Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
Oncology Reports
Join Editorial Board Propose a Special Issue
Print ISSN: 1021-335X Online ISSN: 1791-2431
Journal Cover
November-2025 Volume 54 Issue 5

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
November-2025 Volume 54 Issue 5

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML
Review Open Access

Lymphadenectomy in upper tract urothelial carcinoma: Clinical insights and controversies (Review)

  • Authors:
    • Dihao Lv
    • Yinglong Huang
    • Haihao Li
    • Shi Fu
    • Chen Gong
    • Chadanfeng Yang
    • Jiansong Wang
    • Haifeng Wang
    • Zhiyong Tan
    • Mingxia Ding
  • View Affiliations / Copyright

    Affiliations: Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Yunnan Institute of Urology, Kunming, Yunnan 650101, P.R. China
    Copyright: © Lv et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 137
    |
    Published online on: August 8, 2025
       https://doi.org/10.3892/or.2025.8970
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:



Abstract

Upper tract urothelial carcinoma (UTUC), a relatively rare but highly malignant tumor arising from the renal pelvis and the urothelial layer of the ureter, accounts for 5‑10% of all urothelial malignancies. The lack of specific early symptoms commonly results in a delayed diagnosis, with >60% of cases being diagnosed at an advanced stage of the disease. Lymph node involvement can critically affect the clinical outcomes, thus serving as a key prognostic indicator, guiding both staging protocols and treatment strategies. Lymphadenectomy, typically performed alongside radical nephroureterectomy, not only assists in evaluating the extent of the regional metastases, but also plays a critical role in guiding both staging and treatment planning. Although several studies have suggested that a more extensive lymph node dissection (LND) can improve cancer‑specific survival and disease‑free survival rates, there is still no consensus on the optimal extent of dissection or a universally accepted surgical template. The present review aims to summarize the existing evidence on the efficacy of LND in UTUC, thus emphasizing its potential in staging, outcome prediction and possible survival benefits. The review also aims to address the challenges caused by inconsistent LND practices and the limited availability of robust prospective data. Furthermore, it discusses novel biomarkers that could improve patient classification, and proposes future research directions to improve the management of UTUC through more personalized and evidence‑driven LND strategies.

Introduction

Upper tract urothelial carcinoma (UTUC) is a rare yet clinically aggressive type of cancer, which arises from the transitional epithelium lining the renal pelvis (RP), ureters and proximal urethra (1). Distinct from bladder urothelial carcinoma, UTUC is characterized by unique biological behaviors in terms of clinical presentation, anatomical localization and molecular alterations (2,3). Accounting for 5–10% of all urothelial cancer cases, this disease shows a high prevalence of nodal metastases at initial diagnosis, with lymph node involvement being recorded in 20–30% of cases (4). The frequent absence of early symptoms is considered a marked challenge in the management of UTUC, and commonly leads to late-stage diagnosis, with ~60% of cases being diagnosed at an advanced disease stage (5). In high-grade or locally invasive tumors, the lymph node metastasis (LNM) rate can increase to 30–40% (6). Therefore, LNM is considered a well-established predictor of poor survival outcomes (7).

The treatment approach for non-metastatic UTUC depends on the risk level of the tumor. For patients with low-risk (LR) UTUC, clinicians typically favor minimally invasive approaches, including kidney-preserving endoscopic procedures that maintain renal function. By contrast, high-risk (HR) UTUC typically necessitates a conventional treatment approach, including radical nephroureterectomy (RNU) combined with excision of the bladder cuff. This strategy aids in local disease management and can enhance cancer-specific survival (CSS) outcomes (1,8). It has been reported that in UC, LNM can commonly spread to nearby lymph nodes through the lymphatic drainage pathway (9). Therefore, systemic lymphadenectomy could exert both therapeutic and diagnostic roles by providing more accurate tumor staging, which in turn could improve treatment planning after surgery (10,11). This is particularly important for identifying patients who could benefit from additional therapies (1,12). However, there is still no consensus on the extent of lymph node dissection (LND) in patients with UTUC.

The present review aims to provide an integrative analysis of LND in the management of UTUC, highlighting its evolving clinical applications. The clinical relevance of LND in staging protocols and disease characterization, as well as its prognostic significance in predicting therapeutic efficacy and recurrence, are also discussed. Current controversies regarding surgical strategies and the technical demands associated with LND procedures are critically evaluated. Furthermore, the potential survival benefits associated with the extent of lymph node dissection are analyzed through both clinicopathological and molecular perspectives.

Pathophysiology and lymphatic drainage patterns

In UTUC, lymph nodes are the primary sites for metastasis. Understanding lymphatic drainage patterns is crucial for defining the extent of LND and improving the accuracy of staging and prognosis (13). The drainage variations by tumor location are illustrated in Fig. 1.

Regional lymphovascular drainage
depending on the location of the primary tumors according to the
studies by Matin et al (18)
and Kondo and Tanabe (9). Regional
nodal sites for the renal pelvis and the (A) upper ureter, (B)
middle ureter and (C) distal ureter, with yellow circles
representing the right side and green circles representing the left
side. 1, renal hilar; 2, paracaval; 3, retrocaval; 4, paraaortic;
5, interaortocaval; 6, common iliac; 7, external iliac; 8, internal
iliac; 9, obturator; 10, suprahilar; 11, aortic bifurcation; 12,
presacral.

Figure 1.

Regional lymphovascular drainage depending on the location of the primary tumors according to the studies by Matin et al (18) and Kondo and Tanabe (9). Regional nodal sites for the renal pelvis and the (A) upper ureter, (B) middle ureter and (C) distal ureter, with yellow circles representing the right side and green circles representing the left side. 1, renal hilar; 2, paracaval; 3, retrocaval; 4, paraaortic; 5, interaortocaval; 6, common iliac; 7, external iliac; 8, internal iliac; 9, obturator; 10, suprahilar; 11, aortic bifurcation; 12, presacral.

Although currently no standardized LND templates have been established by major guidelines, such as those provided by the European Association of Urology (EAU) and American Urological Association (AUA) (14–16), a foundational work by Akaza et al (17) was the first to propose a Tumor-Node-Metastasis framework for UTUC. Later, Kondo and Tanabe (9) refined this framework by mapping eight anatomical zones associated with tumor location, namely the RP, and the upper, middle or lower ureter, bilaterally. Regional nodes were defined as those with a metastasis risk of >10%. Proximal segments correspond to areas lying above the inferior mesenteric artery (IMA), the mid-ureteral region spans from the IMA to the common iliac (CI) artery and the distal ureter (DU) extends to the ureteral meatus (9). This mapping laid the foundation for the widely referenced ‘Kondo and Matin templates’ (9,18,19). The LND templates based on the primary tumor location from the studies by Matin et al (18), Kondo et al (19) are illustrated in Fig. 2.

Templates for lymphadenectomy based
on the primary tumor location according to the studies by Matin
et al (18) and Kondo et
al (19). (A) Right renal
pelvis and upper ureter (red box: Renal hilar, paracaval and
retrocaval LNs). (B) Left renal pelvis and upper ureter (yellow
box: Renal hilar and paraaortic LNs). (C) Right middle ureter
(green box: Interaortocaval LNs). (D) Left middle ureter (orange
box: Paraaortic LNs). (E) Right distal ureter (purple box: Common
iliac, external iliac, internal iliac and obturator LNs). (F) Left
distal ureter (blue box: Common iliac, external iliac, internal
iliac, obturator LNs). 1, renal hilar; 2, paracaval; 3, retrocaval;
4, paraaortic; 5, interaortocaval; 6, common iliac; 7, external
iliac; 8, internal iliac; 9, obturator. LN, lymph node.

Figure 2.

Templates for lymphadenectomy based on the primary tumor location according to the studies by Matin et al (18) and Kondo et al (19). (A) Right renal pelvis and upper ureter (red box: Renal hilar, paracaval and retrocaval LNs). (B) Left renal pelvis and upper ureter (yellow box: Renal hilar and paraaortic LNs). (C) Right middle ureter (green box: Interaortocaval LNs). (D) Left middle ureter (orange box: Paraaortic LNs). (E) Right distal ureter (purple box: Common iliac, external iliac, internal iliac and obturator LNs). (F) Left distal ureter (blue box: Common iliac, external iliac, internal iliac, obturator LNs). 1, renal hilar; 2, paracaval; 3, retrocaval; 4, paraaortic; 5, interaortocaval; 6, common iliac; 7, external iliac; 8, internal iliac; 9, obturator. LN, lymph node.

Lymph node status remains a critical prognostic indicator in UTUC. Therefore, previous studies indicated that node-negative (pN0) patients exhibited significantly superior 5-year disease-free survival (DFS) (84.5 vs. 43.6%; P<0.001) and CSS (78.3 vs. 33.2%; P<0.001) rates compared with node-positive (pN+) patients (9,20,21). These findings underscored the significant effect of nodal involvement in the onset of adverse effects and highlighted the importance of accurate nodal assessment in guiding adjuvant treatment decision, as evidenced by clinical trials, such as POUT (22).

Emerging evidence has also suggested that other clinical factors, such as age and gender, and biomarkers, including elevated fibrinogen, cystatin-C, C-reactive protein, neutrophil-to-lymphocyte ratio of >2.7, albumin-to-globulin ratio of <1.45, and preoperative anemia, can be also associated with LNM risk (13,14,23–28).

Clinical application and extent of LND in UTUC

Building on the anatomical principles established by the ‘Kondo and Matin templates’ (9,18,19), several clinical studies have evaluated the therapeutic outcomes of LND. Kondo et al (29) performed retrospective and prospective analyses via comparing the efficacy of systematic complete LND (CLND), incomplete or limited LND (ILND), and no LND (9,29–31). The results demonstrated that in patients with ≥pT2 disease, CLND could display superior CSS compared with ILND/no LND, and more particularly in patients with pT3+ tumors (5-year CSS, 73.2 vs. 43.7 and 47.3%, respectively) (9). For patients with ≥pT2 cN0M0 UTUC, CLND showed improved CSS compared with ILND (86 vs. 71%; P=0.04). Furthermore, CLND could independently predict both CSS and regional recurrence-free survival (RFS) in RP tumors (31). However, the survival benefits were less evident in distal ureteral tumors, possibly due to variations in the extent of lymph node dissection. By contrast, the population-based study by Lughezzani et al (8), and a multi-institutional study (32) reported no significant therapeutic survival benefits for LND, although in these studies the dissection templates were not specified.

Extended LND is increasingly recognized for its prognostic value. However, the majority of RNU procedures remove <8 lymph nodes (33). It has been reported that lymph node density, defined as the number of positive nodes to that of total nodes, can provide higher prognostic value compared with absolute lymph none counts. Therefore, a nodal density ratio of >30% was associated with higher recurrence [hazard ratio (HR), 1.8; P=0.021) and mortality (HR,1.7; P=0.032) risks (34,35). Consistently, lymph node density of >20% could predict worse RFS (HR,1.94) and overall survival (OS; HR, 2.34) (20). The safety of LND has been also well-documented. Therefore, operative duration, estimated blood loss, and hospitalization period were comparable across CLND/ILND/no LND cohorts, with only a minor increase in the incidence of complications reported in CLND (9,36,37).

Due to limited data, the optimal extent of LND in patients with UTUC remains still controversial. The widely adopted ‘Kondo and Matin template’ (9,18) is based on small samples. Furthermore, the genomic differences in UTUC between Chinese and Western populations suggests that the application of LND templates should be region-specific. Notably, to date no studies have directly compared the effect of different LND extents on OS and PFS (38).

The conflicting conclusions between the studies by Kondo et al and Lughezzani et al could be due to critical methodological disparities. Firstly, there was heterogeneity in the surgical techniques applied. Therefore, Kondo et al employed a standardized template-based dissection protocol, including the dissection of interaortocaval (IAC) lymph nodes in right RP tumors. By contrast, the population-based analysis by Lughezzani et al lacked specified dissection templates, potentially leading to omission of HR nodal regions. Secondly, a patient selection bias was recorded. In the study by Kondo et al only patients with cN0 UTUC, who underwent meticulous CLND dissection were included. However, in the study by Lughezzani et al the patient cohort was more heterogeneous, encompassing both clinically node 0 (cN0) and cN+ cases, with variable extents of dissection, thus diluting any therapeutic effects. Thirdly, adjuvant therapy rates varied between the two studies. Therefore, >40% of patients in the study by Lughezzani et al received adjuvant chemotherapy (ChT) compared with only <15% in Kondo's cohort, thus potentially obscuring LND-specific survival benefits (8,9,11,30,31). These factors collectively indicated that the discrepancies in the survival rates could reflect limitations in study design rather than the intrinsic therapeutic value of LND. Future prospective trials should standardize dissection templates and stratify patients based on nodal risk to more accurately evaluate the therapeutic value of LND.

Prognostic significance of LND in UTUC

LNM is associated with adverse clinical outcomes and substantially decreased survival (39). Although evidence remains conflicting regarding the therapeutic value of LND, its potential in achieving definitive pathological staging of UTUC has been well documented. The majority of the available data has been derived from retrospective, single-institution studies, with several of them supporting the prognostic value of LND (12,29,40). These reports demonstrated that pN0 patients exhibited a superior CSS compared with pN+ patients, with 5-year CSS estimates of 56–85 and 0–39%, respectively. These findings indicated that accurate nodal evaluation via LND during RNU could enhance the postoperative risk classification and guide treatment decisions. Notably, two studies identified pNx status as an independent survival risk factor compared with pN0 status (29,41). By contrast, another study did not detect nodal status-related differences in survival endpoints (42). In addition, a population-based study that included 2,824 subjects revealed marked disparities in 5-year CSS, with rates of 34, 78, and 81% recorded for pN+, pNx and pN0 patients, respectively (P<0.001). However, no prognostic significance was reported between the pN0 and pNx groups (43). Only two multicenter cohorts showed differences in CSS rates between the pN0 and pNx groups (12,44). Ouzzane et al (43) found no independent association between CSS and nodal status in adjusted models (pN+ vs. pN0: HR, 1.6; 95% CI, 0.8–3.4; P=0.104; and pNx vs. pN0: HR, 1.14; 95% CI, 0.7–1.9; P=0.592). Subgroup analyses more consistently supported the prognostic value of LND in staging muscle-invasive or locally advanced UTUC (11,32,45). However, the findings of the aforementioned studies have not been verified by a multicenter study (43). Importantly, studies failing to demonstrate staging benefits commonly lack dissection protocol details, thus emphasizing the need for standardized LND templates.

Recent advances in immunotherapy have further underscored the critical role of LND in guiding systemic therapy decisions. According to the 2023 EAU guidelines, adjuvant nivolumab is now recommended for HR patients (≥pT3 or pN+) who are ineligible for or decline platinum-based ChT, provided that their tumors express programmed death-ligand 1 (PD-L1) ≥1% (15). This recommendation is based on proven survival benefits observed in the CheckMate 274 trial in patients with muscle-invasive UC (46). However, UTUC-specific data remain limited. Notably, LND remains indispensable for identifying nodal involvement (pN+ status), which directly determines eligibility for both adjuvant ChT and immunotherapy. While immune checkpoint inhibitors (ICIs) offer novel therapeutic insights, they do not alter the fundamental indications or extent of LND, a template-based dissection approach, which continues to be prioritized for accurate pathological staging and local disease control. Future studies should explore whether genomic differences, such as fibroblast growth factor receptor (FGFR) alterations, between Eastern and Western populations could affect immunotherapy responsiveness and necessitate region-specific LND templates (15).

Therapeutic strategies and controversies in patients with cN+ UTUC

For patients with cN+ UTUC commonly experiencing lymphatic dissemination, neoadjuvant platinum-based ChT prior to RNU/LND merits consideration. However, current guidelines lack UTUC-specific evidence to endorse this strategy, compared with the well-established protocols for bladder cancer (1,47). Regarding ChT timing in operable cN+ disease, the multicenter study by Shigeta et al (48) compared neoadjuvant vs. adjuvant approaches and found that preoperative ChT was associated with significantly improved 3-year DFS (58.2 vs. 37.1%; P=0.004) and CSS (71.3 vs. 47.6%; P=0.002) rates. Notably, selecting adjuvant therapy following RNU critically depends on the pathological findings of LND (49). A previous study suggested that initiating gemcitabine-cisplatin ChT within 90 days after surgery could improve DFS rate (HR, 0.45; 95% CI, 0.30–0.70; P<0.0001) in patients with locally advanced UTUC (22). In metastatic UC, cisplatin-based ChT showed improved response rates compared with carboplatin-based ChT (50). However, as non-responders often switch to immunotherapy, selection bias persists (48). These findings reinforce managing cN+ disease as systemic malignancy requiring comprehensive treatment. Importantly, the majority of studies evaluating RNU by LND excluded cN+ patients, hindering natural history assessment and perpetuating debates regarding the therapeutic value of LND. Although the oncological effect of LND is modest in cN+ cases, accurate nodal staging remains vital for systemic treatment planning (22,51). The current and proposed staging classifications are summarized in Table I (52,53).

Table I.

Stage classification for upper tract urothelial carcinoma: AJCC staging and proposed modifications.

Table I.

Stage classification for upper tract urothelial carcinoma: AJCC staging and proposed modifications.

StageAJCC staging (6th, 7th and 8th editions)Modifications by Li et al (52)Modifications by Abdel-Rahman (53)
IT1N0M0T1N0M0T1N0M0
IIT2N0M0T2N0M0T2N0M0
IIIT3N0M0T3N0M0T3N0M0
IVT4/N+/M1Low grade (G1-2): T4NxM0, TxN1-2M0, TxNxM1IVA: T4/N+M0
High grade (G3-4): T4NxM0, TxN1-2M0, TxNxM1IVB: T1-4/N0/+M1

[i] Nx, regional lymph nodes cannot be assessed; N+, regional lymph node-positive; M1, distant metastasis is present; T4, tumor has invaded into adjacent organs; Tx, primary tumor cannot be assessed; AJCC, American Joint Committee on Cancer.

Controversy of LND in UTUC

Current urological guidelines show considerable variability in their recommendations for LND in UTUC. According to the 2022 EAU guidelines, template-based LND should be considered over node-quantity-based approaches, thus emphasizing its potential survival advantages. Template-based LND has been associated with prolonged CSS time in muscle-invasive cases, reduced local recurrence rates, and improved outcomes in patients without clinical or pathological nodal involvement. Due to the difficulty in preoperatively diagnosing Ta and T1 tumor stages, the EAU strongly recommends concurrently performing template-based LND for all patients undergoing RNU (15). The 2023 AUA guidelines recommend LND during RNU or urethrectomy for HR UTUC, while in patients with LR UTUC, it can be performed under particular conditions, such as intraoperative detection of suspicious nodes, mid/distal ureteral tumors requiring template dissection, discordant biopsy/imaging features or when adjuvant therapy eligibility requires pathological nodal staging (16). The 2023 edition of the National Comprehensive Cancer Network (NCCN) guidelines recommends regional LND for high-grade, large, renal pelvic cancers that invade the renal parenchyma, and high-grade ureteral cancers (54). However, a more scientifically grounded LND template should be developed based on studies of LNM distribution and risk probability.

The standardized LND boundaries recommended by the EAU, NCCN and AUA guidelines are delineated in Table II (15,16,18,19). The EAU criteria integrate findings from three foundational studies, two of which provide precise anatomical resection templates (18,31,55). For malignancies involving the RP, upper ureter (UU) and mid-ureter (MU), Kondo's protocol mandates resection of the renal hilar (RH), paracaval (PC), retrocaval (RC) and IAC lymph nodes (31). By contrast, left-sided tumors necessitate RH and periaortic (PA) lymph node clearance. Lesions affecting the DU require the comprehensive dissection of ipsilateral CI, external iliac (EI), internal iliac (II) and obturator (Ob) nodal basins. Matin's approach differentiates primary and extended dissection zones, particularly for MU tumors, to optimize the detection of metastasis (18). For right RP and UU tumors, RH/PC/RC dissections supplemented with IAC node dissection are recommended to enhance sensitivity. For left counterparts, RH/PA nodes with IAC inclusion are preferred. Right MU tumors need IAC, PC, and RC node dissection for residual disease, while left-sided cases include PA, CI and II nodes. Finally, for right-sided DU tumors, resection of CI, EI, II, Ob and PC nodes is warranted, while for left-sided DU tumors, the resection includes CI, EI, II, Ob and PA nodes (18).

Table II.

Lymph node dissection protocols in upper tract urothelial carcinoma: EAU, AUA and NCCN perspectives.

Table II.

Lymph node dissection protocols in upper tract urothelial carcinoma: EAU, AUA and NCCN perspectives.

GuidelineTumor locationExtent of LNDLymph node groups(Refs.)
EAURP, UU, MU and- Right-sided/left-sided RP, UU and MU tumors:RH, PC, RC,(15,18,19)
DU tumors (right-RH, PC, RC and IAC LNs/RH, and PA LNs.IAC, CI, EI,
sided/left-sided)- Right-sided/left-sided DU tumors: Ipsilateral CI, EI, Ob and II LNs.Ob, II and PA
- Right-sided/left-sided RP and UU tumors (Matin et al): Primarily RP, PC, RC LNs/RH and PA LNs, with additional IAC nodes on both sides to increase the chance of capturing all LNMs.
- Right-sided/left-sided MU tumors (Matin et al): Primarily IAC LNs/PA LNs, with addition of PC, RC, CI and II nodes to remove remaining LNMs.
- Right-sided/left-sided DU tumors (Matin et al): Primarily ipsilateral CI, EI, II and Ob LNs, with addition of PC and PA nodes to remove remaining LNMs.
AUAPyelocaliceal system (upper 2/3 of ureter, distal 1/3 of ureter)- Tumors in the pyelocaliceal system: LNs of the ipsilateral great vessel extending from the RH to at least the inferior mesenteric artery.RH, PA, CI, EI, Ob and II(16)
- Tumors in the upper 2/3 of the ureter: LNs of the ipsilateral great vessel extending from the RH to the aortic bifurcation.
- Tumors in the distal 1/3 of the ureter: Ipsilateral pelvic LND, including at minimum the Ob and EI nodal packets. II and CI nodes may be removed in the appropriate clinical settinga. Higher dissectionb may be considered based on cranial migration of LNM and clinician judgment.
NCCNRP, UU, MU and DU tumors (left-sided/right-sided)- Left-sided RP, UU and MU tumors: Regional LND includes PA LNs from the RH to the aortic bifurcation. Most MU tumors will also include CI, EI, Ob and hypogastric LNs.PA, CI, EI, Ob, hypogastric and PC(54)
- Right-sided RP, UU and MU tumors: Regional LND includes PC LNs from the RH to the inferior vena cava bifurcation. Most MU tumors will also include CI, EI, Ob and hypogastric LNs.
- DU tumors: Regional LND should include CI, EI, Ob and hypogastric LNs.

a Lymph node dissection may be adapted depending on the clinical stage, preoperative imaging (such as computed tomography or magnetic resonance imaging findings), or intraoperative findings, such as suspected or confirmed lymph node metastasis.

b Higher dissection refers to extending the dissection to higher lymph node levels (for example, aortic bifurcation) in cases where lymph node metastases are suspected to migrate cranially, which may be considered based on tumor characteristics (size and stage) or clinician judgment. RH, renal hilar; PA, paraaortic; PC, paracaval; RC, retrocaval; IAC, interaortocaval; CI, common iliac; EI, external iliac; II, internal iliac; Ob, obturator; RP, renal pelvis; UU, upper ureter; MU, middle ureter; DU, distal ureter; LND, lymph node dissection; EAU, European Association of Urology; NCCN, National Comprehensive Cancer Network; AUA, American Urological Association.

According to the NCCN guidelines, the surgical management of the majority of MU neoplasms typically requires nodal dissection extending from the renal hilum to the branching of the inferior vena cava, including the precaval nodes, with concurrent removal of the CI, EI, Ob and pelvic arterial lymph nodes. For PA tumors, nodal resection should encompass the region between the renal pedicle and aortic division, incorporating the CI, EI, Ob and pelvic vascular nodes. In patients with DU tumors, complete unilateral excision of the CI, EI, Ob and pelvic arterial lymph nodes is required (54).

According to AUA recommendations, surgical management of pelvicalyceal malignancies should include unilateral dissection of the major vascular lymph node bundles spanning from the RH to the origin of the IMA. Tumors located to the upper two-thirds of the ureter warrant dissection of lymph nodes extending from the RH to the aortic bifurcation. However, tumors of the third distal ureter require removal of Ob and EI nodes, with optional dissection of the II and CI nodes. Emerging evidence has suggested that for cranial LNM metastasis, extended dissections based on clinical assessment should be considered (18,29,36,37,45,56–63).

Analysis of the Surveillance, Epidemiology and End Results database by Zhai et al (21) and Dong et al (64) suggested a potential survival benefit from LND in patients with cN0 UTUC, particularly in those with T3-4 disease, possibly due to the clearance of micrometastasis. However, to verify the effect of dissection extent on oncological outcomes, further studies are urgently warranted.

Given the ongoing debate regarding the efficacy of LND in UTUC, defining a reasonable dissection range is urgently needed. However, the current research on LND in UTUC remains limited, and the field is still in an exploratory state. Notably, no studies have directly compared the effects of different dissection ranges on clinical outcomes, such as OS and PFS. Although several anatomical templates for LND have been proposed, standardizing LND indications and techniques is challenging due to the retrospective and multicenter nature of the existing studies. In the majority of cases, the decision to perform LND is based on the judgment of the surgeon, considering different factors, such as clinical presentation, tumor location and laterality. The patient outcomes from the reviewed studies according to the lymph node pattern are summarized in Table III (11,14,20,30,32,40,41,45,56,59,61,65,66).

Table III.

Summary of clinical outcomes and treatment approaches in upper tract urothelial carcinoma.

Table III.

Summary of clinical outcomes and treatment approaches in upper tract urothelial carcinoma.

First author, yearDesignYearSample size, npN stage, n (%)5-year DFS, %5-year CSS, %5-year OS, %Median number of LNs removed (range)LN densitySurgical approach, n (%)Post-operative complicationsMedian follow-up (range), months(Refs.)
Brown et al, 2006Single2006184pN0, 89 (48);47.4;78.0;70.3;NSNSOpen, 158 (86);NS30.0(65)
institutional pNx, 71 (39);47.4;78.0;70.3; laparoscopic with
retrospective pN+, 24 (13)NS37.633.9 open bladder-cuff
excision, 26 (14)
KondoSingle2007181pN0, 139 (77);NS85.2;NS6.0 (2–30)Mean,NSNSNS(30)
et al, 2010institutional pNx/pN+, 32 15.5 3.7
retrospective (18)/10 (5) (range,
0-16)
(absolute
number)
BrausiMulti-200782pN0/pN+, 24NSNS80.0;NSNSOpenNS64.7(56)
et al, 2007institutional (29)/16 (20); 30.0 transperitoneal,
retrospective pNx, 42 (51) 74 (90); open
flank, 8 (10)
Secin et al,Single2007252pN0, 105 (41);NS56.0;NS4.0 (2–10)NSOpen (98);NS37.0(40)
2007institutional pNx 28 (11); 73.0; 0 Laparoscopic (2)
retrospective pN+, 119 (48)
RoscignoSingle2008132pN0, 69 (52);72.0;73.0;NS8.0 (2–24)NSOpen (100)NS42.0 (2–191)(41)
et al, 2008institutional pNx, 27 (20);39.0;48.0;
retrospective pN+, 36 (28)35.039.0
RoscignoMulti-20091,130pN0, 412 (36);71.0;77.0;NSNSNSOpen, 924 (82);NS45.0 (1–250)(45)
et al, 2009institutional pNx, 578 (51);66.0;69.0; laparoscopic,
retrospective pN+, 140 (13)29.035.0 206 (18)
LughezzaniMulti-20102,842pN0, 1,835 (64);NS81.2;NSNSNSNSNS43.0 (1–203)(11)
et al, 2010institutional pNx 747 (26); 77.8;
retrospective pN+, 242 (9) 34.2
MasonMulti-20121,029pN0, 199 (20);90.9;72.1;NS4.3 (mean)Mean,Open, 583 (57);NS19.8 (7.2–53.8)(20)
et al, 2012institutional pNx, 753 (73);70.6;74.7; 20%laparoscopic,
retrospective pN+, 77 (7)80.029.8 446 (43)
BurgerMulti-2011785pN0, 136 (17);71.6;79.0;NS3.0 (2–6)NSOpen, 715 (91);NS34.0 (15–65)(32)
et al, 2011institutional pNx, 595 (76);76.9;77.4; laparoscopic,
retrospective pN+, 54 (7)21.326.7 70 (9)
Yoo et al,Single2017418pN0, 116 (29);76.4;NS80.2;7.0 (3–10)NSOpen, 106 (37);NS69.0(60)
2017institutional pNx, 286 (68);73.4; 71.7; minimal invasive,
retrospective pN+, 16 (3)93.7 12.5 180 (63)
IkedaMulti-2017404pN0, 182 (45);78.3;84.5;NS6.0 (3–10)NSOpen, 296 (74);NS43.0 (17–89)(59)
et al, 2017institutional pNx, 177 (45);61.9;73.3; laparoscopic,
retrospective pN+, 40 (10)33.243.6 103 (26)
InokuchiMulti-20172,037pN0, 955 (47);NSNS69.3;6 .0 (3–11)NSOpen, 1,234 (60.5);NS45.8 (21.8–75.9)(14)
et al, 2017institutional pNx, 859 (42); 60.5; laparoscopic,
retrospective pN+, 223 (11) 30.0 787 (38.6)
Li et al,Multi-20211,340pN0, 278 (21);NSNSNSNSNSHand-assisted,Clavien-NS(66)
2021institutional pNx, 1,004 (75); 741 (55); pureDindo >2:
retrospective pN+, 58 (4) laparoscopic,80 patients
458 (34); robotic,(6%)
141 (11)

[i] n (%) values indicate number of patients receiving therapy (percentage of total sample size). NS, not specified; DFS, disease-free survival; CSS, cancer-specific survival; OS, overall survival; LN, lymph node.

Surgical outcomes and complications associated with LND in UTUC

Radical RNU can be performed using open, laparoscopic or robot-assisted techniques, with comparable oncological and safety outcomes (67,68). A study by Pearce et al (62) showed that patients undergoing LND had a 30% higher risk of postoperative complications compared with those without LND. However, no significant differences were observed in intraoperative complications among the different surgical approaches. Robot-assisted NU has been associated with the lowest postoperative complication rate, while open NU exhibits the highest rate, particularly for gastrointestinal and hemorrhagic complications (P<0.001).

In a previous study, Ishiyama et al (69) applied the Clavien-Dindo grading system (70) to evaluate postoperative complications in patients undergoing CLND compared with those in the no LND or ILND group, following propensity matching. The overall adverse event rates were comparable between the two groups (16.7 vs. 20.0%; P=0.7385). However, the incidence of lymphatic leakage was markedly increased in the CLND group (4.76 vs. 0.00%; P=0.0231). Notably, only one high-grade complication, namely Clavien-Dindo grade ≥III, occurred in the CLND cohort. The analysis by Pearce et al (62) of 16,619 RNU patients, including 2,560 who underwent LND, revealed equivalent intraoperative complication rates between the two groups (4%) (62). Postoperative complications were more common in patients who were treated with LND (27–29%). However, statistical significance was not reached (P=0.397). Multivariable analysis revealed a 30% increased risk of surgical morbidity associated with LND [adjusted odds ratio (OR)=1.3]. Winer et al (63) revealed a dose-dependent association between the quantity of lymph nodes removed and 30-day postoperative complication rates (OR=1.18 for each additional five lymph nodes excised; 95% confidence interval, 1.05–1.32; P=0.004). However, no corresponding increase in high-grade complications (grade, ≥III) was observed.

Furthermore, a randomized trial by Blom et al (71) demonstrated no significant differences in morbidity/mortality between different LND extents. However, patients in the LND group exhibited higher rates of blood loss of >1 liter (9.4 vs. 6.5%), thromboembolism (2.2 vs. 1.1%) and lymphatic drainage than the non-LND group. Consistent with other studies, Kondo et al (72) compared template-based LND with non-LND approaches and showed numerically higher, but not statistically significant, complication rates across different Clavien-Dindo grades. Procedure-specific events, including thigh numbness (2.6 vs. 0%) and lymphorrhea (5.2 vs. 1.1%), were more common in patients treated with LND compared with the non-LND group, especially in those undergoing pelvic LND. The overall complication rate was 14.2% (3.9% severe) in the LND group compared with 10.1% (1.1% severe) in the no-LND group. An earlier study by Kondo and Tanabe (9) also documented prolonged operation duration (323 vs. 288 min) and greater intraoperative blood loss (407 vs. 321 ml) in patients who underwent LND. It was therefore hypothesized that although LND could be associated with elevated risks of lymphatic and hemorrhagic complications, these could rarely affect clinical recovery.

Innovative approaches

Limitations of conventional imaging methods

Computed tomography (CT) imaging remains the first-line diagnostic tool for pretherapeutic evaluation of UTUC, with reported sensitivity and specificity rates of 92 and 95%, respectively (73). Research by Millán-Rodríguez et al (74), which included 93 upper tract urothelial tumors from 82 consecutive UTUC patients scheduled for nephroureterectomy, reported promising results for CT, with a sensitivity and specificity of 87.5 and 98%, respectively. Cross-sectional imaging, particularly CT, remains the cornerstone of clinical nodal staging of UTUC. However, a previous multicenter observational trial by Pallauf et al (75), encompassing 865 patients with UTUC who underwent RNU with LND, revealed that conventional imaging exhibited high specificity (91%) but low sensitivity (only 25%), thus resulting in limited detection reliability [area under the curve (AUC), 0.58]. Therefore, the it was proposed that CT imaging should be primarily used to confirm, rather than to exclude, LNM.

Application and limitations of 18F-fluorodeoxyglucose positron emission tomography (18F-FDG-PET)/CT in lymph node staging

Conventional imaging techniques, such as CT and PET, have limited precision in identifying the locations of LNM in UTUC (2,75–77). To enhance nodal staging accuracy, researchers are currently investigating biomarker-based approaches and multimodal imaging combinations. Among them, 18F-FDG/PET/CT imaging has shown emerging utility in identifying metastatic nodal involvement in both UTUC and bladder cancer (78–84). Although early studies indicated comparable detection accuracy between 18F-FDG/PET/CT and conventional magnetic resonance imaging (MRI) (82,84–86), accumulating evidence has supported the superiority of this hybrid modality over MRI, CT or PET alone. The strength of 18F-FDG/PET/CT stems from its capacity to detect metabolically active micrometastases (<2.0 mm), which can be indetectable by CT alone, thus enhancing the sensitivity of nodal staging (82,86). However, its limited specificity creates a challenge, particularly in distinguishing cancerous from inflammatory nodes (81). Several retrospective studies have assessed the utility of 18F-FDG/PET/CT for detecting preoperative LNM in UTUC and bladder cancer (78,79). A study published in 2020 reported a sensitivity rate of 82% and a specificity rate of 84% for preoperative nodal staging in UTUC using 18F-FDG/PET/CT (78).

In addition, a previous systematic review, which included three retrospective studies on the detection of LNM in UTUC, reported sensitivities of 82–95% and specificities of 84–91%, thus highlighting the prognostic value of 18F-FDG/PET/CT (81). A comparative analysis in bladder cancer demonstrated a higher sensitivity rate for 18F-FDG/PET/CT (78%) compared with that for CT (44%) for nodal assessment (87). A key limitation of 18F-FDG/PET/CT involves the interference from urinary excretion of the radiotracer, which can reduce imaging clarity for nodal lesions adjacent to urinary structures (88). Choline PET/CT, which uses (11)C-choline, has also been explored for LNM detection in UTUC. A 2014 study showed high choline uptake in affected lymph nodes in patients with UTUC (89). Additionally, Polom et al (90) attempted to detect sentinel lymph nodes using Technetium-99m injection during ureterorenoscopy and single-photon emission-CT/CT lymphangiography. While theoretically feasible, the method proved challenging due to significant technical constrains during Technetium injection.

Exploration of other imaging methods

Ultrasmall superparamagnetic iron oxide (USPIO) has gained increasing attention as a contrast agent in MRI. Metastatic lymph nodes, lacking macrophages, fail to absorb USPIO, thus resulting in minimal or absent T2-weighted imaging (WI) signal loss. By contrast, benign lymph nodes, which retain the normal activity of macrophages, absorb USPIO, eventually showing low T2WI signals, which aids in distinguishing metastatic nodes from non-metastatic nodes. Emerging evidence has suggested that USPIO-MRI can promote the identification of small nodal metastases, with reported sensitivity ranges of 65–92% and specificity levels of 93–98% (91,92). In an innovative approach, Birkhäuser et al (93) integrated diffusion-weighted imaging sequences with USPIO-MRI, achieving identification rates of 87 and 77% for lymph nodes of ≤8 and ≤3 mm in the short-axis diameter, respectively. The reading time was reduced from 32 to 9 min, with scanning performed 24–36 h post-administration (94). A previous meta-analysis also reported an overall sensitivity of 0.86 for USPIO-MRI in diagnosing pelvic LNM. However, its clinical application remains limited due to safety concerns and high costs (95).

Research into labeled monoclonal antibodies for urothelial neoplasms is ongoing (77). Among them, girentuximab-labeled PET/CT (89Zr-TLX250) has shown promising efficacy in staging renal cell carcinoma and breast cancer. TLX250 targets carbonic anhydrase IX, an enzyme which is significantly overexpressed in urothelial cancer cells (96). A phase I clinical trial is currently underway to assess the potential of 89Zr-TLX250 for imaging urothelial malignancies (87).

Imaging studies are increasingly integrated into preoperative protocols for predicting lymph node metastases in UTUC. A novel diagnostic model incorporating radiological parameters and histopathological characteristics, including pathological staging, lymphovascular invasion (LVI), lesion dimensions and pretreatment nodal status, demonstrated a predictive accuracy of 87.8% for detecting nodal metastasis (AUC=0.878; adjusted concordance index=0.887) (97).

Advances and barriers in molecular biomarkers for UTUC

Novel molecular classification frameworks for UTUC increasingly integrate genomic signatures and transcriptome analysis. Fibulin-2 (FBLN), a significant prognostic indicator, is strongly associated with diminished CSS rates and increased risk of metastasis in urothelial malignancies (P<0.001). It has been reported that FBLN is upregulated in advanced-stage tumors, also being associated with LNM and enhanced cell proliferation activity (93,98–106). The pyruvate dehydrogenase kinase (PDK) family, which is involved in mediating chemotherapeutic resistance and disease advancement in patients with bladder carcinomas (105–107), exhibits parallel pathobiological significance in UTUC. A previous study by Kuo et al (108) indicated that PDK3 upregulation was significantly associated with aggressive clinicopathological features, including metastatic nodal disease, elevated tumor grade and unfavorable survival prognosis (P<0.0001).

Furthermore, other studies demonstrated that cartilage oligomeric matrix protein, another prognostic indicator, was associated with advanced T-stage, LNM, LVI, perineural infiltration, high-grade histology and increased mitotic rates (109–113). Metallothionein 2A was also associated with aggressive behaviors and advanced staging parameters in urothelial malignancies (114).

Laboratory-based biomarkers are also gaining increasing attention. Therefore, the de Ritis ratio (alanine aminotransferase/aspartate aminotransferase) showed prognostic potential in a previous study, since analysis of 135 cases of patients with a de Ritis index of ≥1.3 was strongly predictive of LNM (P=0.0096) (115,116). It has been reported that the systemic inflammatory status can affect metastasis, thus being considered as an additional prognostic tool (115,116). The systemic inflammation index (SII), defined as the number of neutrophils × platelet/lymphocyte count, serves as a low-cost prognostic tool (117–122). A study demonstrated that elevated SII values were associated with LVI positivity and poorer OS rates in UTUC (122). Kobayashi et al (123) incorporated SII >520, Eastern Cooperative Oncology Group Performance Status >0 and clinical stage ≥T3 into a preoperative risk model to guide therapeutic decision-making.

The 2023 EAU guidelines recognize several emerging molecular biomarkers in UTUC. However, their integration into surgical decision-making remains limited. For example, DNA mismatch repair (MMR) deficiency, and particularly germline MSH2/MSH6 mutations associated with Lynch syndrome (LS), were recorded in 9% of patients with UTUC, which was a markedly higher rate compared with that observed in bladder cancer (1%). Although LS screening is recommended for patients <60 years or with suggestive family history, its effect typically comes into practice for surveillance of metachronous cancers rather than in tailoring surgical approaches (124–130). PD-L1 expression (≥1%) serves as a predictive biomarker for ICIs in metastatic UTUC, with pembrolizumab or atezolizumab recommended for cisplatin-ineligible PD-L1-positive patients. Adjuvant nivolumab can be considered for patients with HR (≥pT3/pN+) cisplatin-ineligible UTUC post RNU. However, the aforementioned UTUC-specific survival benefits have not been fully investigated (51,131–133). Alterations in FGFR2/3 guide later-line targeted therapy for platinum-refractory disease (erdafitinib). However, they do affect primary surgical approaches (134). While molecular subtyping (luminal and basal subtypes) holds prognostic potential, its clinical application in surgical planning is limited due to lack of validated treatment response associations (135).

Significant gaps remain in the integration of biomarkers into personalized surgery strategies. Kidney-sparing surgery (KSS) eligibility relies solely on clinical factors, such as tumor size, grade and focality, with no molecular predictors currently used to assess ipsilateral recurrence risk or progression to refine patient selection (136,137). LND templates are determined by tumor location and risk, without biomarkers predictive of occult micrometastasis. Furthermore, neoadjuvant ChT recommendations lack biomarkers, such as ERCC2 mutations for platinum sensitivity, to identify non-responders, which account for ~40% of all cases, risking delayed surgery without therapeutic benefits (138). For patients with LS-associated UTUC, molecular risk stratification, including particular MMR mutations (e.g., MLH1, MSH2, MSH6 and PMS2), does not guide prophylactic surgical interventions. Future efforts should be made to prospectively validate biomarkers in surgical trials, to develop integrated clinical-molecular prognostic models and to investigate biomarker-driven neoadjuvant therapies, such as FGFR inhibitors, to expand KSS eligibility. In summary, while biomarkers increasingly guide systemic therapy and genetic screening, their application in optimizing surgical management, including decisions on KSS candidacy, LND extent and utilization of neoadjuvant ChT, remains an unmet need in the evolution of UTUC precision medicine (55,136–138).

Conclusion and future perspectives

UTUC remains as a challenging oncological entity profoundly affecting a patient's survival and quality of life. The clinical utility of LND during RNU continues to be a subject of debate among urological specialists. While existing evidence supports the diagnostic value and possible therapeutic effects of LND, and particularly muscle-invasive and locally extensive disease, the optimal extent of dissection and its definitive effect on patient survival need further clarification. Although major guidelines vary between LND protocols, accumulating evidence increasingly supports the adoption of standardized anatomical templates to enhance diagnostic accuracy. Therefore, further high-quality studies are necessitated to establish consensus-driven practices. Advancements in functional imaging modalities, such as PET-CT/MRI, and molecular biomarker development, can promote tailored surgical planning. Ultimately, clarifying the therapeutic role of LND in the management of UTUC requires prospective multicenter trials to strengthen evidence-based protocols for optimizing outcomes.

Acknowledgements

Not applicable.

Funding

This study was supported by the National Natural Science Foundation of China (grant no. 82360603), the Yunnan Fundamental Research Projects (grant nos. 202001AY070001-163, 202201AU070220, 202201AY070001-113 and 202401AU070010) and the Yunnan Provincial Department of Education Project (grant no. 2024J0225).

Availability of data and materials

The data generated in the present study may be requested from the corresponding author.

Authors' contributions

HW, MD and ZT were responsible for conceptualization. ZT, CY, JW, YH and SF designed the literature search strategy, screening criteria and evidence synthesis framework.. Formal analysis and data interpretation were performed by YH, SF, HL, DL, CG and JW. ZT, CY and JW helped write the original draft. HW, MD, ZT, CY, JW, YH, SF, HL, DL and CG reviewed and edited the manuscript. Visualization of figures/tables was performed by HL, DL, CG and JW. HW and MD supervised the study. HW and MD acquired funding. All authors have read and approved the manuscript. All authors have made substantial intellectual contributions, critically revised the manuscript for important content, and agree to be accountable for all aspects of the work. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

UTUC

upper tract urothelial carcinoma

LND

lymph node dissection

RNU

radical nephroureterectomy

CSS

cancer-specific survival

DFS

disease-free survival

LNM

lymph node metastasis

EAU

European Association of Urology

NCCN

National Comprehensive Cancer Network

AUA

American Urological Association

CT

computed tomography

PET

positron emission tomography

MRI

magnetic resonance imaging

USPIO

ultrasmall superparamagnetic iron oxide

PDK

pyruvate dehydrogenase kinase

LVI

lymphovascular invasion

AUC

area under the curve

OR

odds ratio

References

1 

Rouprêt M, Babjuk M, Burger M, Capoun O, Cohen D, Compérat EM, Cowan NC, Dominguez-Escrig JL, Gontero P, Hugh Mostafid A, et al: European association of urology guidelines on upper urinary tract urothelial carcinoma: 2020 update. Eur Urol. 79:62–79. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Green DA, Rink M, Xylinas E, Matin SF, Stenzl A, Roupret M, Karakiewicz PI, Scherr DS and Shariat SF: Urothelial carcinoma of the bladder and the upper tract: Disparate twins. J Urol. 189:1214–1221. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Peyrottes A, Ouzaid I, Califano G, Hermieu JF and Xylinas E: Neoadjuvant immunotherapy for muscle-invasive bladder cancer. Medicina (Kaunas). 57:7692021. View Article : Google Scholar : PubMed/NCBI

4 

Giudici N, Bonne F, Blarer J, Minoli M, Krentel F and Seiler R: Characteristics of upper urinary tract urothelial carcinoma in the context of bladder cancer: A narrative review. Transl Androl Urol. 10:4036–4050. 2021. View Article : Google Scholar : PubMed/NCBI

5 

Margulis V, Shariat SF, Matin SF, Kamat AM, Zigeuner R, Kikuchi E, Lotan Y, Weizer A, Raman JD and Wood CG; Upper Tract Urothelial Carcinoma Collaboration The Upper Tract Urothelial Carcinoma Collaboration, : Outcomes of radical nephroureterectomy: A series from the upper tract urothelial carcinoma collaboration. Cancer. 115:1224–1233. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Batata MA, Whitmore WF, Hilaris BS, Tokita N and Grabstald H: Primary carcinoma of the ureter: A prognostic study. Cancer. 35:1626–1632. 1975. View Article : Google Scholar : PubMed/NCBI

7 

Collà Ruvolo C, Nocera L, Stolzenbach LF, Wenzel M, Cucchiara V, Tian Z, Shariat SF, Saad F, Longo N, Montorsi F, et al: Incidence and survival rates of contemporary patients with invasive upper tract urothelial carcinoma. Eur Urol Oncol. 4:792–801. 2021. View Article : Google Scholar : PubMed/NCBI

8 

Lughezzani G, Jeldres C, Isbarn H, Sun M, Shariat SF, 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. 45:3291–3297. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Kondo T and Tanabe K: Role of lymphadenectomy in the management of urothelial carcinoma of the bladder and the upper urinary tract. Int J Urol. 19:710–721. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Chappidi MR, Kates M, Johnson MH, Hahn NM, Bivalacqua TJ and Pierorazio PM: Lymph node yield and tumor location in patients with upper tract urothelial carcinoma undergoing nephroureterectomy affects survival: A U.S. population-based analysis (2004–2012). Urol Oncol. 34:531.e15–531.e24. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Lughezzani G, Jeldres C, Isbarn H, Shariat SF, 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. 75:118–124. 2010. View Article : Google Scholar : PubMed/NCBI

12 

Roscigno M, Brausi M, Heidenreich A, Lotan Y, Margulis V, Shariat SF, Van Poppel H and Zigeuner R: Lymphadenectomy at the time of nephroureterectomy for upper tract urothelial cancer. Eur Urol. 60:776–783. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Deuker M, Rosiello G, Stolzenbach LF, Martin T, Collà Ruvolo C, Nocera L, Tian Z, Roos FC, Becker A, Kluth LA, et al: Sex- and age-related differences in the distribution of metastases in patients with upper urinary tract urothelial carcinoma. J Natl Compr Canc Netw. 19:534–540. 2021. View Article : Google Scholar : PubMed/NCBI

14 

Inokuchi J, Kuroiwa K, Kakehi Y, Sugimoto M, Tanigawa T, Fujimoto H, Gotoh M, Masumori N, Ogawa O, Eto M, et al: Role of lymph node dissection during radical nephroureterectomy for upper urinary tract urothelial cancer: Multi-institutional large retrospective study JCOG1110A. World J Urol. 35:1737–1744. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Rouprêt M, Seisen T, Birtle AJ, Capoun O, Compérat EM, Dominguez-Escrig JL, 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. 84:49–64. 2023. View Article : Google Scholar : PubMed/NCBI

16 

Coleman JA, Clark PE, Bixler BR, Buckley DI, Chang SS, Chou R, Hoffman-Censits J, Kulkarni GS, Matin SF, Pierorazio PM, et al: Diagnosis and management of non-metastatic upper tract urothelial carcinoma: AUA/SUO guideline. J Urol. 209:1071–1081. 2023. View Article : Google Scholar : PubMed/NCBI

17 

Akaza H, Koiso K and Niijima T: Clinical evaluation of urothelial tumors of the renal pelvis and ureter based on a new classification system. Cancer. 59:1369–1375. 1987. View Article : Google Scholar : PubMed/NCBI

18 

Matin SF, Sfakianos JP, Espiritu PN, Coleman JA and Spiess PE: Patterns of lymphatic metastases in upper tract urothelial carcinoma and proposed dissection templates. J Urol. 194:1567–1574. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Kondo T, Hara I, Takagi T, Kodama Y, Hashimoto Y, Kobayashi H, Iizuka J, Omae K, Yoshida K and Tanabe K: Template-based lymphadenectomy in urothelial carcinoma of the renal pelvis: A prospective study. Int J Urol. 21:453–459. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Mason RJ, Kassouf W, Bell DG, Lacombe L, Kapoor A, Jacobsen N, Fairey A, Izawa J, Black P, Tanguay S, et al: The contemporary role of lymph node dissection during nephroureterectomy in the management of upper urinary tract urothelial carcinoma: the Canadian experience. Urology. 79:840–845. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Zhai TS, Jin L, Zhou Z, Liu X, Liu H, Chen W, Lu JY, Yao XD, Feng LM and Ye L: Effect of lymph node dissection on stage-specific survival in patients with upper urinary tract urothelial carcinoma treated with nephroureterectomy. BMC cancer. 19:12072019. View Article : Google Scholar : PubMed/NCBI

22 

Birtle A, Johnson M, Chester J, Jones R, Dolling D, Bryan RT, Harris C, Winterbottom A, Blacker A, Catto JWF, et al: Adjuvant chemotherapy in upper tract urothelial carcinoma (the POUT trial): A phase 3, open-label, randomised controlled trial. Lancet. 395:1268–1277. 2020. View Article : Google Scholar : PubMed/NCBI

23 

Xu H, Ai JZ, Tan P, Lin TH, Jin X, Gong LN, Lei HR, Yang L and Wei Q: Pretreatment elevated fibrinogen level predicts worse oncologic outcomes in upper tract urothelial carcinoma. Asian J Androl. 22:177–183. 2020. View Article : Google Scholar : PubMed/NCBI

24 

Tan P, Shi M, Chen J, Xu H, Xie N, Xu H, Jiang Y, Ai JZ, Liu LR, Yang L and Wei Q: The preoperative serum cystatin-C as an independent prognostic factor for survival in upper tract urothelial carcinoma. Asian J Androl. 21:163–169. 2019. View Article : Google Scholar : PubMed/NCBI

25 

Saito K, Kawakami S, Ohtsuka Y, Fujii Y, Masuda H, Kumagai J, Kobayashi T, Kageyama Y and Kihara K: The impact of preoperative serum C-reactive protein on the prognosis of patients with upper urinary tract urothelial carcinoma treated surgically. BJU Int. 100:269–273. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Xu H, Tan P, Ai J, Huang Y, Lin T, Yang L and Wei Q: Prognostic impact of preoperative albumin-globulin ratio on oncologic outcomes in upper tract urothelial carcinoma treated with radical nephroureterectomy. Clin Genitourin Cancer. 16:e1059–e1068. 2018. View Article : Google Scholar : PubMed/NCBI

27 

Rink M, Sharifi N, Fritsche HM, Aziz A, Miller F, Kluth LA, Ngamsri T, Dahlem R, Chun FK, Shariat SF, et al: Impact of preoperative anemia on oncologic outcomes of upper tract urothelial carcinoma treated with radical nephroureterectomy. J Urol. 191:316–322. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Vartolomei MD, Mathieu R, Margulis V, Karam JA, Rouprêt M, Lucca I, Mbeutcha A, Seitz C, Karakiewicz PI, Fajkovic H, et al: Promising role of preoperative neutrophil-to-lymphocyte ratio in patients treated with radical nephroureterectomy. World J Urol. 35:121–130. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Kondo T, Hashimoto Y, Kobayashi H, Iizuka J, Nakazawa H, Ito F and Tanabe K: Template-based lymphadenectomy in urothelial carcinoma of the upper urinary tract: Impact on patient survival. Int J Urol. 17:848–854. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Kondo T, Nakazawa H, Ito F, Hashimoto Y, Toma H and Tanabe K: Impact of the extent of regional lymphadenectomy on the survival of patients with urothelial carcinoma of the upper urinary tract. J Urol. 178:1212–1217. 2007. View Article : Google Scholar : PubMed/NCBI

31 

Kondo T, Hara I, Takagi T, Kodama Y, Hashimoto Y, Kobayashi H, Iizuka J, Omae K, Ikezawa E, Yoshida K and Tanabe K: Possible role of template-based lymphadenectomy in reducing the risk of regional node recurrence after nephroureterectomy in patients with renal pelvic cancer. Jpn J Clin Oncol. 44:1233–1238. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Burger M, Shariat SF, Fritsche HM, Martinez-Salamanca JI, Matsumoto K, Chromecki TF, Ficarra V, Kassouf W, Seitz C, Pycha A, et al: No overt influence of lymphadenectomy on cancer-specific survival in organ-confined versus locally advanced upper urinary tract urothelial carcinoma undergoing radical nephroureterectomy: A retrospective international, multi-institutional study. World J Urol. 29:465–472. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Duquesne I, Ouzaid I, Loriot Y, Moschini M and Xylinas E: Lymphadenectomy for upper tract urothelial carcinoma: A systematic review. J Clin Med. 8:11902019. View Article : Google Scholar : PubMed/NCBI

34 

Stein JP, Cai J, Groshen S and Skinner DG: Risk factors for patients with pelvic lymph node metastases following radical cystectomy with en bloc pelvic lymphadenectomy: Concept of lymph node density. J Urol. 170:35–41. 2003. View Article : Google Scholar : PubMed/NCBI

35 

Bolenz C, Shariat SF, Fernández MI, Margulis V, Lotan Y, Karakiewicz P, Remzi M, Kikuchi E, Zigeuner R, Weizer A, et al: Risk stratification of patients with nodal involvement in upper tract urothelial carcinoma: Value of lymph-node density. BJU Int. 103:302–306. 2009. View Article : Google Scholar : PubMed/NCBI

36 

Rao SR, Correa JJ, Sexton WJ, Pow-Sang JM, Dickinson SI, Lin HY and Spiess PE: Prospective clinical trial of the feasibility and safety of modified retroperitoneal lymph node dissection at time of nephroureterectomy for upper tract urothelial carcinoma. BJU Int. 110:E475–E480. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Abe T, Takada N, Matsumoto R, Osawa T, Sazawa A, Maruyama S, Tsuchiya K, Harabayashi T, Minami K, Nagamori S, et al: Outcome of regional lymphadenectomy in accordance with primary tumor location on laparoscopic nephroureterectomy for urothelial carcinoma of the upper urinary tract: A prospective study. J Endourol. 29:304–309. 2015. View Article : Google Scholar : PubMed/NCBI

38 

Lu H, Liang Y, Guan B, Shi Y, Gong Y, Li J, Kong W, Liu J, Fang D, Liu L, et al: Aristolochic acid mutational signature defines the low-risk subtype in upper tract urothelial carcinoma. Theranostics. 10:4323–4333. 2020. View Article : Google Scholar : PubMed/NCBI

39 

Novara G, De Marco V, Gottardo F, Dalpiaz O, Bouygues V, Galfano A, Martignoni G, Patard JJ, Artibani W and Ficarra V: Independent predictors of cancer-specific survival in transitional cell carcinoma of the upper urinary tract: multi-institutional dataset from 3 European centers. Cancer. 110:1715–1722. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Secin FP, Koppie TM, Salamanca JI, Bokhari S, Raj GV, Olgac S, Martignoni G, Patard JJ, Artibani W and Ficarra V: Evaluation of regional lymph node dissection in patients with upper urinary tract urothelial cancer. Int J Urol. 14:26–32. 2007. View Article : Google Scholar : PubMed/NCBI

41 

Roscigno M, Cozzarini C, Bertini R, Scattoni V, Freschi M, Da Pozzo LF, Briganti A, Gallina A, Capitanio U, Colombo R, et al: Prognostic value of lymph node dissection in patients with muscle-invasive transitional cell carcinoma of the upper urinary tract. Eur Urol. 53:794–802. 2008. View Article : Google Scholar : PubMed/NCBI

42 

Cho KS, Choi HM, Koo K, Park SJ, Rha KH, Choi YD, Chung BH, Cho NH, Yang SC and Hong SJ: Clinical significance of lymph node dissection in patients with muscle-invasive upper urinary tract transitional cell carcinoma treated with nephroureterectomy. J Korean Med Sci. 24:674–678. 2009. View Article : Google Scholar : PubMed/NCBI

43 

Ouzzane A, Colin P, Ghoneim TP, Zerbib M, De La Taille A, Audenet F, Saint F, Hoarau N, Adam E, Azemar MD, et al: The impact of lymph node status and features on oncological outcomes in urothelial carcinoma of the upper urinary tract (UTUC) treated by nephroureterectomy. World J Urol. 31:189–197. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Abe T, Shinohara N, Muranaka M, Sazawa A, Maruyama S, Osawa T, Harabayashi T, Kubota K, Matsuno Y, Shibata T, et al: Role of lymph node dissection in the treatment of urothelial carcinoma of the upper urinary tract: multi-institutional relapse analysis and immunohistochemical re-evaluation of negative lymph nodes. Eur J Surg Oncol. 36:1085–1091. 2010. View Article : Google Scholar : PubMed/NCBI

45 

Roscigno M, Shariat SF, Margulis V, Karakiewicz P, Remzi M, Kikuchi E, Langner C, Lotan Y, Weizer A, Bensalah K, et al: Impact of lymph node dissection on cancer specific survival in patients with upper tract urothelial carcinoma treated with radical nephroureterectomy. J Urol. 181:2482–2489. 2009. View Article : Google Scholar : PubMed/NCBI

46 

Galsky MD, Witjes JA, Gschwend JE, Milowsky MI, Schenker M, Valderrama BP, Tomita Y, Bamias A, Lebret T, Shariat SF, et al: Adjuvant nivolumab in high-risk muscle-invasive urothelial carcinoma: Expanded efficacy from checkmate 274. J Clin Oncol. 43:15–21. 2025. View Article : Google Scholar : PubMed/NCBI

47 

Abufaraj M, Li R, Meeks J and Shariat SF: Cytoreductive surgery in patients with urothelial bladder cancer. Eur Urol Focus. 9:278–279. 2023. View Article : Google Scholar : PubMed/NCBI

48 

Shigeta K, Matsumoto K, Ogihara K, Murakami T, Anno T, Umeda K, Izawa M, Baba Y, Sanjo T, Shojo K, et al: Does neoadjuvant chemotherapy have therapeutic benefit for node-positive upper tract urothelial carcinoma? Results of a multi-center cohort study. Urol Oncol. 40:105.e19–105.e26. 2022. View Article : Google Scholar : PubMed/NCBI

49 

Jung H, Giusti G, Fajkovic H, Herrmann T, Jones R, Straub M, Baard J, Osther PJS and Brehmer M: Consultation on UTUC, Stockholm 2018: Aspects of treatment. World J Urol. 37:2279–2287. 2019. View Article : Google Scholar : PubMed/NCBI

50 

Galsky MD, Chen GJ, Oh WK, Bellmunt J, Roth BJ, Petrioli R, Dogliotti L, Dreicer R and Sonpavde G: Comparative effectiveness of cisplatin-based and carboplatin-based chemotherapy for treatment of advanced urothelial carcinoma. Ann Oncol. 23:406–410. 2012. View Article : Google Scholar : PubMed/NCBI

51 

Bajorin DF, Witjes JA, Gschwend JE, Schenker M, Valderrama BP, Tomita Y, Bamias A, Lebret T, Shariat SF, Park SH, et al: Adjuvant nivolumab versus placebo in muscle-invasive urothelial carcinoma. N Engl J Med. 384:2102–2114. 2021. View Article : Google Scholar : PubMed/NCBI

52 

Li Z, Li X, Liu Y, Fang J, Zhang X and Xiao K: Can American Joint Committee on Cancer prognostic groups be individualized in patients undergoing surgery for Stage IV invasive upper tract Urothelial Carcinoma? J Cancer. 12:2023–2029. 2021. View Article : Google Scholar : PubMed/NCBI

53 

Abdel-Rahman O: Revisiting the prognostic heterogeneity of AJCC Stage IV carcinomas of the upper urinary tract. Clin Genitourin Cancer. 16:e859–e865. 2018. View Article : Google Scholar : PubMed/NCBI

54 

Flaig TW, Spiess PE, Chair V, Abern M, Agarwal N and Buyyounouski MK: NCCN Guidelines Version 3.2023 Bladder Cancer. National Comprehensive Cancer Network. Available from:. https://www.nccn.org/guidelines/guidelines-detail?category=1&id=1417

55 

Dominguez-Escrig JL, Peyronnet B, Seisen T, Bruins HM, Yuan CY, Babjuk M, Böhle A, Burger M, Compérat EM, Gontero P, et al: Potential benefit of lymph node dissection during radical nephroureterectomy for upper tract urothelial carcinoma: A systematic review by the European association of urology guidelines panel on non-muscle-invasive bladder cancer. Eur Urol Focus. 5:224–241. 2019. View Article : Google Scholar : PubMed/NCBI

56 

Brausi MA, Gavioli M, De Luca G, Verrini G, Peracchia G, Simonini G and Viola M: Retroperitoneal lymph node dissection (RPLD) in conjunction with nephroureterectomy in the treatment of infiltrative transitional cell carcinoma (TCC) of the upper urinary tract: Impact on survival. Eur Urol. 52:1414–1418. 2007. View Article : Google Scholar : PubMed/NCBI

57 

Matsumoto R, Abe T, Takada N, Minami K, Harabayashi T, Nagamori S, Hatanaka KC, Yamashiro K, Kikuchi H, Osawa T, et al: Oncologic outcomes of laparoscopic radical nephroureterectomy in conjunction with template-based lymph node dissection: An extended follow-up study. Urol Oncol. 38:933.e13–933.e18. 2020. View Article : Google Scholar : PubMed/NCBI

58 

Furuse H, Matsushita Y, Yajima T, Kato T, Suzuki T, Matsumoto R, Motoyama D, Sugiyama T, Otsuka A and Ozono S: Systematic regional lymph node dissection for upper tract urothelial carcinoma improves patient survival. Jpn J Clin Oncol. 47:239–246. 2017. View Article : Google Scholar : PubMed/NCBI

59 

Ikeda M, Matsumoto K, Sakaguchi K, Ishii D, Tabata KI, Kurosawa K, Urakami S, Okaneya T and Iwamura M: Effect of lymphadenectomy during radical nephroureterectomy in locally advanced upper tract urothelial carcinoma. Clin Genitourin Cancer. 15:556–562. 2017. View Article : Google Scholar : PubMed/NCBI

60 

Yoo S, You D, Jeong IG, Hong B, Hong JH, Ahn H and Kim CS: Does lymph node dissection during nephroureterectomy affect oncological outcomes in upper tract urothelial carcinoma patients without suspicious lymph node metastasis on preoperative imaging studies? World J Urol. 35:665–673. 2017. View Article : Google Scholar : PubMed/NCBI

61 

Inokuchi J, Eto M, Hara T, Fujimoto H, Nishiyama H, Miyazaki J, Kikuchi E, Hinotsu S, Koie T and Ohyama C; Cancer Registration Committee of the Japanese Urological Association, : Impact of lymph node dissection on clinical outcomes during nephroureterectomy in patients with clinically node-negative upper urinary tract urothelial cancer: Subanalysis of a multi-institutional nationwide case series of the Japanese Urological Association. Jpn J Clin Oncol. 47:652–659. 2017. View Article : Google Scholar : PubMed/NCBI

62 

Pearce SM, Pariser JJ, Patel SG, Steinberg GD, Shalhav AL and Smith ND: The effect of surgical approach on performance of lymphadenectomy and perioperative morbidity for radical nephroureterectomy. Urol Oncol. 34:121.e15–21. 2016. View Article : Google Scholar : PubMed/NCBI

63 

Winer AG, Vertosick EA, Ghanaat M, Corradi RB, Carlsson S, Sjoberg DD, Sankin AI, Sfakianos JP, Cha EK, Dalbagni G and Coleman JA: Prognostic value of lymph node yield during nephroureterectomy for upper tract urothelial carcinoma. Urol Oncol. 35:151.e9–151.e15. 2017. View Article : Google Scholar : PubMed/NCBI

64 

Dong F, Xu T, Wang X, Shen Y, Zhang X, Chen S, Zhong S, Zhang M and Ding Q: Lymph node dissection could bring survival benefits to patients diagnosed with clinically node-negative upper urinary tract urothelial cancer: A population-based, propensity score-matched study. Int J Clin Oncol. 24:296–305. 2019. View Article : Google Scholar : PubMed/NCBI

65 

Brown GA, Busby JE, Wood CG, Pisters LL, Dinney CP, Swanson DA, Grossman HB, Pettaway CA, Munsell MF, Kamat AM and Matin SF: Nephroureterectomy for treating upper urinary tract transitional cell carcinoma: Time to change the treatment paradigm? BJU Int. 98:1176–1180. 2006. View Article : Google Scholar : PubMed/NCBI

66 

Li CC, Chang CH, Huang CP, Hong JH, Huang CY, Chen IA, Lin JT, Lo CW, Yu CC, Tseng JS, et al: Comparing oncological outcomes and surgical complications of hand-assisted, laparoscopic and robotic nephroureterectomy for upper tract urothelial carcinoma. Front Oncol. 11:7314602021. View Article : Google Scholar : PubMed/NCBI

67 

Clements MB, Krupski TL and Culp SH: Robotic-assisted surgery for upper tract urothelial carcinoma: A comparative survival analysis. Ann Surg Oncol. 25:2550–2562. 2018. View Article : Google Scholar : PubMed/NCBI

68 

Peyronnet B, Seisen T, Dominguez-Escrig JL, Bruins HM, Yuan CY, Lam T, Maclennan S, N'dow J, Babjuk M, Comperat E, et al: Oncological outcomes of laparoscopic nephroureterectomy versus open radical nephroureterectomy for upper tract urothelial carcinoma: An European association of urology guidelines systematic review. Eur Urol Focus. 5:205–223. 2019. View Article : Google Scholar : PubMed/NCBI

69 

Ishiyama Y, Kondo T, Kubota S, Shimada K, Yoshida K, Takagi T, Iizuka J and Tanabe K: Therapeutic benefit of lymphadenectomy for older patients with urothelial carcinoma of the upper urinary tract: A propensity score matching study. Jpn J Clin Oncol. 51:802–809. 2021. View Article : Google Scholar : PubMed/NCBI

70 

Dindo D, Demartines N and Clavien PA: Classification of surgical complications: A new proposal with evaluation in a cohort of 6336 patients and results of a survey. Ann Surg. 240:205–213. 2004. View Article : Google Scholar : PubMed/NCBI

71 

Blom JH, van Poppel H, Maréchal JM, Jacqmin D, Schröder FH, de Prijck L and Sylvester R; EORTC Genitourinary Tract Cancer Group, : Radical nephrectomy with and without lymph-node dissection: final results of European Organization for Research and Treatment of Cancer (EORTC) randomized phase 3 trial 30881. Eur Urol. 55:28–34. 2009. View Article : Google Scholar : PubMed/NCBI

72 

Kondo T, Takagi T and Tanabe K: Therapeutic role of template-based lymphadenectomy in urothelial carcinoma of the upper urinary tract. World J Clin Oncol. 6:237–251. 2015. View Article : Google Scholar : PubMed/NCBI

73 

Janisch F, Shariat SF, Baltzer P, Fajkovic H, Kimura S, Iwata T, Korn P, Yang L, Glybochko PV, Rink M and Abufaraj M: Diagnostic performance of multidetector computed tomographic (MDCTU) in upper tract urothelial carcinoma (UTUC): A systematic review and meta-analysis. World J Urol. 38:1165–1175. 2020. View Article : Google Scholar : PubMed/NCBI

74 

Millán-Rodríguez F, Palou J, de la Torre-Holguera P, Vayreda-Martija JM, Villavicencio-Mavrich H and Vicente Rodríguez J: Conventional CT signs in staging transitional cell tumors of the upper urinary tract. Eur Urol. 35:318–322. 1999. View Article : Google Scholar : PubMed/NCBI

75 

Pallauf M, D'Andrea D, König F, Laukhtina E, Yanagisawa T, Rouprêt M, Daneshmand S, Djaladat H, Ghoreifi A, Soria F, et al: Diagnostic accuracy of clinical lymph node staging for upper tract urothelial cancer patients: A multicenter, retrospective, observational study. J Urol. 209:515–524. 2023. View Article : Google Scholar : PubMed/NCBI

76 

Rink M, Ehdaie B, Cha EK, Green DA, Karakiewicz PI, Babjuk M, Margulis V, Raman JD, Svatek RS, Fajkovic H, et al: Stage-specific impact of tumor location on oncologic outcomes in patients with upper and lower tract urothelial carcinoma following radical surgery. Eur Urol. 62:677–684. 2012. View Article : Google Scholar : PubMed/NCBI

77 

Al-Zubaidi M, Viswambaram P, McCombie S, Liow E, Lenzo N, Ferguson T, Redfern AD, Gauci R and Hayne D: 89Zirconium-labelled girentuximab (89Zr-TLX250) PET in Urothelial Cancer Patients (ZiPUP): Protocol for a phase I trial of a novel staging modality for urothelial carcinoma. BMJ Open. 12:e0604782022. View Article : Google Scholar : PubMed/NCBI

78 

Voskuilen CS, Schweitzer D, Jensen JB, Nielsen AM, Joniau S, Muilwijk T, Necchi A, Azizi M, Spiess PE, Briganti A, et al: Diagnostic Value of 18F-fluorodeoxyglucose positron emission tomography with computed tomography for lymph node staging in patients with upper tract urothelial carcinoma. Eur Urol Oncol. 3:73–79. 2020. View Article : Google Scholar : PubMed/NCBI

79 

Zattoni F, Incerti E, Colicchia M, Castellucci P, Panareo S, Picchio M, Fallanca F, Briganti A, Moschini M, Gallina A, et al: Comparison between the diagnostic accuracies of 18F-fluorodeoxyglucose positron emission tomography/computed tomography and conventional imaging in recurrent urothelial carcinomas: A retrospective, multicenter study. Abdom Radiol (NY). 43:2391–2399. 2018. View Article : Google Scholar : PubMed/NCBI

80 

Soubra A, Hayward D, Dahm P, Goldfarb R, Froehlich J, Jha G and Konety BR: The diagnostic accuracy of 18F-fluorodeoxyglucose positron emission tomography and computed tomography in staging bladder cancer: A single-institution study and a systematic review with meta-analysis. World J Urol. 34:1229–1237. 2016. View Article : Google Scholar : PubMed/NCBI

81 

Aydh A, Abufaraj M, Mori K, Quhal F, Pradere B, Motlagh RS, Mostafaei H, Karakiewicz PI and Shariat SF: Performance of fluoro-2-deoxy-D-glucose positron emission tomography-computed tomography imaging for lymph node staging in bladder and upper tract urothelial carcinoma: A systematic review. Arab J Urol. 19:59–66. 2020. View Article : Google Scholar : PubMed/NCBI

82 

Jensen TK, Holt P, Gerke O, Riehmann M, Svolgaard B, Marcussen N and Bouchelouche K: Preoperative lymph-node staging of invasive urothelial bladder cancer with 18F-fluorodeoxyglucose positron emission tomography/computed axial tomography and magnetic resonance imaging: Correlation with histopathology. Scand J Urol Nephrol. 45:122–128. 2011. View Article : Google Scholar : PubMed/NCBI

83 

Asai S, Fukumoto T, Tanji N, Miura N, Miyagawa M, Nishimura K, Yanagihara Y, Shirato A, Miyauchi Y, Kikugawa T and Yokoyama M: Fluorodeoxyglucose positron emission tomography/computed tomography for diagnosis of upper urinary tract urothelial carcinoma. Int J Clin Oncol. 20:1042–1047. 2015. View Article : Google Scholar : PubMed/NCBI

84 

Tanaka H, Yoshida S, Komai Y, Sakai Y, Urakami S, Yuasa T, Yamamoto S, Masuda H, Koizumi M, Kohno A, et al: Clinical value of 18F-fluorodeoxyglucose positron emission tomography/computed tomography in upper tract urothelial carcinoma: Impact on detection of metastases and patient management. Urol Int. 96:65–72. 2016. View Article : Google Scholar : PubMed/NCBI

85 

Takayanagi A, Takahashi A, Yorozuya W, Okabe K, Kaji T and Takagi Y: The usefulness of positron emission tomography/computed tomography in the diagnosis of metastasis in patients with urothelial carcinoma. Nihon Hinyokika Gakkai Zasshi. 113:51–55. 2022.(In Japanese). PubMed/NCBI

86 

Galili U: Cancer immunotherapy by anti-gal-mediated in situ conversion of tumors into autologous vaccines. The Natural Anti-gal Antibody as Foe Turned Friend in Medicine, . 1st edition. Elsevier; Amsterdam: pp. 171–198. 2018

87 

Nayak B, Dogra PN, Naswa N and Kumar R: Diuretic 18F-FDG PET/CT imaging for detection and locoregional staging of urinary bladder cancer: Prospective evaluation of a novel technique. Eur J Nucl Med Mol Imaging. 40:386–393. 2013. View Article : Google Scholar : PubMed/NCBI

88 

Jana S and Blaufox MD: Nuclear medicine studies of the prostate, testes, and bladder. Semin Nucl Med. 36:51–72. 2006. View Article : Google Scholar : PubMed/NCBI

89 

Sassa N, Kato K, Abe S, Iwano S, Ito S, Ikeda M, Shimamoto K, Yamamoto S, Yamamoto T, Gotoh M and Naganawa S: Evaluation of 11C-choline PET/CT for primary diagnosis and staging of urothelial carcinoma of the upper urinary tract: A pilot study. Eur J Nucl Med Mol Imaging. 41:2232–2241. 2014. View Article : Google Scholar : PubMed/NCBI

90 

Polom W, Cytawa W, Polom A, Frankiewicz M, Szurowska E, Lass P and Matuszewski M: Challenging visualization of sentinel lymph nodes in upper urinary tract urothelial carcinoma. J Clin Med. 10:54652021. View Article : Google Scholar : PubMed/NCBI

91 

Lebastchi AH, Gupta N, DiBianco JM, Piert M, Davenport MS, Ahdoot MA, Gurram S, Bloom JB, Gomella PT, Mehralivand S, et al: Comparison of cross-sectional imaging techniques for the detection of prostate cancer lymph node metastasis: A critical review. Transl Androl Urol. 9:1415–1427. 2020. View Article : Google Scholar : PubMed/NCBI

92 

Fortuin AS, Smeenk RJ, Meijer HJ, Witjes AJ and Barentsz JO: Lymphotropic nanoparticle-enhanced MRI in prostate cancer: Value and therapeutic potential. Curr Urol Rep. 15:3892014. View Article : Google Scholar : PubMed/NCBI

93 

Birkhäuser FD, Studer UE, Froehlich JM, Triantafyllou M, Bains LJ, Petralia G, Vermathen P, Fleischmann A and Thoeny HC: Combined ultrasmall superparamagnetic particles of iron oxide-enhanced and diffusion-weighted magnetic resonance imaging facilitates detection of metastases in normal-sized pelvic lymph nodes of patients with bladder and prostate cancer. Eur Urol. 64:953–960. 2013. View Article : Google Scholar : PubMed/NCBI

94 

Caglic I and Barrett T: Diffusion-weighted imaging (DWI) in lymph node staging for prostate cancer. Transl Androl Urol. 7:814–823. 2018. View Article : Google Scholar : PubMed/NCBI

95 

Woo S, Suh CH, Kim SY, Cho JY and Kim SH: The diagnostic performance of MRI for detection of lymph node metastasis in bladder and prostate cancer: An updated systematic review and diagnostic meta-analysis. AJR Am J Roentgenol. 210:W95–W109. 2018. View Article : Google Scholar : PubMed/NCBI

96 

Shamis SAK, Edwards J and McMillan DC: The relationship between carbonic anhydrase IX (CAIX) and patient survival in breast cancer: Systematic review and meta-analysis. Diagn Pathol. 18:462023. View Article : Google Scholar : PubMed/NCBI

97 

Venkat S, Khan AI, Lewicki PJ, Borregales L and Scherr DS: Novel nomograms to predict muscle invasion and lymph node metastasis in upper tract urothelial carcinoma. Urol Oncol. 40:108.e11–108.e17. 2022. View Article : Google Scholar : PubMed/NCBI

98 

Li WM, Chan TC, Huang SK, Wu WJ, Ke HL, Liang PI, Wei YC, Shiue YL and Li CF: Prognostic utility of FBLN2 expression in patients with urothelial carcinoma. Front Oncol. 10:5703402020. View Article : Google Scholar : PubMed/NCBI

99 

Ibrahim AM, Sabet S, El-Ghor AA, Kamel N, Anis SE, Morris JS and Stein T: Fibulin-2 is required for basement membrane integrity of mammary epithelium. Sci Rep. 8:141392018. View Article : Google Scholar : PubMed/NCBI

100 

de Vega S, Iwamoto T and Yamada Y: Fibulins: Multiple roles in matrix structures and tissue functions. Cell Mol Life Sci. 66:1890–1902. 2009. View Article : Google Scholar : PubMed/NCBI

101 

Argraves WS, Greene LM, Cooley MA and Gallagher WM: Fibulins: Physiological and disease perspectives. EMBO Rep. 4:1127–1131. 2003. View Article : Google Scholar : PubMed/NCBI

102 

Timpl R, Sasaki T, Kostka G and Chu ML: Fibulins: A versatile family of extracellular matrix proteins. Nat Rev Mol Cell Biol. 4:479–489. 2003. View Article : Google Scholar : PubMed/NCBI

103 

Baird BN, Schliekelman MJ, Ahn YH, Chen Y, Roybal JD, Gill BJ, Mishra DK, Erez B, O'Reilly M, Yang Y, et al: Fibulin-2 is a driver of malignant progression in lung adenocarcinoma. PLoS One. 8:e670542013. View Article : Google Scholar : PubMed/NCBI

104 

Senapati S, Gnanapragassam VS, Moniaux N, Momi N and Batra SK: Role of MUC4-NIDO domain in the MUC4-mediated metastasis of pancreatic cancer cells. Oncogene. 31:3346–3356. 2012. View Article : Google Scholar : PubMed/NCBI

105 

Mohammad T, Arif K, Alajmi MF, Hussain A, Islam A, Rehman MT and Hassan I: Identification of high-affinity inhibitors of pyruvate dehydrogenase kinase-3: Towards therapeutic management of cancer. J Biomol Struct Dyn. 39:586–594. 2021. View Article : Google Scholar : PubMed/NCBI

106 

Zhu J, Zheng G, Xu H, Jin X, Tang T and Wang X: Expression and prognostic significance of pyruvate dehydrogenase kinase 1 in bladder urothelial carcinoma. Virchows Arch. 477:637–649. 2020. View Article : Google Scholar : PubMed/NCBI

107 

Woolbright BL, Choudhary D, Mikhalyuk A, Trammel C, Shanmugam S, Abbott E, Pilbeam CC and Taylor JA III: The role of pyruvate dehydrogenase kinase-4 (PDK4) in bladder cancer and chemoresistance. Mol Cancer Ther. 17:2004–2012. 2018. View Article : Google Scholar : PubMed/NCBI

108 

Kuo YH, Chan TC, Lai HY, Chen TJ, Wu LC, Hsing CH and Li CF: Overexpression of pyruvate dehydrogenase kinase-3 predicts poor prognosis in urothelial carcinoma. Front Oncol. 11:7491422021. View Article : Google Scholar : PubMed/NCBI

109 

Posey KL, Coustry F and Hecht JT: Cartilage oligomeric matrix protein: COMPopathies and beyond. Matrix Biol. 71-72:161–173. 2018. View Article : Google Scholar : PubMed/NCBI

110 

Englund E, Bartoschek M, Reitsma B, Jacobsson L, Escudero-Esparza A, Orimo A, Leandersson K, Hagerling C, Aspberg A, Storm P, et al: Cartilage oligomeric matrix protein contributes to the development and metastasis of breast cancer. Oncogene. 35:5585–5596. 2016. View Article : Google Scholar : PubMed/NCBI

111 

Liu TT, Liu XS, Zhang M, Liu XN, Zhu FX, Zhu FM, Ouyang SW, Li SB, Song CL, Sun HM, et al: Cartilage oligomeric matrix protein is a prognostic factor and biomarker of colon cancer and promotes cell proliferation by activating the Akt pathway. J Cancer Res Clin Oncol. 144:1049–1063. 2018. View Article : Google Scholar : PubMed/NCBI

112 

Norman GL, Gatselis NK, Shums Z, Liaskos C, Bogdanos DP, Koukoulis GK and Dalekos GN: Cartilage oligomeric matrix protein: A novel non-invasive marker for assessing cirrhosis and risk of hepatocellular carcinoma. World J Hepatol. 7:1875–1883. 2015. View Article : Google Scholar : PubMed/NCBI

113 

Kuo YH, Lai HY, Chan TC, Hsing CH, Huang SK, Hsieh KL, Chen TJ, Li WS, Lu JC and Li CF: Upregulation of cartilage oligomeric matrix protein predicts poor prognosis in urothelial carcinoma. Onco Targets Ther. 15:727–740. 2022. View Article : Google Scholar : PubMed/NCBI

114 

Li WM, Ke HL, Kuo YH, Lai HY, Chan TC, Hsing CH, Hsieh KL, Li WS, Chen TJ, Wei YC, et al: High MT2A expression predicts worse prognosis in patients with urothelial carcinoma. Oncology. 100:485–497. 2022. View Article : Google Scholar : PubMed/NCBI

115 

Nishikawa M, Miyake H, Kurahashi T and Fujisawa M: Significance of multiple preoperative laboratory abnormalities as prognostic indicators in patients with urothelial carcinoma of the upper urinary tract following radical nephroureterectomy. Int J Clin Oncol. 23:151–157. 2018. View Article : Google Scholar : PubMed/NCBI

116 

Mori K, Janisch F, Mostafaei H, Kimura S, Lysenko I, Karakiewicz PI, Briganti A, Enikeev DV, Rouprêt M, Margulis V, et al: Prognostic role of preoperative De Ritis ratio in upper tract urothelial carcinoma treated with nephroureterectomy. Urol Oncol. 38:601.e17–601.e24. 2020. View Article : Google Scholar : PubMed/NCBI

117 

Mantovani A, Allavena P, Sica A and Balkwill F: Cancer-related inflammation. Nature. 454:436–444. 2008. View Article : Google Scholar : PubMed/NCBI

118 

Coussens LM and Werb Z: Inflammation and cancer. Nature. 420:860–867. 2002. View Article : Google Scholar : PubMed/NCBI

119 

Wang K, Diao F, Ye Z, Zhang X, Zhai E, Ren H, Li T, Wu H, He Y, Cai S and Chen J: Prognostic value of systemic immune-inflammation index in patients with gastric cancer. Chin J Cancer. 36:752017. View Article : Google Scholar : PubMed/NCBI

120 

Zhang Y, Chen B, Wang L, Wang R and Yang X: Systemic immune-inflammation index is a promising noninvasive marker to predict survival of lung cancer: A meta-analysis. Medicine (Baltimore). 98:e137882019. View Article : Google Scholar : PubMed/NCBI

121 

Wang L, Wang C, Wang J, Huang X and Cheng Y: A novel systemic immune-inflammation index predicts survival and quality of life of patients after curative resection for esophageal squamous cell carcinoma. J Cancer Res Clin Oncol. 143:2077–2086. 2017. View Article : Google Scholar : PubMed/NCBI

122 

Jan HC, Wu KY, Tai TY, Weng HY, Yang WH, Ou CH and Hu CY: The systemic immune-inflammation index (SII) increases the prognostic significance of lymphovascular invasion in upper tract urothelial carcinoma after radical nephroureterectomy. Cancer Manag Res. 14:3139–3149. 2022. View Article : Google Scholar : PubMed/NCBI

123 

Kobayashi S, Ito M, Takemura K, Suzuki H, Yonese I and Koga F: Preoperative models incorporating the systemic immune-inflammation index for predicting prognosis and muscle invasion in patients with non-metastatic upper tract urothelial carcinoma. Int J Clin Oncol. 27:574–584. 2022. View Article : Google Scholar : PubMed/NCBI

124 

Siegel RL, Miller KD, Fuchs HE and Jemal A: Cancer statistics, 2021. CA Cancer J Clin. 71:7–33. 2021.PubMed/NCBI

125 

Therkildsen C, Eriksson P, Höglund M, Jönsson M, Sjödahl G, Nilbert M and Liedberg F: Molecular subtype classification of urothelial carcinoma in Lynch syndrome. Mol Oncol. 12:1286–1295. 2018. View Article : Google Scholar : PubMed/NCBI

126 

Pradere B, Lotan Y and Roupret M: Lynch syndrome in upper tract urothelial carcinoma: Significance, screening, and surveillance. Curr Opin Urol. 27:48–55. 2017. View Article : Google Scholar : PubMed/NCBI

127 

Audenet F, Colin P, Yates DR, Ouzzane A, Pignot G, Long JA, Soulie M, Phé V, Bensadoun H, Guy L, et al: A proportion of hereditary upper urinary tract urothelial carcinomas are misclassified as sporadic according to a multi-institutional database analysis: Proposal of patient-specific risk identification tool. BJU Int. 110:E583–E589. 2012. View Article : Google Scholar : PubMed/NCBI

128 

Ju JY, Mills AM, Mahadevan MS, Fan J, Culp SH, Thomas MH and Cathro HP: Universal lynch syndrome screening should be performed in all upper tract urothelial carcinomas. Am J Surg Pathol. 42:1549–1555. 2018. View Article : Google Scholar : PubMed/NCBI

129 

Gayhart MG, Johnson N, Paul A, Quillin JM, Hampton LJ, Idowu MO and Smith SC: Universal mismatch repair protein screening in upper tract urothelial carcinoma. Am J Clin Pathol. 154:792–801. 2020. View Article : Google Scholar : PubMed/NCBI

130 

Rasmussen M, Madsen MG and Therkildsen C: Immunohistochemical screening of upper tract urothelial carcinomas for lynch syndrome diagnostics: A systematic review. Urology. 165:44–53. 2022. View Article : Google Scholar : PubMed/NCBI

131 

Vuky J, Balar AV, Castellano D, O'Donnell PH, Grivas P, Bellmunt J, Powles T, Bajorin D, Hahn NM, Savage MJ, et al: Long-term outcomes in KEYNOTE-052: Phase II study investigating first-line pembrolizumab in cisplatin-ineligible patients with locally advanced or metastatic urothelial cancer. J Clin Oncol. 38:2658–2666. 2020. View Article : Google Scholar : PubMed/NCBI

132 

Balar AV, Galsky MD, Rosenberg JE, Powles T, Petrylak DP, Bellmunt J, Loriot Y, Necchi A, Hoffman-Censits J, Perez-Gracia JL, et al: Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: A single-arm, multicentre, phase 2 trial. Lancet. 389:67–76. 2017. View Article : Google Scholar : PubMed/NCBI

133 

Bellmunt J, de Wit R, Vaughn DJ, Fradet Y, Lee JL, Fong L, Vogelzang NJ, Climent MA, Petrylak DP, Choueiri TK, et al: Pembrolizumab as second-line therapy for advanced urothelial carcinoma. N Engl J Med. 376:1015–1026. 2017. View Article : Google Scholar : PubMed/NCBI

134 

Loriot Y, Necchi A, Park SH, Garcia-Donas J, Huddart R, Burgess E, Fleming M, Rezazadeh A, Mellado B, Varlamov S, et al: Erdafitinib in locally advanced or metastatic urothelial carcinoma. N Engl J Med. 381:338–348. 2019. View Article : Google Scholar : PubMed/NCBI

135 

Fujii Y, Sato Y, Suzuki H, Kakiuchi N, Yoshizato T, Lenis AT, Maekawa S, Yokoyama A, Takeuchi Y, Inoue Y, et al: Molecular classification and diagnostics of upper urinary tract urothelial carcinoma. Cancer Cell. 39:793–809.e8. 2021. View Article : Google Scholar : PubMed/NCBI

136 

Seisen T, Colin P, Hupertan V, Yates DR, Xylinas E, Nison L, Cussenot O, Neuzillet Y, Bensalah K, Novara G, et al: Postoperative nomogram to predict cancer-specific survival after radical nephroureterectomy in patients with localised and/or locally advanced upper tract urothelial carcinoma without metastasis. BJU Int. 114:733–740. 2014. View Article : Google Scholar : PubMed/NCBI

137 

Seisen T, Peyronnet B, Dominguez-Escrig JL, Bruins HM, Yuan CY, Babjuk M, Böhle A, Burger M, Compérat EM, Cowan NC, 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. 70:1052–1068. 2016. View Article : Google Scholar : PubMed/NCBI

138 

Leow JJ, Chong YL, Chang SL, Valderrama BP, Powles T and 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. 79:635–654. 2021. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Lv D, Huang Y, Li H, Fu S, Gong C, Yang C, Wang J, Wang H, Tan Z, Ding M, Ding M, et al: Lymphadenectomy in upper tract urothelial carcinoma: Clinical insights and controversies (Review). Oncol Rep 54: 137, 2025.
APA
Lv, D., Huang, Y., Li, H., Fu, S., Gong, C., Yang, C. ... Ding, M. (2025). Lymphadenectomy in upper tract urothelial carcinoma: Clinical insights and controversies (Review). Oncology Reports, 54, 137. https://doi.org/10.3892/or.2025.8970
MLA
Lv, D., Huang, Y., Li, H., Fu, S., Gong, C., Yang, C., Wang, J., Wang, H., Tan, Z., Ding, M."Lymphadenectomy in upper tract urothelial carcinoma: Clinical insights and controversies (Review)". Oncology Reports 54.5 (2025): 137.
Chicago
Lv, D., Huang, Y., Li, H., Fu, S., Gong, C., Yang, C., Wang, J., Wang, H., Tan, Z., Ding, M."Lymphadenectomy in upper tract urothelial carcinoma: Clinical insights and controversies (Review)". Oncology Reports 54, no. 5 (2025): 137. https://doi.org/10.3892/or.2025.8970
Copy and paste a formatted citation
x
Spandidos Publications style
Lv D, Huang Y, Li H, Fu S, Gong C, Yang C, Wang J, Wang H, Tan Z, Ding M, Ding M, et al: Lymphadenectomy in upper tract urothelial carcinoma: Clinical insights and controversies (Review). Oncol Rep 54: 137, 2025.
APA
Lv, D., Huang, Y., Li, H., Fu, S., Gong, C., Yang, C. ... Ding, M. (2025). Lymphadenectomy in upper tract urothelial carcinoma: Clinical insights and controversies (Review). Oncology Reports, 54, 137. https://doi.org/10.3892/or.2025.8970
MLA
Lv, D., Huang, Y., Li, H., Fu, S., Gong, C., Yang, C., Wang, J., Wang, H., Tan, Z., Ding, M."Lymphadenectomy in upper tract urothelial carcinoma: Clinical insights and controversies (Review)". Oncology Reports 54.5 (2025): 137.
Chicago
Lv, D., Huang, Y., Li, H., Fu, S., Gong, C., Yang, C., Wang, J., Wang, H., Tan, Z., Ding, M."Lymphadenectomy in upper tract urothelial carcinoma: Clinical insights and controversies (Review)". Oncology Reports 54, no. 5 (2025): 137. https://doi.org/10.3892/or.2025.8970
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team