Diagnostic status and new explorations in upper urinary tract urothelial carcinoma: a literature review
Review Article

Diagnostic status and new explorations in upper urinary tract urothelial carcinoma: a literature review

Zujuan Shan ORCID logo, Xin Deng, Liansheng Yang, Guifu Zhang

Department of Urology, The First People’s Hospital of Honghe Prefecture, Gejiu, China

Contributions: (I) Conception and design: Z Shan; (II) Administrative support: L Yang, G Zhang; (III) Provision of study materials or patients: Z Shan, X Deng; (IV) Collection and assembly of data: Z Shan, L Yang; (V) Data analysis and interpretation: Z Shan, G Zhang; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Liansheng Yang, MD; Guifu Zhang, MD. Department of Urology, The First People’s Hospital of Honghe Prefecture, No. 1 Xiyuan Road, Gejiu 661017, China. Email: hhzyykyk@163.com; scuroyl@163.com.

Background and Objective: Upper urinary tract urothelial carcinomas (UTUCs) are characterized by their obscure location, insidious onset, and propensity for early muscle-invasive and lymph node metastasis, resulting in a generally poor prognosis. Clinically, diagnosis primarily relies on urine cytology, computed tomography urography (CTU), and white light ureteroscopy (WL-URS). These methods, however, suffer from low sensitivity, high cost, inefficient testing, patient discomfort, surgery-related complications, and potential tumor implantation. This literature review summarizes the advantages and disadvantages of the existing diagnostic tools for UTUC and discusses promising advancements in the field.

Methods: A comprehensive computer-assisted search, supplemented by literature tracing, was conducted to review publications on UTUCs diagnostics from the PubMed, Embase, and Web of Science databases covering the period from 2000 to August 2024. The research focused on synthesizing findings from the selected publications.

Key Content and Findings: Over 100 studies were reviewed, spanning from traditional diagnostic methods like urine cytology, CTU, and WL-URS, as well as newer methods including endoscopic advances, biopsy methods, radiomics, immunocytology [urinary tumor cell (UTC) assay], urinary genetic tests (identifying chromosomal abnormalities, DNA mutations, DNA methylation, and RNA), and hematological and urinary tests for microRNAs (miRNAs) and proteins (plasma protein classifiers). While traditional methods have been extensively evaluated, they remain suboptimal in terms of effectiveness, safety, patient satisfaction, and cost. Emerging diagnostics and molecular markers show promise, such as the high sensitivity of the urinary UTC assay for early-stage tumors and the non-invasive nature of various urinary molecular tests, along with the high accuracy of plasma protein classifiers. However, these new tools generally lack thorough evaluation and are often restricted to reports from single institutions or simple differentiation of cancerous cases from controls under experimental conditions.

Conclusions: Traditional diagnostic methods for UTUCs exhibit significant limitations, which newer tools and molecular markers are poised to address. However, comprehensive evaluations of these innovations are required to confirm their efficacy.

Keywords: Upper urinary tract urothelial carcinomas (UTUCs); diagnosis; endoscopy; molecular markers


Submitted Jul 03, 2024. Accepted for publication Sep 30, 2024. Published online Nov 28, 2024.

doi: 10.21037/tau-24-326


Introduction

Background

Upper urinary tract urothelial carcinomas (UTUCs), including renal pyelocaliceal and ureteral cancers, are part of the broader category of urothelial cancers (UCs) alongside bladder cancers (BCs). In Western countries, UTUCs represent 5–10% of UCs (1), and in East Asia, their prevalence can reach up to 43% due to the use of aristolochic acid in herbal medicine (2-5). These carcinomas are characterized by their deep location and insidious onset, complicating early detection. About 50–60% of cases are muscularly invasive at diagnosis (6), and 20–30% present with lymph node metastasis (7,8). Extensive statistical analysis shows that advanced stages and lymph node involvement often correlate with a poor prognosis; 5-year cancer-specific survival rates are below 50% for pT2/pT3 patients and under 10% for pT4 cases (6).

In clinical practice, current diagnostic standards primarily depend on urinary cytology, computed tomographic urography (CTU), and traditional white light ureteroscopy (WL-URS). However, these methods face significant challenges: urinary cytology suffers from low sensitivity; CTU struggles to effectively detect flat lesions and to distinguish between benign and malignant conditions; WL-URS, despite its diagnostic potential, is costly, technically challenging, and associated with patient discomfort as well as risks of surgical complications and tumor implantation in the bladder (9-11). These challenges underscore the necessity for more efficacious diagnostic techniques.

Recent advancements have introduced a variety of new diagnostic tools and molecular markers, including innovative endoscopic and biopsy methods, radiomics, immunocytology, molecular diagnostics, and plasma protein classifiers. These innovations aim to rectify the deficiencies of traditional methods, offering a more thorough diagnostic framework. This review aims to offer a concise overview of these new diagnostic methods and markers, their effectiveness, and their potential integration into clinical practice. We present this article in accordance with the Narrative Review reporting checklist (available at https://tau.amegroups.com/article/view/10.21037/tau-24-326/rc).


Methods

Two authors (Z.S. and X.D.) performed an exhaustive literature review using the PubMed, Embase, and Web of Science databases, employing a mix of subject terms and free words. The search strategy is elaborated in Appendix 1 (using PubMed as an example) and in Table 1. All studies on the epidemiologic and diagnostic evaluation of UTUCs within the period from 2000 to August 2024 were included. Other literature was obtained from lists of references and similar literature. Exclusions were made for non-English papers, inaccessible full texts, and types such as commentaries, opinion pieces, editorials, letters, and case reports. Studies whose methods and results were judged unreliable by two corresponding authors (L.Y. and G.Z.) were also excluded. Figure 1 shows a flowchart summarizing the selection process. Given the heterogeneity of the findings, a narrative synthesis was conducted instead of a quantitative meta-analysis.

Table 1

The search strategy summary

Items Specification
Date of search First searched in November 2023; second searched in August 2024
Databases and other sources searched PubMed, Embase, Web of Science
Search terms used “Carcinoma”; “Transitional Cell/diagnosis”; “Ureteral Neoplasms/diagnosis”; “UTUC”; “Dignosis”; “Molecular”
Timeframe 2000 to August 2024
Exclusion criteria (I) Non-English papers; (II) full text not available; (III) commentaries, opinion pieces, editorials, letters, and case reports; (IV) research methods and results were unreliable; (V) outdated diagnostic tools
Selection process Two authors (Z.S. and X.D.) independently searched and screened the literature. The corresponding authors then reassessed the selected articles, excluding those with controversial scope or quality

UTUC, upper urinary tract urothelial carcinoma.

Figure 1 Literature screening flowchart. UTUC, upper urinary tract urothelial carcinoma.

Results

Through a comprehensive database search and literature tracking, we initially obtained 1,647 papers. After a systematic screening, over 100 papers were included in this study. Preliminary analysis revealed that while some studies continue to assess traditional diagnostic tools like urocytology, CTU, and WL-URS, there is a growing interest in exploring and developing new diagnostic techniques and molecular markers. These include radiomics, immunocytology, WL-URS, and various DNA-, RNA-, and protein-based markers.

Urinary cytology and immunocytology

Urinary cytology

Urinary cytology is valued for its simplicity and non-invasive nature. It often indicates high-grade UTUCs when positive results are observed, excluding BCs and other urothelial carcinomas (12). However, its diagnostic sensitivity varies, with sensitivities reported between 11% and 71.1%, and specificities from 80% to 100% (9,13-30). Techniques such as selective collection of upper urinary tract (UUT) specimens via ureteral cannulae (31,32) or repeated saline flushes of the UUT mucosa (barbotage cytology) (33) have shown potential to enhance sensitivity, though these methods require further research for validation.

Immunocytology

Immunocytology is an advanced form of urinary cytology. Originating from a small 2001 study, it employed three monoclonal antibodies (19A211, M344, LDQ10) targeting two mucins and one carcinoembryonic antigen for UTUC diagnosis. This approach demonstrated a notable increase in sensitivity compared to traditional urinary cytology, particularly in detecting low-grade and early-stage tumors, and when combined, these methods further improved sensitivity (14). More recently, a novel urinary tumor cell (UTC) assay developed by Wang’s team utilized a nanostructured polystyrene substrate to capture over 90% of urinary exfoliated cells from morning urine samples. These cells were immunostained with CK20, CD45, and CD11b, and analyzed through a fully automated, high-resolution cytological imaging system. A sample was deemed positive if it contained one or more urinary cells (34). This test achieved an overall sensitivity of 85% and a specificity of 92.5% for UTC detection, outperforming urinary fluorescence in situ hybridization (FISH), which recorded sensitivities of 37.5% and 61.5% in low-grade and low-stage (≤ pT1) tumors, respectively (35). The test successfully identified all seven pTa cases, highlighting its potential for early UTUC diagnosis and suggesting promising prospects for widespread clinical application.

CTU/magnetic resonance urography (MRU) and radiomics

CTU has replaced intravenous pyelography as the preferred imaging technique for suspected UTUCs, due to its superior ability to detect non-enhancing stones, blood clots, and severe obstructions (36,37). The introduction of multidetector CTU has further enhanced diagnostic capabilities, providing more detailed images with reduced ionizing radiation exposure and shorter acquisition times (38). Meta-analyses indicate that CTU maintains high specificity, exceeding 88%, with pooled sensitivities of 92% and 96%, and specificities of 99% and 95%, respectively (38,39). It also demonstrates robust T-staging accuracy, with understaging and overstaging rates reported at 8% and 11.2%, respectively (40). However, CTU faces challenges in effectively detecting carcinoma in situ (CIS) and superficial tumor extensions (41), with reports indicating that CIS and non-papillary tumors comprise up to 21.6–28.7% and 15.1–27.5% of UTUCs cases, respectively (42,43). Challenges also arise in distinguishing some benign conditions like chronic inflammation, ureteral endometriosis, and cystic ureteropelvic inflammation (36,37).

MRU is considered an alternative for patients allergic to iodinated contrast agents or unable to undergo CTU. Despite this, the use of gadolinium-based contrast agents in MRU poses a risk of nephrogenic systemic fibrosis, especially in patients with renal impairment. Furthermore, MRU does not match CTU in terms of diagnostic and staging accuracy, with reported sensitivities generally below 76% (44-46).

Radiomics analyzes multidimensional information reflecting tumor characteristics that are difficult to discern with the naked eye by utilizing high-throughput mining to diagnose the disease. Recently, three studies on UTUCs demonstrated that radiomics is effective for preoperative pathologic staging and grading, showing marked superiority to URS biopsy (47-49). Goto et al. analyzed CT texture in 86 patients, noting a significant difference in the histogram of CT attenuation numbers: muscle-invasive tumors exhibited a multi-peaked shape, whereas non-muscle invasive tumors displayed a single-peaked shape (47). Alqahtani et al. conducted a CTU-based radiomics analysis with 106 patients, achieving 84% and 83% sensitivity and 93% and 76% specificity, respectively, in predicting tumor grade (low-grade and high-grade) and staging (early stage, Ta-T1, and late stage T2-T4) (48). Similarly, Zheng’s study involving 140 patients used a radiomics-based machine learning model to predict tumor grading, achieving an area under the curve (AUC) of 0.914 and 0.903 in the training and validation sets, respectively (49).

URS and biopsies

Traditional WL-URS

WL-URS allows direct visualization of the location, structure, and size of suspicious lesions, and facilitates biopsy, offering higher diagnostic accuracy and specificity compared to CTU (41). Despite its advantages, WL-URS has several limitations and risks, including: (I) a lower detection rate for CIS compared to CTU (41,43,50); (II) conventional ureteral forceps often fail to grasp subepithelial connective and deep muscular tissues, leading to inadequate staging and grading. A multicenter study reported only a 34.5% concordance rate between URS staging and grading and postoperative pathology results (51), with another meta-analysis highlighting high rates of understaging and undergrading at 46% and 32%, respectively (52); (III) URS is costly, technically demanding, and associated with significant risks, including infection, hemorrhage, and local injury. The literature indicates that the complication rate of URS is approximately 9–11% (53), with ureteral perforation accounting for 1–4% of cases, although most are localized (10); (IV) URS biopsies may cause tumor implantation in the bladder, with several studies indicating that URS biopsies significantly increase the rate of intravesical recurrence (11,54-57).

New endoscopic and biopsy techniques

Narrow-band imaging (NBI)

NBI enhances the visualization of tumor boundaries and vascular structures by filtering broadband light to select narrow bands at 415 and 540 nm. This technique increases the detection rate of early lesions like pTa and CIS but may also result in a higher rate of false positives, leading to unnecessary biopsies (58,59).

Optical coherence tomography (OCT)

OCT provides imaging up to a depth of 2 mm, proving useful for preoperative staging and grading (60). A prospective study reported preoperative grading and staging accuracies of 83% and 88%, respectively, outperforming conventional biopsy (61). However, its limited depth restricts its applicability in advanced diseases.

Confocal laser endomicroscopy (CLE)

Originally employed in tracheal and intestinal examinations, CLE provides dynamic images of cellular structures and morphology using fluorescein staining (62). Its efficacy and safety in diagnosing UTUCs have been validated in studies with small sample sizes (62,63). Recent evaluations by Freund et al. reported CLE grading accuracies between 72% and 88% (64). However, CLE’s effectiveness relies heavily on image quality and necessitates specialized training.

Photodynamic diagnosis (PDD)

PDD utilizes 5-aminolevulinic acid (5-ALA), which is metabolized to protoporphyrin IX (PpIX) in vivo. Cancer cells deficient in ferrous chelatase accumulate PpIX, which fluoresces red under violet light (65). PDD significantly enhances diagnostic sensitivity, especially in flat lesions, CIS, and dysplasia. Nevertheless, it is susceptible to false positives due to local inflammation and experiences a considerable incidence of adverse effects, such as transient hypotension, photosensitized facial rash, and deranged liver enzymes, affecting up to 25% of patients (66-69).

New biopsy techniques

In clinical practice, devices for pelvic ureteral sampling include anterior and posterior biopsy forceps and tipless anterior and lateral grasping basket sets. Posterior biopsy forceps overcome the challenges of small-caliber anterior biopsy forceps, such as low tissue volume and high fragmentation, thus enhancing sample collection. The basket sets are particularly effective at grasping papillary tumors (70,71). Moreover, posterior large-caliber biopsy forceps and nested baskets secure higher quality tissue than anterior biopsy forceps, although nested baskets are somewhat less efficient and tend to produce larger artifacts (71,72). Klein et al. recently introduced a novel frozen biopsy technique that achieves the highest biopsy tissue volume and quality with the smallest area of artifacts in isolated renal pelvic ureteral tissues. This technique uses CO2 to form ice crystals at the tip of a metal probe under a pressure drop, which then envelop the target tissue and adhere to the metal, facilitating tissue retrieval by suddenly withdrawing the prob (72). Additionally, Klett et al. developed a “formal processing” biopsy technique, which involves placing a cold cup clamp at the lesion’s base, closing it, and advancing it by 3–10 mm before withdrawal. This method has significantly improved tissue yields and diagnostic accuracy for lesions of 1 cm or larger (73).

Recently, Fukumoto’s phosphorylated ribosomal protein S6-based 3D imaging and pathology analysis of URS biopsy tissues has enabled a more accurate predictive grading of tumors while avoiding cumbersome tissue sectioning, particularly beneficial in UTUCs, which are typically smaller and more friable (74).

Molecular diagnostics

Urine genetic information testing

In UCs, urine directly contacts the tumor and is less complex than blood, facilitating UC diagnosis by detecting abnormalities such as tumor-derived DNA, mRNAs, and microRNAs (miRNAs). Research indicates that first-morning urine contains the most genetic information. The supernatant, comprising fragmented cell-free tumor nucleic acids and other derivatives, proves more effective than the sediment, which mainly holds detached cancer cells and their debris (75,76). Several studies have demonstrated that urine more accurately reflects the genetic abnormalities of tumor tissues in patients with UTUCs (3,42,77,78). Disparities in genetic abnormalities between urine and tumor samples may be due to inter-tumor and intra-tumor heterogeneity, clonal proliferation of the urinary epithelium, and insufficient aberrant cells or DNA in the urine (42,77). Significant advancements in urinary genetic abnormality tests have been made in diagnosing BCs, with tests such as Urovysion, EpiCheck test, and Xpert® BC test receiving approval from the US Food and Drug Administration (FDA) and Conformitè Europëenne (CE) marking (79), advances in these tests for UTUCs are outlined below:

Chromosomes
(I) Urovysion®-FISH

FISH utilizes fluorescently labeled specific oligonucleotide fragments as probes to detect abnormalities in chromosomes 3, 7, 17, and 9p21 in urinary exfoliated cells, aiding in the diagnosis of urothelial carcinomas (UCs). FISH’s diagnostic value for UTUCs has been extensively explored, showing a wide range of sensitivities from 35% to 100%, generally outperforming urocytology, with specificities from 80% to 100% (9,15,18-20,22-29,80,81). However, most FISH studies involve small sample sizes and rarely evaluate low-grade and early-stage tumors (15,35,82). Noteworthy studies include the Sassa study and Aalami’s meta-analysis. The Sassa study, involving 75 patients with high-grade UTUCs, reported FISH sensitivities and specificities of 60% and 84%, respectively, compared to urocytology’s 28% sensitivity and 100% specificity (83). Aalami’s meta-analysis, including data from 1,067 patients, presented pooled sensitivity and specificity rates of 72% and 95%, respectively (84). Additionally, several studies have emphasized FISH’s role in predicting tumor pathological grading and intravesical recurrence (85-87).

(II) Aneuploidy detection

Springer et al. investigated aneuploidy by analyzing copy number changes on 39 chromosome arms in urine from 56 UTUCs patients. Alterations were found in 22 cases, with a detection sensitivity of 39%. The most frequently altered chromosome arms included 1q, 7q, 8q, 17p, and 18q (77).

DNA mutation and methylation
(I) DNA mutation

High-throughput sequencing of UTUCs tissues and urinary DNA has revealed significant mutations in genes such as FGFR3, TERT, KMT2D, KDM6A, CDKN2A, TP53, ARID1A, RAS, and CCND1 (42,77,78,88). FGFR3, HRAS, and TERT mutations are more common in patients with non-muscle-invasive UTUCs, while TP53 and CCND1 mutations occur more frequently in muscle-invasive UTUCs. TERT and PIK3CA mutations are more prevalent in lower-grade tumors, while TP53 mutations are significantly higher in higher-grade tumors (42,78). TERT mutations in UTUCs urine primarily occur at two hotspots, 66 and 88 bp upstream of its transcription start site (g.1295228C>T and g.1295250C>T) (16,77). The frequency of mutations varies among different populations, with FGFR3 mutations being less frequent in the Chinese Han population (3,89).

The distribution and frequency of these gene mutations assist in the diagnosis and staging of UTUCs. However, relying solely on single-gene or multi-gene testing does not fully address clinical needs. Recently, Fujii et al. achieved a sensitivity of 78% by sequencing urinary sediment-derived DNA in 43 UTUCs cases, significantly surpassing the 29.3% sensitivity of urinary cytology. This sensitivity remained consistent in 35 newly recruited patients, unaffected by previous UCs or the severity of pyuria and hematuria (42). When patients with severe urinary flow obstructions were excluded, sensitivity increased to 92.3%, with a specificity of 100% at a mutation detection rate of 74.5% (42).

(II) DNA methylation

Urine-derived DNA methylation assays have been established for diagnosing BCs, such as the bladder EpiCheck assay, which utilizes 15 urinary DNA methylation biomarkers alongside a complex algorithm (79,90,91). However, these assays remain under-researched in UTUCs. To date, only the methylation of genes such as CDH1, HSPA2, RASSF1A, TMEFF2, VIM, GDF15, NRN1, ONECUT2, and bladder EpiCheck assays have been evaluated in UTUCs (78,92-97), with results detailed in Table 2. Additionally, methylation in these genes is referenced against BCs, and extensive gene sequencing has shown significant differences in mutation frequency and burden between the two tumor types (42,88). Recently, Fujimoto et al. screened UTUCs tissues to identify 10 cytosine-phosphate-guanine (CPG) loci effective in differentiating BCs from normal uroepithelium. A diagnostic panel comprising these loci and neighboring sites achieved sensitivity, specificity, and AUC values of over 86.6%, 93.5%, and 0.959, respectively (99). The diagnostic efficacy was unaffected by clinicopathologic parameters such as gender, age, stage, grading, and lymph node metastasis, but it has not yet been validated in urine specimens.

Table 2

Diagnostic performance of urine genetic information abnormality testing in UTUCs

Author, country/region, year Population and specimen Molecular target Detection scheme Sensibility (%) Specificity (%) PPV (%) NPV (%) AUC Accuracy (%)
Hayashi (16), Japan, 2019 UTUC =56 (17 Tis/Ta, 11 T1, 28 ≥T2; 7 LG, 47 HG, 2 others), non-UC hematuria =50, no evidence of tumor after surgery =21, health =26, urine Mutations of TERT and FGFR3 ddPCR 46.4 (TERT) 100 100 62.5 NA NA
16.1 (FGFR3) 100 100 51.5 NA NA
55.4 (TERT + FGFR3) 100 100 66.7 NA NA
78.6 (urinary cytology + TERT + FGFR3) 96 95.7 80 NA NA
Springer (77), United States and Taiwan, 2018 UTUC =56 (11 Ta, 8 T1, 10 T2, 24 T3, 3 T4; 6 LG, 50 HG), urine Mutations in 10 genes (FGFR3, TP53, CDKN2A, ERBB2, HRAS, KRAS, PIK3CA, MET, VHL, and MLL), mutations in the TERT promoter, and aneuploidy assays (copy number variations on 39 chromosome arms) PCR, Fast-SeqS 64 (mutations of 10 genes) NA NA NA NA NA
29 (mutations of TERT) NA NA NA NA NA
39 (aneuploidy assays) NA NA NA NA NA
75 (10 genes + TERT + aneuploidy assays, UroSEEK) NA NA NA NA NA
79 (UroSEEK, Ta/T1) NA NA NA NA NA
73 (UroSEEK, T2/T3/T4) NA NA NA NA NA
80 (UroSEEK, LG) NA NA NA NA NA
73 (UroSEEK, HG) NA NA NA NA NA
Xu (78), China, 2020 NMIUC =22, MIUC =39, LG =17, HG =47, non-UTUC =86, urine Mutations of AKT1, ASXL2, CREBBP, ERBB2, ERBB3, ERCC2, FBXW7, FGFR3, HRAS, KDM6A, KRAS, PIK3CA, RHOA, SF3B1, TP53, TERT, U2AF1 and methylation status of ONECUT2 NGS + BS-RT-PCR 71.9 (mutations) 95.4 92 82 NA NA
89.1 (methylation) 94.2 91.9 92 0.93 NA
92.2 (mutations + methylation) 91.9 85 94.1 NA NA
Territo (92), Spain, 2022 Suspicious UTUC =86, 83 UUT urine, 73 voided urine, 47 and 42 cases were confirmed respectively 15 DNA methylations (EpiCheck) RT-PCR 83 (UUT urine) 81 79 78 NA 82
55 (voided urine) 81 NA 57 NA 66
68 (UUT urine, LG) NA NA 80 NA NA
95 (UUT urine, HG) NA NA 97 NA NA
100 (UUT urine, CIS) NA NA 100 NA NA
36 (voided urine, LG) NA NA 67 NA NA
80 (voided urine, HG) NA NA 93 NA NA
0 (voided urine, CIS) NA 85 95 NA NA
Guo (93), China, 2018 UTUC =98, control =113, urine Methylation status of CDH1, HSPA2, RASSF1A, TMEFF2, VIM and GDF15 promoters qMSP 82 68 NA NA 0.836 NA
Pierconti (94), Italy, 2021 UTUC =82 (64 G3T1N0, 3 G3T2N1, 15 G2T1N0), hematuria =52, UUT urine 15 DNA methylations (EpiCheck) RT-PCR 97.44 100 100 96.23 NA 98.45
Ouyang (95), China, 2022 UTUC =95 (17 LG, 76 HG, 2 others; 64 Ta-2, 29 T3-4, 2 Tx), non-UTUC =307 (102 other tumors, 205 benign), external validation set =76, midstream urine Mutations of TERT and FGFR3, methylation status of NRN1 qPCR 91.58 (methylation) 94.79 84.58 97.36 0.932 NA
34.74 (mutations) 96.74 76.73 82.74 0.657 NA
91.58 (mutations + methylation) 94.79 84.58 97.36 0.958 NA
Monteiro-Reis (96), Portugal, 2014 UTUC tissue =57 (4 pTa, 22 pT1, 13 pT2, 17 pT3, 1 pT4; 15 LG, 42 HG), kidney cancer tissue =36, UTUC urine =22 (3 LG, 19 HG), non-UTUC urine =20, tissue and urine Methylation status of GDF15, TMEFF2 and VIM promoter qMSP Tissue
-95 (VIM) 100 100 92 0.974 97
-98 (VIM/GDF15) 100 100 97 NA 99
-100 (VIM/GDF15/TMEFF2) 100 100 100 1 100
Urine
-82 (VIM) 100 100 83 NA 91
-91 (VIM/GDF15) 100 100 91 NA 95
-91 (VIM/GDF15/TMEFF2) 100 100 91 0.919 100
Pycha (97), Italy, 2023 Suspicious UTUC =79 (31 cases were confirmed: 14 G1, 12 G2, 5 G3), 97 analyses, UUT urine 15 DNA methylations (EpiCheck), 5 mRNAs (ABL1, CRH, IGF2, UPK1B, ANXA10) (Xpert® BC) RT-PCR 64.5 (EpiCheck) 78.8 58.8 82.5 NA NA
57.7 (EpiCheck, 26 LG) NA NA NA NA NA
100 (EpiCheck, HG) NA NA NA NA NA
100 (Xpert® BC) 4.5 33 100 NA NA
100 (Xpert® BC, 26 LG) NA NA NA NA NA
100 (Xpert® BC, HG) NA NA NA NA NA
D’Elia (98), Italy, 2022 Suspicious UTUC =82 (27 cases were confirmed: 20 LG, 7 HG), 87 analyses, UUT urine 5 mRNAs (ABL1, CRH, IGF2, UPK1B, ANXA10) (Xpert® BC) RT-PCR 100 16.7 35 100 NA NA
100 (LG) NA NA NA NA NA
100 (HG) NA NA NA NA NA

UTUCs, upper urinary tract urothelial carcinomas; PPV, positive predictive value; NPV, negative predictive value; AUC, area under the curve; LG, low grade; HG, high grade; UC, urothelial cancer; ddPCR, digital droplet polymerase chain reaction; NA, not assessed; PCR, polymerase chain reaction; Fast-SeqS, Fast-Sequencing System; NGS, next generation sequencing; NMIUC, non-muscle-invasive urothelial carcinoma; MIUC, muscle-invasive urothelial carcinoma; BS-RT-PCR, bisulfite-specific-RT-PCR; RT-PCR, real time-polymerase chain reaction; UUT, upper urinary tract; CIS, carcinoma in situ; qMSP, quantitative methylation-specific polymerase chain reaction; qPCR, quantitative polymerase chain reaction.

From the limited study data available, the diagnostic efficacy of urine-derived DNA methylation testing for UTUCs is significantly better than that of urine genetic testing. Combining these two approaches can further enhance diagnostic accuracy. It is crucial to determine whether to proceed with CTU or URS based on methylation testing results, or to consider reducing the frequency of these procedures during follow-up. However, further evaluation is required.

Xpert® BC testing

The Xpert® BC assay is designed for the early diagnosis of BCs by qualitatively detecting the levels of five target mRNAs (ABL1, CRH, IGF2, UPK1B, and ANXA10) in urine. The Xpert® BC assay’s performance in diagnosing UTUCs was assessed in two similar studies featuring predominantly small samples of low-grade cases (97,98). In these studies, UUT urine was selected as the test specimen. The sensitivity reached 100% for both low-grade and high-grade patients, significantly higher than that of urocytology, FISH, and bladder EpiCheck tests, although the specificity was lower, at 4.5% and 16.7%, respectively.

miRNAs detection

miRNAs are small non-coding, single-stranded RNA molecules. For example, serum miRNA371 is utilized in the diagnostic monitoring of testicular germ cell tumors and is included in various European clinical guidelines (100). In UTUCs, Tao et al. identified 13 significantly different miRNAs in 46 patients compared to 30 patients with hematuria. Ten of these miRNAs could differentiate between cancerous and hematuric patients with an AUC >0.8, with miR-664a-3p, miR-431-5p, and let-7c demonstrating the strongest performance (101). Kriebel et al. reported that miR-141 was overexpressed in both UTUCs serum and matched tissues (102). A larger study showed that miR-210 was effective in identifying oncogenic diseases in tissues (103). Recently, Urabe et al. identified 12 differential miRNAs in the serum of UC patients, with miR-4433a-3p and miR-6778-5p performing best in UTUCs (104). The diagnostic performance of these miRNAs for UTUCs is summarized in Table 3.

Table 3

Diagnostic performance of miRNAs detection in UTUCs

Author, country/region, year Population Detection scheme miRNAs Sensibility (%) Specificity (%) PPV (%) NPV (%) AUC
Urabe (104), Japan, 2022 UTUC =20 (10 discovery set, 10 validation set), control =50 Chip-based PCR miR-4433a-3p 86 96 NA NA 0.94
miR-6778-5p 86 96 NA NA 0.95
Tao (101), China, 2015 UTUC =46, hematuria =30 qRT-PCR miR-664 a-3p NA NA NA NA 0.998
miR-431-5p NA NA NA NA 0.933
let-7c NA NA NA NA 0.905
Kriebel (102), Germany, 2015 UTUC =44, control =36 qRT-PCR miR-141 70.5 73.5 NA NA 0.726
Browne (105), United States, 2019 Screening cohort (n=35) (19 pTa/pT1, 16 ≥ pT2, LG 42.9%), secondary cohort (n=123) (76 pTa/pT1, 47 ≥ pT2, LG 37%) qRT-PCR ① miR-29b-2-5p + miR-18a-5p + miR-223-3p + miR-199a-5p 83 (① predict HG) 85 91 73 0.86
② miR-10b-5p + miR-26a-5p + miR-31-5p + miR-146b-5p 64 (② predict ≥ pT2) 96 90 81 0.90
Ke (103), Taiwan, 2017 UTUC =83 (50 T1-2, 33 T3-4), control =50 qRT-PCR miR-210 80 90 NA NA 0.904

miRNAs, microRNAs; UTUCs, upper urinary tract urothelial carcinomas; PPV, positive predictive value; NPV, negative predictive value; AUC, area under the curve; PCR, polymerase chain reaction; NA, not assessed; qRT-PCR, quantitative reverse transcription polymerase chain reaction; LG, low grade; HG, high grade.

In addition to their diagnostic applications, miRNAs also contribute to staging and grading. Tao et al. and Kriebel et al. observed that miR-664a-3p, miR-431-5p, miR-423-5p, miR-191-5p, miR-92a-3p, miR-16-5p, and let-7b-5p were significantly upregulated in patients with myxoid invasion, while miR-10a and miR-135 were downregulated (101,102). Browne et al. developed diagnostic models to predict high-grade (model ①) and myeloid invasion (model ②) in patients, by optimizing combinations of miRNAs identified in 158 UTUC tissues, achieving AUCs of 0.86 and 0.90, respectively (Table 3) (105).

Proteins and post-translational modification detection

Protein-based tumor markers are the most traditional and widely used diagnostic tools because they are easily accessible and minimally invasive. In UCs, the direct contact between the tumor and urine makes urine protein detection in urine a viable method. Recently, with advances in high-throughput sequencing technology, several UTUC-associated proteins and post-translational modifications have been detected and evaluated. However, these findings are mostly reported by single institutions. Below are some of the key proteins and modifications identified:

Plasma secreted phosphoprotein 1 (SPP1)

SPP1 is part of the small integrin-binding ligand N-linked glycoprotein family. Li et al. discovered that SPP1 in UTUC tissues was the most significantly upregulated protein-coding gene (over 70-fold) among 948 differentially regulated genes compared to matched normal urinary tract epithelium (106). Enzyme-Linked Immunosorbent Assay (ELISA) tests on larger serum samples demonstrated that SPP1 was significantly correlated with pathological grade and T-stage, achieving an AUC of 0.838 for distinguishing cancerous from noncancerous cases, with sensitivities of 87% for muscle-invasive and 77.8% for high-grade disease (98).

N-glycan modification of serum immunoglobulins

N-glycan modifications are common post-translational modifications. Kodama et al. used fast capillary electrophoresis with light-emitting diode-induced fluorescence to identify 26 N-glycan signals on serum immunoglobulins. UTUC scores constructed using these signals outperformed urocytology in UTUC detection, with sensitivity, positive predictive value (PPV), negative predictive value (NPV), and AUC at 90% specificity of 67.6%, 86.8%, 76.3%, and 0.868, respectively (107).

Plasma multiprotein classifiers

Recently, Qu et al. conducted high-throughput measurements of plasma proteins from 362 UTUC patients and 239 healthy individuals, developing a 10-protein classifier model (ANXA6, CTSB, CD44, SAA1, HBZ, S100A8, DPH5, PAM, RAP1A, and THUMPD1) that effectively differentiated cancerous from non-cancerous cases, with sensitivity, specificity, and AUC of 99.7%, 99.8%, and 0.942, respectively (101). This model also performed well in an independent cohort with an AUC of 0.925 (108). Additionally, various 12-protein classifiers were constructed to predict the presence of muscle-invasive disease, all achieving AUCs greater than 0.73.

Urine nuclear matrix protein 22 (NMP22) and bladder tumor antigen (BTA)

The BTA test, an economical and rapid assay, detects human complement factor H-associated proteins in urine and is available in both qualitative and quantitative formats. Walsh et al. reported sensitivities and specificities of 82% and 89%, respectively, using the qualitative assay, significantly outperforming urinary cytology (13). A recent study of a Chinese population indicated sensitivities of 98% and 93%, and specificities of 63% and 81% for qualitative and quantitative tests, respectively, respectively (109).

The NMP22 assay, which measures the concentration of NMP22 significantly higher in UC cells than in normal cells, showed sensitivities between 70–73.2% and specificities from 43.2–92%. Notably, this assay’s performance was consistent across samples from the UUT or bladder, although results may be influenced by hematuria, urinary tract infections, and instrumentation (17,22,109).

Urinary p16INK4a (p16)/Ki-67 double immunolabeling

The urinary p16/Ki-67 double immunolabeling technique uses biomarkers p16 and Ki-67 for diagnostic purposes. p16, a product of the CDKN2a gene on chromosome 9p21, regulates the cell cycle, while Ki-67 is a marker of cell proliferation. Sun et al. reported that the diagnostic sensitivity of simultaneous positivity of p16 and Ki-67 in UTUC was 53.1% (for low-grade and high-grade tumors 12.5% and 66.7%, respectively) and specificity was 100% (15).


Discussion

Given the high incidence of certain diseases in East Asian countries and their characteristics—insidious onset, difficult diagnosis, easy invasion and metastasis, and poor prognosis—it is highly valuable to search for and develop safe, effective, and simple diagnostic tools. Historically, urine cytology, CTU/MRU and WL-URS have been common in clinical settings, yet they exhibit numerous drawbacks: urine cytology has low sensitivity; CTU can miss superficial extension diseases and carries the potential risk of contrast agents; and URS is costly, involves complications related to invasive procedures, and poses a risk of tumor implantation in the bladder. Fortunately, new techniques have partially addressed these shortcomings. For example, UTC tests have significantly enhanced diagnostic sensitivity for early-stage tumors by enriching urinary exfoliated cells with special materials; various new light source endoscopes have improved tumor visualization and detection, especially for CIS; and advancements in biopsy techniques and devices have enhanced the quality of biopsy specimens and preoperative staging and grading. Additionally, the performance of plasma protein classifiers has been notably impressive.

However, we must acknowledge that new technologies also have drawbacks, such as: (I) many new techniques and markers have only been reported by single institutions or shown differentiation between cancerous cases and controls under experimental conditions. (II) Certain methods, such as the NBI test and urine mRNA tests, exhibit a high rate of false positives, which could lead to unnecessary further investigations or surgeries. (III) The efficacy of urinary molecular markers may be affected by hydration status, renal pathology, and medication use. (IV) Many molecular marker tests require sophisticated expertise and advanced laboratory facilities, demanding substantial financial resources.


Conclusions

In this review, we have summarized the benefits, risks, and limitations of UTUC-related diagnostic tools (Table 4). Looking forward, our expectations for future research and the application of new technologies, particularly in molecular diagnostics, include: (I) expanded research and validation across multiple institutions and diverse populations, with performance evaluations in actual clinical settings. (II) Focused evaluations on patients with low-grade, low-stage, CIS, postoperative recurrences, and those with concurrent BCs. (III) Prospective multicenter comparative studies conducted alongside traditional methods such as CTU and WL-URS. (IV) Economic analyses via model construction to assess cost-effectiveness. (V) The development of high-throughput assays and analyses of genetic, protein, and epigenetic modifications in UTUCs to create diagnostic panels that are better tailored to the unique needs and characteristics of UTUCs.

Table 4

Summary of benefits, risks, and limitations of various UTUC diagnostic measures

Diagnostic methods Categorizations Benefits Limitations and risks
Urinary cytology Cytology Non-invasive, simple, and economical Low sensitivity
Immunocytology Cytology Non-invasive, simple, effective for early-stage tumors Limited to single-agency assessment, requires further validation
CTU Radiographic High sensitivity and specificity, widely used clinically High cost, low efficiency, potential risks associated with contrast agents, difficulty in identifying superficial tumor extensions and differentiating benign from malignant disease
MRU Radiographic Suitable for patients allergic to iodine contrast or unable to undergo CTU Costly, less efficient, and sensitive than CTU
Radiomics Radiographic and artificial intelligence Provides machine-readable data, more accurate in preoperative staging and grading than URS biopsy Requires large-scale data training and software development
WL-URS URS Direct tumor visualization, allows biopsy and treatment, accurate preoperative staging and grading High cost, low efficiency, invasive procedures, surgery-related complications, patient pain, risk of tumor implantation in the bladder, missed diagnosis of CIS
NBI URS Higher detection rate for CIS and pTa than WL-URS High false positive rate, requires specialized equipment
OCT URS More accurate in preoperative staging and grading than WL-URS Limited by 2 mm imaging depth, requires specialized equipment
CLE URS More accurate in preoperative staging and grading than WL-URS Requires high-quality images, highly skilled personnel, and specialized equipment
PDD URS Higher detection rate for CIS and dysplasia than WL-URS Risks of transient hypotension and photosensitivity
Urovysion®-FISH Genetic information Non-invasive, higher sensitivity than urine cytology Multiple influencing factors and wide variation in sensitivity
DNA mutation and DNA methylation Genetic information Non-invasive, promising for future diagnostics Most of the relevant DNA mutations and DNA methylations that received evaluation originated from BCs and lacked sufficient specificity for UTUC; mostly simple identification of cancerous versus noncancerous cases under experimental conditions; and effectiveness may be influenced by hydration status, renal pathology, and dosing
Xpert® BC testing Genetic information Non-invasive, highly sensitive High false positive rate, pending further evaluation
miRNAs detection Genetic information Non-invasive or minimally invasive, effective for diagnostic performance and staging Wide variation between studies, variable specimen sources, lacks thorough assessment
SPP1 Protein Minimally invasive, effective performance Limited to single-agency assessment, requires further validation
N-glycan modification of serum immunoglobulins Protein modification Minimally invasive, effective performance Limited to single-agency assessment, requires further validation
Plasma multiprotein classifiers Proteins Minimally invasive, effective performance Limited to single-agency assessment, requires further validation
Urine NMP22 Protein Non-invasive, economical, effective performance Performance can be affected by infections and hematuria
Urine BTA Protein Non-invasive, economical, effective performance Requires further assessment
Urinary p16/Ki-67 double immunolabeling Immunolabeling Non-invasive Poor performance, lacks thorough assessment

UTUC, upper urinary tract urothelial carcinoma; CTU, computed tomography urography; MRU, magnetic resonance urography; URS, ureteroscopy; WL-URS, white light ureteroscopy; CIS, carcinoma in situ; NBI, narrow-band imaging; OCT, optical coherence tomography; CLE, confocal laser endomicroscopy; PDD, photodynamic diagnosis; FISH, fluorescence in situ hybridization; BCs, bladder cancers; miRNAs, microRNAs; SPP1, plasma secreted phosphoprotein 1; NMP22, nuclear matrix protein 22; BTA, bladder tumor antigen.


Acknowledgments

Funding: This study was funded by the Science and Technology Program of Yunnan Provincial Department of Science and Technology (Project No. 202101AY070001-231).


Footnote

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://tau.amegroups.com/article/view/10.21037/tau-24-326/rc

Peer Review File: Available at https://tau.amegroups.com/article/view/10.21037/tau-24-326/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://tau.amegroups.com/article/view/10.21037/tau-24-326/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Rouprêt M, Seisen T, Birtle AJ, et al. European Association of Urology Guidelines on Upper Urinary Tract Urothelial Carcinoma: 2023 Update. Eur Urol 2023;84:49-64. [Crossref] [PubMed]
  2. Chen CH, Dickman KG, Moriya M, et al. Aristolochic acid-associated urothelial cancer in Taiwan. Proc Natl Acad Sci U S A 2012;109:8241-6. [Crossref] [PubMed]
  3. Lu H, Liang Y, Guan B, et al. Aristolochic acid mutational signature defines the low-risk subtype in upper tract urothelial carcinoma. Theranostics 2020;10:4323-33. [Crossref] [PubMed]
  4. Huang J, Zhang X. Guidelines for diagnosis and treatment of urothelial carcinoma of upper urinary tract. Beijing: Science Press; 2022.
  5. Liao CI, Fang HC, Lee PT, et al. Trends in the incidence of urothelial carcinoma in Taiwan after the ban on aristolochic acid-containing Chinese herbal preparations, 2001-2018: a national population-based cohort study. J Cancer Res Clin Oncol 2023;149:8201-11. [Crossref] [PubMed]
  6. Margulis V, Shariat SF, Matin SF, et al. Outcomes of radical nephroureterectomy: a series from the Upper Tract Urothelial Carcinoma Collaboration. Cancer 2009;115:1224-33. [Crossref] [PubMed]
  7. Kondo T, Nakazawa H, Ito F, et al. Primary site and incidence of lymph node metastases in urothelial carcinoma of upper urinary tract. Urology 2007;69:265-9. [Crossref] [PubMed]
  8. Roscigno M, Shariat SF, Margulis V, et al. The extent of lymphadenectomy seems to be associated with better survival in patients with nonmetastatic upper-tract urothelial carcinoma: how many lymph nodes should be removed? Eur Urol 2009;56:512-8. [Crossref] [PubMed]
  9. Białek Ł, Bilski K, Dobruch J, et al. Non-Invasive Biomarkers in the Diagnosis of Upper Urinary Tract Urothelial Carcinoma-A Systematic Review. Cancers (Basel) 2022;14:1520. [Crossref] [PubMed]
  10. Schoenthaler M, Buchholz N, Farin E, et al. The Post-Ureteroscopic Lesion Scale (PULS): a multicenter video-based evaluation of inter-rater reliability. World J Urol 2014;32:1033-40. [Crossref] [PubMed]
  11. Marchioni M, Primiceri G, Cindolo L, et al. Impact of diagnostic ureteroscopy on intravesical recurrence in patients undergoing radical nephroureterectomy for upper tract urothelial cancer: a systematic review and meta-analysis. BJU Int 2017;120:313-9. [Crossref] [PubMed]
  12. Bitaraf M, Ghafoori Yazdi M, Amini E. Upper Tract Urothelial Carcinoma (UTUC) Diagnosis and Risk Stratification: A Comprehensive Review. Cancers (Basel) 2023;15:4987. [Crossref] [PubMed]
  13. Walsh IK, Keane PF, Ishak LM, et al. The BTA stat test: a tumor marker for the detection of upper tract transitional cell carcinoma. Urology 2001;58:532-5. [Crossref] [PubMed]
  14. Lodde M, Mian C, Wiener H, et al. Detection of upper urinary tract transitional cell carcinoma with ImmunoCyt: a preliminary report. Urology 2001;58:362-6. [Crossref] [PubMed]
  15. Sun X, Liu X, Xia M, et al. The combined application of urinary liquid-based cytology with fluorescence in situ hybridization and p16/Ki-67 dual immunostaining is valuable for improving the early diagnosis of upper tract urothelial carcinomas. Diagn Cytopathol 2017;45:895-902. [Crossref] [PubMed]
  16. Hayashi Y, Fujita K, Matsuzaki K, et al. Diagnostic potential of TERT promoter and FGFR3 mutations in urinary cell-free DNA in upper tract urothelial carcinoma. Cancer Sci 2019;110:1771-9. [Crossref] [PubMed]
  17. Jovanovic M, Soldatovic I, Janjic A, et al. Diagnostic value of the nuclear matrix protein 22 test and urine cytology in upper tract urothelial tumors. Urol Int 2011;87:134-7. [Crossref] [PubMed]
  18. Shan Z, Wu P, Zheng S, et al. Evaluation of upper urinary tract tumors by FISH in Chinese patients. Cancer Genet Cytogenet 2010;203:238-46. [Crossref] [PubMed]
  19. Gomella LG, Mann MJ, Cleary RC, et al. Fluorescence in situ hybridization (FISH) in the diagnosis of bladder and upper tract urothelial carcinoma: the largest single-institution experience to date. Can J Urol 2017;24:8620-6.
  20. Akkad T, Brunner A, Pallwein L, et al. Fluorescence in situ hybridization for detecting upper urinary tract tumors--a preliminary report. Urology 2007;70:753-7. [Crossref] [PubMed]
  21. Yamamichi G, Nakata W, Tani M, et al. High diagnostic efficacy of 5-aminolevulinic acid-induced fluorescent urine cytology for urothelial carcinoma. Int J Clin Oncol 2019;24:1075-80. [Crossref] [PubMed]
  22. Todenhöfer T, Hennenlotter J, Tews V, et al. Impact of different grades of microscopic hematuria on the performance of urine-based markers for the detection of urothelial carcinoma. Urol Oncol 2013;31:1148-54. [Crossref] [PubMed]
  23. Chen AA, Grasso M. Is there a role for FISH in the management and surveillance of patients with upper tract transitional-cell carcinoma? J Endourol 2008;22:1371-4. [Crossref] [PubMed]
  24. Yu Q, Li Y, Li G, et al. Prospective evaluation of FISH for detecting upper tract urothelial carcinoma in voided urine specimens. Oncol Lett 2016;12:183-8. [Crossref] [PubMed]
  25. Lin T, Liu Z, Liu L, et al. Prospective evaluation of fluorescence in situ hybridization for diagnosing urothelial carcinoma. Oncol Lett 2017;13:3928-34. [Crossref] [PubMed]
  26. Xu C, Zeng Q, Hou J, et al. Utility of a modality combining FISH and cytology in upper tract urothelial carcinoma detection in voided urine samples of Chinese patients. Urology 2011;77:636-41. [Crossref] [PubMed]
  27. Yu J, Xiong H, Wei C, et al. Utility of fluorescence in situ hybridization analysis for detecting upper urinary tract-urothelial carcinoma. J Cancer Res Ther 2017;13:647-50. [Crossref] [PubMed]
  28. Marín-Aguilera M, Mengual L, Ribal MJ, et al. Utility of fluorescence in situ hybridization as a non-invasive technique in the diagnosis of upper urinary tract urothelial carcinoma. Eur Urol 2007;51:409-15; discussion 415. [Crossref] [PubMed]
  29. Luo B, Li W, Deng CH, et al. Utility of fluorescence in situ hybridization in the diagnosis of upper urinary tract urothelial carcinoma. Cancer Genet Cytogenet 2009;189:93-7. [Crossref] [PubMed]
  30. Zhao Y, Deng FM, Melamed J, et al. Diagnostic role of urine cytology and ureteroscopic biopsies in detection of high grade upper tract urothelial carcinoma. Am J Clin Exp Urol 2021;9:221-8.
  31. Potretzke AM, Knight BA, Vetter JM, et al. Diagnostic Utility of Selective Upper Tract Urinary Cytology: A Systematic Review and Meta-analysis of the Literature. Urology 2016;96:35-43. [Crossref] [PubMed]
  32. Messer J, Shariat SF, Brien JC, et al. Urinary cytology has a poor performance for predicting invasive or high-grade upper-tract urothelial carcinoma. BJU Int 2011;108:701-5. [Crossref] [PubMed]
  33. Malm C, Grahn A, Jaremko G, et al. Diagnostic accuracy of upper tract urothelial carcinoma: how samples are collected matters. Scand J Urol 2017;51:137-45. [Crossref] [PubMed]
  34. Wang X, Gu Y, Zhang S, et al. Unbiased enrichment of urine exfoliated cells on nanostructured substrates for sensitive detection of urothelial tumor cells. Cancer Med 2020;9:290-301. [Crossref] [PubMed]
  35. Wang X, Zhang S, Wu L, et al. Diagnostic performance of an immunoassay based on urine exfoliated cell enrichment nanotechnology for upper tract urothelial carcinoma: a retrospective, monocentric study. BMC Urol 2022;22:194. [Crossref] [PubMed]
  36. Jinzaki M, Kikuchi E, Akita H, et al. Role of computed tomography urography in the clinical evaluation of upper tract urothelial carcinoma. Int J Urol 2016;23:284-98. [Crossref] [PubMed]
  37. Jinzaki M, Matsumoto K, Kikuchi E, et al. Comparison of CT urography and excretory urography in the detection and localization of urothelial carcinoma of the upper urinary tract. AJR Am J Roentgenol 2011;196:1102-9. [Crossref] [PubMed]
  38. Janisch F, Shariat SF, Baltzer P, et al. Diagnostic performance of multidetector computed tomographic (MDCTU) in upper tract urothelial carcinoma (UTUC): a systematic review and meta-analysis. World J Urol 2020;38:1165-75. [Crossref] [PubMed]
  39. Chlapoutakis K, Theocharopoulos N, Yarmenitis S, et al. Performance of computed tomographic urography in diagnosis of upper urinary tract urothelial carcinoma, in patients presenting with hematuria: Systematic review and meta-analysis. Eur J Radiol 2010;73:334-8. [Crossref] [PubMed]
  40. Yu SH, Hur YH, Hwang EC, et al. Does multidetector computed tomographic urography (MDCTU) T staging classification correspond with pathologic T staging in upper tract urothelial carcinoma? Int Urol Nephrol 2021;53:69-75. [Crossref] [PubMed]
  41. Grahn A, Melle-Hannah M, Malm C, et al. Diagnostic accuracy of computed tomography urography and visual assessment during ureterorenoscopy in upper tract urothelial carcinoma. BJU Int 2017;119:289-97. [Crossref] [PubMed]
  42. Fujii Y, Sato Y, Suzuki H, et al. Molecular classification and diagnostics of upper urinary tract urothelial carcinoma. Cancer Cell 2021;39:793-809.e8. [Crossref] [PubMed]
  43. Gillan A, El-Mokadem I, Rai B, et al. Carcinoma in situ is significantly underdetected by prenephroureterectomy ureteroscopy in the management of upper tract urothelial cancers. Biomed Res Int 2015;2015:547586. [Crossref] [PubMed]
  44. Wu GY, Lu Q, Wu LM, et al. Comparison of computed tomographic urography, magnetic resonance urography and the combination of diffusion weighted imaging in diagnosis of upper urinary tract cancer. Eur J Radiol 2014;83:893-9. [Crossref] [PubMed]
  45. Obuchi M, Ishigami K, Takahashi K, et al. Gadolinium-enhanced fat-suppressed T1-weighted imaging for staging ureteral carcinoma: correlation with histopathology. AJR Am J Roentgenol 2007;188:W256-61. [Crossref] [PubMed]
  46. Razavi SA, Sadigh G, Kelly AM, et al. Comparative effectiveness of imaging modalities for the diagnosis of upper and lower urinary tract malignancy: a critically appraised topic. Acad Radiol 2012;19:1134-40. [Crossref] [PubMed]
  47. Goto K, Honda Y, Ikeda K, et al. Tumor heterogeneity evaluated by computed tomography detects muscle-invasive upper tract urothelial carcinoma that is associated with inflammatory tumor microenvironment. Sci Rep 2021;11:14251. [Crossref] [PubMed]
  48. Alqahtani A, Bhattacharjee S, Almopti A, et al. Radiomics-based machine learning approach for the prediction of grade and stage in upper urinary tract urothelial carcinoma: a step towards virtual biopsy. Int J Surg 2024;110:3258-68. [Crossref] [PubMed]
  49. Zheng Y, Shi H, Fu S, et al. A computed tomography urography-based machine learning model for predicting preoperative pathological grade of upper urinary tract urothelial carcinoma. Cancer Med 2024;13:e6901. [Crossref] [PubMed]
  50. Yamany T, van Batavia J, Ahn J, et al. Ureterorenoscopy for upper tract urothelial carcinoma: how often are we missing lesions? Urology 2015;85:311-5. [Crossref] [PubMed]
  51. Mori K, Katayama S, Laukhtina E, et al. Discordance Between Clinical and Pathological Staging and Grading in Upper Tract Urothelial Carcinoma. Clin Genitourin Cancer 2022;20:95.e1-6. [Crossref] [PubMed]
  52. Subiela JD, Territo A, Mercadé A, 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;46:1989-97. [Crossref] [PubMed]
  53. De Coninck V, Keller EX, Somani B, et al. Complications of ureteroscopy: a complete overview. World J Urol 2020;38:2147-66. [Crossref] [PubMed]
  54. Katims AB, Say R, Derweesh I, et al. Risk Factors for Intravesical Recurrence after Minimally Invasive Nephroureterectomy for Upper Tract Urothelial Cancer (ROBUUST Collaboration). J Urol 2021;206:568-76. [Crossref] [PubMed]
  55. Yonese I, Ito M, Waseda Y, et al. Adverse Prognostic Impact of Diagnostic Ureterorenoscopy in a Subset of Patients with High-Risk Upper Tract Urothelial Carcinoma Treated with Radical Nephroureterectomy. Cancers (Basel) 2022;14:3962. [Crossref] [PubMed]
  56. Nowak Ł, Krajewski W, Chorbińska J, et al. The Impact of Diagnostic Ureteroscopy Prior to Radical Nephroureterectomy on Oncological Outcomes in Patients with Upper Tract Urothelial Carcinoma: A Comprehensive Systematic Review and Meta-Analysis. J Clin Med 2021;10:4197. [Crossref] [PubMed]
  57. Sharma V, Miest TS, Juvet TS, et al. The Impact of Upper Tract Urothelial Carcinoma Diagnostic Modality on Intravesical Recurrence after Radical Nephroureterectomy: A Single Institution Series and Updated Meta-Analysis. J Urol 2021;206:558-67. [Crossref] [PubMed]
  58. Traxer O, Geavlete B, de Medina SG, et al. Narrow-band imaging digital flexible ureteroscopy in detection of upper urinary tract transitional-cell carcinoma: initial experience. J Endourol 2011;25:19-23. [Crossref] [PubMed]
  59. Iordache VF, Geavlete PA, Georgescu DA, et al. NBI-assisted digital flexible ureteroscopy in transitional renal cell carcinoma - an evidence-based assessment "through the looking glass" of the pathological analysis. Rom J Morphol Embryol 2018;59:1091-6.
  60. Bus MT, Muller BG, de Bruin DM, et al. Volumetric in vivo visualization of upper urinary tract tumors using optical coherence tomography: a pilot study. J Urol 2013;190:2236-42. [Crossref] [PubMed]
  61. Bus MT, de Bruin DM, Faber DJ, et al. Optical Coherence Tomography as a Tool for In Vivo Staging and Grading of Upper Urinary Tract Urothelial Carcinoma: A Study of Diagnostic Accuracy. J Urol 2016;196:1749-55. [Crossref] [PubMed]
  62. Bui D, Mach KE, Zlatev DV, et al. A Pilot Study of In Vivo Confocal Laser Endomicroscopy of Upper Tract Urothelial Carcinoma. J Endourol 2015;29:1418-23. [Crossref] [PubMed]
  63. Breda A, Territo A, Guttilla A, et al. Correlation Between Confocal Laser Endomicroscopy (Cellvizio(®)) and Histological Grading of Upper Tract Urothelial Carcinoma: A Step Forward for a Better Selection of Patients Suitable for Conservative Management. Eur Urol Focus 2018;4:954-9. [Crossref] [PubMed]
  64. Freund JE, Liem EIML, Savci-Heijink CD, et al. Confocal laser endomicroscopy for upper tract urothelial carcinoma: validation of the proposed criteria and proposal of a scoring system for real-time tumor grading. World J Urol 2019;37:2155-64. [Crossref] [PubMed]
  65. Harada Y, Murayama Y, Takamatsu T, et al. 5-Aminolevulinic Acid-Induced Protoporphyrin IX Fluorescence Imaging for Tumor Detection: Recent Advances and Challenges. Int J Mol Sci 2022;23:6478. [Crossref] [PubMed]
  66. Fukuhara H, Kurabayashi A, Furihata M, et al. 5-aminolevulinic acid-mediated photodynamic diagnosis using fluorescence ureterorenoscopy for urinary upper tract urothelial carcinoma ∼Preliminary prospective single centre trial∼. Photodiagnosis Photodyn Ther 2020;29:101617. [Crossref] [PubMed]
  67. Liu Q, Zhang X, Zhou F, et al. Diagnostic accuracy of photodynamic diagnosis for upper urinary tract urothelial carcinoma: A systematic review and meta-analysis. Photodiagnosis Photodyn Ther 2020;32:102067. [Crossref] [PubMed]
  68. Kata SG, Aboumarzouk OM, Zreik A, et al. Photodynamic diagnostic ureterorenoscopy: A valuable tool in the detection of upper urinary tract tumour. Photodiagnosis Photodyn Ther 2016;13:255-60. [Crossref] [PubMed]
  69. Osman E, Alnaib Z, Kumar N. Photodynamic diagnosis in upper urinary tract urothelial carcinoma: A systematic review. Arab J Urol 2017;15:100-9. [Crossref] [PubMed]
  70. Breda A, Territo A, Sanguedolce F, et al. Comparison of biopsy devices in upper tract urothelial carcinoma. World J Urol 2019;37:1899-905. [Crossref] [PubMed]
  71. Lama DJ, Safiullah S, Patel RM, et al. Multi-institutional Evaluation of Upper Urinary Tract Biopsy Using Backloaded Cup Biopsy Forceps, a Nitinol Basket, and Standard Cup Biopsy Forceps. Urology 2018;117:89-94. [Crossref] [PubMed]
  72. Klein JT, John A, Bohnert L, et al. Improving the Quality of Human Upper Urinary Tract Specimens by Cryobiopsy. Front Oncol 2022;12:810367. [Crossref] [PubMed]
  73. Klett DE, Alom M, Wymer K, et al. A novel upper tract ureteroscopic biopsy technique: the "form tackle". Int Braz J Urol 2022;48:367-8. [Crossref] [PubMed]
  74. Fukumoto K, Kanatani S, Jaremko G, et al. Three-dimensional imaging of upper tract urothelial carcinoma improves diagnostic yield and accuracy. JCI Insight 2024;9:e175751. [Crossref] [PubMed]
  75. Michela B. Liquid Biopsy: A Family of Possible Diagnostic Tools. Diagnostics (Basel) 2021;11:1391. [Crossref] [PubMed]
  76. Satyal U, Srivastava A, Abbosh PH. Urine Biopsy-Liquid Gold for Molecular Detection and Surveillance of Bladder Cancer. Front Oncol 2019;9:1266. [Crossref] [PubMed]
  77. Springer SU, Chen CH, Rodriguez Pena MDC, et al. Non-invasive detection of urothelial cancer through the analysis of driver gene mutations and aneuploidy. Elife 2018;7:e32143. [Crossref] [PubMed]
  78. Xu Y, Ma X, Ai X, et al. A Urine-Based Liquid Biopsy Method for Detection of Upper Tract Urinary Carcinoma. Front Oncol 2020;10:597486. [Crossref] [PubMed]
  79. Crocetto F, Barone B, Ferro M, et al. Liquid biopsy in bladder cancer: State of the art and future perspectives. Crit Rev Oncol Hematol 2022;170:103577. [Crossref] [PubMed]
  80. Wang J, Wu J, Peng L, et al. Distinguishing urothelial carcinoma in the upper urinary tract from benign diseases with hematuria using FISH. Acta Cytol 2012;56:533-8. [Crossref] [PubMed]
  81. Huang WT, Li LY, Pang J, et al. Fluorescence in situ hybridization assay detects upper urinary tract transitional cell carcinoma in patients with asymptomatic hematuria and negative urine cytology. Neoplasma 2012;59:355-60. [Crossref] [PubMed]
  82. Yang T, Li Y, Li J, et al. Diagnostic Value Comparison of Urothelium Carcinoma Among Urine Exfoliated Cells Fluorescent In Situ Hybridization (FISH) Examination, Computerized Tomography (CT) Scan, and Urine Cytologic Examination. Med Sci Monit 2018;24:5788-92. [Crossref] [PubMed]
  83. Sassa N, Iwata H, Kato M, et al. Diagnostic Utility of UroVysion Combined With Conventional Urinary Cytology for Urothelial Carcinoma of the Upper Urinary Tract. Am J Clin Pathol 2019;151:469-78. [Crossref] [PubMed]
  84. Aalami AH, Aalami F. Diagnostic performance of fluorescence in situ hybridization (FISH) in upper tract urothelial carcinoma (UTUC): a systematic review and meta-analysis. Int J Clin Oncol 2022;27:1605-15. [Crossref] [PubMed]
  85. Guan B, Du Y, Su X, et al. Positive urinary fluorescence in situ hybridization indicates poor prognosis in patients with upper tract urothelial carcinoma. Oncotarget 2018;9:14652-60. [Crossref] [PubMed]
  86. Xu B, Zhang JE, Ye L, et al. Evaluation of the diagnostic efficiency of voided urine fluorescence in situ hybridization for predicting the pathology of preoperative "low-risk" upper tract urothelial carcinoma. Front Oncol 2023;13:1225428. [Crossref] [PubMed]
  87. Iwata H, Sassa N, Kato M, et al. UroVysion(®) predicts intravesical recurrence after radical nephroureterectomy for urothelial carcinoma of the upper urinary tract: a prospective study. Int J Clin Oncol 2021;26:178-85. [Crossref] [PubMed]
  88. Audenet F, Isharwal S, Cha EK, et al. Clonal Relatedness and Mutational Differences between Upper Tract and Bladder Urothelial Carcinoma. Clin Cancer Res 2019;25:967-76. [Crossref] [PubMed]
  89. Yuan X, Liu C, Wang K, et al. The genetic difference between Western and Chinese urothelial cell carcinomas: infrequent FGFR3 mutation in Han Chinese patients. Oncotarget 2016;7:25826-35. [Crossref] [PubMed]
  90. D'Andrea D, Soria F, Zehetmayer S, et al. Diagnostic accuracy, clinical utility and influence on decision-making of a methylation urine biomarker test in the surveillance of non-muscle-invasive bladder cancer. BJU Int 2019;123:959-67. [Crossref] [PubMed]
  91. Maas M, Todenhöfer T, Black PC. Urine biomarkers in bladder cancer - current status and future perspectives. Nat Rev Urol 2023;20:597-614. [Crossref] [PubMed]
  92. Territo A, Gallioli A, Diana P, 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;208:570-9. [Crossref] [PubMed]
  93. Guo RQ, Xiong GY, Yang KW, et al. Detection of urothelial carcinoma, upper tract urothelial carcinoma, bladder carcinoma, and urothelial carcinoma with gross hematuria using selected urine-DNA methylation biomarkers: A prospective, single-center study. Urol Oncol 2018;36:342.e15-23. [Crossref] [PubMed]
  94. Pierconti F, Martini M, Fiorentino V, et al. Upper urothelial tract high-grade carcinoma: comparison of urine cytology and DNA methylation analysis in urinary samples. Hum Pathol 2021;118:42-8. [Crossref] [PubMed]
  95. Ouyang W, Luo L, Zhang J, et al. Urine Cellular DNA Point Mutation and Methylation for Identifying Upper Tract Urinary Carcinoma. Cancers (Basel) 2022;14:3537. [Crossref] [PubMed]
  96. Monteiro-Reis S, Leça L, Almeida M, et al. Accurate detection of upper tract urothelial carcinoma in tissue and urine by means of quantitative GDF15, TMEFF2 and VIM promoter methylation. Eur J Cancer 2014;50:226-33. [Crossref] [PubMed]
  97. Pycha S, Trenti E, Mian C, et al. Diagnostic value of Xpert® BC Detection, Bladder Epicheck®, Urovysion® FISH and cytology in the detection of upper urinary tract urothelial carcinoma. World J Urol 2023;41:1323-8. [Crossref] [PubMed]
  98. D'Elia C, Trenti E, Krause P, et al. Xpert® bladder cancer detection as a diagnostic tool in upper urinary tract urothelial carcinoma: preliminary results. Ther Adv Urol 2022;14:17562872221090320. [Crossref] [PubMed]
  99. Fujimoto M, Arai E, Tsumura K, et al. Establishment of diagnostic criteria for upper urinary tract urothelial carcinoma based on genome-wide DNA methylation analysis. Epigenetics 2020;15:1289-301. [Crossref] [PubMed]
  100. Nestler T, Schoch J, Belge G, et al. MicroRNA-371a-3p-The Novel Serum Biomarker in Testicular Germ Cell Tumors. Cancers (Basel) 2023;15:3944. [Crossref] [PubMed]
  101. Tao J, Yang X, Li P, et al. Identification of circulating microRNA signatures for upper tract urothelial carcinoma detection. Mol Med Rep 2015;12:6752-60. [Crossref] [PubMed]
  102. Kriebel S, Schmidt D, Holdenrieder S, et al. Analysis of tissue and serum microRNA expression in patients with upper urinary tract urothelial cancer. PLoS One 2015;10:e0117284. [Crossref] [PubMed]
  103. Ke HL, Li WM, Lin HH, et al. Hypoxia-regulated MicroRNA-210 Overexpression is Associated with Tumor Development and Progression in Upper Tract Urothelial Carcinoma. Int J Med Sci 2017;14:578-84. [Crossref] [PubMed]
  104. Urabe F, Matsuzaki J, Takeshita F, et al. Independent verification of circulating miRNA as diagnostic biomarkers for urothelial carcinoma. Cancer Sci 2022;113:3510-7. [Crossref] [PubMed]
  105. Browne BM, Stensland KD, Patel CK, et al. MicroRNA Expression Profiles in Upper Tract Urothelial Carcinoma Differentiate Tumor Grade, Stage, and Survival: Implications for Clinical Decision-Making. Urology 2019;123:93-100. [Crossref] [PubMed]
  106. Li Y, He S, He A, et al. Identification of plasma secreted phosphoprotein 1 as a novel biomarker for upper tract urothelial carcinomas. Biomed Pharmacother 2019;113:108744. [Crossref] [PubMed]
  107. Kodama H, Yoneyama T, Tanaka T, et al. N-glycan signature of serum immunoglobulins as a diagnostic biomarker of urothelial carcinomas. Cancer Med 2021;10:1297-313. [Crossref] [PubMed]
  108. Qu Y, Yao Z, Xu N, et al. Plasma proteomic profiling discovers molecular features associated with upper tract urothelial carcinoma. Cell Rep Med 2023;4:101166. [Crossref] [PubMed]
  109. Gong YW, Wang YR, Fan GR, et al. Diagnostic and prognostic role of BTA, NMP22, survivin and cytology in urothelial carcinoma. Transl Cancer Res 2021;10:3192-205. [Crossref] [PubMed]
Cite this article as: Shan Z, Deng X, Yang L, Zhang G. Diagnostic status and new explorations in upper urinary tract urothelial carcinoma: a literature review. Transl Androl Urol 2024;13(11):2570-2586. doi: 10.21037/tau-24-326

Download Citation