multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2021, 22, 525–537. [Google Scholar] [CrossRef]
  62. Bajorin, D.F.; Witjes, J.A.; Gschwend, J.E.; Schenker, M.; Valderrama, B.P.; Tomita, Y.; Bamias, A.; Lebret, T.; Shariat, S.F.; Park, S.H.; et al. Adjuvant nivolumab versus placebo in muscle-invasive urothelial carcinoma. N. Engl. J. Med. 2021, 384, 2102–2114. [Google Scholar] [CrossRef] [PubMed]
  63. Rose, T.L.; Harrison, M.R.; Deal, A.M.; Osterman, C.K.; Ramalingam, S.; Whang, Y.E.; Brower, B.Y.; Bjurlin, M.; Smith, A.B.; Nielsen, M.E.; et al. Phase II study of gemcitabine and split-dose cisplatin plus pembrolizumab as neoadjuvant therapy prior to radical cystectomy (RC) in patients with muscle-invasive bladder cancer (MIBC). J. Clin. Oncol. 2021, 39, 396. [Google Scholar] [CrossRef]
  64. Lopez-Beltran, A.; Cimadamore, A.; Blanca, A.; Massari, F.; Vau, N.; Scarpelli, M.; Cheng, L.; Montironi, R. Immune checkpoint inhibitors for the treatment of bladder cancer. Cancers 2021, 13, 131. [Google Scholar] [CrossRef] [PubMed]
  65. Califano, G.; Ouzaid, I.; Verze, P.; Hermieu, J.-F.; Mirone, V.; Xylinas, E. Immune checkpoint inhibition in upper tract urothelial carcinoma. World J. Urol. 2020, 39, 1357–1367. [Google Scholar] [CrossRef] [PubMed]
  66. Balar, A.; Galsky, M.D.; Rosenberg, J.E.; Powles, T.; Petrylak, D.P.; Bellmunt, J.; Loriot, Y.; Necchi, A.; Hoffman-Censits, J.; Perez-Gracia, J.L.; 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 2016, 389, 67–76. [Google Scholar] [CrossRef] [Green Version]
  67. Balar, A.V.; Castellano, D.; O’Donnell, P.H.; Grivas, P.; Vuky, J.; Powles, T.; Plimack, E.R.; Hahn, N.M.; de Wit, R.; Pang, L.; et al. First-line pembrolizumab in cisplatin-ineligible patients with locally advanced and unresectable or metastatic urothelial cancer (KEYNOTE-052): A multicentre, single-arm, phase 2 study. Lancet Oncol. 2017, 18, 1483–1492. [Google Scholar] [CrossRef]
  68. Galsky, M.D.; Arija, J.A.; Bamias, A.; Davis, I.D.; De Santis, M.; Kikuchi, E.; Garcia-Del-Muro, X.; De Giorgi, U.; Mencinger, M.; Izumi, K.; et al. Atezolizumab with or without chemotherapy in metastatic urothelial cancer (IMvigor130): A multicentre, randomised, placebo-controlled phase 3 trial. Lancet 2020, 395, 1547–1557. [Google Scholar] [CrossRef]
  69. Powles, T.; Csőszi, T.; Özgüroğlu, M.; Matsubara, N.; Géczi, L.; Cheng, S.Y.-S.; Fradet, Y.; Oudard, S.; Vulsteke, C.; Barrera, R.M.; et al. Pembrolizumab alone or combined with chemotherapy versus chemotherapy as first-line therapy for advanced urothelial carcinoma (KEYNOTE-361): A randomised, open-label, phase 3 trial. Lancet Oncol. 2021, 22, 931–945. [Google Scholar] [CrossRef]
  70. Powles, T.; Durán, I.; van der Heijden, M.S.; Loriot, Y.; Vogelzang, N.J.; De Giorgi, U.; Oudard, S.; Retz, M.M.; Castellano, D.; Bamias, A.; et al. Atezolizumab versus chemotherapy in patients with platinum-treated locally advanced or metastatic urothelial carcinoma (IMvigor211): A multicentre, open-label, phase 3 randomised controlled trial. Lancet 2018, 391, 748–757. [Google Scholar] [CrossRef]
  71. Loriot, Y.; Necchi, A.; Park, S.H.; Garcia-Donas, J.; Huddart, R.; Burgess, E.; Fleming, M.; Rezazadeh, A.; Mellado, B.; Varlamov, S.; et al. Erdafitinib in locally advanced or metastatic urothelial carcinoma. N. Engl. J. Med. 2019, 381, 338–348. [Google Scholar] [CrossRef]
  72. Ding, X.; Zong, J.; Li, X.; Bai, X.; Tan, B.; Sun, W.; Wang, R.; Ding, Y. Dramatic responses of recurrent upper urinary tract urothelial carcinoma harboring FGFR3 and TP53 activating mutations to pembrolizumab in combination with erdafitinib: A case report. OncoTargets Ther. 2021, 14, 2177–2183. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Proposal of molecular subtypes classification of upper-tract urothelial carcinomas adapted from Su et al. [12]. Broadly, upper-tract urothelial carcinomas can be divided into two subtypes, namely EpiC-high and EpiC-low. Epic-low subtype is hypomethylated, immune-desert, and characterized by FGFR3 somatic mutations with potential efficacy of the combination of FGFR3 immunotherapy and immune checkpoint inhibitors (ICI). Conversely, EpiC-high subtype is hypermethylated, immune-inflamed, and enriched with somatic mutations of SWI/SNF genes with potential benefit for ICI.
Figure 1. Proposal of molecular subtypes classification of upper-tract urothelial carcinomas adapted from Su et al. [12]. Broadly, upper-tract urothelial carcinomas can be divided into two subtypes, namely EpiC-high and EpiC-low. Epic-low subtype is hypomethylated, immune-desert, and characterized by FGFR3 somatic mutations with potential efficacy of the combination of FGFR3 immunotherapy and immune checkpoint inhibitors (ICI). Conversely, EpiC-high subtype is hypermethylated, immune-inflamed, and enriched with somatic mutations of SWI/SNF genes with potential benefit for ICI.
Cancers 13 04341 g001
Table 1. Adjuvant and neoadjuvant systemic treatment for UTUC patients.
Table 1. Adjuvant and neoadjuvant systemic treatment for UTUC patients.
TrialDrugStudy DesignLineOverall pts n, UTUC pts n. (%)Outcomes
(Primary Endpoint)
IMvigor 010 [61]AtezolizumabPhase 3 RCTAdjuvant809; 54 (6.7%)Median disease-free survival, 19.4 months (95% CI 15.9–24.8)
Checkmate 274 [62]NivolumabPhase 3 RCTAdjuvant709; 149 (21%)Median disease-free survival 20.8 months (95% confidence interval [CI], 16.5 to 27.6)
NCT02690558 [63]Cisplatin, gemcitabine, pembrolizumabPhase 2Neoadjuvant39; napCR:36%
POUT [43]Cisplatin or carboplatin + gemcitabinePhase 3 RCTAdjuvant261Disease-free survival (hazard ratio 0.45, 95% CI 0.30–0.68; p = 0.0001)
Abbreviations: RCT: randomized controlled trial, pts: patients; na: non available; pCR: pathologic complete response.
Table 2. Studies assessing ICI in patients with locally advanced or metastatic UC (only trials reporting data of UTUC patients were selected).
Table 2. Studies assessing ICI in patients with locally advanced or metastatic UC (only trials reporting data of UTUC patients were selected).
TrialDrug/Control ArmStudy
Design
LineOverall pts n, UTUC pts n. (%)Outcomes
(Primary Endpoint)
JAVELIN-100 [55]Avelumab/BSCPhase 3 RCT1L700, 187 (27%)median OS: 21.4 months vs. 14.3 months; hazard ratio for death, 0.69; 95% confidence interval [CI], 0.56 to 0.86; p = 0.001
KEYNOTE 052 [67]PembrolizumabPhase 21L370, 69 (19%)ORR: 24%, 95% CI 20–29)
IMvigor 130 [68]Atezolizumab + platinum-based chemotherapy (A)/Atezolizumab (B)/Platinum-based chemotherapyPhase 3 RCT1L1213, 312 (26%)median PFS: 8.2 months (95% CI 6.5–8.3) in group A and 6.3 months (6.2–7.0) in group C (stratified hazard ratio [HR] 0.82, 95% CI 0.70–0.96; one-sided p = 0.007). median OS: 16.0 months (13.9–18.9) in group A and 13.4 months (12.0–15.2) in group C (0.83, 0.69–1.00; one-sided p = 0.027). Median overall survival was 15.7 months (13.1–17.8) for group B and 13.1 months (11.7–15.1) for group C (1.02, 0.83–1.24)
KEYNOTE 361 [69]Cisplatin or Carboplatin + Gemcitabine + Pembrolizumab/Pembrolizumab/Cisplatin or Carboplatin + GemcitabinePhase 3 RCT1L1010, 211 (21%)median OS: 17·0 months (14.5–19.5) in the pembrolizumab plus chemotherapy group versus 14.3 months (12.3–16.7) in the chemotherapy group (0.86, 0.72–1.02; p = 0.0407) median PFS: 8.3 months (95% CI 7.5–8.5) in the pembrolizumab plus chemotherapy group versus 7.1 months (6.4–7.9) in the chemotherapy group (hazard ratio [HR] 0.78, 95% CI 0.65–0.93; p = 0.0033)
KEYNOTE-045 [56]Pembrolizumab/Paclitaxel or Docetaxel or VinfluninePhase 3 RCT2L748, 75 (10%)median OS: 10.3 months (95% CI 8.0 to 11.8) vs. 7.4 months (95% CI, 6.1 to 8.3) (hazard ratio for death, 0.73; 95% CI, 0.59 to 0.91; p = 0.002) median PFS: 2.1 months (95% CI, 2.0 to 2.2) vs. 3.3 months (95% CI, 2.3 to 3.5) (HR 0.98; 95% CI, 0.81 to 1.19; p = 0.42)
IMvigor 211 [70]Atezolizumab/Paclitaxel or Docetaxel or VinfluninePhase 3 RCT2L931, 236 (25%)median OS: 11.1 (95% CI 8.6–15.5) vs. 10.6 months (95% CI 8.4–12.2) p = 0.41
IMvigor 210 [66]AtezolizumabPhase 22L119, 33 (28%)ORR: 23% (95% CI 16–31)
Abbreviations: BSC: best supportive care; RCT: randomized controlled trial; OS: overall survival; PFS: progression free survival; ORR: objective response rate.
Table 3. Ongoing trials assessing immune checkpoint inhibitors-based combinations therapies in the metastatic setting.
Table 3. Ongoing trials assessing immune checkpoint inhibitors-based combinations therapies in the metastatic setting.
Trial
Identification
DrugsComparative ArmAdministrationStudy DesignLinePrimary Endpoint
NCT03513952Atezolizumab/CYT107AtezolizumabIVPhase 2≥2ORR
NCT03237780Atezolizumab/eribulinEribulinIVPhase 2>2ORR
NCT02496208Cabozantinib/Nivolumab ± IpilimumabNAPO/IVPhase 1>1RP2D/safety
NCT04940299Tocilizumab/Ipilimumab/NivolumabNAIVPhase 21Safety/DLT
NCT03606174Sitravatinib/Nivolumab and Sitravatinib/Pembrolizumab/Enfortumab vedotinNAPO/IV and PO/IV/IVPhase 21, ≥2ORR
NCT04602078Atezolizumab/Gemcitabine/CisplatinNAIVPhase 21ORR
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Thouvenin, J.; Martínez Chanzá, N.; Alhalabi, O.; Lang, H.; Tannir, N.M.; Barthélémy, P.; Malouf, G.G. Efficacy of Immune Checkpoint Inhibitors in Upper Tract Urothelial Carcinomas: Current Knowledge and Future Directions. Cancers 2021, 13, 4341. https://doi.org/10.3390/cancers13174341

AMA Style

Thouvenin J, Martínez Chanzá N, Alhalabi O, Lang H, Tannir NM, Barthélémy P, Malouf GG. Efficacy of Immune Checkpoint Inhibitors in Upper Tract Urothelial Carcinomas: Current Knowledge and Future Directions. Cancers. 2021; 13(17):4341. https://doi.org/10.3390/cancers13174341

Chicago/Turabian Style

Thouvenin, Jonathan, Nieves Martínez Chanzá, Omar Alhalabi, Hervé Lang, Nizar M. Tannir, Philippe Barthélémy, and Gabriel G. Malouf. 2021. "Efficacy of Immune Checkpoint Inhibitors in Upper Tract Urothelial Carcinomas: Current Knowledge and Future Directions" Cancers 13, no. 17: 4341. https://doi.org/10.3390/cancers13174341

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop