Next Article in Journal
The Immunocytokine FAP-IL-2v Enhances Anti-Neuroblastoma Efficacy of the Anti-GD2 Antibody Dinutuximab Beta
Next Article in Special Issue
A Review of How Lasers Are Used in UTUC Surgery: Can the Choice of Laser Affect Outcomes?
Previous Article in Journal
c-MYC Protein Stability Is Sustained by MAPKs in Colorectal Cancer
Previous Article in Special Issue
Prognostic Value of Comorbidity for Patients with Upper Tract Urothelial Carcinoma after Radical Nephroureterectomy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Neoadjuvant Chemotherapy before Nephroureterectomy in High-Risk Upper Tract Urothelial Cancer: A Systematic Review and Meta-Analysis

1
Department of Urology and Andrology, University Hospital Salzburg, Paracelsus Medical University, 5020 Salzburg, Austria
2
Department of Urology, The James Buchanan Brady Urological Institute, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
*
Author to whom correspondence should be addressed.
Cancers 2022, 14(19), 4841; https://doi.org/10.3390/cancers14194841
Submission received: 19 August 2022 / Revised: 25 September 2022 / Accepted: 27 September 2022 / Published: 4 October 2022
(This article belongs to the Special Issue Urothelial Carcinoma of the Upper Urinary Tract: What Changed?)

Abstract

:

Simple Summary

In muscle-invasive bladder cancer, platinum-based neoadjuvant chemotherapy is a well-established concept, since it contributes to pathological downstaging and improves overall survival. Immunotherapy has become increasingly important in adjuvant and palliative treatment for metastatic urothelial carcinoma, and phase II studies have shown assuring data in neoadjuvant treatment before cystectomy. Although upper urinary tract urothelial cancer usually presents as more invasive at diagnosis than bladder cancer, there is no recommendation for neoadjuvant therapy before nephroureterectomy. This meta-analysis comprises eleven comparative trials on neoadjuvant chemotherapy in this setting and analyzes pathological downstaging as well as oncological outcomes. However, no comparative studies investigating immunotherapy in upper tract urothelial cancer were found. The pathological downstaging and complete response were significantly improved in patients who received neoadjuvant chemotherapy. The data also suggested a benefit in overall survival and progression-free survival in these patients. Prospective studies are needed to confirm these findings and assess the role of immunotherapy in this setting.

Abstract

Neoadjuvant chemotherapy is a well-established concept in muscle-invasive bladder cancer with known advantages in overall survival. Phase II trials show encouraging response rates for neoadjuvant immunotherapy before radical surgery in urothelial cancer. There is no recommendation for neoadjuvant therapy in upper tract urothelial carcinoma before nephroureterectomy. Our aim was to assess the available data on neoadjuvant chemotherapy and immunotherapy before nephroureterectomy in patients with high-risk upper tract urothelial carcinoma in terms of pathological downstaging and oncological outcomes. Two investigators screened PubMed/Medline for comparative trials in the English language. We identified 368 studies and included eleven investigations in a systematic review and meta-analysis for neoadjuvant chemotherapy and control groups. There were no comparative trials investigating immunotherapy in this setting. All 11 studies reported on overall pathological downstaging with a significant effect in favor of neoadjuvant chemotherapy (OR 5.17; 95%CI 3.82; 7.00). Pathological complete response and non-muscle invasive disease were significantly higher in patients receiving neoadjuvant chemotherapy (OR 12.07; 95%CI 4.16; 35.03 and OR 1.62; 95%CI 1.05; 2.49). Overall survival and progression-free survival data analysis showed a slight benefit for neoadjuvant chemotherapy. Our results show that neoadjuvant chemotherapy is effective in downstaging in upper urinary tract urothelial carcinoma. The selection of patients and chemotherapy regimens are unclear.

1. Introduction

Urothelial carcinoma (UC) is the 6th most common tumor entity in developed countries. Upper urinary tract urothelial carcinoma (UTUC) accounts for 5–10% of all UCs. The rest mostly comprises bladder cancer (BC), accounting for approximately 90–95%, as well as a few cases of urethral cancer [1,2]. At first diagnosis, approximately 60% of UTUC shows invasive pathology [3,4], as opposed to only 25% in BC [5]. Prognosis is poor, especially for high grade and advanced diseases with overall 5-year disease-specific survival (DSS) rates ranging from 57% to 73% [3,6,7,8].
There are different environmental and pathological risk factors influencing oncological outcomes in UTUC [3,8,9,10,11,12,13,14,15]. High-risk disease has been defined by the European Association of Urology as any criteria of the following: multifocal disease, tumor size ≥2 cm, high-grade cytology, high-grade ureterorenoscopic biopsy, local invasion on computed tomography (CT), hydronephrosis, previous radical cystectomy for high grade BC, and variant histology [2].
Diagnostics rely on radiologic imaging, cytology and ureterorenoscopy (URS) with biopsy [2,10]. In case of localized, high-risk UTUC, radical nephroureterectomy (RNU) with template lymph node dissection is recommended as the therapy of choice, whilst low-risk tumors can be approached with kidney-sparing strategies [2].
In patients with muscle-invasive diseases, adjuvant platinum-based chemotherapy is currently recommended, since it increases disease-free survival [2,16]. However, there is no actual recommendation on neoadjuvant therapy for UTUC in the European Association of Urology (EAU) Guidelines, aside from a flow chart implying +/− “perioperative” platinum-based chemotherapy for high-risk UTUC [2]. In localized, clinically lymph node negative muscle-invasive bladder cancer (MIBC), neoadjuvant cisplatin-based chemotherapy is a well-known and guideline-recommended strategy, because it improves overall survival (OS) by 5–8% at 5 years postoperatively [17,18,19]. Phase II trials using checkpoint-inhibitors (CPIs) pembrolizumab and atezolizumab as neoadjuvant approach before radical surgery also have shown favorable results in terms of response and pathological downstaging with an acceptable safety profile [20,21].
Although management of UTUC often is derived from findings in BC [22,23,24], it is well known that UTUC acts differently than lower tract tumors and shows distinct molecular patterns [25,26]. Biologically, a predominance of the luminal subtype of UC as well as a high FGFR-3 density have been shown [27,28,29,30], which might indicate worse responses to checkpoint inhibition [31]. Adversely, Moss et al. found an upregulation of CPI target CTLA4, PD1 and PDL1 expression in a subset of patients with clinically aggressive diseases, which might indicate an increased effect of immunotherapy in this collective [29]. However, this finding has to be interpreted with great care since only 31 patients were included in the overall analysis, with even less patients included in the sub-groups. Balar et al. investigated the CPI atezolizumab in a first line setting for cisplatin-ineligible locally advanced and metastatic UC. They found higher response rates in patients with UTUC as a primary than patients with lower tract primaries [32].
On the other hand D’Andrea et al. showed comparable results in terms of pathological downstaging for neoadjuvant chemotherapy in UTUC and MIBC in a multi-centric retrospective analysis [33].
The rationale for neoadjuvant therapy in UTUC comprises pathological downstaging, treatment of micro-metastases and eligibility for cisplatin and higher possible doses of chemotherapy due to better kidney function before RNU, respectively [34]. Downstaging to T0 seems to be the best surrogate for survival [35], as for MIBC [36].
An obvious benefit of neoadjuvant therapy regimens compared to the adjuvant setting is eligibility for chemotherapy, which decreases distinctly after RNU due to renal insufficiency [37,38,39,40]. In terms of drawbacks, there might be a delay to definitive surgery depending on local surgery waiting periods, which might have an impact especially on non-responders [41,42]. Since there usually is no pathological evidence of muscular invasion before surgery [43], patients with T1 disease or lower might be overtreated with neoadjuvant therapy [19].
Data for neoadjuvant therapy in UTUC has always been scarce. Nevertheless, due to increasing insights in chemotherapy and experiences with new agents in BC, preoperative systemic therapy has been suggested to be an option for some patients [8,33,44,45]. In 1995 Igawa et al. showed a CR (complete response) rate of 13% and an overall response rate of 53% in a collective of 15 patients undergoing neoadjuvant chemotherapy (NAC) with methotrexate, vinblastine, adriamycin/epirubicin and cisplatin (MVAC/MVEC) [46] before RNU. A phase II trial using sequential platinum-containing NAC for UC included five cases of UTUC. The results showed good tolerability and pathological downstaging in 60% [35]. In daily practice, the proportion of patients undergoing RNU treated with NAC is low, but increasing [8,47].
Our aim was to assess the available data on different neoadjuvant concepts, including chemotherapy and CPIs, in a systematic review and meta-analysis.

2. Material and Methods

The systematic review and meta-analysis was performed according to the 2015 PRISMA-P Consensus statement [48]. The study was registered in the International prospective register of systematic reviews (PROSPERO) [49] with the unique identification number CRD42022327241.

2.1. Eligibility Criteria

Inclusion criteria: We included controlled observational trials, controlled retrospective trials and randomized controlled trials in English language. In terms of PICO criteria (participants, interventions, comparators, outcomes) studies must research neoadjuvant therapy regimens including either chemotherapy or immunotherapy in patients with high-risk UTUC undergoing RNU. Clinically positive locoregional lymph nodes were allowed. Predominant urothelial tumor histology throughout the study population was required for inclusion. Oncological outcomes in terms of pathological or clinical response (CR, partial remission (PR), stable disease (SD), progressive disease (PD)); pathological downstaging and/or progression free survival (PFS), DSS and OS, respectively, should have been reported. The data cutoff was 02/2022.
Exclusion Criteria: Articles in languages other than English, editorials, letters to the editor, and case reports were excluded from the study. Additionally, single-arm studies without a comparator were excluded from systematic review. Collectives including patients with non-locoregional metastases were excluded from analysis.

2.2. Information Sources

PubMed/Medline was searched systematically. Reference lists of screened publications were also screened for additional publications, which could not be identified by the pre-specified keywords.

2.3. Search Strategy

The pre-specified keywords were used to search the pre-specified databases in different combinations, including a synonym for UTUC and neoadjuvant therapy each.

2.4. Study Records

Data management: All identified studies were recorded using Mendeley [50]. The data retrieved during data extraction phase was saved using Microsoft Excel [51] spread sheets.
Selection Process: Two independent reviewers screened the databases according to the keywords for according studies, excluded duplicates and checked for eligibility by review of title and abstract. Full text read was performed for unclear cases. The selection of both authors was then compared for discrepancies. In such cases, a third reviewer was asked to aid in decision-making. A detailed flowchart of the selection process is depicted in Figure 1.
Data Collection Process: Data was extracted according to a pre-specified Excel sheet by both authors to avoid typing errors. Recording of additional interesting data, which had not been pre-specified, was allowed. In such a case, the spreadsheet was adapted and the items in question had to be checked, with all studies already screened for missed data.

2.5. Data Items

Baseline parameters: Pre-specified baseline data included type of neoadjuvant therapy, dose, cycles, number of patients, tumor location, multifocality, gender, tumor size, smoking status, hydronephrosis, baseline glomerular filtration rate (GFR), Eastern Cooperative Oncology Group (ECOG) performance score, previous or concurrent BC, diagnostic measures, clinical/radiological (re-)staging according to TNM classification [52], staging modalities, time to surgery from diagnosis and from end of chemotherapy, performed lymphadenectomy and open or laparoscopic surgery.
Outcomes and prioritization: Clinical and pathological response (CR, PR, SD, PD), pathological staging according to TNM classification [52], pathological downstaging, lymphovascular invasion (LVI), PFS, DSS, OS, and adverse events (per category and per Clavien-Dindo classification) were recorded to assess them effectivity and safety. The main prioritization was to analyze effectivity of neoadjuvant therapy in terms of pathological and clinical response as well as long time oncological outcome.
Due to extensive differences in data presentation throughout the publications, some outcomes were grouped for analysis. For example, some studies only presented data on T3 and T4 tumors as pooled results, so we grouped data into a T3/T4 category for the rest of the included studies as well, in order to achieve an overall more representative comparison.
OS, PFS and DSS data were only presented in a proportion (percent), but not in total numbers throughout all studies. For these parameters, rounding errors have to be accounted for.

2.6. Risk of Bias Assessment

Risk of bias assessment was performed according to the ROBINS-I tool for non-randomized studies [53] after study selection. It showed overall low risk of bias for the select papers.

2.7. Data Synthesis

A meta-analysis based on 11 studies was created to compare the effects of “study” vs. “control”. Studies with double zeros are included in the meta-analysis using a continuity correction. The effect size was estimated based on odds-ratios (ORs) and hazard ratios (HRs) including their corresponding two-sided 95% confidence intervals (CIs). Based on the estimated heterogeneity between the studies, which was assessed using Higgins I2 and Cochran’s Q, a common fixed effect or random effects model was calculated. Funnel plots and Egger’s test for asymmetry were used to assess potential publication bias.
A p-value < 0.05 was taken as the uncorrected statistical significance level (two-sided), therefore, all inferential results are only descriptive. For statistical analysis, the statistical computing software R Version 4.1.2 (R Foundation for Statistical Computing, Vienna, Austria; URL: http://www.R-project.org, accessed on 22 April 2022) was used. For conducting the meta-analysis, the R packages meta [54] and metafor were used.
Data not appropriate for quantitative synthesis were presented narratively, in the context of current literature.

3. Results

3.1. Search Results

Overall, 368 studies were found using the pre-specified keywords. No additional publications were identified through other sources. After elimination of duplicates, 297 studies remained for title and abstract screening. A full-text read was performed on 42 studies. Finally, eleven studies met the eligibility criteria and were included for systematic review and meta-analysis.

3.2. Baseline Parameters

Baseline study characteristics are shown in Table 1. The analyzed studies comprised a broad range of different chemotherapy agents, as well as schedules. No controlled studies were available for immunotherapy in the neoadjuvant setting for high-risk UTUC. Some publications excluded clinically locoregional lymph node positive disease from analysis (n = 5), while others included clinically locoregional lymph node positive disease (n = 3) or only analyzed tumors with clinically locoregional lymph node invasion (n = 3).
The selected baseline population characteristics, as far as reported, are depicted in Table 2. Clinical tumor and lymph node staging were similar throughout study and control groups. There was a tendency towards laparoscopic surgery in the NAC groups.

3.3. Pathological Downstaging

Pathological downstaging was assessed in different ways depending on the available data. Firstly, meta-analyses of pathological T stages and N stages in patients with and without neoadjuvant chemotherapy are presented as a surrogate marker of difference in downstaging. Secondly, the clinical to pathological downstaging, if available, was analyzed. Thirdly, the overall pathological downstaging and LVI as presented by the studies were assessed.
Six studies reported on CR in terms of pathological T0 stage. Meta-analysis showed a significantly higher proportion of pathological CR in the populations that received neoadjuvant therapy (OR 12.07; 95% CI 4.16; 35.03) (Figure 2).
Five studies presented a pathological T1 stage of neoadjuvant chemotherapy and control groups (non-muscle invasive). Results showed borderline significantly higher numbers within the preoperatively treated patients (OR 1.62; 95% CI 1.05; 2.49) (Figure 3).
Five studies reported on a pathological T2 stage (muscle-invasive). No significant difference could be shown in between the groups (OR 1.15; 95% CI 0.73; 1.80) (Figure 4).
Six studies presented data on clinical T3 and T4 stages, which were grouped as locally invasive. There were significantly higher numbers of advanced disease stages within the control groups, which had not received neoadjuvant therapy (OR 0.27; 95% CI 0.19; 0.38) (Figure 5).
Five studies assessed pathologically positive lymph nodes (pN+). There was no significant difference between the study and the control groups (OR 0.90; 95% CI 0.56; 1.44) (Figure 6).
Only two studies explicitly reported on reduction of clinical to pathological stage throughout both groups. These showed a significant difference in favor of neoadjuvant therapy (OR 11.98; 95% CI 2.04; 70.33) (Figure 7).
The same studies also presented data on the downstaging of clinically positive to pathologically negative lymph node stage (OR 8.49; 95% CI 2.76; 26.14) (Figure 8).
All studies assessed overall pathological downstaging with a significant difference in favor of neoadjuvant therapy (OR 5.17; 95% CI 3.82; 7.00) (Figure 9).
Seven studies reported on lymphovascular invasion (LVI) in a pathological specimen. A significant difference, with a higher proportion of LVI positive tumors within the control groups, could be shown (OR 0.48; 95% CI 0.38; 0.62) (Figure 10).

3.4. Oncological Outcomes

Two studies presented three-year OS, showing a significant benefit for the study groups (OR 3.80; 95% CI 1.80; 8.01) (Figure 11).
Six studies reported on five-year OS, also showing a significant difference in favor of the study groups (OR 2.46; 95% CI 1.45; 4.16) (Figure 12).
In terms of five-year DSS, five studies presented information showing a significant benefit in favor of neoadjuvant therapy (OR 2.53; 95% CI 1.78; 3.59). However, Egger’s graph was significant for publication bias (p = 0.031) (Figure 13).
Three studies reported on five-year PFS with a positive trend in favor of neoadjuvant therapy, but without reaching significance (OR 1.78; 95% CI 0.92; 3.44) (Figure 14).
Two studies reported PFS data adjusted for multivariate analysis (Intuition for Inverse Probability of Treatment Weighting (IPTW) adjusted PFS). Meta-analysis showed a significant risk reduction in disease progression in patients treated with NAC (HR 0.42; 95% CI 0.19; 0.91) (Figure 15).
Four studies reported DSS data adjusted for multivariate analysis (IPTW adjusted DSS). A significant risk reduction with NAC could be shown in the analysis (HR 0.44; 95% CI 0.32; 0.61) (Figure 16).
Six studies reported OS data adjusted for multivariate analysis (IPTW adjusted OS). A significant risk reduction with NAC could be shown in the analysis (HR 0.51; 95% CI 0.40; 0.66). However, Egger’s graph for publication bias was positive (p = 0.034) (Figure 17).

4. Discussion

We assessed eleven comparative studies, which represents an update to the latest work from Leow et al. 2020, who included seven controlled studies [66]. All studies comprised representative populations in terms of age and gender. UTUC is known to be more frequent in men, with a male to female ratio of 2:1 [67]. The same is true for the location of the primary tumor with a known ratio of 2:1 in the renal pelvis versus the ureter [68]. Clinical staging data was presented descriptively with balanced numbers between the groups, and therefore represents a fair starting point for downstaging analysis. Due to differences in timelines, markedly more laparoscopic and robotic RNUs were performed in patients who received neoadjuvant chemotherapy. We did not present further patient characteristics due to the extensive differences in data reporting and presentation.
Pathological downstaging was assessed differently throughout the studies, but all of them showed a significant difference in favor of neoadjuvant therapy (OR 5.17). However, details of assessment, such as clinical and pathological staging data per patient, were not presented throughout all trials. In terms of the T0 stage, six studies showed significantly higher numbers of pathologically complete responses within the study groups. The same was true for the T1 stage, with significantly higher numbers of non-invasive tumors within the study groups, albeit less unequivocal. Reversely significantly higher numbers of locally advanced tumors (T3/4) could be found in the control groups. No differences could be shown in localized invasive disease (T2). Pathological T0 disease is a clear surrogate for complete response and the pathological T3/4 stage (locally advanced disease) helps to address the response or failure to response. However, T1 (non-muscle-invasive) and T2 (muscle-invasive, non-locally advanced disease) are more difficult to interpret. We believe they might have a role in the assessment of partial response. For example, our finding of more T1 tumors within the study group could be a hint for increased partial response within these patients. However, this notion is based on balanced selection of patients’ initial clinical tumor stage and subjected to major selection bias. It might be interesting to gather this data on an individual patient basis, thereby a thorough analysis of actual downstaging as well as degree of downstaging is possible.
Two studies explicitly presented data on pathological downstaging from clinical staging, with a significant benefit for the study groups. Additionally, LVI was significantly more often present within control groups, which had not received neoadjuvant therapy.
Altogether, these results show a relevant benefit in terms of tumor downstaging after NAC before RNU. This is especially true since clinical tumor stages were distributed rather equally throughout the groups (Table 2).
Interestingly, there was no significant difference in pathologically positive lymph nodes with or without NAC. However, two studies presented data explicitly on downstaging from clinical positive to negative lymph nodes with significant benefit in favor of the study groups. These results highlight the need for prospective trials with more differentiated study populations, which distinguish clinically locoregional lymph node positive and negative populations. This question is crucial considering MIBC, in which clinically lymph node positive disease is excluded from neoadjuvant therapy according to the EAU guideline [17], and inductive chemotherapy is to be considered.
Three- and five-years OS data showed a benefit for neoadjuvant therapy. This was consistent with our results in the IPTW adjusted outcomes. However, Egger’s Test yielded a positive result for publication bias, so these results should be interpreted with caution. A five-years DSS also showed a benefit for neoadjuvant therapy, and Egger’s Test was positive as well, so we would recommend waiting for more solid data on this issue. For five-years PFS, the study results did not show significant results, although there seemed to be a trend towards increased PFS within the study groups. An analysis of IPTW-adjusted PFS showed significant risk reduction for patients who received neoadjuvant therapy.
All in all, retrospective data on pathological downstaging shows a benefit for NAC, whilst oncological follow-up data was too equivocal to allow a final conclusion. Nevertheless, data on OS and PFS seems promising.
We could not identify any controlled trials investigating CPIs in the neoadjuvant setting for UTUC. There are several phase II trials showing significantly increased complete response rates for neoadjuvant therapy with different CPIs in invasive UC before radical surgery, ranging from 31 to 46% [20,21,69,70]. This effect was more pronounced in PD-L1 positive patients in all trials. Notably, in some studies, patients with UTUC were included, but subgroup analyses are missing [21,70].
The CPI Nivolumab showed significantly increased PFS in high-risk muscle-invasive UC patients, including those with UTUC, in an adjuvant setting [71]. However, subgroup analysis still has to be conducted before treatment recommendations can be given, according to EAU guidelines [2].
Some single-arm trials present data on neoadjuvant CPI therapy for UTUC cohorts. The PURE-02 study comprises a collective of only ten patients undergoing neoadjuvant therapy with three cycles of pembrolizumab. Only one patient achieved CR, and two patients progressed and received chemotherapy. The authors concluded that pembrolizumab as single-agent therapy is not suitable in this setting [72].
Additionally, multiple trials are in progress or being set up for neoadjuvant concepts in MIBC [73]. The EV-103 study (ClinicalTrials.gov Identifier: NCT03288545) includes experimental arms investigating the drug-antibody conjugate Enfortumab-Vedotin with or without the CPI Pembrolizumab. In SURE-01, the same is done for a similar drug, Sacituzumab-Govitecan, with or without Pembrolizumab for patients who cannot or do not want to receive cisplatin-based therapy (ClinicalTrials.gov Identifier: NCT05226117). However, neither of these trials includes patients with UTUC. All in all, the role of neoadjuvant immunotherapy in UTUC has yet to be determined.
Considering the high amount of FGFR-3 expression in UTUC, another targeted agent class to be investigated in the future is FGFR-3 inhibitors, such as erdafitinib. It has shown efficacy in receptor positive UC and is approved for therapy in locally advanced and metastatic UC [17,74].
Our results represent an update to previous systematic analyses attending to the same matter [66,75,76,77]. Yang et al. found a significant improvement of DSS with a HR of 0.25 (95% CI 0.06–0.61) in patients receiving NAC for UTUC before RNU [77]. Gregg et al. found an OS benefit with an HR of 0.36 (95% CI 0.19–0.69, p = 0.002), albeit only two retrospective comparative trials could be included [76]. Kim et al. investigated four retrospective comparative trials in their analysis. They found an overall OS benefit for NAC with a pooled HR of 0.46 (95% CI 0.27–0.79, p = 0.005). Furthermore, they showed improved DSS with a pooled HR of 0,41 (95% CI 0.26–0.65, p = 0.0001) and PFS with a pooled HR of 0.53 (95% CI 0.39–0.73, p ≤ 0.0001) within the study groups. The pooled odds ratio (OR) for pathological downstaging was 0.21 (95% CI 0.09–0.06, p = 0.004), indicating a strong advantage of NAC. In contrast to our analysis, the proportion of pathologically positive lymph nodes was significantly lower within the study groups [75]. Leow et al. performed the biggest analysis so far, comprising seven comparative and nine single-arm studies. Five of the single-arm studies were prospective in nature, the rest had a retrospective design. The pooled HR of six studies for OS was 0.44 (95% CI 0.32–0.59, p < 0.001). DSS was improved with a pooled HR of three studies of 0,38 (95% CI 0.24–0.61, p < 0.001). The pooled pathological CR rate was 11% (95% CI 8–15%) and the pooled rate of downstaging was 33% (95% CI 14–52%) [66]. In contrast to the work of Leow et al., who also analyzed an adjuvant treatment regimen, we only concentrated on neoadjuvant therapies. Additionally, we looked at controlled studies only. In return, our meta-analysis included multiple different parameters such as lymph node state, lymphovascular invasion and different categories of pathological downstaging according to the different trial designs. Even though prospective evidence is still needed to draw tangible conclusions for clinical decision making, our data help to pose the correct questions in these trials and find the appropriate patient collective, which might profit from neoadjuvant therapy. As an example, our finding of equivalent numbers of patients with pathologically positive lymph nodes in both groups highlights the necessity of stratification for clinically positive lymph nodes. Concerning OS and CSS data, we analyzed three-year and five-year data, depending on the available results. Additionally, we analyzed PFS with two different analyses, suggesting a benefit for NAC. Furthermore, our work highlights the need for additional research on immunotherapy in this setting.

5. Limitations

The major limitations of our results are the retrospective nature of the analyzed trials, the inhomogeneous study populations and treatment regimens, as well as major differences in data presentation. Most notably, the criteria for administration of NAC throughout the different trials cannot be reproduced, which might represent a major selection bias.
A challenge in UTUC as compared to BC is that there rarely is representative histology before surgery. Histology acquired by URS and forceps-biopsy usually only helps to determine tumor grade and get a visual impression of tumor architecture [26]. Biopsy was shown to be highly accurate in the determination of grading, but not staging with significant correlation of high-grade diseases on biopsy with high-grade and muscle-invasive features of the pathological specimen [78,79,80]. Clinical staging is mostly radiological and relies on imaging (usually CT) [81,82]. A partial pathological response to neoadjuvant treatment is therefore difficult to assess, since it is impossible to know whether a patient had pT1 diseases in the first place or because of downstaging after neoadjuvant treatment. None of the investigated studies reported on radiological re-staging after neoadjuvant therapy before surgery, which would have helped to assess the radiological response.
CT urography represents the most accurate means of clinical staging with correct prediction of the pathological stage in 88% in a case series [83]. The staging of organ confined diseases was correct in 97%, but only 67% of cases in locally advanced diseases in a different collective [84]. Thus, it might be a worthwhile strategy for future trials to repeat radiographic staging after neoadjuvant therapy for assessment of the response.
Due to the retrospective data analysis, chemotherapy regimens varied widely across the studies, and often different schemata with different number of cycles were used within the same populations (Table 2). Nevertheless, platinum-containing regimens were predominating, with Gemcitabine/Cisplatinum and MVAC being the most frequently used schedules analogous to MIBC [23]. Future trials will need to assess standardized approaches in terms of therapy and the number of cycles.
There was no evidence for significant publication bias according to the ROBINS-I tool. Egger’s graph and funnel plots were positive for two endpoints, as depicted in the Results section (5-year DSS and IPTW adjusted OS). Thus, these results have to be interpreted with care.

6. Conclusions

NAC before RNU in UTUC is effective in pathological downstaging and seems to have some oncological benefit in terms of OS and PFS. Exact therapy regimens and patient characteristics to identify those profiting are unclear. Randomized controlled prospective trials are needed to determine whether NAC could become part of a standardized approach. Stratification for parameters like positive locoregional lymph nodes and radiological tumor stage could help to establish selection criteria.
Aside from clinical trials, neoadjuvant immunotherapy does not play a role in the treatment of high-risk UTUC at the moment.

Author Contributions

Conceptualization, D.O. and C.E.; methodology, D.O., M.S., C.E. and S.D.; software, D.O. and P.T.; validation, D.O., P.T. and C.E.; formal analysis, D.O., C.E., M.P. and S.D.; investigation, D.O. and C.E.; resources, D.O. and C.E.; data curation, D.O., M.P. and C.E.; writing—original draft preparation, D.O., M.P. and S.D.; writing—review and editing, P.T., M.S. and S.D; visualization, D.O. and P.T.; supervision, S.D., M.S. and P.T.; project administration, C.E. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

Biostatistical analyses were performed by Wolfgang Schimetta, Department of Applied Systems Research and Statistics, Johannes Kepler University, 4020 Linz, Austria.

Conflicts of Interest

All authors state that they have no conflict of interest in regard to the reviewed topic.

Abbreviations

UCurothelial carcinoma
UTUCupper urinary tract urothelial carcinoma
BCbladder cancer
DSSdisease-specific survival
CTcomputed tomography
URSureterorenoscopy
RNUradical nephroureterectomy
EAUEuropean Association of Urology
MIBCmuscle-invasive bladder cancer
OSoverall survival
CPIcheckpoint-Inhibitor
CRcomplete remission
NACneoadjuvant chemotherapy
MVAC/MVECmethotrexate, vinblastine, adriamycin/epirubicin and cisplatin
PICOparticipants, interventions, comparators, outcomes
PRpartial remission
SDstable disease
PDprogressive disease
PFSprogression-free survival
GFRglomerular filtration rate
ECOGEastern Cooperative Oncology Group
LVIlymphovascular invasion
ORodds ratio
HRhazard ratio
CIconfidence interval
IPTWintuition for inverse probability of treatment weighting

References

  1. Siegel, R.L.; Miller, K.D.; Fuchs, H.E.; Jemal, A. Cancer Statistics, 2021. CA Cancer J. Clin. 2021, 71, 7–33. [Google Scholar] [CrossRef] [PubMed]
  2. Rouprêt, M.; Babjuk, M.; Burger, M.; Capoun, O.; Cohen, D.; Compérat, E.M.; Cowan, N.C.; Dominguez-Escrig, J.L.; Gontero, P.; Hugh Mostafid, A.; et al. European Association of Urology Guidelines on Upper Urinary Tract Urothelial Carcinoma: 2020 Update. Eur. Urol. 2021, 79, 62–79. [Google Scholar] [CrossRef] [PubMed]
  3. Margulis, V.; Shariat, S.F.; Matin, S.F.; Kamat, A.M.; Zigeuner, R.; Kikuchi, E.; Lotan, Y.; Weizer, A.; Raman, J.D.; 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]
  4. Lughezzani, G.; Jeldres, C.; Isbarn, H.; Sun, M.; Shariat, S.F.; Widmer, H.; Arjane, P.; Graefen, M.; Perrotte, P.; Montorsi, F.; et al. Temporal stage and grade migration in surgically treated patients with upper tract urothelial carcinoma. BJU Int. 2010, 105, 799–804. [Google Scholar] [CrossRef] [PubMed]
  5. Compérat, E.; Larré, S.; Roupret, M.; Neuzillet, Y.; Pignot, G.; Quintens, H.; Houéde, N.; Roy, C.; Durand, X.; Varinot, J.; et al. Clinicopathological characteristics of urothelial bladder cancer in patients less than 40 years old. Virchows Arch. 2015, 466, 589–594. [Google Scholar] [CrossRef]
  6. Rosiello, G.; Palumbo, C.; Knipper, S.; Pecoraro, A.; Luzzago, S.; Deuker, M.; Mistretta, F.A.; Tian, Z.; Fossati, N.; Gallina, A.; et al. Contemporary conditional cancer-specific survival after radical nephroureterectomy in patients with nonmetastatic urothelial carcinoma of upper urinary tract. J. Surg. Oncol. 2020, 121, 1154–1161. [Google Scholar] [CrossRef]
  7. van Doeveren, T.; van der Mark, M.; van Leeuwen, P.J.; Boormans, J.L.; Aben, K.K.H. Rising incidence rates and unaltered survival rates for primary upper urinary tract urothelial carcinoma: A Dutch population-based study from 1993 to 2017. BJU Int. 2021, 128, 343–351. [Google Scholar] [CrossRef]
  8. Cohen, A.; Kuchta, K.; Park, S. Neoadjuvant and adjuvant chemotherapy use in upper tract urothelial carcinoma. Urol. Oncol. 2017, 35, 322–327. [Google Scholar] [CrossRef]
  9. Leow, J.J.; Orsola, A.; Chang, S.L.; Bellmunt, J. A contemporary review of management and prognostic factors of upper tract urothelial carcinoma. Cancer Treat. Rev. 2015, 41, 310–319. [Google Scholar] [CrossRef]
  10. Soria, F.; Shariat, S.F.; Lerner, S.P.; Fritsche, H.M.; Rink, M.; Kassouf, W.; Spiess, P.E.; Lotan, Y.; Ye, D.; Fernández, M.I.; et al. Epidemiology, diagnosis, preoperative evaluation and prognostic assessment of upper-tract urothelial carcinoma (UTUC). World J. Urol. 2017, 35, 379–387. [Google Scholar] [CrossRef]
  11. Seisen, T.; Granger, B.; Colin, P.; Léon, P.; Utard, G.; Renard-Penna, R.; Compérat, E.; Mozer, P.; Cussenot, O.; Shariat, S.F.; et al. A Systematic Review and Meta-analysis of Clinicopathologic Factors Linked to Intravesical Recurrence After Radical Nephroureterectomy to Treat Upper Tract Urothelial Carcinoma. Eur. Urol. 2015, 67, 1122–1133. [Google Scholar] [CrossRef] [PubMed]
  12. Cha, E.K.; Shariat, S.F.; Kormaksson, M.; Novara, G.; Chromecki, T.F.; Scherr, D.S.; Lotan, Y.; Raman, J.D.; Kassouf, W.; Zigeuner, R.; et al. Predicting clinical outcomes after radical nephroureterectomy for upper tract urothelial carcinoma. Eur. Urol. 2012, 61, 818–825. [Google Scholar] [CrossRef] [PubMed]
  13. Lughezzani, G.; Burger, M.; Margulis, V.; Matin, S.F.; Novara, G.; Roupret, M.; Shariat, S.F.; Wood, C.G.; Zigeuner, R. Prognostic factors in upper urinary tract urothelial carcinomas: A comprehensive review of the current literature. Eur. Urol. 2012, 62, 100–114. [Google Scholar] [CrossRef] [PubMed]
  14. Ruvolo, C.C.; Nocera, L.; Stolzenbach, L.F.; Wenzel, M.; Cucchiara, V.; Tian, Z.; Shariat, S.F.; Saad, F.; Longo, N.; Montorsi, F.; et al. Incidence and Survival Rates of Contemporary Patients with Invasive Upper Tract Urothelial Carcinoma. Eur. Urol. Oncol. 2021, 4, 792–801. [Google Scholar] [CrossRef]
  15. Colin, P.; Koenig, P.; Ouzzane, A.; Berthon, N.; Villers, A.; Biserte, J.; Rouprêt, M. Environmental factors involved in carcinogenesis of urothelial cell carcinomas of the upper urinary tract. BJU Int. 2009, 104, 1436–1440. [Google Scholar] [CrossRef]
  16. Birtle, A.; Johnson, M.; Chester, J.; Jones, R.; Dolling, D.; Bryan, R.T.; 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]
  17. 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]
  18. Grossman, H.B.; Natale, R.B.; Tangen, C.M.; Speights, V.O.; Vogelzang, N.J.; Trump, D.L.; de Vere White, R.W.; Sarosdy, M.F.; Wood, D.P., Jr.; Raghavan, D.; et al. Neoadjuvant chemotherapy plus cystectomy compared with cystectomy alone for locally advanced bladder cancer. N. Engl. J. Med. 2003, 349, 859–866. [Google Scholar] [CrossRef]
  19. Vale, C.L. Neoadjuvant chemotherapy in invasive bladder cancer: Update of a systematic review and meta-analysis of individual patient data advanced bladder cancer (ABC) meta-analysis collaboration. Eur. Urol. 2005, 48, 202–206. [Google Scholar] [CrossRef]
  20. Powles, T.; Kockx, M.; Rodriguez-Vida, A.; Duran, I.; Crabb, S.J.; Van Der Heijden, M.S.; Szabados, B.; Pous, A.F.; Gravis, G.; Herranz, U.A.; et al. Clinical efficacy and biomarker analysis of neoadjuvant atezolizumab in operable urothelial carcinoma in the ABACUS trial. Nat. Med. 2019, 25, 1706–1714. [Google Scholar] [CrossRef]
  21. 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. Off. J. Am. Soc. Clin. Oncol. 2018, 36, 3353–3360. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Campbell, M.T.; Shah, A.Y.; Matin, S.F.; Siefker-Radtke, A.O. Optimizing management of upper tract urothelial carcinoma. Urol. Oncol. 2017, 35, 492–498. [Google Scholar] [CrossRef] [PubMed]
  23. O’Donnell, P.H.; Stadler, W.M. The role of chemotherapy in upper tract urothelial carcinoma. Adv. Urol. 2009, 2009, 419028. [Google Scholar] [CrossRef] [PubMed]
  24. Raman, J.D.; Scherr, D.S. Management of patients with upper urinary tract transitional cell carcinoma. Nat. Clin. Pract. Urol. 2007, 4, 432–443. [Google Scholar] [CrossRef]
  25. Green, D.A.; Rink, M.; Xylinas, E.; Matin, S.F.; Stenzl, A.; Roupret, M.; Karakiewicz, P.I.; Scherr, D.S.; Shariat, S.F. Urothelial carcinoma of the bladder and the upper tract: Disparate twins. J. Urol. 2013, 189, 1214–1221. [Google Scholar] [CrossRef]
  26. 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 2017, 1, e000126. [Google Scholar] [CrossRef]
  27. Compérat, E.; Egevad, L.; Lopez-Beltran, A.; Camparo, P.; Algaba, F.; Amin, M.; Epstein, J.I.; Hamberg, H.; Hulsbergen-van de Kaa, C.; Kristiansen, G.; et al. An interobserver reproducibility study on invasiveness of bladder cancer using virtual microscopy and heatmaps. Histopathology 2013, 63, 756–766. [Google Scholar] [CrossRef]
  28. Sfakianos, J.P.; Cha, E.K.; Iyer, G.; Scott, S.N.; Zabor, E.C.; Shah, R.H.; 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] [Green Version]
  29. 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]
  30. 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. 2019, 25, 967–976. [Google Scholar] [CrossRef]
  31. Necchi, A.; Raggi, D.; Gallina, A.; Ross, J.S.; Fare, E.; Giannatempo, P.; Marandino, L.; Colecchia, M.; Luciano, R.; Bianchi, M.; et al. Impact of Molecular Subtyping and Immune Infiltration on Pathological Response and Outcome Following Neoadjuvant Pembrolizumab in Muscle-invasive Bladder Cancer. Eur. Urol. 2020, 77, 701–710. [Google Scholar] [CrossRef] [PubMed]
  32. Balar, A.V.; 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 2017, 389, 67–76. [Google Scholar] [CrossRef] [Green Version]
  33. D’Andrea, D.; Matin, S.; Black, P.C.; Petros, F.G.; Zargar, H.; Dinney, C.P.; Cookson, M.S.; Kassouf, W.; Dall’Era, M.A.; McGrath, J.S.; et al. Comparative effectiveness of neoadjuvant chemotherapy in bladder and upper urinary tract urothelial carcinoma. BJU Int. 2021, 127, 528–537. [Google Scholar] [CrossRef] [PubMed]
  34. Audenet, F.; Yates, D.R.; Cussenot, O.; Rouprêt, M. The role of chemotherapy in the treatment of urothelial cell carcinoma of the upper urinary tract (UUT-UCC). Urol. Oncol. 2013, 31, 407–413. [Google Scholar] [CrossRef]
  35. Siefker-Radtke, A.O.; Dinney, C.P.; Shen, Y.; Williams, D.L.; Kamat, A.M.; Grossman, H.B.; Millikan, R.E. A phase 2 clinical trial of sequential neoadjuvant chemotherapy with ifosfamide, doxorubicin, and gemcitabine followed by cisplatin, gemcitabine, and ifosfamide in locally advanced urothelial cancer: Final results. Cancer 2013, 119, 540–547. [Google Scholar] [CrossRef]
  36. Peyton, C.C.; Tang, D.; Reich, R.R.; Azizi, M.; Chipollini, J.; Pow-Sang, J.M.; Manley, B.; Spiess, P.E.; Poch, M.A.; Sexton, W.J.; et al. Downstaging and Survival Outcomes Associated With Neoadjuvant Chemotherapy Regimens Among Patients Treated With Cystectomy for Muscle-Invasive Bladder Cancer. JAMA Oncol. 2018, 4, 1535–1542. [Google Scholar] [CrossRef]
  37. 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] [Green Version]
  38. Xylinas, E.; Rink, M.; Margulis, V.; Clozel, T.; Lee, R.K.; Comploj, E.; Novara, G.; Raman, J.D.; Lotan, Y.; Weizer, A.; et al. Impact of renal function on eligibility for chemotherapy and survival in patients who have undergone radical nephro-ureterectomy. BJU Int. 2013, 112, 453–461. [Google Scholar] [CrossRef] [Green Version]
  39. 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]
  40. Raman, J.D.; Lin, Y.K.; Kaag, M.; Atkinson, T.; Crispen, P.; Wille, M.; Smith, N.; Hockenberry, M.; Guzzo, T.; Peyronnet, B.; et al. High rates of advanced disease, complications, and decline of renal function after radical nephroureterectomy. Urol. Oncol. 2014, 32, 47.e9–47.e14. [Google Scholar] [CrossRef]
  41. Gayed, B.A.; Thoreson, G.R.; Margulis, V. The role of systemic chemotherapy in management of upper tract urothelial cancer. Curr. Urol. Rep. 2013, 14, 94–101. [Google Scholar] [CrossRef] [PubMed]
  42. Waldert, M.; Karakiewicz, P.I.; Raman, J.D.; Remzi, M.; Isbarn, H.; Lotan, Y.; Capitanio, U.; Bensalah, K.; Marberger, M.J.; Shariat, S.F. A delay in radical nephroureterectomy can lead to upstaging. BJU Int. 2010, 105, 812–817. [Google Scholar] [CrossRef] [PubMed]
  43. Subiela, J.D.; Territo, A.; Mercadé, A.; Balañà, J.; Aumatell, J.; Calderon, J.; Gallioli, A.; González-Padilla, D.A.; Gaya, J.M.; Palou, J.; et al. Diagnostic accuracy of ureteroscopic biopsy in predicting stage and grade at final pathology in upper tract urothelial carcinoma: Systematic review and meta-analysis. Eur. J. Surg. Oncol. 2020, 46, 1989–1997. [Google Scholar] [CrossRef] [PubMed]
  44. Leow, J.J.; Liu, Z.; Tan, T.W.; Lee, Y.M.; Yeo, E.K.; Chong, Y.L. Optimal Management of Upper Tract Urothelial Carcinoma: Current Perspectives. Onco Targets Ther. 2020, 13, 1–15. [Google Scholar] [CrossRef] [Green Version]
  45. Tabayoyong, W.; Li, R.; Gao, J.; Kamat, A. Optimal Timing of Chemotherapy and Surgery in Patients with Muscle-Invasive Bladder Cancer and Upper Urinary Tract Urothelial Carcinoma. Urol. Clin. North Am. 2018, 45, 155–167. [Google Scholar] [CrossRef]
  46. Igawa, M.; Urakami, S.; Shiina, H.; Kishi, H.; Himeno, Y.; Ishibe, T.; Kadena, H.; Usui, T. Neoadjuvant chemotherapy for locally advanced urothelial cancer of the upper urinary tract. Urol. Int. 1995, 55, 74–77. [Google Scholar] [CrossRef]
  47. Browne, B.M.; Stensland, K.D.; Moynihan, M.J.; Canes, D. An Analysis of Staging and Treatment Trends for Upper Tract Urothelial Carcinoma in the National Cancer Database. Clin. Genitourin. Cancer 2018, 16, e743–e750. [Google Scholar] [CrossRef]
  48. Moher, D.; Shamseer, L.; Clarke, M.; Ghersi, D.; Liberati, A.; Petticrew, M.; Shekelle, P.; Stewart, L.A.; PRISMA-P Group. Preferred reporting items for systematic review and meta-analysis protocols (PRISMA-P) 2015 statement. Syst. Rev. 2015, 4, 1. [Google Scholar] [CrossRef] [Green Version]
  49. P.R.O.S.P.E.R.O. Available online: https://www.crd.york.ac.uk/prospero/ (accessed on 10 May 2022).
  50. Mendeley Ltd. Mendeley. 2021. Available online: https://www.mendeley.com (accessed on 1 March 2022).
  51. Microsoft Corporation. Microsoft Excel. 2018. Available online: https://office.microsoft.com/excel (accessed on 1 March 2022).
  52. Brierley, J.D.; Gospodarowicz, M.K.; Wittekind, C. TNM Classification of Malignant Tumors. UICC International Union Against Cancer, 85th ed.; Wiley-Blackwell and UICC: New York, NY, USA, 2017. [Google Scholar]
  53. Sterne, J.A.; Hernán, M.A.; Reeves, B.C.; Savović, J.; Berkman, N.D.; Viswanathan, M.; Henry, D.; Altman, D.G.; Ansari, M.T.; Boutron, I.; et al. ROBINS-I: A tool for assessing risk of bias in non-randomised studies of interventions. BMJ 2016, 355, i4919. [Google Scholar] [CrossRef] [Green Version]
  54. Balduzzi, S.; Rücker, G.; Schwarzer, G. How to perform a meta-analysis with R: A practical tutorial. Evid. Based Ment. Health 2019, 22, 153–160. [Google Scholar] [CrossRef]
  55. Matin, S.F.; Margulis, V.; Kamat, A.; Wood, C.G.; Grossman, H.B.; Brown, G.A.; Dinney, C.P.N.; Millikan, R.; Siefker-Radtke, A.O. Incidence of downstaging and complete remission after neoadjuvant chemotherapy for high-risk upper tract transitional cell carcinoma. Cancer 2010, 116, 3127–3134. [Google Scholar] [CrossRef] [PubMed]
  56. Porten, S.; Siefker-Radtke, A.O.; Xiao, L.; Margulis, V.; Kamat, A.M.; Wood, C.G.; Jonasch, E.; Dinney, C.P.N.; Matin, S.F. Neoadjuvant chemotherapy improves survival of patients with upper tract urothelial carcinoma. Cancer 2014, 120, 1794–1799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. 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. 2021, 127, 332–339. [Google Scholar] [CrossRef] [PubMed]
  58. Kitamura, H.; Igarashi, M.; Tanaka, T.; Shindo, T.; Masumori, N.; Tamakawa, M.; Kawaai, Y.; Tsukamoto, T. A role for preoperative systemic chemotherapy in node-positive upper tract urothelial carcinoma treated with radical nephroureterectomy. Jpn. J. Clin. Oncol. 2012, 42, 1192–1196. [Google Scholar] [CrossRef] [Green Version]
  59. Kobayashi, K.; Saito, T.; Kitamura, Y.; Bilim, V.; Toba, T.; Kawasaki, T.; Hara, N.; Tanikawa, T.; Tomita, Y. Effect of preoperative chemotherapy on survival of patients with upper urinary tract urothelial carcinoma clinically involving regional lymph nodes. Int. J. Urol. 2016, 23, 153–158. [Google Scholar] [CrossRef]
  60. Hosogoe, S.; Hatakeyama, S.; Kusaka, A.; Hamano, I.; Iwamura, H.; Fujita, N.; Yamamoto, H.; Tobisawa, Y.; Yoneyama, T.; Yoneyama, T.; et al. Platinum-based Neoadjuvant Chemotherapy Improves Oncological Outcomes in Patients with Locally Advanced Upper Tract Urothelial Carcinoma. Eur. Urol. Focus 2018, 4, 946–953. [Google Scholar] [CrossRef] [Green Version]
  61. Chen, L.; Ou, Z.; Wang, R.; Zhang, M.; He, W.; Zhang, J.; Zu, X.; Yi, L.; Xu, R.; Jiang, S.; et al. Neoadjuvant Chemotherapy Benefits Survival in High-Grade Upper Tract Urothelial Carcinoma: A Propensity Score-Based Analysis. Ann. Surg. Oncol. 2020, 27, 1297–1303. [Google Scholar] [CrossRef]
  62. Hamaya, T.; Hatakeyama, S.; Tanaka, T.; Kubota, Y.; Togashi, K.; Hosogoe, S.; Fujita, N.; Kusaka, A.; Tokui, N.; Okamoto, 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, 128, 468–476. [Google Scholar] [CrossRef]
  63. Kubota, Y.; Hatakeyama, S.; Tanaka, T.; Fujita, N.; Iwamura, H.; Mikami, J.; Yamamoto, H.; Tobisawa, Y.; Yoneyama, T.; Yoneyama, T.; et al. Oncological outcomes of neoadjuvant chemotherapy in patients with locally advanced upper tract urothelial carcinoma: A multicenter study. Oncotarget 2017, 8, 101500–101508. [Google Scholar] [CrossRef] [Green Version]
  64. Rajput, M.Z.; Kamat, A.M.; Clavell-Hernandez, J.; Siefker-Radtke, A.O.; Grossman, H.B.; Dinney, C.P.N.; Matin, S.F. Perioperative outcomes of laparoscopic radical nephroureterectomy and regional lymphadenectomy in patients with upper urinary tract urothelial carcinoma after neoadjuvant chemotherapy. Urology 2011, 78, 61–67. [Google Scholar] [CrossRef]
  65. Shigeta, K.; Matsumoto, K.; Ogihara, K.; Murakami, T.; Anno, T.; Umeda, K.; Izawa, M.; Baba, Y.; Sanjo, T.; Shojo, K.; et al. Does neoadjuvant chemotherapy have therapeutic benefit for node-positive upper tract urothelial carcinoma? Results of a multi-center cohort study. Urol. Oncol. 2022, 40, 105.e19–105.e26. [Google Scholar] [CrossRef] [PubMed]
  66. 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]
  67. Shariat, S.F.; Favaretto, R.L.; Gupta, A.; Fritsche, H.M.; Matsumoto, K.; Kassouf, W.; Walton, T.J.; Tritschler, S.; Baba, S.; Matsushita, K.; et al. Gender differences in radical nephroureterectomy for upper tract urothelial carcinoma. World J. Urol. 2011, 29, 481–486. [Google Scholar] [CrossRef]
  68. Favaretto, R.L.; Shariat, S.F.; Chade, D.C.; Godoy, G.; Adamy, A.; Kaag, M.; Bochner, B.H.; Coleman, J.; Dalbagni, G. The effect of tumor location on prognosis in patients treated with radical nephroureterectomy at Memorial Sloan-Kettering Cancer Center. Eur. Urol. 2010, 58, 574–580. [Google Scholar] [CrossRef] [Green Version]
  69. van Dijk, N.; Gil-Jimenez, A.; Silina, K.; Hendricksen, K.; Smit, L.A.; de Feijter, J.M.; van Montfoort, M.L.; van Rooijen, C.; Peters, D.; Broeks, A.; et al. Preoperative ipilimumab plus nivolumab in locoregionally advanced urothelial cancer: The NABUCCO trial. Nat. Med. 2020, 26, 1839–1844. [Google Scholar] [CrossRef]
  70. Gao, J.; Navai, N.; Alhalabi, O.; Siefker-Radtke, A.; Campbell, M.T.; Tidwell, R.S.; Guo, C.C.; Kamat, A.M.; Matin, S.F.; Araujo, J.C.; et al. Neoadjuvant PD-L1 plus CTLA-4 blockade in patients with cisplatin-ineligible operable high-risk urothelial carcinoma. Nat. Med. 2020, 26, 1845–1851. [Google Scholar] [CrossRef] [PubMed]
  71. 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]
  72. Necchi, A.; Martini, A.; Raggi, D.; Cucchiara, V.; Colecchia, M.; Lucianò, R.; Villa, L.; Mazzone, E.; Basile, G.; Scuderi, S.; et al. A feasibility study of preoperative pembrolizumab before radical nephroureterectomy in patients with high-risk, upper tract urothelial carcinoma: PURE-02. Urol. Oncol. 2022, 40, 10.e1–10.e6. [Google Scholar] [CrossRef]
  73. Iacovino, M.L.; Miceli, C.C.; De Felice, M.; Barone, B.; Pompella, L.; Chiancone, F.; Di Zazzo, E.; Tirino, G.; Della Corte, C.M.; Imbimbo, C.; et al. Novel Therapeutic Opportunities in Neoadjuvant Setting in Urothelial Cancers: A New Horizon Opened by Molecular Classification and Immune Checkpoint Inhibitors. Int. J. Mol. Sci. 2022, 23, 1133. [Google Scholar] [CrossRef]
  74. 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]
  75. Kim, D.K.; Lee, J.Y.; Kim, J.W.; Hah, Y.S.; Cho, K.S. Effect of neoadjuvant chemotherapy on locally advanced upper tract urothelial carcinoma: A systematic review and meta-analysis. Crit. Rev. Oncol. Hematol. 2019, 135, 59–65. [Google Scholar] [CrossRef] [PubMed]
  76. Gregg, R.W.; Vera-Badillo, F.E.; Booth, C.M.; Mahmud, A.; Brundage, M.; Leveridge, M.J.; Hanna, T.P. Perioperative chemotherapy for urothelial carcinoma of the upper urinary tract: A systematic review and meta-analysis. Crit. Rev. Oncol. Hematol. 2018, 128, 58–64. [Google Scholar] [CrossRef] [PubMed]
  77. Yang, X.; Li, P.; Deng, X.; Dong, H.; Cheng, Y.; Zhang, X.; Yang, C.; Tang, J.; Yuan, W.; Xu, X.; et al. Perioperative treatments for resected upper tract urothelial carcinoma: A network meta-analysis. Oncotarget 2017, 8, 3568–3580. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Clements, T.; Messer, J.C.; Terrell, J.D.; Herman, M.P.; Ng, C.K.; Scherr, D.S.; Scoll, B.; Boorjian, S.A.; Uzzo, R.G.; Wille, M.; et al. High-grade ureteroscopic biopsy is associated with advanced pathology of upper-tract urothelial carcinoma tumors at definitive surgical resection. J. Endourol. 2012, 26, 398–402. [Google Scholar] [CrossRef]
  79. Rojas, C.P.; Castle, S.M.; Llanos, C.A.; Cortes, J.A.S.; Bird, V.; Rodriguez, S.; Reis, I.M.; Zhao, W.; Gomez-Fernandez, C.; Leveillee, R.J.; et al. Low biopsy volume in ureteroscopy does not affect tumor biopsy grading in upper tract urothelial carcinoma. Urol. Oncol. 2013, 31, 1696–1700. [Google Scholar] [CrossRef]
  80. Brown, G.A.; Matin, S.F.; Busby, J.E.; Dinney, C.P.N.; Grossman, H.B.; Pettaway, C.A.; Munsell, M.F.; Kamat, A.M. Ability of clinical grade to predict final pathologic stage in upper urinary tract transitional cell carcinoma: Implications for therapy. Urology 2007, 70, 252–256. [Google Scholar] [CrossRef] [Green Version]
  81. Seisen, T.; Colin, P.; Rouprêt, M. Risk-adapted strategy for the kidney-sparing management of upper tract tumours. Nat. Rev. Urol. 2015, 12, 155–166. [Google Scholar] [CrossRef]
  82. Honda, Y.; Nakamura, Y.; Teishima, J.; Goto, K.; Higaki, T.; Narita, K.; Akagi, M.; Terada, H.; Kaichi, Y.; Fujii, S.; et al. Clinical staging of upper urinary tract urothelial carcinoma for T staging: Review and pictorial essay. Int. J. Urol. 2019, 26, 1024–1032. [Google Scholar] [CrossRef]
  83. Millan-Rodriguez, F.; Palou, J.; De la Torre-Holguera, P.; Vayreda-Martija, J.M.; Villavicencio-Mavrich, H.; Vicente-Rodriguez, J. Conventional CT signs in staging transitional cell tumors of the upper urinary tract. Eur. Urol. 1999, 35, 318–322. [Google Scholar] [CrossRef]
  84. Fritz, G.A.; Schoellnast, H.; Deutschmann, H.A.; Quehenberger, F.; Tillich, M. Multiphasic multidetector-row CT (MDCT) in detection and staging of transitional cell carcinomas of the upper urinary tract. Eur. Radiol. 2006, 16, 1244–1252. [Google Scholar] [CrossRef]
Figure 1. Flowchart of study selection process.
Figure 1. Flowchart of study selection process.
Cancers 14 04841 g001
Figure 2. Pathological stage T0 (pathological complete response).
Figure 2. Pathological stage T0 (pathological complete response).
Cancers 14 04841 g002
Figure 3. Pathological stage T1 (non-muscle invasive).
Figure 3. Pathological stage T1 (non-muscle invasive).
Cancers 14 04841 g003
Figure 4. Pathological stage T2 (muscle-invasive).
Figure 4. Pathological stage T2 (muscle-invasive).
Cancers 14 04841 g004
Figure 5. Pathological stage T3/4 (locally advanced).
Figure 5. Pathological stage T3/4 (locally advanced).
Cancers 14 04841 g005
Figure 6. Pathologically positive locoregional lymph nodes.
Figure 6. Pathologically positive locoregional lymph nodes.
Cancers 14 04841 g006
Figure 7. Downstaging from clinical to pathological T stage.
Figure 7. Downstaging from clinical to pathological T stage.
Cancers 14 04841 g007
Figure 8. Downstaging from clinical to pathological lymph node stage.
Figure 8. Downstaging from clinical to pathological lymph node stage.
Cancers 14 04841 g008
Figure 9. Overall pathological downstaging.
Figure 9. Overall pathological downstaging.
Cancers 14 04841 g009
Figure 10. Lymphovascular invasion.
Figure 10. Lymphovascular invasion.
Cancers 14 04841 g010
Figure 11. Three-year overall survival.
Figure 11. Three-year overall survival.
Cancers 14 04841 g011
Figure 12. Five-year overall survival.
Figure 12. Five-year overall survival.
Cancers 14 04841 g012
Figure 13. Five-year disease-specific survival.
Figure 13. Five-year disease-specific survival.
Cancers 14 04841 g013
Figure 14. Five-year progression-free survival.
Figure 14. Five-year progression-free survival.
Cancers 14 04841 g014
Figure 15. IPTW adjusted progression-free survival.
Figure 15. IPTW adjusted progression-free survival.
Cancers 14 04841 g015
Figure 16. IPTW adjusted cancer specific survival (CSS).
Figure 16. IPTW adjusted cancer specific survival (CSS).
Cancers 14 04841 g016
Figure 17. IPTW adjusted overall survival.
Figure 17. IPTW adjusted overall survival.
Cancers 14 04841 g017
Table 1. Basic study characteristics.
Table 1. Basic study characteristics.
StudyAuthorsJournalYearStudy TypeTimelinePair-MatchingAgentCyclesNStaging ModalitiescN+
1 [55]Matin et al.Cancer2010retrospective single-center2004–2008noMVAC (19), CGI (9), GTA (6), GC (5), other (4)2–9 (median 4)43CT (Recist Criteria)no
1993–2004 107
2 [56]Porten et al.Cancer2014retrospective single-center2004–2008yesCisplatin containing (MVAC, Gem/Cis, Cis/Gem/Ifos; 21), high dose ifosf/doxo/gem (3), kidney sparing (primarily gem/pac/doxo; 7)2–6 (median 4)31 no
1993–2003 81
3 [57]Zennamni et al.BJU Int.2021retrospective single-center2005–2019yesGem/Cis, MVAC2117CTno
67
4 [58]Kitamura et al.Jpn J Clin Oncol2012retrospective single-center1995–2010noMVAC (14), Gem/Cis (1)2–3 (median 2)15CT (Recist Criteria)yes (only)
14
5 [59]Kobayashi et al.Int J Urol2016retrospective single-center1991–2013no1991–1995 MEP (3), 1996–2009 MVAC (9), 2010- Gem/Cis (14)2–4 (median 3)24CT (Recist Criteria)yes (only)
31
6 [60]Hosogoe et al.Eur Urol Focus2018retrospective single-center1995–2016yesGem/Cis (16), Gem/Carbo (35)2–4 q3w51CT (Recist Criteria)yes
51
7 [61]Chen et al.Ann Surg Oncol2020retrospective multi-center2012–2015yesGem/Cis2–4 q3w37CT (Recist Criteria)no
37
8 [62]Hamaya et al.BJU Int2021retrospective multi-center2000–2020noGem/Cis, Gem/Carbo, MVAC, docetaxel-based regimen2–4144CT (Recist Criteria)yes
145
9 [63]Kubota et al.Onco-target2017retrospective multi-center1995–2017noGem/Cis (21), Gem/Carbo (76), MVAC (4)2–4 q3w101CT (Recist Criteria)yes
133
10 [64]Rajput et al.Urology2011retrospective single-center2003–2010novariable (MVAC, CGI, MVAC + Bevacizumab, GTA, IAG, CG, GT)1–7 (median 4)26CT or MRIno
56
11 [65]Shigeta et al.Urol Oncol2021retrospective single-center1990–2016noGC (25), MVAC (11) 36CT (Recist Criteria) or MRIyes (only)
53
Table 2. Descriptive presentation of patient characteristics.
Table 2. Descriptive presentation of patient characteristics.
StudyControl
cT3/T4ntotal229246
naverage3841
# of studies66
%weighted84.90%87.22%
cT2ntotal7457
naverage1210
# of studies66
%weighted40.84%37.59%
c T1ntotal4027
naverage107
# of studies44
%weighted31.13%32.92%
cN+ntotal132134
naverage2222
# of studies66
%weighted65.65%76.12%
surgery: openntotal188336
naverage3867
# of studies55
%weighted76.76%89.96%
surgery: laparoscopic/roboticntotal6739
naverage138
# of studies55
%weighted34.79%13.49%
Abbreviation: c (clinical staging).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Oswald, D.; Pallauf, M.; Deininger, S.; Törzsök, P.; Sieberer, M.; Eiben, C. Neoadjuvant Chemotherapy before Nephroureterectomy in High-Risk Upper Tract Urothelial Cancer: A Systematic Review and Meta-Analysis. Cancers 2022, 14, 4841. https://doi.org/10.3390/cancers14194841

AMA Style

Oswald D, Pallauf M, Deininger S, Törzsök P, Sieberer M, Eiben C. Neoadjuvant Chemotherapy before Nephroureterectomy in High-Risk Upper Tract Urothelial Cancer: A Systematic Review and Meta-Analysis. Cancers. 2022; 14(19):4841. https://doi.org/10.3390/cancers14194841

Chicago/Turabian Style

Oswald, David, Maximilian Pallauf, Susanne Deininger, Peter Törzsök, Manuela Sieberer, and Christian Eiben. 2022. "Neoadjuvant Chemotherapy before Nephroureterectomy in High-Risk Upper Tract Urothelial Cancer: A Systematic Review and Meta-Analysis" Cancers 14, no. 19: 4841. https://doi.org/10.3390/cancers14194841

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