Next Article in Journal
Chimeric Antigen Receptor-T Cell Therapy for Lymphoma: New Settings and Future Directions
Next Article in Special Issue
Overcoming Understaging and Undergrading in Upper Tract Urothelial Carcinoma. Comment on Ghoreifi et al. Modern Kidney-Sparing Management of Upper Tract Urothelial Carcinoma. Cancers 2023, 15, 4495
Previous Article in Journal
Congenital Tumors—Magnetic Resonance Imaging Findings with Focus on Rare Tumors
Previous Article in Special Issue
Clinical and Biological Differences between Upper Tract Carcinoma and Bladder Urothelial Cancer, Including Implications for Clinical Practice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Upper Tract Urothelial Carcinoma: A Narrative Review of Current Surveillance Strategies for Non-Metastatic Disease

by
Jakob Klemm
1,2,*,†,
Kensuke Bekku
2,3,†,
Mohammad Abufaraj
2,4,
Ekaterina Laukhtina
2,5,
Akihiro Matsukawa
2,6,
Mehdi Kardoust Parizi
2,
Pierre I. Karakiewicz
7 and
Shahrokh F. Shariat
2,5,8,9,10,11,12
1
Department of Urology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany
2
Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
3
Department of Urology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama 700-8525, Japan
4
Division of Urology, Department of Special Surgery, Jordan University Hospital, The University of Jordan, Amman 11733, Jordan
5
Institute for Urology and Reproductive Health, Sechenov University, 119991 Moscow, Russia
6
Department of Urology, Jikei University School of Medicine, Tokyo 105-8461, Japan
7
Cancer Prognostics and Health Outcomes Unit, University of Montreal Health Centre, Montreal, QC H2X 3E4, Canada
8
Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman 11942, Jordan
9
Karl Landsteiner Institute of Urology and Andrology, 1090 Vienna, Austria
10
Department of Urology, Weill Cornell Medical College, New York, NY 10065, USA
11
Department of Urology, University of Texas Southwestern, Dallas, TX 75390, USA
12
Department of Urology, Second Faculty of Medicine, Charles University, 252 50 Prague, Czech Republic
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2024, 16(1), 44; https://doi.org/10.3390/cancers16010044 (registering DOI)
Submission received: 17 November 2023 / Revised: 11 December 2023 / Accepted: 19 December 2023 / Published: 20 December 2023

Abstract

:

Simple Summary

Upper tract urinary carcinoma (UTUC) is a rare type of cancer affecting the urinary system. Patients with UTUC often undergo surgeries like kidney-sparing surgery or radical nephroureterectomy. However, even after treatment, there remains a risk of the cancer recurring in different parts of the body. This narrative review aims to better understand the frequency and locations of such recurrences, which is crucial for effectively monitoring patients after their initial treatment. Currently, there is limited information on the optimal methods for tracking patients post-surgery, and on how early detection of cancer, before the appearance of symptoms, might improve health outcomes. This article presents the most important current guideline recommendations and elucidates the evidence behind them. Exploring new imaging technologies and improving methods for assessing patient risk, can potentially lead to more personalized and effective monitoring plans in the near future.

Abstract

Non-metastatic upper urinary tract carcinoma (UTUC) is a comparatively rare condition, typically managed with either kidney-sparing surgery (KSS) or radical nephroureterectomy (RNU). Irrespective of the chosen therapeutic modality, patients with UTUC remain at risk of recurrence in the bladder; in patients treated with KSS, the risk of recurrence is high in the remnant ipsilateral upper tract system but there is a low but existent risk in the contralateral system as well as in the chest and in the abdomen/pelvis. For patients treated with RNU for high-risk UTUC, the risk of recurrence in the chest, abdomen, and pelvis, as well as the contralateral UT, depends on the tumor stage, grade, and nodal status. Hence, implementing a risk-stratified, location-specific follow-up is indicated to ensure timely detection of cancer recurrence. However, there are no data on the type and frequency/schedule of follow-up or on the impact of the recurrence type and site on outcomes; indeed, it is not well known whether imaging-detected asymptomatic recurrences confer a better outcome than recurrences detected due to symptoms/signs. Novel imaging techniques and more precise risk stratification methods based on time-dependent probabilistic events hold significant promise for making a cost-efficient individualized, patient-centered, outcomes-oriented follow-up strategy possible. We show and discuss the follow-up protocols of the major urologic societies.

1. Introduction

Upper urinary tract carcinoma (UTUC) is a relatively rare malady, constituting approximately 5–10% of urothelial carcinomas [1]. The treatment of non-metastatic UTUC is based on risk stratification into low- vs. high-risk tumors, with preferred therapeutic options being kidney-sparing surgery (KSS) or radical nephroureterectomy (RNU) with perioperative platinum-based chemotherapy when possible and indicated [2]. Indeed, the current guidelines from the European Association of Urology (EAU), the American Urological Association (AUA), and the National Comprehensive Cancer Network (NCCN) recommend offering neoadjuvant or adjuvant chemotherapy to high-risk UTUC patients, as they have been shown to improve disease-free survival (DFS), at least in non-metastatic UTUC patients [2,3,4,5]. Recognizing the potential for metachronous bladder tumors, local recurrences, or distant metastases after RNU or KSS, the EAU, AUA, and NCCN guidelines advocate robust surveillance protocols for UTUC patients [2,3,4]. However, evidence on follow-up strategies after definitive treatment for non-metastatic UTUC is low. For this reason, this article aims to summarize the available evidence on surveillance after surgery for non-metastatic UTUC with curative intent.

2. Evidence Acquisition

We searched the PubMed database up to 1 October 2023 using pre-defined search criteria as follows: (Upper Tract Urothelial Carcinoma) OR (Renal Pelvic Urothelial Cancer) OR (Ureteral Urothelial Cancer) AND (Surveillance) OR (Follow-up). In addition, we assessed the references in the EAU, AUA, and NCCN guidelines.

3. Prognostic Factors for UTUC

Assessment of established prognostic factors can help better understand which patients are at risk for disease recurrence and/or progression after definitive surgical therapy with curative intent for non-metastatic UTUC. However, it is essential to recognize that UTUC and bladder urothelial carcinoma are distinct entities, characterized by unique clinical, pathological, practical, and molecular factors [6].

3.1. Patient Related Factors

Advanced age and worse performance status have been significantly associated with decreased CSS in several studies including a systematic review including all published articles until December 2014 (Hazard ratio (HR) 1.02) [7]. Studies including a SEER database UTUC cohort analysis revealed that stage, grade, age, and sex were significantly associated with Cancer-Specific Survival (CSS) in 9208 non-metastatic UTUC patients treated with RNU [8,9,10,11]. However, as opposed to bladder cancer, sex was not associated with CSS in UTUC patients according to individual studies and a meta-analysis of 39,759 UTUC patients (pooled HR 0.94, 95% confidence interval (CI) = 0.89–1.00) [11,12]. Moreover, being a smoker at diagnosis has been shown to increase the risk of recurrence and mortality. In a retrospective study involving 864 clinically non-metastatic UTUC patients treated with RNU, of whom 202 were identified as heavy long-term smokers, the study found that heavy long-term smoking was significantly associated with advanced disease, disease recurrence, and worse CSS [13].
Although Lynch syndrome patients are at higher risk of developing UTUC [14], high microsatellite instability (MSI), which is a screening tool for Lynch syndrome in UTUC patients, seems to be associated with better overall survival [15].
Furthermore, residing in Balkan endemic nephropathy (BEN) areas has been independently linked to an increased risk of bladder recurrence following RNU for UTUC, with patients from these regions experiencing higher rates compared to those outside BEN areas (HR 1.81; p = 0.01) [16]. Additionally, a propensity-matched survival analysis revealed that patients in Taiwan and China who have undergone kidney transplantation are at a higher risk of developing UTUC than those without such a transplant history [17].

3.2. Tumor Stage and Grade

Despite the insights gained from these patient-related prognostic factors, the TNM stage and tumor grade continue to be the most influential prognostic indicators for UTUC [8,9,18,19,20]. A large retrospective study, including 13,314 patients with primary UTUC in the Netherlands between 1993 and 2017, reported five-year CSSs of 86% (95% CI = 84–87), 70% (95% CI = 68–72), and 44% for Ta/Tis, T1-T2, and non-organ confined tumors, respectively [21]. Simultaneously, a survival analysis of 6826 patients who underwent RNU for non-metastatic UTUC from the SEER database revealed a decreasing five-year CSS with increasing T stage: 86% for T1 high-grade N0 disease, 78% for T2 N0 disease, 63% for T3 N0, and 39% for T4 N0 or any N1–3 disease [22].
Moreover, M0 UTUC patients with lymph node involvement experience very poor five-year OS rates of approximately 15–30% [23,24,25]. In addition, extracapsular extension and lymph node density have been reported to be strong predictors of survival outcomes in N+ UTUC patients [26].

3.3. Tumor Characteristics

Tumor location has been shown to be associated with worse outcomes in univariable analyses [27]. According to a systematic review of 14,895 RNU patients, ureteral tumor location has a negative impact on CSS compared to pelvicalyceal tumors after adjusting for covariates (pooled HR of 1.52, p < 0.001) [28]. Multifocality is another factor that has been associated with disease recurrence and CSS in 2492 RNU patients, of whom 590 had multifocal tumors (HR 1.43, p = 0.019 and HR 1.46, p = 0.027, respectively) [29]. Furthermore, clinical tumor size has been linked to T stage, as shown by a large multi-institutional retrospective study of 932 RNUs for non-metastatic UTUC. The study demonstrated that a tumor size of 2 cm was the optimal cutoff to identify patients at risk for >T2 disease (decision curve analysis: clinical net benefit of 0.09 and a net reduction of 8 per 100 patients) [30]. An analysis of 4657 patients from the SEER database confirmed these findings: each 1 cm increase in tumor size translated into an adjusted odds ratio of 1.25 (p > 0.001) [31].

3.4. Other Pathological Features

Similarly to bladder UC, histological subtypes including micropapillary, squamous, and/or sarcomatoid were associated with worse CSS in a systematic review including 12,865 UTUC patients (pooled HR 2.00, 95% CI = 1.57–2.56) [32]. Additionally, lymphovascular invasion (LVI) was demonstrated to be an independent predictor of CSS in a large multicenter series including 763 UTUC patients who underwent RNU without neoadjuvant chemotherapy (HR 3.3, p = 0.005) [33]. Finally, as with other malignancies, positive surgical margins are associated with an increased risk of disease recurrence after RNU (HR 2.7, p = 0.001) [34].
In summary, risk factors serve as key indicators in forecasting the likelihood for a patient to experience recurrence of a disease, and, as such, they are integral to risk stratification [35], which in turn can help in patient counseling and shared decision-making based on evidence regarding intensification or deintensification of adjuvant therapy and surveillance. Indeed, prognostic tools integrating the above risk factors are the data-driven backbone to the development of effective and cost-serving surveillance protocols.

4. Current Surveillance Protocols after RNU

Leading urological and oncological associations such as the NCCN, AUA, and EAU propose surveillance protocols for both KSS- and RNU-treated patients. These protocols typically encompass a combination of regular cystoscopy, cytology, and imaging (Table 1, Table 2 and Table 3) [2,3,4]. The specific follow-up protocol for UTUC is generally determined based on the risk stratification group and the type of definitive therapy performed (i.e., KSS or RNU). Table 1, Table 2 and Table 3 provide an overview of the exact surveillance protocols of the EAU, AUA, and NCCN guidelines. Although little evidence exists on the value of these follow-up protocols, there is a rationale behind each of the surveillance modalities and regimens. In general, evidence shows that patients with asymptomatically detected recurrence have better overall survival, CSS, and recurrence-free survival than symptomatic UTUC patients [36].

4.1. Bladder Recurrence

Since bladder recurrence is common after definitive treatment for UTUC, cystoscopy is an integral part of the follow-up. The risk of bladder recurrence was assessed by two studies that created a nomogram to predict bladder recurrence at different time points. The UTUC collaboration group analyzed 1839 UTUC patients treated with open or laparoscopic RNU and found an intravesical recurrence in 31% of the patients with a median follow-up of 45 months [19]. Similarly, a retrospective study from Japan showed an intravesical recurrence rate of 29% after 5 years in 754 UTUC patients treated with RNU [37]. Nonetheless, no specific thresholds were found regarding intravesical recurrence risk and endoscopic surveillance.
Conditional survival analyses take into account that a patient’s likelihood of bladder recurrence decreases with increasing recurrence-free survival. For example, Shigeta et al. analyzed 364 UTUC patients treated with open or laparoscopic RNU and found a 5-year intravesical recurrence-free survival (IVRFS) rate of 41.5%. However, the 5-year conditional IVRFS rate after 4 years of survivorship was 96.7% [38]. Similarly, the 5-year IVRFS rate of 3544 UTUC patients who underwent RNU was 55% but the conditional 5-year IVRFS rate increased to 90% after 4 years of survivorship [39]. These data support that the guideline recommendations cover a timeframe of 5 years. However, the retrospective study design and the relatively small patient cohort of the Shigeta study represent major limitations, meaning that no clear conclusions can be drawn regarding endoscopic surveillance protocols.
Martini et al. recommended continuing cystoscopy follow-up for more than 10 years, especially for patients with a prior history of bladder cancer. This recommendation is supported by Weibull regression models for the hazard rate of recurrence and other-cause mortality, which indicate a higher risk of recurrence than other-cause mortality for patients under 70 years of age [40]. The impact of a single dose of intravesical post-operative chemotherapy lowers the risk of intravesical recurrence [41] and could lead to a de-escalation of cystoscopies, but it is unclear to what extent. After KSS, the rate of IVR is likely to be higher due to many interventions with ureteroscopies, which have been shown to lead to higher rate of IVR due to presumed seeding [42].

4.2. Local and Distant Recurrence

Unlike intravesical recurrences, which can be effectively monitored through frequent cystoscopies, the emergence of loco-regional and distant recurrences necessitates regular abdominopelvic and chest imaging. Generally, local recurrence rates after RNU are reported to range from 5 to 32% in retrospective studies [43,44,45,46,47,48]. In a study conducted by Martini et al., the risk of non-bladder recurrence was assessed in 1378 prospectively collected UTUC patients treated with RNU across various European academic centers. Patients were classified into two groups based on their prior history of bladder cancer. After 2 years, the risk of non-bladder recurrence was 42% for patients without and 47% for those with a history of prior bladder cancer. Considering that European guidelines advocate a deintensification of imaging after 2 years, the authors suggest maintaining a semiannual imaging schedule until after the fourth year when the risk curve for non-bladder recurrence post RNU significantly plateaus [40].
A study from Japan that followed 733 UTUC patients post RNU found a non-bladder recurrence rate of 34% within 5 years, with most recurrences occurring within the first 3 years following treatment [49]. The study also indicated a correlation between the location of the primary tumor and patterns of recurrence or metastasis. Lower and middle ureter tumors were more prone to local recurrence in the pelvic cavity, while tumors in the renal pelvis or upper ureter were more likely to metastasize to the lungs or liver [49].
In terms of metastatic patterns in UTUC patients, primary metastatic UTUC patients are most likely to experience lung (36.1%) bone (27%), or liver (19.1%) metastasis, with brain metastasis only in 1.6% of cases (SEER: 9436 primary UTUC patients) [50]. These patterns were corroborated by Tanaka et al., who found that 30% of 733 low- and high-risk UTUC patients experienced recurrence within 3 years post RNU, with distant recurrences accounting for 56% of these cases. The predominant sites of metastasis were the lungs, liver, and bones [49]. Other studies have reported similar metastasis rates post RNU, ranging from 8.3% to 46% [44,45,46,47,48,51]. Consequently, abdominal and chest imaging are part of the surveillance protocols of the NCCN, AUA, and EAU guidelines, allowing for the detection of these metastasis patterns.

4.3. Risk Stratification and Surveillance

Risk stratification plays a crucial role in striking the right equilibrium between the intensity and frequency of surveillance modalities tailored for each patient. A retrospective analysis of 1029 UTUC patients who underwent RNU in Canada revealed that 73% of patients experienced recurrence at any site (urothelial, local, and distant recurrences) within the first two years [52]. Based on these findings, the authors suggested a surveillance protocol based on three risk groups: low risk (pTa-T1, pN0, low grade, no LVI, unifocality), intermediate risk (pTa-T1, pN0, +/− high grade, +/− LVI, +/− multifocality), and high risk (≥pT2 and/or pN+), with the first two years post RNU demanding heightened surveillance, particularly for high-risk patients [52].
In a retrospective analysis of 426 UTUC patients who underwent RNU, Momota et al. sought to assess the cost-effectiveness of surveillance protocols. Initially, a pathology-based surveillance protocol (normal risk: ≤pT2N0; high risk: N0 with pT3 or LVI+; very high risk: pT4, positive surgical margin, or lymph node involvement (cN+ or pN+)) was utilized; however, it fell short in its ability to effectively distinguish between patients with a high risk of recurrence and those without. The authors subsequently improved cost-effectiveness by implementing a risk-score-based surveillance protocol which weighted different risk factors, resulting in a 55% reduction in the cost of 5-year surveillance [53]. However, it should be noted that the authors did not include grading in their risk stratification, resulting in risk groups with mixed grading.
In a retrospective analysis of 714 UTUC patients who underwent RNU, Shigeta et al. found that smokers had a higher risk of UTUC-related death compared to non-smokers according to Weibull model estimates. The authors suggest that extending surveillance may be necessary for this population to detect and manage potential recurrences or metastases [54]. Yet, similar to the Momota study, this study also had a major limitation in risk stratification, as it was not based on the standard pathological features or risk groups recommended by guidelines.
The key questions are the optimal protocol and the duration of surveillance. Lindner et al. studied time-to-tumor recurrence in 54 UTUC patients post RNU and 14 UTUC patients post KSS. They discovered that 38.9% of patients post RNU developed distant metastasis, with the vast majority (85.7%) occurring within the first year post surgery. Only 9.5% and 4.8% occurred in the second and third years, respectively [55]. These findings underscore the rationale of the EAU guidelines’ recommendation to de-intensify imaging after the first two years post RNU.
Finally, the delivery of adjuvant systemic therapy with platinum-based combination has shown to lower recurrence rates and may impact the intensity of surveillance imaging [56].
In conclusion, considering their elevated risk of recurrence, patients with UTUC post RNU necessitate a meticulous follow-up regimen. This should encompass routine chest and abdominopelvic imaging, supplemented with periodic cystoscopy. It is particularly crucial to maintain an intensified monitoring schedule during the initial two years of surveillance.

5. Current Surveillance Protocols after Endoscopic Treatment

A kidney-sparing approach is generally recommended to reduce morbidity associated with radical surgery [2] for UTUC tumors with low-risk features including all of the following: unifocal disease, tumor size < 2 cm, negative cytology, low-grade ureterorenoscopy (URS) biopsy, and no invasive aspect on computed tomography (CT). Current surveillance strategies for UTUC patients after KSS (either for low-risk tumors or for imperative indications such as solitary kidney, bilateral UTUC, chronic kidney disease, or any other comorbidity compromising the use of RNU) include imaging and periodic cystoscopy but also URS as a standard (Table 1, Table 2 and Table 3) [2].

5.1. Recurrence Rates

The design of surveillance strategies is fundamentally shaped by recurrence rates and patterns. This principle is illustrated in a recent systematic review that scrutinized the oncologic outcome of endoscopic surgeries for UTUC including 1091 patients from twenty studies with mostly low-grade tumors. The authors found a pooled bladder recurrence rate of 35% (95% CI 28–42.3: I2 = 48%) after retrograde URS and 17.7% (95% CI 6.5–32.1: I2 = 29%) after antegrade URS. Additionally, the pooled rate of upper urinary tract recurrence was 56.4% (95% CI 41.2–70.9: I2 = 93%) after retrograde treatment and 36.2% (95% CI 25.5–47.6: I2 = 57%) after antegrade treatment [57].
Interestingly, Mohapatra et al. retrospectively analyzed patients who initially received endoscopic treatment at two large tertiary referral centers in the U.S. for low- and high-risk UTUC and found that 80% of their cohort experienced disease progression to high-risk UTUC, triggering an RNU within 5 years [58].
Given these results, it is crucial to implement a rigorous follow-up schedule involving repeated cystoscopy and ureteroscopy, particularly after endoscopic treatment. This strategy ensures timely detection of any disease recurrence or progression, and keeps the option for RNU open, preserving the critical window of opportunity for intervention if required.

5.2. Cytology

Urinary cytology serves as a crucial tool for diagnosing UTUC and its subsequent surveillance, particularly in high-risk tumors, regardless of the definitive therapy approach. This tool is widely recommended across various guidelines due to its high specificity and non-invasive nature [2,3,4]. The adoption of the Paris System for Reporting Urinary Cytology (TPS) has led to varying results with respect to sensitivity and specificity. Studies have reported sensitivity ranging from 19% to 82%, while specificity ranges from 86% to 100% for primary diagnosis [59]. Interestingly, these results align with a meta-analysis from the pre-TPS era, which demonstrated a pooled sensitivity of 53% (95% CI = 42–64; I2 = 86%) and a pooled specificity of 90% (95% CI = 85–93; I2 = 0%) for UTUC detection in upper urinary tract cytology during primary diagnosis [60].
Given these findings, cytology is also recommended for UTUC follow-up. However, it is important to note that the impact of the Paris System on sensitivity and specificity during surveillance is yet to be determined.

5.3. Second-Look URS and Endoscopic Follow-Up

EAU and AUA guidelines agree on an early repeated URS which should be performed one to three and six months after KSS [2,3]. Villa et al. demonstrated a high cancer detection rate of 51.2% during the second URS 6–8 weeks after initial URS treatment in 41 UTUC patients with high- or low-risk UTUC. Patients who had a positive result during the second URS had an 81.3% likelihood of also having a positive result during the third URS, in contrast to a cancer detection rate of 41.2% for the third URS following a negative second URS result [61]. The authors therefore strongly recommend a second-look URS within 6–8 weeks after initial endoscopic management.
Several studies provide further evidence for the importance of close endoscopic follow-up in UTUC patients who undergo KSS. Kawada et al. conducted a systematic review and found that endoscopically managed tumors had similar oncologic outcomes to those managed with RNU, but recurrence in the upper urothelial tract was observed in 28–85% of patients across the studies [62]. Lindner et al. reported a stable recurrence rate after KSS (endoscopic treatment and ureterectomy) at 12.5% to 20.5% per year during the first 5 years after surgery, with six upper urinary tract recurrences, two bladder recurrences, and two lymph node and/or distant metastases in 14 KSS patients (57.1% high-grade tumors). As most of the recurrences after KSS concerned the bladder or upper urinary tract (six upper urinary tract recurrences, two bladder recurrences), the authors suggest, contrary to the current EAU, AUA, and NCCN guidelines, that cystoscopies and URS should not be deintensified after 2 years of follow-up [55].
This is further emphasized by Seisen et al., who analyzed 42 primary endoscopically treated patients with mostly low-grade tumors. The local recurrence-free survival defined as recurrence in the operation site was 35.7% after endoscopic treatment. Additionally, Kaplan–Meier curves showed a consistent incidence of local recurrence over the 5-year follow-up period [63]. Moreover, Hendriks et al. found a higher IVR rate for high-risk UTUC patients treated with KSS (endoscopic treatment and ureterectomy) compared to RNU in a propensity-score-matched cohort based on EAU risk stratification (52% for KSS vs. 32% for RNU; p = 0.029) [64].
In conclusion, while the existing evidence may be of a low level, it, nonetheless, underscores the necessity for rigorous surveillance, including URS, following the endoscopic management of UTUC.
Specifically, because KSS is an alternative to RNU, it has promise to be safe while retaining the renal unit.

6. Current Surveillance Protocols after Segmental or Distal Ureterectomy

Segmental and distal ureterectomy represent non-endoscopic kidney-sparing approaches that are recommended for low-risk UTUC tumors [2]. Seisen et al. assessed oncologic outcomes of ureterectomies compared with RNU in a systematic review including 586 segmental ureterectomy patients from comparative studies and reported no differences in CSS, OS, and RFS between the two groups [65]. Seisen also showed that the 5-year local RFS, defined as recurrence in the operation site, ranged from 37% to 91% across studies [66,67,68,69]. Similarly, Fang et al. included 983 ureterectomy patients from comparative studies and reported no differences in oncologic outcomes between ureterectomy and RNU (CSS: HR 0.90, p = 0.33, OS: HR 0.98, p = 0.93, and RFS: HR 1.06, p = 0.72). However, patients undergoing ureterectomy were more likely to harbor favorable pathological features. The cumulative recurrence rate 5 years after surgery ranged from 16% to 72% across three studies comprising 165 patients [66,68,70,71].
Due to ureteroureterostomy or ureteroneocystostomy, which complicate endoscopic follow-up, surveillance after ureterectomy was frequently based on the follow-up regimen of RNUs across retrospective studies [66,68]. Yet, Kim et al. retrospectively analyzed 394 RNU patients and 44 segmental ureterectomy patients and found no significant differences regarding 3-year PFS and IVRFS (68% vs. 73%: p = 0.9 and 42% vs. 37%: p = 0.8, respectively). Interestingly, the authors stated the use of semiannual ureteroscopy in their follow-up regimen for patients treated with segmental ureterectomy [72]. While ureterectomies are considered kidney-sparing approaches for UTUC, the surveillance protocols recommended by guidelines do not distinguish between endoscopic management and ureterectomy, leading to non-compliance with guideline protocols, especially after ureterectomy. As of now, this issue has not been addressed in the guidelines.

7. Discussion

The existing guideline recommendations for surveillance following definitive therapy for UTUC largely rely on expert opinions and low-level evidence due to the relative rarity of this disease, which has led to a dearth of large prospective randomized clinical trials that could bolster the evidence. The available data frequently hail from retrospective studies involving small cohorts. Most existing data on surveillance, recurrence, and progression center around RNU. Given that UTUC patients, particularly those in the high-risk category, are generally at substantial risk of recurrence or progression, frequent follow-up examinations seem necessary.
The challenge for healthcare providers lies in delivering an appropriate surveillance protocol with the correct frequency to suit each individual patient and tumor type. As highlighted by Momota et al., risk stratification plays a pivotal role in selecting the appropriate patient and surveillance protocol, and it significantly impacts healthcare system costs, resource utilization, and patient convenience/quality of life [53]. Currently, risk stratification largely depends on tumor T and N stage as well as grade. However, the advent of next-generation sequencing (NGS) in recent years has led to the discovery of molecular subtypes of various tumor entities, which are gradually being incorporated into clinical practice. Additionally, liquid biopsies, which utilize blood or other body fluids like urine, are being explored and show promise in the detection of UTUC [73]. The utilization of biomarkers, in general, has the potential to enhance surveillance protocols. The integration of biomarkers and liquid biopsies could lead to less-invasive disease monitoring, improved detection accuracy, and the identification of novel therapeutic targets, thereby potentially reducing the risk of recurrence. Incorporating such biomarkers into routine surveillance could transform patient management, allowing for more personalized and effective monitoring strategies.
A systematic review assessing UTUC alterations revealed significant differences between UTUC and urothelial bladder cancer, particularly in areas such as activated FGFR3 signaling, the extent of altered somatic expression of DNA mismatch repair genes, and individual UTUC molecular subtypes [74]. The impact of these discoveries on future treatments and, consequently, future surveillance protocols in UTUC is in its burgeoning phase and much is expected.
Furthermore, emerging urinary biomarkers might offer the potential to reduce patient discomfort by providing equivalent diagnostic value without necessitating invasive ureterorenoscopy during follow-up. Territo et al. evaluated the diagnostic worth of EpiCheck, a urine test based on the analysis of 15 DNA methylation biomarkers, and the results were promising with a sensitivity/negative predictive value (NPV) for high-grade tumors of 96%/97%, compared to 71%/86% for cytology [75]. However, additional evidence is required to integrate these novel urinary biomarkers into surveillance protocols.
Additionally, by enhancing precise imaging techniques, patients can not only be directed towards the treatment approach that best suits their individual needs, but also the accuracy of risk stratification can be improved. This enhancement may help circumvent unnecessary and invasive surveillance methods such as URS. Moreover, advancements in imaging such as PET/CT and PET/MRI, which seem to be promising imaging tools for the detection of lymph nodes and distant metastases in urothelial bladder cancer, hold the potential to change surveillance protocols and risk stratification by offering a more accurate nodal and distant assessment of the disease [76].

8. Conclusions

The evidence supporting surveillance protocols following definitive therapy for UTUC is currently sparse, and predominantly reliant on low-level evidence and expert opinion. Given the rarity of UTUC, conducting large prospective randomized clinical trials may prove challenging, underscoring the need for refined risk stratification methods. Surveillance protocols may be optimized in the future to meet the individual needs of each patient by enhancing risk stratification accuracy through more precise imaging and/or the implementation of novel urine and blood-based biomarkers.

Author Contributions

Conceptualization J.K., K.B., S.F.S. and P.I.K.; methodology J.K., K.B., E.L., A.M., M.A. and M.K.P.; literature research J.K., K.B. and E.L.; writing—original draft preparation J.K., K.B. and M.A.; writing—review and editing S.F.S., P.I.K., E.L., A.M. and M.K.P.; supervision S.F.S., M.A. and P.I.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Conflicts of Interest

The authors declare no conflict of interest.

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]
  2. Rouprêt, M.; Seisen, T.; Birtle, A.J.; Capoun, O.; Compérat, E.M.; Dominguez-Escrig, J.L.; Gürses Andersson, I.; Liedberg, F.; Mariappan, P.; Hugh Mostafid, A.; et al. European Association of Urology Guidelines on Upper Urinary Tract Urothelial Carcinoma: 2023 Update. Eur. Urol. 2023, 84, 49–64. [Google Scholar] [CrossRef]
  3. Coleman, J.A.; Clark, P.E.; Bixler, B.R.; Buckley, D.I.; Chang, S.S.; Chou, R.; Hoffman-Censits, J.; Kulkarni, G.S.; Matin, S.F.; Pierorazio, P.M.; et al. Diagnosis and Management of Non-Metastatic Upper Tract Urothelial Carcinoma: AUA/SUO Guideline. J. Urol. 2023, 209, 1071–1081. [Google Scholar] [CrossRef]
  4. Flaig, T.W.; Spiess, P.E.; Agarwal, N.; Bangs, R.; Boorjian, S.A.; Buyyounouski, M.K.; Chang, S.; Downs, T.M.; Efstathiou, J.A.; Friedlander, T.; et al. Bladder Cancer, Version 3.2020, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2020, 18, 329–354. [Google Scholar] [CrossRef]
  5. 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]
  6. Robinson, B.D.; Vlachostergios, P.J.; 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, 2977. [Google Scholar] [CrossRef]
  7. Kim, H.S.; Jeong, C.W.; Kwak, C.; Kim, H.H.; Ku, J.H. Association between demographic factors and prognosis in urothelial carcinoma of the upper urinary tract: A systematic review and meta-analysis. Oncotarget 2017, 8, 7464–7476. [Google Scholar] [CrossRef]
  8. Collà Ruvolo, 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]
  9. Lughezzani, G.; Jeldres, C.; Isbarn, H.; Sun, M.; Shariat, S.F.; Alasker, A.; Pharand, D.; Widmer, H.; Arjane, P.; Graefen, M.; et al. Nephroureterectomy and segmental ureterectomy in the treatment of invasive upper tract urothelial carcinoma: A population-based study of 2299 patients. Eur. J. Cancer 2009, 45, 3291–3297. [Google Scholar] [CrossRef]
  10. Lughezzani, G.; Sun, M.; Perrotte, P.; Shariat, S.F.; Jeldres, C.; Budäus, L.; Latour, M.; Widmer, H.; Duclos, A.; Bénard, F.; et al. Gender-related differences in patients with stage I to III upper tract urothelial carcinoma: Results from the Surveillance, Epidemiology, and End Results database. Urology 2010, 75, 321–327. [Google Scholar] [CrossRef]
  11. 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] [PubMed]
  12. Mori, K.; Mostafaei, H.; Enikeev, D.V.; Lysenko, I.; Quhal, F.; Kimura, S.; Karakiewicz, P.I.; Egawa, S.; Shariat, S.F. Differential Effect of Sex on Outcomes after Radical Surgery for Upper Tract and Bladder Urothelial Carcinoma: A Systematic Review and Meta-Analysis. J. Urol. 2020, 204, 58–62. [Google Scholar] [CrossRef] [PubMed]
  13. Rink, M.; Xylinas, E.; Margulis, V.; Cha, E.K.; Ehdaie, B.; Raman, J.D.; Chun, F.K.; Matsumoto, K.; Lotan, Y.; Furberg, H.; et al. Impact of smoking on oncologic outcomes of upper tract urothelial carcinoma after radical nephroureterectomy. Eur. Urol. 2013, 63, 1082–1090. [Google Scholar] [CrossRef] [PubMed]
  14. Lonati, C.; Necchi, A.; Gómez Rivas, J.; Afferi, L.; Laukhtina, E.; Martini, A.; Ventimiglia, E.; Colombo, R.; Gandaglia, G.; Salonia, A.; et al. Upper Tract Urothelial Carcinoma in the Lynch Syndrome Tumour Spectrum: A Comprehensive Overview from the European Association of Urology—Young Academic Urologists and the Global Society of Rare Genitourinary Tumors. Eur. Urol. Oncol. 2022, 5, 30–41. [Google Scholar] [CrossRef] [PubMed]
  15. 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]
  16. Milojevic, B.; Dzamic, Z.; Grozdic Milojevic, I.; Bumbasirevic, U.; Santric, V.; Kajmakovic, B.; Janicic, A.; Durutovic, O.; Dragicevic, D.; Bojanic, N.; et al. Prognostic value of Balkan endemic nephropathy and gender on upper tract urothelial carcinoma outcomes after radical nephroureterectomy: A cohort study. Urol. Oncol. 2021, 39, 786.e9–786.e16. [Google Scholar] [CrossRef] [PubMed]
  17. Luo, H.L.; Chiang, P.H.; Cheng, Y.T.; Chen, Y.T. Propensity-Matched Survival Analysis of Upper Urinary Tract Urothelial Carcinomas between End-Stage Renal Disease with and without Kidney Transplantation. BioMed Res. Int. 2019, 2019, 2979142. [Google Scholar] [CrossRef]
  18. Mbeutcha, A.; Rouprêt, M.; Kamat, A.M.; Karakiewicz, P.I.; Lawrentschuk, N.; Novara, G.; Raman, J.D.; Seitz, C.; Xylinas, E.; Shariat, S.F. Prognostic factors and predictive tools for upper tract urothelial carcinoma: A systematic review. World J. Urol. 2017, 35, 337–353. [Google Scholar] [CrossRef]
  19. Xylinas, E.; Kluth, L.; Passoni, N.; Trinh, Q.-D.; Rieken, M.; Lee, R.K.; Fajkovic, H.; Novara, G.; Margulis, V.; Raman, J.D.; et al. Prediction of Intravesical Recurrence After Radical Nephroureterectomy: Development of a Clinical Decision-making Tool. Eur. Urol. 2014, 65, 650–658. [Google Scholar] [CrossRef]
  20. Compérat, E.M.; Burger, M.; Gontero, P.; Mostafid, A.H.; Palou, J.; Rouprêt, M.; van Rhijn, B.W.G.; Shariat, S.F.; Sylvester, R.J.; Zigeuner, R.; et al. Grading of Urothelial Carcinoma and The New “World Health Organisation Classification of Tumours of the Urinary System and Male Genital Organs 2016”. Eur. Urol. Focus 2019, 5, 457–466. [Google Scholar] [CrossRef]
  21. 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] [PubMed]
  22. 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] [PubMed]
  23. Pelcovits, A.; Mueller-Leonhard, C.; Mega, A.; Amin, A.; Kim, S.P.; Golijanin, D.; Gershman, B. Outcomes of upper tract urothelial carcinoma with isolated lymph node involvement following surgical resection: Implications for multi-modal management. World J. Urol. 2020, 38, 1243–1252. [Google Scholar] [CrossRef] [PubMed]
  24. Roscigno, M.; Brausi, M.; Heidenreich, A.; Lotan, Y.; Margulis, V.; Shariat, S.F.; Van Poppel, H.; Zigeuner, R. Lymphadenectomy at the time of nephroureterectomy for upper tract urothelial cancer. Eur. Urol. 2011, 60, 776–783. [Google Scholar] [CrossRef] [PubMed]
  25. Lughezzani, G.; Jeldres, C.; Isbarn, H.; Shariat, S.F.; Sun, M.; Pharand, D.; Widmer, H.; Arjane, P.; Graefen, M.; Montorsi, F.; et al. A critical appraisal of the value of lymph node dissection at nephroureterectomy for upper tract urothelial carcinoma. Urology 2010, 75, 118–124. [Google Scholar] [CrossRef] [PubMed]
  26. Fajkovic, H.; Cha, E.K.; Jeldres, C.; Donner, G.; Chromecki, T.F.; Margulis, V.; Novara, G.; Lotan, Y.; Raman, J.D.; Kassouf, W.; et al. Prognostic value of extranodal extension and other lymph node parameters in patients with upper tract urothelial carcinoma. J. Urol. 2012, 187, 845–851. [Google Scholar] [CrossRef]
  27. Isbarn, H.; Jeldres, C.; Shariat, S.F.; Liberman, D.; Sun, M.; Lughezzani, G.; Widmer, H.; Arjane, P.; Pharand, D.; Fisch, M.; et al. Location of the primary tumor is not an independent predictor of cancer specific mortality in patients with upper urinary tract urothelial carcinoma. J. Urol. 2009, 182, 2177–2181. [Google Scholar] [CrossRef] [PubMed]
  28. Kaczmarek, K.; Lemiński, A.; Gołąb, A.; Słojewski, M. Survival differences of patients with ureteral versus pelvicalyceal tumours: A systematic review and meta-analysis. Arch. Med. Sci. 2021, 17, 603–612. [Google Scholar] [CrossRef]
  29. Chromecki, T.F.; Cha, E.K.; Fajkovic, H.; Margulis, V.; Novara, G.; Scherr, D.S.; Lotan, Y.; Raman, J.D.; Kassouf, W.; Bensalah, K.; et al. The impact of tumor multifocality on outcomes in patients treated with radical nephroureterectomy. Eur. Urol. 2012, 61, 245–253. [Google Scholar] [CrossRef]
  30. Foerster, B.; Abufaraj, M.; Mari, A.; Seisen, T.; Bandini, M.; Schweitzer, D.; Czech, A.K.; Moschini, M.; D’Andrea, D.; Bianchi, M.; et al. The Performance of Tumor Size as Risk Stratification Parameter in Upper Tract Urothelial Carcinoma (UTUC). Clin. Genitourin. Cancer 2021, 19, 272.e1–272.e7. [Google Scholar] [CrossRef]
  31. Collà Ruvolo, C.; Nocera, L.; Stolzenbach, L.F.; Wenzel, M.; Califano, G.; Tian, Z.; Verze, P.; Shariat, S.F.; Saad, F.; Briganti, A.; et al. Tumor Size Predicts Muscle-invasive and Non–organ-confined Disease in Upper Tract Urothelial Carcinoma at Radical Nephroureterectomy. Eur. Urol. Focus 2022, 8, 498–505. [Google Scholar] [CrossRef] [PubMed]
  32. Mori, K.; Janisch, F.; Parizi, M.K.; Mostafaei, H.; Lysenko, I.; Kimura, S.; Enikeev, D.V.; Egawa, S.; Shariat, S.F. Prognostic Value of Variant Histology in Upper Tract Urothelial Carcinoma Treated with Nephroureterectomy: A Systematic Review and Meta-Analysis. J. Urol. 2020, 203, 1075–1084. [Google Scholar] [CrossRef] [PubMed]
  33. Novara, G.; Matsumoto, K.; Kassouf, W.; Walton, T.J.; Fritsche, H.M.; Bastian, P.J.; Martínez-Salamanca, J.I.; Seitz, C.; Lemberger, R.J.; Burger, M.; et al. Prognostic role of lymphovascular invasion in patients with urothelial carcinoma of the upper urinary tract: An international validation study. Eur. Urol. 2010, 57, 1064–1071. [Google Scholar] [CrossRef] [PubMed]
  34. Colin, P.; Ouzzane, A.; Yates, D.R.; Audenet, F.; Pignot, G.; Arvin-Berod, A.; Merigot de Treigny, O.; Laurent, G.; Valeri, A.; Irani, J.; et al. Influence of positive surgical margin status after radical nephroureterectomy on upper urinary tract urothelial carcinoma survival. Ann. Surg. Oncol. 2012, 19, 3613–3620. [Google Scholar] [CrossRef] [PubMed]
  35. Shariat, S.F.; Capitanio, U.; Jeldres, C.; Karakiewicz, P.I. Can nomograms be superior to other prediction tools? BJU Int. 2009, 103, 492–495; discussion 495–497. [Google Scholar] [CrossRef] [PubMed]
  36. Horiguchi, H.; Hatakeyama, S.; Anan, G.; Kubota, Y.; Kodama, H.; Momota, M.; Kido, K.; Yamamoto, H.; Tobisawa, Y.; Yoneyama, T.; et al. Detecting asymptomatic recurrence after radical nephroureterectomy contributes to better prognosis in patients with upper urinary tract urothelial carcinoma. Oncotarget 2018, 9, 8746–8755. [Google Scholar] [CrossRef] [PubMed]
  37. Ishioka, J.; Saito, K.; Kijima, T.; Nakanishi, Y.; Yoshida, S.; Yokoyama, M.; Matsuoka, Y.; Numao, N.; Koga, F.; Masuda, H.; et al. Risk stratification for bladder recurrence of upper urinary tract urothelial carcinoma after radical nephroureterectomy. BJU Int. 2015, 115, 705–712. [Google Scholar] [CrossRef] [PubMed]
  38. Shigeta, K.; Kikuchi, E.; Hagiwara, M.; Ando, T.; Mizuno, R.; Abe, T.; Mikami, S.; Miyajima, A.; Nakagawa, K.; Oya, M. The Conditional Survival with Time of Intravesical Recurrence of Upper Tract Urothelial Carcinoma. J. Urol. 2017, 198, 1278–1285. [Google Scholar] [CrossRef]
  39. Ploussard, G.; Xylinas, E.; Lotan, Y.; Novara, G.; Margulis, V.; Rouprêt, M.; Matsumoto, K.; Karakiewicz, P.I.; Montorsi, F.; Remzi, M.; et al. Conditional Survival After Radical Nephroureterectomy for Upper Tract Carcinoma. Eur. Urol. 2015, 67, 803–812. [Google Scholar] [CrossRef]
  40. Martini, A.; Lonati, C.; Nocera, L.; Fallara, G.; Raggi, D.; Herout, R.; Zamboni, S.; Ploussard, G.; Predere, B.; Mattei, A.; et al. Oncologic Surveillance After Radical Nephroureterectomy for High-risk Upper Tract Urothelial Carcinoma. Eur. Urol. Oncol. 2022, 5, 451–459. [Google Scholar] [CrossRef]
  41. Hwang, E.C.; Sathianathen, N.J.; Jung, J.H.; Kim, M.H.; Dahm, P.; Risk, M.C. Single-dose intravesical chemotherapy after nephroureterectomy for upper tract urothelial carcinoma. Cochrane Database Syst. Rev. 2019, 5, Cd013160. [Google Scholar] [CrossRef]
  42. Yonese, I.; Ito, M.; Waseda, Y.; Kobayashi, S.; Toide, M.; Takazawa, R.; Koga, F. Impact of diagnostic ureteral catheterization on intravesical tumour recurrence following radical nephroureterectomy for upper tract urothelial carcinoma. World J. Urol. 2023, 41, 1869–1875. [Google Scholar] [CrossRef]
  43. 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]
  44. Gao, J.; Liu, J.; Liu, J.; Lin, S.; Ding, D. Survival and risk factors among upper tract urothelial carcinoma patients after radical nephroureterectomy in Northeast China. Front. Oncol. 2022, 12, 1012292. [Google Scholar] [CrossRef]
  45. Taweemonkongsap, T.; Nualyong, C.; Amornvesukit, T.; Leewansangtong, S.; Srinualnad, S.; Chaiyaprasithi, B.; Sujijantararat, P.; Tantiwong, A.; Soontrapa, S. Outcomes of surgical treatment for upper urinary tract transitional cell carcinoma: Comparison of retroperitoneoscopic and open nephroureterectomy. World J. Surg. Oncol. 2008, 6, 3. [Google Scholar] [CrossRef]
  46. Yoo, S.; You, D.; Jeong, I.G.; Hong, B.; Hong, J.H.; Ahn, H.; Kim, C.-S. Impact of Tumor Location on Local Recurrence After Nephroureterectomy for Upper Tract Urothelial Carcinoma: Implications for Adjuvant Radiotherapy. Clin. Genitourin. Cancer 2017, 15, e199–e204. [Google Scholar] [CrossRef]
  47. Li, X.; Cui, M.; Gu, X.; Fang, D.; Li, H.; Qin, S.; Yang, K.; Zhu, T.; Li, X.; Zhou, L.; et al. Pattern and risk factors of local recurrence after nephroureterectomy for upper tract urothelial carcinoma. World J. Surg. Oncol. 2020, 18, 114. [Google Scholar] [CrossRef]
  48. Chen, T.S.; Chen, Y.T.; Wang, H.J.; Chiang, P.H.; Yang, W.C.; Lee, W.C.; Chuang, Y.C.; Cheng, Y.T.; Kang, C.H.; Lee, W.C.; et al. The Prognostic Impact of Tumor Location in pT3N0M0 Upper Urinary Tract Urothelial Carcinoma: A Retrospective Cohort Study. Front. Oncol. 2022, 12, 850874. [Google Scholar] [CrossRef]
  49. Tanaka, N.; Kikuchi, E.; Kanao, K.; Matsumoto, K.; Kobayashi, H.; Ide, H.; Miyazaki, Y.; Obata, J.; Hoshino, K.; Shirotake, S.; et al. Metastatic Behavior of Upper Tract Urothelial Carcinoma After Radical Nephroureterectomy: Association with Primary Tumor Location. Ann. Surg. Oncol. 2014, 21, 1038–1045. [Google Scholar] [CrossRef]
  50. Hu, X.H.; Miao, J.; Qian, L.; Zhang, D.H.; Wei, H.B. The predictors and surgical outcomes of different distant metastases patterns in upper tract urothelial carcinoma: A SEER-based study. Front. Surg. 2022, 9, 1045831. [Google Scholar] [CrossRef]
  51. Colin, P.; Ghoneim, T.P.; Nison, L.; Seisen, T.; Lechevallier, E.; Cathelineau, X.; Ouzzane, A.; Zerbib, M.; Long, J.A.; Ruffion, A.; et al. Risk stratification of metastatic recurrence in invasive upper urinary tract carcinoma after radical nephroureterectomy without lymphadenectomy. World J. Urol. 2014, 32, 507–512. [Google Scholar] [CrossRef]
  52. Locke, J.A.; Hamidizadeh, R.; Kassouf, W.; Rendon, R.A.; Bell, D.; Izawa, J.; Chin, J.; Kapoor, A.; Shayegen, B.; Lattouf, J.B.; et al. Surveillance guidelines based on recurrence patterns for upper tract urothelial carcinoma. Can. Urol. Assoc. J. 2018, 12, 243–251. [Google Scholar] [CrossRef]
  53. Momota, M.; Hatakeyama, S.; Yamamoto, H.; Iwamura, H.; Tobisawa, Y.; Yoneyama, T.; Yoneyama, T.; Hashimoto, Y.; Koie, T.; Iwabuchi, I.; et al. Risk-stratified surveillance protocol improves cost-effectiveness after radical nephroureterectomy in patients with upper tract urothelial carcinoma. Oncotarget 2018, 9, 23047–23057. [Google Scholar] [CrossRef]
  54. Shigeta, K.; Kikuchi, E.; Abe, T.; Hagiwara, M.; Ogihara, K.; Anno, T.; Umeda, K.; Baba, Y.; Sanjo, T.; Shojo, K.; et al. A Novel Risk-based Approach Simulating Oncological Surveillance After Radical Nephroureterectomy in Patients with Upper Tract Urothelial Carcinoma. Eur. Urol. Oncol. 2020, 3, 756–763. [Google Scholar] [CrossRef]
  55. Lindner, A.K.; Pichler, M.; Maier, S.; Ulmer, H.; Gorreri, T.; Luger, A.K.; Barth, D.A.; Seeber, A.; Kocher, F.; Pichler, R. Optimization of postoperative surveillance protocols in upper tract urothelial cancer: A retrospective cohort study. Front. Oncol. 2023, 13, 1143030. [Google Scholar] [CrossRef]
  56. 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]
  57. Laukhtina, E.; Kawada, T.; Quhal, F.; Yanagisawa, T.; Rajwa, P.; von Deimling, M.; Pallauf, M.; Bianchi, A.; Majdoub, M.; Enikeev, D.; et al. Oncologic and Safety Outcomes for Retrograde and Antegrade Endoscopic Surgeries for Upper Tract Urothelial Carcinoma: A Systematic Review and Meta-analysis. Eur. Urol. Focus 2023, 9, 258–263. [Google Scholar] [CrossRef]
  58. Mohapatra, A.; Strope, S.A.; Liu, N.; Winer, A.; Benfante, N.E.; Coleman, J.A.; Vetter, J.; Murray, K.S. Importance of long-term follow-up after endoscopic management for upper tract urothelial carcinoma and factors leading to surgical management. Int. Urol. Nephrol. 2020, 52, 1465–1469. [Google Scholar] [CrossRef]
  59. Zhang, M.L.; Miki, Y.; Hang, J.-F.; Vohra, M.; Peyton, S.; McIntire, P.J.; VandenBussche, C.J.; Vohra, P. A review of upper urinary tract cytology performance before and after the implementation of The Paris System. Cancer Cytopathol. 2021, 129, 264–274. [Google Scholar] [CrossRef]
  60. Potretzke, A.M.; Knight, B.A.; Vetter, J.M.; Anderson, B.G.; Hardi, A.C.; Bhayani, S.B.; Figenshau, R.S. Diagnostic Utility of Selective Upper Tract Urinary Cytology: A Systematic Review and Meta-analysis of the Literature. Urology 2016, 96, 35–43. [Google Scholar] [CrossRef]
  61. Villa, L.; Cloutier, J.; Letendre, J.; Ploumidis, A.; Salonia, A.; Cornu, J.N.; Montorsi, F.; Traxer, O. Early repeated ureteroscopy within 6-8 weeks after a primary endoscopic treatment in patients with upper tract urothelial cell carcinoma: Preliminary findings. World J. Urol. 2016, 34, 1201–1206. [Google Scholar] [CrossRef]
  62. Kawada, T.; Laukhtina, E.; Quhal, F.; Yanagisawa, T.; Rajwa, P.; Pallauf, M.; von Deimling, M.; Bianchi, A.; Pradere, B.; Fajkovic, H.; et al. Oncologic and Safety Outcomes for Endoscopic Surgery Versus Radical Nephroureterectomy for Upper Tract Urothelial Carcinoma: An Updated Systematic Review and Meta-analysis. Eur. Urol. Focus 2023, 9, 236–240. [Google Scholar] [CrossRef]
  63. Seisen, T.; Nison, L.; Remzi, M.; Klatte, T.; Mathieu, R.; Lucca, I.; Bozzini, G.; Capitanio, U.; Novara, G.; Cussenot, O.; et al. Oncologic Outcomes of Kidney Sparing Surgery versus Radical Nephroureterectomy for the Elective Treatment of Clinically Organ Confined Upper Tract Urothelial Carcinoma of the Distal Ureter. J. Urol. 2016, 195, 1354–1361. [Google Scholar] [CrossRef]
  64. Hendriks, N.; Baard, J.; Beerlage, H.P.; Schout, B.M.A.; Doherty, K.S.G.; Pelger, R.C.M.; Kamphuis, G.M. Survival and Long-term Effects of Kidney-sparing Surgery Versus Radical Nephroureterectomy on Kidney Function in Patients with Upper Urinary Tract Urothelial Carcinoma. Eur. Urol. Open Sci. 2022, 40, 104–111. [Google Scholar] [CrossRef]
  65. Seisen, T.; Peyronnet, B.; Dominguez-Escrig, J.L.; Bruins, H.M.; Yuan, C.Y.; Babjuk, M.; Böhle, A.; Burger, M.; Compérat, E.M.; Cowan, N.C.; et al. Oncologic Outcomes of Kidney-sparing Surgery Versus Radical Nephroureterectomy for Upper Tract Urothelial Carcinoma: A Systematic Review by the EAU Non-muscle Invasive Bladder Cancer Guidelines Panel. Eur. Urol. 2016, 70, 1052–1068. [Google Scholar] [CrossRef]
  66. Dalpiaz, O.; Ehrlich, G.; Quehenberger, F.; Pummer, K.; Zigeuner, R. Distal ureterectomy is a safe surgical option in patients with urothelial carcinoma of the distal ureter. Urol. Oncol. 2014, 32, 34.e3–34.e8. [Google Scholar] [CrossRef]
  67. Fukushima, H.; Saito, K.; Ishioka, J.; Matsuoka, Y.; Numao, N.; Koga, F.; Masuda, H.; Fujii, Y.; Sakai, Y.; Arisawa, C.; et al. Equivalent survival and improved preservation of renal function after distal ureterectomy compared with nephroureterectomy in patients with urothelial carcinoma of the distal ureter: A propensity score-matched multicenter study. Int. J. Urol. 2014, 21, 1098–1104. [Google Scholar] [CrossRef]
  68. Bagrodia, A.; Kuehhas, F.E.; Gayed, B.A.; Wood, C.G.; Raman, J.D.; Kapur, P.; Derweesh, I.H.; Bensalah, K.; Sagalowsky, A.I.; Shariat, S.F.; et al. Comparative analysis of oncologic outcomes of partial ureterectomy vs radical nephroureterectomy in upper tract urothelial carcinoma. Urology 2013, 81, 972–977. [Google Scholar] [CrossRef]
  69. Colin, P.; Ouzzane, A.; Pignot, G.; Ravier, E.; Crouzet, S.; Ariane, M.M.; Audouin, M.; Neuzillet, Y.; Albouy, B.; Hurel, S.; et al. Comparison of oncological outcomes after segmental ureterectomy or radical nephroureterectomy in urothelial carcinomas of the upper urinary tract: Results from a large French multicentre study. BJU Int. 2012, 110, 1134–1141. [Google Scholar] [CrossRef]
  70. Fang, D.; Seisen, T.; Yang, K.; Liu, P.; Fan, X.; Singla, N.; Xiong, G.; Zhang, L.; Li, X.; Zhou, L. A systematic review and meta-analysis of oncological and renal function outcomes obtained after segmental ureterectomy versus radical nephroureterectomy for upper tract urothelial carcinoma. Eur. J. Surg. Oncol. 2016, 42, 1625–1635. [Google Scholar] [CrossRef]
  71. Hung, S.Y.; Yang, W.C.; Luo, H.L.; Hsu, C.C.; Chen, Y.T.; Chuang, Y.C. Segmental ureterectomy does not compromise the oncologic outcome compared with nephroureterectomy for pure ureter cancer. Int. Urol. Nephrol. 2014, 46, 921–926. [Google Scholar] [CrossRef] [PubMed]
  72. Kim, T.H.; Lee, C.U.; Kang, M.; Jeon, H.G.; Jeong, B.C.; Seo, S.I.; Jeon, S.S.; Lee, H.M.; Sung, H.H. Comparison of oncologic and functional outcomes between radical nephroureterectomy and segmental ureterectomy for upper urinary tract urothelial carcinoma. Sci. Rep. 2021, 11, 7828. [Google Scholar] [CrossRef] [PubMed]
  73. Duquesne, I.; Abou Chakra, M.; Hage, L.; Pinar, U.; Loriot, Y. Liquid biopsies for detection, surveillance, and prognosis of urothelial cancer: A future standard? Expert. Rev. Anticancer Ther. 2023, 23, 995–1007. [Google Scholar] [CrossRef] [PubMed]
  74. Hassler, M.R.; Bray, F.; Catto, J.W.F.; 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]
  75. Territo, A.; Gallioli, A.; Diana, P.; Boissier, R.; Fontana, M.; Gaya, J.M.; Sanguedolce, F.; Calderón, J.; Piana, A.; Fontanet, S.; et al. DNA Methylation Urine Biomarkers Test in the Diagnosis of Upper Tract Urothelial Carcinoma: Results from a Single-Center Prospective Clinical Trial. J. Urol. 2022, 208, 570–579. [Google Scholar] [CrossRef]
  76. Muin, D.; Laukhtina, E.; Hacker, M.; Shariat, S.F. PET in bladder cancer imaging. Curr. Opin. Urol. 2023, 33, 206–210. [Google Scholar] [CrossRef]
Table 1. Displaying the current EAU guideline surveillance protocol.
Table 1. Displaying the current EAU guideline surveillance protocol.
EAUMonths 3691215182124303642485460
low risk after RNUcytologynot mandatory
cystoscopy
CT/MR urographynot mandatory
high risk after RNUcytologyannually thereafter
cystoscopyannually thereafter
CT/MR urography annually thereafter
Chest CT
low risk after KSScytology
cystoscopy
CT/MR urography
URS
high risk after KSScytology
cystoscopy
CT/MR urography
URS
low riskunifocal, tumor size < 2 cm, low-grade cytology, low-grade URS biopsy, no invasive aspect on CT urography (all of these)
high riskhydronephrosis, tumor size ≥ 2 cm, high-grade cytology, high-grade URS biopsy, multifocal, previous RC for MIBC, variant histology (any of these)
CT = computed tomography; MR = magnet resonance; URS = ureterorenoscopy. ◯ = recommended.
Table 2. Displaying the current AUA guideline surveillance protocol.
Table 2. Displaying the current AUA guideline surveillance protocol.
AUAMonths3691215182124303642485460
<pT2 N0/M0 after RNUcystoscopy, cytology
cross-sectional imaging * x x
chest imaging
BMP **
>pT2 Nx/0 after RNUcystoscopy, cytology
cross-sectional imaging *
chest imaging
BMP **
low risk after KSScystoscopy, cytology
URS
cross-sectional imaging *
chest imaging
BMP **
high risk after KSScystoscopy, cytology
URS
cross-sectional imaging *
chest imaging
BMP **
low riskBx: low-grade; cytology: no HGUC, <cT2 N0M0, no sessile or flat tumors
high riskBX: high-grade; cytology: HGUC, ≥cT2 N+
* Cross-sectional imaging of the abdomen and pelvis should be performed with contrast when possible; ** basic metabolic panel. ◯ = recommended; x = optional.
Table 3. Displaying the current NCCN guideline surveillance protocol.
Table 3. Displaying the current NCCN guideline surveillance protocol.
NCCN 3691215182124303642485460months
pT0–1 after RNUcytologylonger intervals not specified
cystoscopy
cross-sectional imaging *not specifically recommended
pT2–4, pN+ after RNUcytologylonger intervals not specified
cystoscopy
cross-sectional imaging *not specifically recommended
chest imaging
pT0–1 after KSScytologylonger intervals not specified
cystoscopy
cross-sectional imaging *3–12-month intervals
URS
pT2–4, pN+ after KSScytologylonger intervals not specified
cystoscopy
cross-sectional imaging *3–12-month intervals
chest imaging
URS
* Abdominal/pelvic CT or MRI with and without contrast. ◯ = recommended.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Klemm, J.; Bekku, K.; Abufaraj, M.; Laukhtina, E.; Matsukawa, A.; Parizi, M.K.; Karakiewicz, P.I.; Shariat, S.F. Upper Tract Urothelial Carcinoma: A Narrative Review of Current Surveillance Strategies for Non-Metastatic Disease. Cancers 2024, 16, 44. https://doi.org/10.3390/cancers16010044

AMA Style

Klemm J, Bekku K, Abufaraj M, Laukhtina E, Matsukawa A, Parizi MK, Karakiewicz PI, Shariat SF. Upper Tract Urothelial Carcinoma: A Narrative Review of Current Surveillance Strategies for Non-Metastatic Disease. Cancers. 2024; 16(1):44. https://doi.org/10.3390/cancers16010044

Chicago/Turabian Style

Klemm, Jakob, Kensuke Bekku, Mohammad Abufaraj, Ekaterina Laukhtina, Akihiro Matsukawa, Mehdi Kardoust Parizi, Pierre I. Karakiewicz, and Shahrokh F. Shariat. 2024. "Upper Tract Urothelial Carcinoma: A Narrative Review of Current Surveillance Strategies for Non-Metastatic Disease" Cancers 16, no. 1: 44. https://doi.org/10.3390/cancers16010044

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