Attention: Restrictions on use of AUA, AUAER, and UCF content in third party applications, including artificial intelligence technologies, such as large language models and generative AI.
You are prohibited from using or uploading content you accessed through this website into external applications, bots, software, or websites, including those using artificial intelligence technologies and infrastructure, including deep learning, machine learning and large language models and generative AI.

Diagnosis and Management of Non-Metastatic Upper Tract Urothelial Carcinoma: AUA/SUO Guideline (2023)

Using AUA Guidelines

This AUA guideline is provided free of use to the general public for academic and research purposes. However, any person or company accessing AUA guidelines for promotional or commercial use must obtain a licensed copy. To obtain the licensable copy of this guideline, please contact Keith Price at kprice@auanet.org.

To cite this guideline:
Coleman JA, Clark PE, Bixler BR, et al. Diagnosis and management of non-metastatic upper tract urothelial carcinoma: AUA/SUO guideline. J Urol. 2023;209(6):1071-1081.

Unabridged version of this Guideline

Appendix for this Guidelines I

Appendix for this Guidelines II

Appendix for this Guidelines III

Appendix for this Guidelines IV

Appendix for this Guidelines V

Panel Members

Jonathan A. Coleman, MD; Peter E. Clark, MD; David I. Buckley, MD, MPH; Sam S. Chang MD, MBA; Roger Chou, MD; Jean Hoffman-Centsis, MD; Girish S. Kulkarni, MD, PhD; Surena F. Matin, MD; Phillip M. Pierorazio, MD; Aaron M. Potretzke, MD; Sarah P. Psutka, MD; Jay D. Raman, MD; Angela B. Smith, MD; Laura Smith

SUMMARY

Purpose

The purpose of this guideline is to provide a useful reference on the effective evidence-based diagnoses and management of non-metastatic upper tract urothelial carcinoma (UTUC).

Methodology

The Pacific Northwest Evidence-based Practice Center of Oregon Health & Science University (OHSU) team conducted searches in Ovid MEDLINE (1946 to March 3rd, 2022), Cochrane Central Register of Controlled Trials (through January 2022), and Cochrane Database of Systematic Reviews (through January 2022). The searches were updated August 2022 and January 2023. When sufficient evidence existed, the body of evidence was assigned a strength rating of A (high), B (moderate), or C (low) for support of Strong, Moderate, or Conditional Recommendations. In the absence of sufficient evidence, additional information is provided as Clinical Principles and Expert Opinions.

GUIDELINE STATEMENTS

Diagnosis and Evaluation

  1. For patients with suspected UTUC, a cystoscopy and cross- sectional imaging of the upper tract with contrast including delayed images of the collecting system and ureter should be performed. (Strong Recommendation; Evidence Level: Grade B)
  2. Clinicians should evaluate patients with suspected UTUC with diagnostic ureteroscopy and biopsy of any identified lesion and cytologic washing from the upper tract system being inspected. (Strong Recommendation; Evidence Level: Grade C)
  3. In patients who have concomitant lower tract tumors (bladder/urethra) discovered at the time of ureteroscopy, the lower tract tumors should be managed in the same setting as ureteroscopy. (Expert Opinion)
  4. In cases of existing ureteral strictures or difficult access to the upper tract, clinicians should minimize risk of ureteral injury by using gentle dilation techniques such as temporary stenting (pre-stenting) and limit use of aggressive dilation access techniques such as ureteral access sheaths. (Expert Opinion)
  5. In cases where ureteroscopy cannot be safely performed or is not possible, an attempt at selective upper tract washing or barbotage for cytology may be made and pyeloureterography performed in cases where good quality imaging such as CT or MR urography cannot be obtained. (Conditional Recommendation; Evidence Level: Grade C)
  6. At the time of ureteroscopy for suspected UTUC, clinicians should not perform ureteroscopic inspection of a radiographically and clinically normal contralateral upper tract. (Expert Opinion)
  7. For patients with suspected/ diagnosed UTUC, clinicians should obtain a personal and family history to identify known hereditary risk factors for familial diseases associated with Lynch Syndrome (LS) (colorectal, ovarian, endometrial, gastric, biliary, small bowel, pancreatic, prostate, skin and brain cancer) for which referral for genetic counseling should be offered. (Expert Opinion)
  8. Universal histologic testing of UTUC with additional studies, such as immunohistochemical (IHC) or microsatellite instability (MSI), should be performed to identify patients with high probability of Lynch-related cancers whom clinicians should refer for genetic counseling and germline testing. (Strong Recommendation; Evidence Level: Grade B)

Risk Stratification

  1. At the time of identified UTUC, clinicians should perform a standardized assessment documenting clinically meaningful endoscopic (focality, location, appearance, size) and radiographic (invasion, obstruction, and lymphadenopathy) features to facilitate clinical staging and risk assessment. (Strong Recommendation; Evidence Level: Grade B)
  2. Following standardized assessment, clinicians should risk-stratify patients as “low-” or “high” risk for invasive disease (pT2 or greater) based on obtained endoscopic, cytologic, pathologic, and radiographic findings. Further stratification into favorable and unfavorable risk groups should then be based on standard identified features (Table 5). (Strong Recommendation; Evidence Level: Grade B)
  3. Patients with UTUC should be assessed prior to surgery for the risk of post-NU CKD or dialysis. (Expert Opinion)

Treatment

  1. Clinicians should provide patients with a description of the short- and long-term risks associated with recommended diagnostic and therapeutic options. This includes the need for endoscopic follow-up, clinically significant strictures, toxicities associated with surgical treatment and side effects from neoadjuvant and adjuvant therapies. (Clinical Principle)

Kidney Sparing Management

  1. Tumor ablation should be the initial management option for patients with LR favorable UTUC. (Strong Recommendation; Evidence Level: Grade B)
  2. Tumor ablation may be the initial management option offered to patients with LR unfavorable UTUC and select patients with HR favorable disease who have low-volume tumors or cannot undergo RNU. (Conditional Recommendation; Evidence Level: Grade C)
  3. Tumor ablation may be accomplished via a retrograde or antegrade percutaneous approach and repeat endoscopic evaluation should be performed within three months. (Expert Opinion)
  4. Following ablation of UTUC tumors and after confirming there is no perforation of the bladder or upper tract, clinicians may instill adjuvant pelvicalyceal chemotherapy (Conditional Recommendation; Evidence Level: Grade C) or intravesical chemotherapy (Expert Opinion) to decrease the risk of urothelial cancer recurrence.
  5. Pelvicalyceal therapy with BCG may be offered to patients with HR favorable UTUC after complete tumor ablation or patients with upper tract carcinoma in situ (CIS). (Expert Opinion)
  6. When tumor ablation is not feasible or evidence of risk group progression is identified in patients with LR UTUC, surgical resection of all involved sites either by RNU or segmental resection of the ureter should be offered. (Moderate Recommendation; Evidence Level: Grade C)
  7. Clinicians may offer watchful waiting or surveillance alone to select patients with UTUC with significant comorbidities, competing risks of mortality, or at significant risk of End-Stage Renal Disease (ESRD) with any intervention resulting in dialysis. (Expert Opinion)

Surgical Management

  1. Clinicians should recommend RNU or SU for surgically eligible patients with HR UTUC. (Strong Recommendation; Evidence Level: Grade B)
  2. For surgically eligible patients with HR and unfavorable LR cancers endoscopically confirmed as confined to the lower ureter in a functional renal unit, distal ureterectomy and ureteral reimplantation is the preferred treatment. (Expert Opinion)
  3. When performing NU or distal ureterectomy, the entire distal ureter including the intramural ureteral tunnel and ureteral orifice should be excised, and the urinary tract should be closed in a watertight fashion. (Strong Recommendation, Evidence Level: Grade B)
  4. In patients undergoing RNU or SU (including distal ureterectomy) for UTUC, a single dose of perioperative intravesical chemotherapy should be administered in eligible patients to reduce the risk of bladder recurrence. (Strong Recommendation; Evidence Level: Grade A)

Lymph Node Dissection (LND)

  1. For patients with LR UTUC, clinicians may perform LND at time of NU or ureterectomy. (Conditional Recommendation; Evidence Level: Grade C)
  2. For patients with HR UTUC, clinicians should perform LND at the time of NU or ureterectomy. (Strong Recommendation; Evidence Level: Grade B)

Neoadjuvant/Adjuvant Chemotherapy and Immunotherapy

  1. Clinicians should offer cisplatin-based NAC to patients undergoing RNU or ureterectomy with HR UTUC, particularly in those patients whose post-operative eGFR is expected to be less than 60 mL/min/1.73m2 or those with other medical comorbidities that would preclude platinum-based chemotherapy in the post-operative setting. (Strong Recommendation; Evidence Level: Grade B)
  2. Clinicians should offer platinum-based adjuvant chemotherapy to patients with advanced pathological stage (pT2–T4 pN0–N3 M0 or pTany N1–3 M0) UTUC after RNU or ureterectomy who have not received neoadjuvant platinum-based therapy. (Strong Recommendation; Evidence Level: Grade A)
  3. Adjuvant nivolumab therapy may be offered to patients who received neoadjuvant platinum-based chemotherapy (ypT2–T4 or ypN+) or who are ineligible for or refuse perioperative cisplatin (pT3, pT4a, or pN+). (Conditional Recommendation; Evidence Level: Grade B)
  4. In patients with metastatic (M+) UTUC, RNU or ureterectomy should not be offered as initial therapy. (Expert Opinion)
  5. Patients with clinical, regional node-positive (cN1-3, M0) UTUC should initially be treated with systemic therapy. Consolidative RNU or ureterectomy with lymph-node dissection may be performed in those with a partial or complete response. (Expert Opinion)
  6. Patients with unresectable UTUC (including those who are ineligible or refuse surgery [RNU or ureterectomy]) should be offered a clinical trial or best supportive care including palliative management (radiation, systemic approach, endoscopic, or ablative) for refractory symptoms such as hematuria. (Expert Opinion)

Surveillance and Survivorship

Post-Treatment Surveillance

Surveillance After Kidney Sparing
  1. Low-risk patients managed with kidney sparing treatment should undergo a follow-up cystoscopy and upper tract endoscopy within one to three months to confirm successful treatment. Once confirmed, these patients should undergo continued cystoscopic surveillance of the bladder at least every six to nine months for the first two years and then at least annually thereafter. Endoscopy should be repeated at six months and one year. Upper tract imaging should be performed at least every six to nine months for two years, then annually up to five years. surveillance after five years in the absence of recurrence should be based on shared decision-making between the patient and clinician. (Expert Opinion)
  2. High-risk patients managed with kidney sparing treatment should undergo a follow-up cystoscopy and upper tract endoscopy with cytology within one to three months. Patients with no evidence of disease should undergo cystoscopic surveillance of the bladder and cytology at least every three to six months for the first three years and then at least annually thereafter. Endoscopy should be repeated at least at six months and one year. Upper tract imaging should be performed every three to six months for three years, then annually up to five years. surveillance after five years in the absence of recurrence should be encouraged and based on shared decision-making between the patient and clinician. (Expert Opinion)
  3. Patients who develop urothelial recurrence in the bladder or urethra or positive cytology following treatment for UTUC should be evaluated for possible ipsilateral recurrence or development of new contralateral upper tract disease. (Expert Opinion)
Surveillance after Radical NU
  1. After NU, patients with <pT2 N0/M0 disease should undergo surveillance with cystoscopy and cytology within three months after surgery, then repeated based on pathologic grade. For LG this should repeated at least every six to nine months for the first two years and then at least annually thereafter. For HG, this should be repeated at least every three to six months for the first three years and then at least annually thereafter. Due to the metastasis risk and estimated 5% probability for contralateral disease, cross-sectional imaging of the abdomen and pelvis should be done within 6 months after surgery and then at least annually for a minimum of 5 years. Surveillance after five years in the absence of recurrence should be encouraged and based on shared decision-making between the patient and clinician (See Table 6). (Expert Opinion)
T2+ managed with NU
  1. For Patients who have undergone NU for >pT2 Nx/0 disease, a clinician should perform surveillance cystoscopy with cytology at three months after surgery, then every three to six months for 3 years, and then annually thereafter. Cross-sectional imaging of the abdomen and pelvis with multiphasic contrast-enhanced CT urography should be performed every three to six months for years one and two, every six months at year three, and annually thereafter to year five. A clinician should perform chest imaging, preferably with chest CT, every 6-12 months for the first 5 years. Beyond five years after surgery in patients without recurrence, ongoing surveillance with cystoscopy and upper tract imaging may be continued on an annual basis according to principles of shared/informed decision-making. (Expert Opinion)

Survivorship

  1. For patients with reduced or deteriorating renal function following NU or other intervention, clinicians should consider referral to nephrology. (Expert Opinion)
  2. Clinicians should discuss disease-related stresses and risk factors and encourage patients with urothelial cancer to adopt healthy lifestyle habits, including smoking cessation, exercise, and a healthy diet, to promote long-term health benefits and quality of life. (Expert Opinion)

INTRODUCTION

Purpose

Upper Tract urothelial cancer (UTUC) is a rare disease, posing unique challenges to clinical management and significant risks to patients – both from the disease and treatment forms. UTUC is often considered analogous to urothelial cancer of the bladder, yet pathogenic, genomic, biologic, and clinical distinctions between these entities have been identified.1, 2 As a clinically clear example, the diagnosis of UTUC of the renal pelvis is associated with a 5-year mortality rate >50%, comparatively worse than the <25% rate for bladder cancer.3 The risk of renal functional loss and associated patient morbidity places patients at an additional clinical disadvantage, warranting specialized approaches and instrumentation for disease assessment, clinical staging and management. Such aspects highlight the clear need for well-designed, multi-disciplinary strategies to guide optimal management for this vulnerable patient population to control variability and reduce the risks from under- and over-treatment. Emerging data from standardized paradigms for evaluation, counseling, and management provide a basis for appropriate risk stratified approaches to optimize patient care, limit toxicity, and improve cancer control and survival. Curation and dissemination of this information, especially in a rare disease prone to clinical complexity, is critical to well-informed patient care and the consideration for referral to experienced, multi-disciplinary teams in more challenging cases.

Methodology

Panel Formation and Process

The UTUC Panel was created in 2021 by the American Urological Association Education and Research, Inc. (AUAER) to develop a clinical guideline addressing management of localized or regionally advanced UTUC. This guideline was developed in collaboration with the Society of Urologic Oncology (SUO). The Practice Guidelines Committee (PGC) of the AUA selected the Panel Chair who in turn appointed the additional panel members based on an open nomination process. The Panel included specialists from urology and oncology.

Peer Review and Document Approval

An integral part of the guideline development process at the AUA is external peer review. The AUA conducted a thorough peer review process to ensure that the document was reviewed by experts in the diagnosis and treatment of UTUC. In addition to reviewers from the AUA PGC, Science and Quality Council (SQC), and Board of Directors (BOD), the document was reviewed by representatives from the American Society of Clinical Oncology (ASCO), American Society for Radiation Oncology (ASTRO), and SUO as well as external content experts. Additionally, a call for reviewers was placed on the AUA website from November 18- December 2, 2022, to allow any additional interested parties to request a copy of the document for review. The guideline was also sent to the Urology Care Foundation and the AUA Public Policy & Advocacy team to open the document further to the patient perspective. The draft guideline document was distributed to 114 peer reviewers. All peer review comments were blinded and sent to the Panel for review. In total, 46 reviewers provided comments, including 34 external reviewers. At the end of the peer review process, a total of 681 comments were received. Following comment discussion, the Panel revised the draft as needed. Once finalized, the guideline was submitted for approval to the AUA PGC, SQC, and BOD for final approval.

Search Strategy

The Pacific Northwest Evidence-based Practice Center of Oregon Health & Science University (OHSU) team conducted searches in Ovid MEDLINE (1946 to March 3rd, 2022), Cochrane Central Register of Controlled Trials (through January 2022), and Cochrane Database of Systematic Reviews (through January 2022). The searches were updated August 2022. The team developed a search strategy by using medical subject headings terms and key words relevant to the diagnosis and treatment UTUC. The evidence review team also reviewed relevant systematic reviews and references provided by the Panel to identify articles that may have been missed by the database searches.

Study Selection and Data Abstraction

Study selection was based on predefined eligibility criteria for the patient populations, interventions, outcomes, and study designs of interest. Two reviewers independently screened titles, abstracts, and full text for inclusion. Differences between reviewers regarding eligibility were resolved through consensus.

Assessment of Risk of Bias (ROB) of Individual Studies

Two investigators independently assessed ROB using predefined criteria. Disagreements were resolved by consensus. For randomized trials and cohort studies, criteria for assessing ROB were adapted from the U.S. Preventive Services Task Force. 4  Criteria for randomized trials included use of appropriate randomization and allocation concealment methods, baseline comparability of groups, blinding, attrition, and use of intention-to-treat analysis. For cohort studies on prognostic factors, criteria included methods for assembling cohorts, attrition, blinding assessment of outcomes, and adjustment for potential confounding. Systematic reviews were assessed using AMSTAR 2 (Assessing the Methodological Quality of Systematic Reviews) criteria. 5  Criteria included use of pre-specified methods, appropriate search methods, assessment of risk of bias, and appropriate synthesis methods. Studies were rated as “low ROB,” “moderate ROB,” or “high ROB” based on the presence and seriousness of methodological shortcomings. The evidence review team graded strength of evidence on outcomes by adapting the AUA’s three predefined levels (A, B, or C) of strength of evidence.

Determination of Evidence Strength

The categorization of evidence strength is conceptually distinct from the quality of individual studies. Evidence strength refers to the body of evidence available for a particular question and includes not only the quality of individual studies but consideration of study design; consistency of findings across studies; adequacy of sample sizes; and generalizability of study populations, settings, and interventions for the purposes of the guideline. The AUA categorizes body of evidence strength as Grade A (well-conducted and highly-generalizable randomized control trial (RCTs) or exceptionally strong observational studies with consistent findings), Grade B (RCTs with some weaknesses of procedure or generalizability or moderately strong observational studies with consistent findings), or Grade C (RCTs with serious deficiencies of procedure or generalizability or extremely small sample sizes or observational studies that are inconsistent, have small sample sizes, or have other problems that potentially confound interpretation of data). By definition, Grade A evidence has a high level of certainty, Grade B evidence has a moderate level of certainty, and Grade C evidence has a low level of certainty (Table 1).6

Table 1: Strength of Evidence Definitions

AUA Nomenclature: Linking Statement Type to Evidence Strength

The AUA nomenclature system explicitly links statement type to body of evidence strength, level of certainty, magnitude of benefit or risk/burdens, and the Panel’s judgment regarding the balance between benefits and risks/burdens (Table 2). Strong Recommendations are directive statements that an action should (benefits outweigh risks/burdens) or should not (risks/burdens outweigh benefits) be undertaken because net benefit or net harm is substantial. Moderate Recommendations are directive statements that an action should (benefits outweigh risks/burdens) or should not (risks/burdens outweigh benefits) be undertaken because net benefit or net harm is moderate. Conditional Recommendations are non-directive statements used when the evidence indicates there is no apparent net benefit or harm or when the balance between benefits and risks/burden is unclear. All three statement types may be supported by any body of evidence strength grade. Body of evidence strength Grade A in support of a Strong or Moderate Recommendation indicates the statement can be applied to most patients in most circumstances and that future research is unlikely to change confidence. Body of evidence strength Grade B in support of a Strong or Moderate Recommendation indicates the statement can be applied to most patients in most circumstances, but better evidence could change confidence. Body of evidence strength Grade C in support of a Strong or Moderate Recommendation indicates the statement can be applied to most patients in most circumstances, but better evidence is likely to change confidence. Body of evidence strength Grade C is only rarely used in support of a Strong Recommendation. Conditional Recommendations can also be supported by any evidence strength. When body of evidence strength is Grade A, the statement indicates benefits and risks/burdens appear balanced, the best action depends on patient circumstances, and future research is unlikely to change confidence. When body of evidence strength Grade B is used, benefits and risks/burdens appear balanced, the best action also depends on individual patient circumstances, and better evidence could change confidence. When body of evidence strength Grade C is used, there is uncertainty regarding the balance between benefits and risks/burdens, alternative strategies may be equally reasonable, and better evidence is likely to change confidence.

Where gaps in the evidence existed, Clinical Principles or Expert Opinions are provided via consensus of the Panel. A Clinical Principle is a statement about a component of clinical care widely agreed upon by urologists or other clinicians for which there may or may not be evidence in the medical literature. Expert Opinion refers to a statement based on members' clinical training, experience, knowledge, and judgment for which  there may or may not be evidence in the medical literature.

Table 2: AUA Nomenclature Linking Statement Type to Level of Certainty, Magnitude of Benefit or Risk/Burden, and Body of Evidence Strength

BACKGROUND

UTUC refers to urothelial tumors that originate from the inner lining of the ureter, calyces, or renal pelvis.7 These anatomic structures derive embryologically from mesoderm and the ureteric bud associated with the wolffian duct, separate and distinct from the bladder and urethra, which are endodermal structures developed from the cloaca. Although related in pathogenesis to lower tract urothelial cancer (bladder and urethra), UTUC is much less common, only affecting 5-10% of all patients with urothelial carcinoma though poorly documented such that true estimates of incidence are difficult to track.8 According to the American Cancer Society, approximately 4,010 Americans will be diagnosed with cancer of the ureter/other urinary organs in 2022. Surveillance, Epidemiology, and End Results (SEER) estimates report a consistent incidence of renal pelvic tumors between 0.9 - 1.0 cases per 100,000 in the U.S. through 2019, equaling between 2,980-3,280 cases per year.7 Together, these data indicate an estimated total incidence of UTUC of just over 7,000 U.S. cases per year – slightly less than the annual incidence of testis cancer (8,000 – 10,000 cases).

As a rare disease with complex management paradigms, clinicians should have knowledge of patient demographics, staging distribution and causative factors when evaluating patients with suspected UTUC. According to SEER population data, approximately 25% of cases will present as localized disease, over 50% will have regionally advanced cancers, and nearly 20% will have distant disease at the time of diagnosis. Peak incidence is seen in adults aged >70 years and is three times more common in men than women in western countries.3, 9 Risk factors include occupational exposure, geographic location, Balkan endemic nephropathy associated with aristolochia herbal ingestion, chronic upper tract inflammation, and hereditary factors such as Lynch and Lynch-like syndromes.10

GUIDELINE STATEMENTS

Diagnosis and Evaluation

Guideline Statement 1

For patients with suspected UTUC, a cystoscopy and cross- sectional imaging of the upper tract with contrast including delayed images of the collecting system and ureter should be performed. (Strong Recommendation; Evidence Level: Grade B)

Discussion


Guideline Statement 2

Clinicians should evaluate patients with suspected UTUC with diagnostic ureteroscopy and biopsy of any identified lesion and cytologic washing from the upper tract system being inspected. (Strong Recommendation; Evidence Level: Grade C)

Discussion


Guideline Statement 3

In patients who have concomitant lower tract tumors (bladder/urethra) discovered at the time of ureteroscopy, the lower tract tumors should be managed in the same setting as ureteroscopy. (Expert Opinion)

Discussion


Guideline Statement 4

In cases of existing ureteral strictures or difficult access to the upper tract, clinicians should minimize risk of ureteral injury by using gentle dilation techniques such as temporary stenting (pre-stenting) and limit use of aggressive dilation access techniques such as ureteral access sheaths. (Expert Opinion)

Discussion


Guideline Statement 5

In cases where ureteroscopy cannot be safely performed or is not possible, an attempt at selective upper tract washing or barbotage for cytology may be made and pyeloureterography performed in cases where good quality imaging such as CT or MR urography cannot be obtained. (Conditional Recommendation; Evidence Level: Grade C)

Discussion


Guideline Statement 6

At the time of ureteroscopy for suspected UTUC, clinicians should not perform ureteroscopic inspection of a radiographically and clinically normal contralateral upper tract. (Expert Opinion)

Discussion


Guideline Statement 7

For patients with suspected/ diagnosed UTUC, clinicians should obtain a personal and family history to identify known hereditary risk factors for familial diseases associated with Lynch Syndrome (LS) (colorectal, ovarian, endometrial, gastric, biliary, small bowel, pancreatic, prostate, skin and brain cancer) for which referral for genetic counseling should be offered. (Expert Opinion)

Discussion


Guideline Statement 8

Universal histologic testing of UTUC with additional studies, such as immunohistochemical (IHC) or microsatellite instability (MSI), should be performed to identify patients with high probability of Lynch-related cancers whom clinicians should refer for genetic counseling and germline testing. (Strong Recommendation; Evidence Level: Grade B)

Discussion


Risk Stratification

Guideline Statement 9

At the time of identified UTUC, clinicians should perform a standardized assessment documenting clinically meaningful endoscopic (focality, location, appearance, size) and radiographic (invasion, obstruction, and lymphadenopathy) features to facilitate clinical staging and risk assessment. (Strong Recommendation; Evidence Level: Grade B)

Discussion


Guideline Statement 10

Following standardized assessment, clinicians should risk-stratify patients as “low-” or “high” risk for invasive disease (pT2 or greater) based on obtained endoscopic, cytologic, pathologic, and radiographic findings. Further stratification into favorable and unfavorable risk groups should then be based on standard identified features (Table 5). (Strong Recommendation; Evidence Level: Grade B)

Discussion


Guideline Statement 11

Patients with UTUC should be assessed prior to surgery for the risk of post-NU CKD or dialysis. (Expert Opinion)

Discussion


Treatment

Guideline Statement 12

Clinicians should provide patients with a description of the short- and long-term risks associated with recommended diagnostic and therapeutic options. This includes the need for endoscopic follow-up, clinically significant strictures, toxicities associated with surgical treatment and side effects from neoadjuvant and adjuvant therapies. (Clinical Principle)

Discussion


Kidney Sparing Management

Guideline Statement 13

Tumor ablation should be the initial management option for patients with LR favorable UTUC. (Strong Recommendation; Evidence Level: Grade B)

Discussion


Guideline Statement 14

Tumor ablation may be the initial management option offered to patients with LR unfavorable UTUC and select patients with HR favorable disease who have low-volume tumors or cannot undergo RNU. (Conditional Recommendation; Evidence Level: Grade C)

Discussion


Guideline Statement 15

Tumor ablation may be accomplished via a retrograde or antegrade percutaneous approach and repeat endoscopic evaluation should be performed within three months. (Expert Opinion)

Discussion


Guideline Statement 16

Following ablation of UTUC tumors and after confirming there is no perforation of the bladder or upper tract, clinicians may instill adjuvant pelvicalyceal chemotherapy (Conditional Recommendation; Evidence Level: Grade C) or intravesical chemotherapy (Expert Opinion) to decrease the risk of urothelial cancer recurrence.

Discussion


Guideline Statement 17

Pelvicalyceal therapy with BCG may be offered to patients with HR favorable UTUC after complete tumor ablation or patients with upper tract carcinoma in situ (CIS). (Expert Opinion)

Discussion


Guideline Statement 18

When tumor ablation is not feasible or evidence of risk group progression is identified in patients with LR UTUC, surgical resection of all involved sites either by RNU or segmental resection of the ureter should be offered. (Moderate Recommendation; Evidence Level: Grade C)

Discussion


Guideline Statement 19

Clinicians may offer watchful waiting or surveillance alone to select patients with UTUC with significant comorbidities, competing risks of mortality, or at significant risk of End-Stage Renal Disease (ESRD) with any intervention resulting in dialysis. (Expert Opinion)

Discussion


Surgical Management

Guideline Statement 20

Clinicians should recommend RNU or SU for surgically eligible patients with HR UTUC. (Strong Recommendation; Evidence Level: Grade B)

Discussion


Guideline Statement 21

For surgically eligible patients with HR and unfavorable LR cancers endoscopically confirmed as confined to the lower ureter in a functional renal unit, distal ureterectomy and ureteral reimplantation is the preferred treatment. (Expert Opinion)

Discussion


Guideline Statement 22

When performing NU or distal ureterectomy, the entire distal ureter including the intramural ureteral tunnel and ureteral orifice should be excised, and the urinary tract should be closed in a watertight fashion. (Strong Recommendation, Evidence Level: Grade B)

Discussion


Guideline Statement 23

In patients undergoing RNU or SU (including distal ureterectomy) for UTUC, a single dose of perioperative intravesical chemotherapy should be administered in eligible patients to reduce the risk of bladder recurrence. (Strong Recommendation; Evidence Level: Grade A)

Discussion


Lymph Node Dissection (LND)

Guideline Statement 24

For patients with LR UTUC, clinicians may perform LND at time of NU or ureterectomy. (Conditional Recommendation; Evidence Level: Grade C)

Discussion


Guideline Statement 25

For patients with HR UTUC, clinicians should perform LND at the time of NU or ureterectomy. (Strong Recommendation; Evidence Level: Grade B)

Discussion


Neoadjuvant/Adjuvant Chemotherapy and Immunotherapy

Guideline Statement 26

Clinicians should offer cisplatin-based NAC to patients undergoing RNU or ureterectomy with HR UTUC, particularly in those patients whose post-operative eGFR is expected to be less than 60 mL/min/1.73m2 or those with other medical comorbidities that would preclude platinum-based chemotherapy in the post-operative setting. (Strong Recommendation; Evidence Level: Grade B)

Discussion


Guideline Statement 27

Clinicians should offer platinum-based adjuvant chemotherapy to patients with advanced pathological stage (pT2–T4 pN0–N3 M0 or pTany N1–3 M0) UTUC after RNU or ureterectomy who have not received neoadjuvant platinum-based therapy. (Strong Recommendation; Evidence Level: Grade A)

Discussion


Guideline Statement 28

Adjuvant nivolumab therapy may be offered to patients who received neoadjuvant platinum-based chemotherapy (ypT2–T4 or ypN+) or who are ineligible for or refuse perioperative cisplatin (pT3, pT4a, or pN+). (Conditional Recommendation; Evidence Level: Grade B)

Discussion


Guideline Statement 29

In patients with metastatic (M+) UTUC, RNU or ureterectomy should not be offered as initial therapy. (Expert Opinion)

Discussion


Guideline Statement 30

Patients with clinical, regional node-positive (cN1-3, M0) UTUC should initially be treated with systemic therapy. Consolidative RNU or ureterectomy with lymph-node dissection may be performed in those with a partial or complete response. (Expert Opinion)

Discussion


Guideline Statement 31

Patients with unresectable UTUC (including those who are ineligible or refuse surgery [RNU or ureterectomy]) should be offered a clinical trial or best supportive care including palliative management (radiation, systemic approach, endoscopic, or ablative) for refractory symptoms such as hematuria. (Expert Opinion)

Discussion


Surveillance and Survivorship

Post-Treatment Surveillance
Surveillance After Kidney Sparing

Guideline Statement 32

Low-risk patients managed with kidney sparing treatment should undergo a follow-up cystoscopy and upper tract endoscopy within one to three months to confirm successful treatment. Once confirmed, these patients should undergo continued cystoscopic surveillance of the bladder at least every six to nine months for the first two years and then at least annually thereafter. Endoscopy should be repeated at six months and one year. Upper tract imaging should be performed at least every six to nine months for two years, then annually up to five years. surveillance after five years in the absence of recurrence should be based on shared decision-making between the patient and clinician. (Expert Opinion)

Discussion


Guideline Statement 33

High-risk patients managed with kidney sparing treatment should undergo a follow-up cystoscopy and upper tract endoscopy with cytology within one to three months. Patients with no evidence of disease should undergo cystoscopic surveillance of the bladder and cytology at least every three to six months for the first three years and then at least annually thereafter. Endoscopy should be repeated at least at six months and one year. Upper tract imaging should be performed every three to six months for three years, then annually up to five years. surveillance after five years in the absence of recurrence should be encouraged and based on shared decision-making between the patient and clinician. (Expert Opinion)

Discussion


Guideline Statement 34

Patients who develop urothelial recurrence in the bladder or urethra or positive cytology following treatment for UTUC should be evaluated for possible ipsilateral recurrence or development of new contralateral upper tract disease. (Expert Opinion)

Discussion


Surveillance after Radical NU

Guideline Statement 35

After NU, patients with <pT2 N0/M0 disease should undergo surveillance with cystoscopy and cytology within three months after surgery, then repeated based on pathologic grade. For LG this should repeated at least every six to nine months for the first two years and then at least annually thereafter. For HG, this should be repeated at least every three to six months for the first three years and then at least annually thereafter. Due to the metastasis risk and estimated 5% probability for contralateral disease, cross-sectional imaging of the abdomen and pelvis should be done within 6 months after surgery and then at least annually for a minimum of 5 years. Surveillance after five years in the absence of recurrence should be encouraged and based on shared decision-making between the patient and clinician (See Table 6). (Expert Opinion)

Discussion


T2+ managed with NU

Guideline Statement 36

For Patients who have undergone NU for >pT2 Nx/0 disease, a clinician should perform surveillance cystoscopy with cytology at three months after surgery, then every three to six months for 3 years, and then annually thereafter. Cross-sectional imaging of the abdomen and pelvis with multiphasic contrast-enhanced CT urography should be performed every three to six months for years one and two, every six months at year three, and annually thereafter to year five. A clinician should perform chest imaging, preferably with chest CT, every 6-12 months for the first 5 years. Beyond five years after surgery in patients without recurrence, ongoing surveillance with cystoscopy and upper tract imaging may be continued on an annual basis according to principles of shared/informed decision-making. (Expert Opinion)

Discussion


Survivorship

Guideline Statement 37

For patients with reduced or deteriorating renal function following NU or other intervention, clinicians should consider referral to nephrology. (Expert Opinion)

Discussion


Guideline Statement 38

Clinicians should discuss disease-related stresses and risk factors and encourage patients with urothelial cancer to adopt healthy lifestyle habits, including smoking cessation, exercise, and a healthy diet, to promote long-term health benefits and quality of life. (Expert Opinion)

Discussion


FUTURE DIRECTIONS

Urothelial cancers can arise anywhere in the urinary tract and anatomical features can affect management. Techniques and approaches for addressing tumors in the lower urinary tract (bladder and urethra) have several advantages for standardizing management strategies since they are more easily accessed, clinically staged, locally treated, and readily followed than tumors arising in the upper tract. The large variety of clinical scenarios encountered in upper tract disease coupled with limited access and instrumentation as well as risks of significant comorbidities and organ dysfunction present major challenges and barriers to management that are only recently being recognized and confronted through concerted collaborative efforts required in this rare disease. This last feature also underscores the most serious unmet need that this guideline seeks to address, which are the large education gaps and variation in clinical care surrounding a highly lethal malignancy, rarer than testis cancer, with concentrated expertise in few dedicated centers. Educating clinicians about the current state of medical knowledge, highlighting important nuances of management, and teaching specialized techniques necessary for safe and successful treatment is a pressing priority.

Biology and biomarkers

There is no one-size-fits-all approach to treating UTUC, and further refinements are needed for characterizing aspects of disease risk and biology to help direct care. Recent studies have identified significant genomic distinctions between primary UTUC and primary bladder cancers, namely a higher prevalence of activating FGFR3 mutations (fibroblast growth factor receptor 3) in UTUC as a key driver for tumorigenesis. Investigating the key question as to why this occurs more in upper tract tumors may help lead toward identifying causative factors and the development of preventative strategies, particularly in HR populations such as LS. Genomic markers may also prove useful as less non-invasive biomarkers of tumor grade and stage and for identifying potential pathways for directed treatment, such as FGFR3 inhibition. Other urinary biomarkers investigated to identify UTUC have suggested improved accuracy over urinary cytology, such as DNA methylation assays, RNA panels and cell-free DNA.156-160 Further evaluation of these panels in the clinically relevant setting of screening, evaluation and surveillance seem warranted. Enhancing diagnostic capabilities utilizing the limited tissue samples yielded in UTUC would improve risk stratification and refine treatment planning while facilitating less invasive follow-up approaches to monitor for recurrence or response to treatment. Like surveillance for lower tract disease, urinary biomarkers may provide a less invasive and easily accessible means to refine post-treatment follow-up for urothelial recurrence with better-informed indications and timing for endoscopic surveillance procedures.

Instrumentation and ablative treatments

Improvements in flexible digital endoscopes have greatly improved visualization and access to the upper urinary tract to reach and identify tumors. Instrumentation to allow for effective and safe tissue sampling has been much slower to develop – leaving clinicians to struggle using techniques that are highly skill-dependent and inefficient. Newer devices are in development that may leverage the ability of robotic endoscopy with snake-like instruments to offer better and more precise endoscopic surgical capabilities. The advent of new therapies such as reverse thermo-hydrogel preparation of mitomycin have provided an important new means of treating low-risk tumors. Additional treatments to support kidney sparing approaches are yet needed, especially for small volume HG cancers. Energy devices such as the thulium:YAG laser have recently been approved and added to thermal ablative capabilities. New photodynamic treatments are also now in Phase III clinical trials to offer additional options for treatment. While these primary treatment options have great therapeutic potential, urothelial recurrences are a subsequent issue in follow-up which other groups are also addressing through clinical trials using approaches such as instilled topical chemotherapeutics.

Multi-disciplinary care

Managing patients with UTUC requires a multi-disciplinary team approach to optimize overall care. Access to medical genetics specialists is important for screening and counseling patients with LS – a population just beginning to be recognized and gain appropriate attention for the challenges in care. Improvements in surgical management have limits when disease biology exceeds localized treatment requiring systemic therapies. The integration of medical oncology expertise is therefore critical to provide risk-appropriate adjunctive care to improve cancer specific outcomes and quality of life. Clinical trials with close collaboration between medical oncologist and urologist are addressing some of the key issues of multi-disciplinary care and listed below. The developing field of nephro-oncology also plays a significant role in treatment planning for these vulnerable patient populations who face the prospect of severe renal functional decline and require special attention. Partnerships among these specialties are developing in centers with dedicated UTUC programs to centralize and standardize care – a strategy that has proven effective in optimizing outcomes for other rare cancers that are prone to mismanagement.

Tools and Resources

ABBREVIATIONS

AEAdverse Event
ASCOAmerican Society of Clinical Oncology
AUAAmerican Urological Association
AUAERAmerican Urological Association Education and Research
BCGBacillus Calmette–Guérin
BMPBasic Metabolic Panel
BCEBladder Cuff Excision
BODBoard of Directors
CSSCancer-Specific Survival
CSMCancer-Specific Mortality
CISCarcinoma in Situ
CDCCenters for Disease Control and Prevention
CKDChronic Kidney Disease
CRCColorectal Cancers
CTComputerized Tomography
CIConfidence Interval
DMDiabetes Mellitus
DFSDisease-Free Survival
ESRDEnd-Stage Renal Disease
eGFREstimated Glomerular Filtration Rate
FNAFine-Needle Aspiration
FISHFluorescence In Situ Hybridization
HNPCCHereditary Nonpolyposis Colorectal Cancer
HGHigh-Grade
HRHigh-Risk
HSHigh-Stage
HTNHypertension
IHCImmunohistochemical
LGLow-Grade
LRLow-Risk
LNDLymph Node Dissection
LVILymphovascular Invasion
LSLynch Syndrome
MRMagnetic resonance
MMRMechanisms of Mismatch Repair
MSIMicrosatellite Instability
MMCMitomycin-C
MDCTUMultidetector Computed Tomography Urography
NCCNNational Comprehensive Cancer Network
NPVNegative Predictive Value
NACNeoadjuvant Chemotherapy
NUNephroureterectomy
OHSUOregon Health & Science University
OSOverall Survival
PPVPositive Predictive Value
PGCPractice Guidelines Committee
PFSProgression-Free Survival
RNURadical Nephroureterectomy
RCTRandomized Control Trial
RFSRecurrence-Free Survival
ROBRisk of Bias
SQCScience and Quality Council
SUSegmental Ureterectomy
SEERSurveillance, Epidemiology, and End Results
TURBTTrans Urethral Resection of Bladder Tumor
USUltrasound
UTUCUpper Tract Urothelial Cancer

REFERENCES

  1. Green DA, Rink M, Xylinas E et al: Urothelial carcinoma of the bladder and the upper tract: Disparate twins. J Urol 2013; 189: 1214.
  2. Audenet F, Isharwal S, Cha EK et al: Clonal relatedness and mutational differences between upper tract and bladder urothelial carcinoma. Clin Cancer Res 2019; 25: 967.
  3. SEER: Renal pelvis seer 5-year relative survival rates, 2012-2018: National Cancer Institute, vol. 2022, 2018
  4. Harris RP, Helfand M, Woolf SH et al: Current methods of the us preventive services task force: A review of the process. Am J Prev Med 2001; 20: 21.
  5. Shea BJ, Reeves BC, Wells G et al: Amstar 2: A critical appraisal tool for systematic reviews that include randomised or non-randomised studies of healthcare interventions, or both. Bmj 2017; 358: j4008.
  6. Faraday M, Hubbard H, Kosiak B et al: Staying at the cutting edge: A review and analysis of evidence reporting and grading; the recommendations of the american urological association. BJU Int 2009; 104: 294.
  7. Counsil C: Upper tract urothelial cancer (utuc), vol. 2022, 2022
  8. Siegel RL, Miller KD, Fuchs HE et al: Cancer statistics, 2022. CA Cancer J Clin 2022; 72: 7.
  9. Wu J, Chen S, Wu X et al: Trends of incidence and prognosis of upper tract urothelial carcinoma. Bosn J Basic Med Sci 2021; 21: 607.
  10. Mohammad NS, Nazli R, Zafar H et al: Effects of lipid based multiple micronutrients supplement on the birth outcome of underweight pre-eclamptic women: A randomized clinical trial. Pak J Med Sci 2022; 38: 219.
  11. Janisch F, Shariat SF, Baltzer P et al: Diagnostic performance of multidetector computed tomographic (mdctu) in upper tract urothelial carcinoma (utuc): A systematic review and meta-analysis. World J Urol 2020; 38: 1165.
  12. Rud E, Galtung KF, Lauritzen PM et al: Examining the upper urinary tract in patients with hematuria-time to revise the ct urography protocol? Eur Radiol 2020; 30: 1664.
  13. David RA, James B, Adeloye D et al: Accuracy of ultrasound vs computed tomography scan for upper urinary tract malignancies and development of a risk-based diagnostic algorithm for haematuria in a uk tertiary centre. Int Urol Nephrol 2021; 53: 49.
  14. Takahashi N, Glockner JF, Hartman RP et al: Gadolinium enhanced magnetic resonance urography for upper urinary tract malignancy. J Urol 2010; 183: 1330.
  15. Cowan NC, Turney BW, Taylor NJ et al: Multidetector computed tomography urography for diagnosing upper urinary tract urothelial tumour. BJU Int 2007; 99: 1363.
  16. Guarnizo E, Pavlovich CP, Seiba M et al: Ureteroscopic biopsy of upper tract urothelial carcinoma: Improved diagnostic accuracy and histopathological considerations using a multi-biopsy approach. J Urol 2000; 163: 52.
  17. Kleinmann N, Healy KA, Hubosky SG et al: Ureteroscopic biopsy of upper tract urothelial carcinoma: Comparison of basket and forceps. J Endourol 2013; 27: 1450.
  18. Vashistha V, Shabsigh A and Zynger DL: Utility and diagnostic accuracy of ureteroscopic biopsy in upper tract urothelial carcinoma. Arch Pathol Lab Med 2013; 137: 400.
  19. Dodd LG, Johnston WW, Robertson CN et al: Endoscopic brush cytology of the upper urinary tract. Evaluation of its efficacy and potential limitations in diagnosis. Acta Cytol 1997; 41: 377.
  20. Low RK, Moran ME and Anderson KR: Ureteroscopic cytologic diagnosis of upper tract lesions. J Endourol 1993; 7: 311.
  21. Sheline M, Amendola MA, Pollack HM et al: Fluoroscopically guided retrograde brush biopsy in the diagnosis of transitional cell carcinoma of the upper urinary tract: Results in 45 patients. AJR Am J Roentgenol 1989; 153: 313.
  22. Potretzke AM, Knight BA, Vetter JM et al: Diagnostic utility of selective upper tract urinary cytology: A systematic review and meta-analysis of the literature. Urology 2016; 96: 35.
  23. Barkan GA, Wojcik EM, Nayar R et al: The paris system for reporting urinary cytology: The quest to develop a standardized terminology. Acta Cytol 2016; 60: 185.
  24. Jin H, Lin T, Hao J et al: A comprehensive comparison of fluorescence in situ hybridization and cytology for the detection of upper urinary tract urothelial carcinoma: A systematic review and meta-analysis. Medicine (Baltimore) 2018; 97: e13859.
  25. van Doeveren T, Nakauma-Gonzalez JA, Mason AS et al: The clonal relation of primary upper urinary tract urothelial carcinoma and paired urothelial carcinoma of the bladder. Int J Cancer 2021; 148: 981.
  26. Lynch HT, Lynch PM, Lanspa SJ et al: Review of the lynch syndrome: History, molecular genetics, screening, differential diagnosis, and medicolegal ramifications. Clin Genet 2009; 76: 1.
  27. Latham A, Srinivasan P, Kemel Y et al: Microsatellite instability is associated with the presence of lynch syndrome pan-cancer. J Clin Oncol 2019; 37: 286.
  28. Lipton LR, Johnson V, Cummings C et al: Refining the amsterdam criteria and bethesda guidelines: Testing algorithms for the prediction of mismatch repair mutation status in the familial cancer clinic. Journal of Clinical Oncology 2004; 22: 4934.
  29. Metcalfe MJ, Petros FG, Rao P et al: Universal point of care testing for lynch syndrome in patients with upper tract urothelial carcinoma. J Urol 2018; 199: 60.
  30. Bartley AN, Luthra R, Saraiya DS et al: Identification of cancer patients with lynch syndrome: Clinically significant discordances and problems in tissue-based mismatch repair testing. Cancer Prev Res (Phila) 2012; 5: 320.
  31. Teo MY, Bambury RM, Zabor EC et al: DNA damage response and repair gene alterations are associated with improved survival in patients with platinum-treated advanced urothelial carcinoma. Clin Cancer Res 2017; 23: 3610.
  32. Teo MY, Seier K, Ostrovnaya I et al: Alterations in DNA damage response and repair genes as potential marker of clinical benefit from pd-1/pd-l1 blockade in advanced urothelial cancers. J Clin Oncol 2018; 36: 1685.
  33. Umar A, Boland CR, Terdiman JP et al: Revised bethesda guidelines for hereditary nonpolyposis colorectal cancer (lynch syndrome) and microsatellite instability. J Natl Cancer Inst 2004; 96: 261.
  34. Vasen HF, Watson P, Mecklin JP et al: New clinical criteria for hereditary nonpolyposis colorectal cancer (hnpcc, lynch syndrome) proposed by the international collaborative group on hnpcc. Gastroenterology 1999; 116: 1453.
  35. Favaretto RL, Shariat SF, Savage C et al: Combining imaging and ureteroscopy variables in a preoperative multivariable model for prediction of muscle-invasive and non-organ confined disease in patients with upper tract urothelial carcinoma. BJU Int 2012; 109: 77.
  36. Messer JC, Terrell JD, Herman MP et al: Multi-institutional validation of the ability of preoperative hydronephrosis to predict advanced pathologic tumor stage in upper-tract urothelial carcinoma. Urol Oncol 2013; 31: 904.
  37. Brien JC, Shariat SF, Herman MP et al: Preoperative hydronephrosis, ureteroscopic biopsy grade and urinary cytology can improve prediction of advanced upper tract urothelial carcinoma. J Urol 2010; 184: 69.
  38. Petros FG, Qiao W, Singla N et al: Preoperative multiplex nomogram for prediction of high-risk nonorgan-confined upper-tract urothelial carcinoma. Urol Oncol 2019; 37: 292.e1.
  39. Ma R, Xia H, Qiu M et al: A diagnostic nomogram of pathologic grade for preoperative risk stratification in upper tract urothelial carcinoma. Clin Med Insights Oncol 2020; 14: 1179554920927662.
  40. Yoshida T, Kobayashi T, Kawaura T et al: Development and external validation of a preoperative nomogram for predicting pathological locally advanced disease of clinically localized upper urinary tract carcinoma. Cancer Med 2020; 9: 3733.
  41. Mori K, Katayama S, Laukhtina E et al: Discordance between clinical and pathological staging and grading in upper tract urothelial carcinoma. Clin Genitourin Cancer 2022; 20: 95.e1.
  42. Subiela JD, Territo A, Mercadé A et al: Diagnostic accuracy of ureteroscopic biopsy in predicting stage and grade at final pathology in upper tract urothelial carcinoma: Systematic review and meta-analysis. Eur J Surg Oncol 2020; 46: 1989.
  43. Malm C, Grahn A, Jaremko G et al: Diagnostic accuracy of upper tract urothelial carcinoma: How samples are collected matters. Scand J Urol 2017; 51: 137.
  44. Su X, Hao H, Li X et al: Fluorescence in situ hybridization status of voided urine predicts invasive and high-grade upper tract urothelial carcinoma. Oncotarget 2017; 8: 26106.
  45. Wang J, Wu J, Peng L et al: Distinguishing urothelial carcinoma in the upper urinary tract from benign diseases with hematuria using fish. Acta Cytol 2012; 56: 533.
  46. Mammen S, Krishna S, Quon M et al: Diagnostic accuracy of qualitative and quantitative computed tomography analysis for diagnosis of pathological grade and stage in upper tract urothelial cell carcinoma. J Comput Assist Tomogr 2018; 42: 204.
  47. Ng CK, Shariat SF, Lucas SM et al: Does the presence of hydronephrosis on preoperative axial ct imaging predict worse outcomes for patients undergoing nephroureterectomy for upper-tract urothelial carcinoma? Urol Oncol 2011; 29: 27.
  48. Scolieri MJ, Paik ML, Brown SL et al: Limitations of computed tomography in the preoperative staging of upper tract urothelial carcinoma. Urology 2000; 56: 930.
  49. Almås B, Øverby S, Halvorsen OJ et al: Preoperative predictors of pathological tumour stage and prognosis may be used when selecting candidates for intensified treatment in upper tract urothelial carcinoma. Scand J Urol 2021; 55: 100.
  50. Yu SH, Hur YH, Hwang EC et al: Does multidetector computed tomographic urography (mdctu) t staging classification correspond with pathologic t staging in upper tract urothelial carcinoma? Int Urol Nephrol 2021; 53: 69.
  51. Goto K, Honda Y, Ikeda K et al: Tumor heterogeneity evaluated by computed tomography detects muscle-invasive upper tract urothelial carcinoma that is associated with inflammatory tumor microenvironment. Sci Rep 2021; 11: 14251.
  52. Yoshida R, Yoshizako T, Maruyama M et al: The value of adding diffusion-weighted images for tumor detection and preoperative staging in renal pelvic carcinoma for the reader's experience. Abdom Radiol (NY) 2017; 42: 2297.
  53. Roy C, Labani A, Alemann G et al: Dwi in the etiologic diagnosis of excretory upper urinary tract lesions: Can it help in differentiating benign from malignant tumors? A retrospective study of 98 patients. AJR Am J Roentgenol 2016; 207: 106.
  54. Milojevic B, Djokic M, Sipetic-Grujicic S et al: Prognostic significance of non-muscle-invasive bladder tumor history in patients with upper urinary tract urothelial carcinoma. Urol Oncol 2013; 31: 1615.
  55. Zeng S, Ying Y, Yu X et al: Impact of previous, simultaneous or intravesical recurrence bladder cancer on prognosis of upper tract urothelial carcinoma after nephroureterectomy: A large population-based study. Transl Androl Urol 2021; 10: 4365.
  56. Venkat S, Khan AI, Lewicki PJ et al: Novel nomograms to predict muscle invasion and lymph node metastasis in upper tract urothelial carcinoma. Urol Oncol 2022; 40: 108.e11.
  57. Kaag M, Trost L, Thompson RH et al: Preoperative predictors of renal function decline after radical nephroureterectomy for upper tract urothelial carcinoma. BJU Int 2014; 114: 674.
  58. Xylinas E, Rink M, Margulis V et al: Impact of renal function on eligibility for chemotherapy and survival in patients who have undergone radical nephro-ureterectomy. BJU Int 2013; 112: 453.
  59. Schwartzmann I, Pastore AL, Saccà A et al: Upper urinary tract urothelial carcinoma tumor seeding along percutaneous nephrostomy track: Case report and review of the literature. Urol Int 2017; 98: 115.
  60. Wu Z, Chen Q, Djaladat H et al: A preoperative nomogram to predict renal function insufficiency for cisplatin-based adjuvant chemotherapy following minimally invasive radical nephroureterectomy (robuust collaborative group). Eur Urol Focus 2022; 8: 173.
  61. Aguilar Palacios D, Wilson B, Ascha M et al: New baseline renal function after radical or partial nephrectomy: A simple and accurate predictive model. J Urol 2021; 205: 1310.
  62. Fajkovic H, Klatte T, Nagele U et al: Results and outcomes after endoscopic treatment of upper urinary tract carcinoma: The austrian experience. World Journal of Urology 2013; 31: 37.
  63. Wen J, Ji ZG and Li HZ: Treatment of upper tract urothelial carcinoma with ureteroscopy and thulium laser: A retrospective single center study. BMC Cancer 2018; 18: 196.
  64. Shenhar C, Veredgorn Y, Bulis S et al: Endoscopic management of low-grade upper tract urothelial carcinoma: Characterizing the long-term burden of care in comparison to radical nephroureterectomy. Urology 2022; 159: 152.
  65. Campi R, Cotte J, Sessa F et al: Robotic radical nephroureterectomy and segmental ureterectomy for upper tract urothelial carcinoma: A multi-institutional experience. World J Urol 2019; 37: 2303.
  66. Kim TH, Lee CU, Kang M et al: Comparison of oncologic and functional outcomes between radical nephroureterectomy and segmental ureterectomy for upper urinary tract urothelial carcinoma. Sci Rep 2021; 11: 7828.
  67. Zhang J, Yang F, Wang M et al: Comparison of radical nephroureterectomy and partial ureterectomy for the treatment of upper tract urothelial carcinoma. Biomed Res Int 2018; 2018: 2793172.
  68. Campbell SC, Clark PE, Chang SS et al: Renal mass and localized renal cancer: Evaluation, management, and follow-up: Aua guideline: Part i. J Urol 2021; 206: 199.
  69. Campbell SC, Uzzo RG, Karam JA et al: Renal mass and localized renal cancer: Evaluation, management, and follow-up: Aua guideline: Part ii. J Urol 2021; 206: 209.
  70. Jeon HG, Jeong IG, Lee JW et al: Prognostic factors for chronic kidney disease after curative surgery in patients with small renal tumors. Urology 2009; 74: 1064.
  71. Hung PH, Tsai HB, Hung KY et al: Increased risk of end-stage renal disease in patients with renal cell carcinoma: A 12-year nationwide follow-up study. Medicine (Baltimore) 2014; 93: e52.
  72. Li L, Lau WL, Rhee CM et al: Risk of chronic kidney disease after cancer nephrectomy. Nat Rev Nephrol 2014; 10: 135.
  73. Malcolm JB, Bagrodia A, Derweesh IH et al: Comparison of rates and risk factors for developing chronic renal insufficiency, proteinuria and metabolic acidosis after radical or partial nephrectomy. BJU Int 2009; 104: 476.
  74. Stiles KP, Moffatt MJ, Agodoa LY et al: Renal cell carcinoma as a cause of end-stage renal disease in the united states: Patient characteristics and survival. Kidney Int 2003; 64: 247.
  75. Jeon HG, Choo SH, Sung HH et al: Small tumour size is associated with new-onset chronic kidney disease after radical nephrectomy in patients with renal cell carcinoma. Eur J Cancer 2014; 50: 64.
  76. Cho A, Lee JE, Kwon GY et al: Post-operative acute kidney injury in patients with renal cell carcinoma is a potent risk factor for new-onset chronic kidney disease after radical nephrectomy. Nephrol Dial Transplant 2011; 26: 3496.
  77. Thadhani R, Pascual M and Bonventre JV: Acute renal failure. N Engl J Med 1996; 334: 1448.
  78. Matin SF, Pierorazio PM, Kleinmann N et al: Durability of response to primary chemoablation of low-grade upper tract urothelial carcinoma using ugn-101, a mitomycin-containing reverse thermal gel: Olympus trial final report. J Urol 2022; 207: 779.
  79. Kleinmann N, Matin SF, Pierorazio PM et al: Primary chemoablation of low-grade upper tract urothelial carcinoma using ugn-101, a mitomycin-containing reverse thermal gel (olympus): An open-label, single-arm, phase 3 trial. Lancet Oncol 2020; 21: 776.
  80. Bin X, Roy OP, Ghiraldi E et al: Impact of tumour location and surgical approach on recurrence-free and cancer-specific survival analysis in patients with ureteric tumours. BJU International 2012; 110: E514.
  81. Chen YT, Yu CC, Yeh HC et al: Endoscopic management versus radical nephroureterectomy for localized upper tract urothelial carcinoma in a high endemic region. Scientific Reports 2021; 11: 4040.
  82. Grasso M, Fishman AI, Cohen J et al: Ureteroscopic and extirpative treatment of upper urinary tract urothelial carcinoma: A 15-year comprehensive review of 160 consecutive patients. BJU International 2012; 110: 1618.
  83. Lee BR, Jabbour ME, Marshall FF et al: 13-year survival comparison of percutaneous and open nephroureterectomy approaches for management of transitional cell carcinoma of renal collecting system: Equivalent outcomes. J Endourol 1999; 13: 289.
  84. Raymundo EM, Lipkin ME, Banez LB et al: Third prize: The role of endoscopic nephron-sparing surgery in the management of upper tract urothelial carcinoma. Journal of Endourology 2011; 25: 377.
  85. Rouprêt M, Hupertan V, Traxer O et al: Comparison of open nephroureterectomy and ureteroscopic and percutaneous management of upper urinary tract transitional cell carcinoma. Urology 2006; 67: 1181.
  86. Seisen T, Nison L, Remzi M et al: Oncologic outcomes of kidney sparing surgery versus radical nephroureterectomy for the elective treatment of clinically organ confined upper tract urothelial carcinoma of the distal ureter. Journal of Urology 2016; 195: 1354.
  87. Hoffman A, Yossepowitch O, Erlich Y et al: Oncologic results of nephron sparing endoscopic approach for upper tract low grade transitional cell carcinoma in comparison to nephroureterectomy - a case control study. BMC Urol 2014; 14: 97.
  88. Upfill-Brown A, Lenis AT, Faiena I et al: Treatment utilization and overall survival in patients receiving radical nephroureterectomy versus endoscopic management for upper tract urothelial carcinoma: Evaluation of updated treatment guidelines. World Journal of Urology 2019; 37: 1157.
  89. Vemana G, Kim EH, Bhayani SB et al: Survival comparison between endoscopic and surgical management for patients with upper tract urothelial cancer: A matched propensity score analysis using surveillance, epidemiology and end results-medicare data. Urology 2016; 95: 115.
  90. Cutress ML, Stewart GD, Zakikhani P et al: Ureteroscopic and percutaneous management of upper tract urothelial carcinoma (utuc): Systematic review. BJU Int 2012; 110: 614.
  91. Cho KS, Hong SJ, Cho NH et al: Grade of hydronephrosis and tumor diameter as preoperative prognostic factors in ureteral transitional cell carcinoma. Urology 2007; 70: 662.
  92. Foerster B, Abufaraj M, Matin SF et al: Pretreatment risk stratification for endoscopic kidney-sparing surgery in upper tract urothelial carcinoma: An international collaborative study. Eur Urol 2021; 80: 507.
  93. Scotland KB, Kleinmann N, Cason D et al: Ureteroscopic management of large ≥2 cm upper tract urothelial carcinoma: A comprehensive 23-year experience. Urology 2018; 121: 66.
  94. Douglawi A, Ghoreifi A, Lee R et al: Bladder recurrence following diagnostic ureteroscopy in patients undergoing nephroureterectomy for upper tract urothelial cancer: Is ureteral access sheath protective? Urology 2022; 160: 142.
  95. Villa L, Cloutier J, Letendre J et al: Early repeated ureteroscopy within 6-8 weeks after a primary endoscopic treatment in patients with upper tract urothelial cell carcinoma: Preliminary findings. World J Urol 2016; 34: 1201.
  96. Oosterlinck W, Kurth KH, Schröder F et al: A prospective european organization for research and treatment of cancer genitourinary group randomized trial comparing transurethral resection followed by a single intravesical instillation of epirubicin or water in single stage ta, t1 papillary carcinoma of the bladder. J Urol 1993; 149: 749.
  97. Sylvester RJ, Oosterlinck W and van der Meijden AP: A single immediate postoperative instillation of chemotherapy decreases the risk of recurrence in patients with stage ta t1 bladder cancer: A meta-analysis of published results of randomized clinical trials. J Urol 2004; 171: 2186.
  98. Gallioli A, Boissier R, Territo A et al: Adjuvant single-dose upper urinary tract instillation of mitomycin c after therapeutic ureteroscopy for upper tract urothelial carcinoma: A single-centre prospective non-randomized trial. J Endourol 2020; 34: 573.
  99. Cutress ML, Stewart GD, Wells-Cole S et al: Long-term endoscopic management of upper tract urothelial carcinoma: 20-year single-centre experience. BJU Int 2012; 110: 1608.
  100. Labbate C, Woldu S, Murray K et al: Efficacy and safety of mitomycin gel (ugn-101) as an adjuvant therapy after complete endoscopic management of upper tract urothelial carcinoma. J Urol 2023: 101097ju0000000000003185.
  101. Territo A, Fontanet S, Meneghetti I et al: Management of primary upper urinary tract carcinoma in situ diagnosed by ureteroscopic biopsy: Is bacillus calmette-guerin an alternative to nephroureterectomy? Actas Urol Esp (Engl Ed) 2022.
  102. Redrow GP, Guo CC, Brausi MA et al: Upper urinary tract carcinoma in situ: Current knowledge, future direction. J Urol 2017; 197: 287.
  103. Metcalf M and Pierorazio PM: Future strategies to enhance kidney preservation in upper urinary tract urothelial carcinoma. Transl Androl Urol 2020; 9: 1831.
  104. Katims AB, Tam AW, Rosen DC et al: Novel treatment of upper tract urothelial carcinoma in situ with docetaxel in bcg refractory patients. Urol Oncol 2021; 39: 234.e9.
  105. Fontanet S, Gallioli A, Baboudjian M et al: Topical instillation of bcg immunotherapy for biopsy-proven primary upper urinary tract carcinoma in situ: A single institution series and systematic review. Urol Oncol 2022.
  106. Foerster B, D'Andrea D, Abufaraj M et al: Endocavitary treatment for upper tract urothelial carcinoma: A meta-analysis of the current literature. Urol Oncol 2019; 37: 430.
  107. Giannarini G, Kessler TM, Birkhäuser FD et al: Antegrade perfusion with bacillus calmette-guérin in patients with non-muscle-invasive urothelial carcinoma of the upper urinary tract: Who may benefit? Eur Urol 2011; 60: 955.
  108. Raman JD, Lin YK, Shariat SF et al: Preoperative nomogram to predict the likelihood of complications after radical nephroureterectomy. BJU Int 2017; 119: 268.
  109. Syed JS, Nguyen KA, Suarez-Sariemento A et al: Outcomes of upper tract urothelial cancer managed non-surgically. Canadian Journal of Urology 2019; 26: 9699.
  110. Syed JS, Nguyen KA, Suarez-Sarmiento A et al: Survival outcomes for patients with localised upper tract urothelial carcinoma managed with non-definitive treatment. BJU International 2018; 121: 124.
  111. Chou R, Jungbauer RM and Cheney TP: Management of upper tract urothelial carcinoma: A systematic evidence review, 2022
  112. Simone G, Papalia R, Guaglianone S et al: Laparoscopic versus open nephroureterectomy: Perioperative and oncologic outcomes from a randomised prospective study. Eur Urol 2009; 56: 520.
  113. Xylinas E, Rink M, Cha EK et al: Impact of distal ureter management on oncologic outcomes following radical nephroureterectomy for upper tract urothelial carcinoma. Eur Urol 2014; 65: 210.
  114. Abrate A, Sessa F, Sebastianelli A et al: Segmental resection of distal ureter with termino-terminal ureteric anastomosis vs bladder cuff removal and ureteric re-implantation for upper tract urothelial carcinoma: Results of a multicentre study. BJU International 2019; 124: 116.
  115. Capitanio U, Shariat SF, Isbarn H et al: Comparison of oncologic outcomes for open and laparoscopic nephroureterectomy: A multi-institutional analysis of 1249 cases. European Urology 2009; 56: 1.
  116. Ha YS, Chung JW, Choi SH et al: Impact of a bladder cuff excision during radical nephroureterectomy on cancer specific survival in patients with upper tract urothelial cancer in korea: A retrospective, multi-institutional study. Minerva Urologica e Nefrologica 2017; 69: 466.
  117. Jeldres C, Sun M, Isbarn H et al: A population-based assessment of perioperative mortality after nephroureterectomy for upper-tract urothelial carcinoma. Urology 2010; 75: 315.
  118. Kang M, Jeong CW, Kwak C et al: The characteristics of recurrent upper tract urothelial carcinoma after radical nephroureterectomy without bladder cuff excision. Yonsei Medical Journal 2015; 56: 375.
  119. Lughezzani G, Sun M, Perrotte P et al: Should bladder cuff excision remain the standard of care at nephroureterectomy in patients with urothelial carcinoma of the renal pelvis? A population-based study. European Urology 2010; 57: 956.
  120. Nazzani S, Preisser F, Mazzone E et al: Nephroureterectomy with or without bladder cuff excision for localized urothelial carcinoma of the renal pelvis. European Urology Focus 2020; 6: 298.
  121. O'Brien T, Ray E, Singh R et al: Prevention of bladder tumours after nephroureterectomy for primary upper urinary tract urothelial carcinoma: A prospective, multicentre, randomised clinical trial of a single postoperative intravesical dose of mitomycin c (the odmit-c trial). Eur Urol 2011; 60: 703.
  122. Ito A, Shintaku I, Satoh M et al: Prospective randomized phase ii trial of a single early intravesical instillation of pirarubicin (thp) in the prevention of bladder recurrence after nephroureterectomy for upper urinary tract urothelial carcinoma: The thp monotherapy study group trial. J Clin Oncol 2013; 31: 1422.
  123. Freifeld Y, Ghandour R, Singla N et al: Intraoperative prophylactic intravesical chemotherapy to reduce bladder recurrence following radical nephroureterectomy. Urol Oncol 2020; 38: 737.e11.
  124. Yoo SH, Jeong CW, Kwak C et al: Intravesical chemotherapy after radical nephroureterectomy for primary upper tract urothelial carcinoma: A systematic review and network meta-analysis. J Clin Med 2019; 8.
  125. Guo R, Zhu Y, Xiong G et al: Role of lymph node dissection in the management of upper tract urothelial carcinomas: A meta-analysis. BMC Urology 2018; 18: 24.
  126. Chan VW, Wong CHM, Yuan Y et al: Lymph node dissection for upper tract urothelial carcinoma: A systematic review. Arab J Urol 2020; 19: 37.
  127. Zhai TS, Jin L, Zhou Z et al: Effect of lymph node dissection on stage-specific survival in patients with upper urinary tract urothelial carcinoma treated with nephroureterectomy. BMC Cancer 2019; 19: 1207.
  128. Piraino JA, Snow ZA, Edwards DC et al: Nephroureterectomy vs. Segmental ureterectomy of clinically localized, high-grade, urothelial carcinoma of the ureter: Practice patterns and outcomes. Urologic Oncology 2020; 38: 851.e1.
  129. Leow JJ, Chong YL, Chang SL et al: Neoadjuvant and adjuvant chemotherapy for upper tract urothelial carcinoma: A 2020 systematic review and meta-analysis, and future perspectives on systemic therapy. Eur Urol 2021; 79: 635.
  130. Grossman HB, Natale RB, Tangen CM et al: Neoadjuvant chemotherapy plus cystectomy compared with cystectomy alone for locally advanced bladder cancer. N Engl J Med 2003; 349: 859.
  131. Neoadjuvant chemotherapy in invasive bladder cancer: Update of a systematic review and meta-analysis of individual patient data advanced bladder cancer (abc) meta-analysis collaboration. Eur Urol 2005; 48: 202.
  132. Porena M, Mearini E, Mearini L et al: Voiding dysfunction after radical retropubic prostatectomy: More than external urethral sphincter deficiency. Eur Urol 2007; 52: 38.
  133. Margulis V, Puligandla M, Trabulsi EJ et al: Phase ii trial of neoadjuvant systemic chemotherapy followed by extirpative surgery in patients with high grade upper tract urothelial carcinoma. J Urol 2020; 203: 690.
  134. Coleman JA, Yip W, Wong NC et al: Multicenter phase ii clinical trial of gemcitabine and cisplatin as neoadjuvant chemotherapy for patients with high-grade upper tract urothelial carcinoma. J Clin Oncol 2023: Jco2200763.
  135. Birtle A, Johnson M, Chester J et al: Adjuvant chemotherapy in upper tract urothelial carcinoma (the pout trial): A phase 3, open-label, randomised controlled trial. Lancet 2020; 395: 1268.
  136. Birtle A, Chester J, Jones R et al: Updated outcomes of pout: A phase iii randomized trial of peri-operative chemotherapy versus surveillance in upper tract urothelial cancer (utuc). Journal of Clinical Oncology 2021; 39:455.
  137. Bellmunt J, Hussain M, Gschwend JE et al: Adjuvant atezolizumab versus observation in muscle-invasive urothelial carcinoma (imvigor010): A multicentre, open-label, randomised, phase 3 trial. Lancet Oncol 2021; 22: 525.
  138. Bajorin DF, Witjes JA, Gschwend JE et al: Adjuvant nivolumab versus placebo in muscle-invasive urothelial carcinoma. N Engl J Med 2021; 384: 2102.
  139. Powles T, Assaf ZJ, Davarpanah N et al: Ctdna guiding adjuvant immunotherapy in urothelial carcinoma. Nature 2021; 595: 432.
  140. Testing mk-3475 (pembrolizumab) after surgery for localized muscle-invasive bladder cancer and locally advanced urothelial cancer (ambassador), vol. 2022
  141. Zhang X, Wang P, Qi K et al: The role of surgery on primary site in metastatic upper urinary tract urothelial carcinoma and a nomogram for predicting the survival of patients with metastatic upper urinary tract urothelial carcinoma. Cancer Med 2021; 10: 8079.
  142. Nazzani S, Preisser F, Mazzone E et al: Survival effect of nephroureterectomy in metastatic upper urinary tract urothelial carcinoma. Clin Genitourin Cancer 2019; 17: e602.
  143. Chakiryan N, Martinez A, Gao L et al: Optimizing the sequence of chemotherapy for upper tract urothelial carcinoma with clinically positive regional lymph nodes. Journal of Urology 2019; 202: 76.
  144. Khriguian J, Patrocinio H, Andonian S et al: Stereotactic ablative radiation therapy for the treatment of upper urinary tract urothelial carcinoma. Pract Radiat Oncol 2022; 12: e34.
  145. Liu MZ, Gao XS, Qin SB et al: Radiation therapy for nonmetastatic medically inoperable upper-tract urothelial carcinoma. Transl Androl Urol 2021; 10: 2929.
  146. Brown N, Olayos E, Elmer S et al: Renal embolization and urothelial sclerotherapy for recurrent obstructive urosepsis and intractable haematuria from upper tract urothelial carcinoma. Cardiovasc Intervent Radiol 2016; 39: 467.
  147. Kapoor A, Allard CB, Black P et al: Canadian guidelines for postoperative surveillance of upper urinary tract urothelial carcinoma. Can Urol Assoc J 2013; 7: 306.
  148. Seisen T, Granger B, Colin P et al: A systematic review and meta-analysis of clinicopathologic factors linked to intravesical recurrence after radical nephroureterectomy to treat upper tract urothelial carcinoma. Eur Urol 2015; 67: 1122.
  149. Locke JA, Hamidizadeh R, Kassouf W et al: Surveillance guidelines based on recurrence patterns for upper tract urothelial carcinoma. Can Urol Assoc J 2018; 12: 243.
  150. Chang SS, Boorjian SA, Chou R et al: Diagnosis and treatment of non-muscle invasive bladder cancer: Aua/suo guideline. J Urol 2016; 196: 1021.
  151. Katims AB, Say R, Derweesh I et al: Risk factors for intravesical recurrence after minimally invasive nephroureterectomy for upper tract urothelial cancer (robuust collaboration). J Urol 2021; 206: 568.
  152. Li X, Cui M, Gu X et al: Pattern and risk factors of local recurrence after nephroureterectomy for upper tract urothelial carcinoma. World Journal of Surgical Oncology 2020; 18: 114.
  153. Rink M, Xylinas E, Margulis V et al: Impact of smoking on oncologic outcomes of upper tract urothelial carcinoma after radical nephroureterectomy. Eur Urol 2013; 63: 1082.
  154. Lu Y, Zhang W, Fan S et al: Metabolic syndrome and risk of upper tract urothelial carcinoma: A case-control study from surveillance, epidemiology and end results-medicare-linked database. Front Oncol 2020; 10: 613366.
  155. Ehdaie B, Chromecki TF, Lee RK et al: Obesity adversely impacts disease specific outcomes in patients with upper tract urothelial carcinoma. J Urol 2011; 186: 66.
  156. Territo A, Gallioli A, Diana P et al: DNA methylation urine biomarkers test in the diagnosis of upper tract urothelial carcinoma: Results from a single-center prospective clinical trial. J Urol 2022; 208: 570.
  157. Ghoreifi A, Seyedian SL, Piatti P et al: A urine-based DNA methylation marker test to detect upper tract urothelial carcinoma: A prospective cohort study. J Urol 2023: 101097ju0000000000003188.
  158. Pierconti F, Martini M, Fiorentino V et al: Upper urothelial tract high-grade carcinoma: Comparison of urine cytology and DNA methylation analysis in urinary samples. Hum Pathol 2021; 118: 42.
  159. D'Elia C, Trenti E, Krause P et al: Xpert® bladder cancer detection as a diagnostic tool in upper urinary tract urothelial carcinoma: Preliminary results. Ther Adv Urol 2022; 14: 17562872221090320.
  160. Harsanyi S, Novakova ZV, Bevizova K et al: Biomarkers of bladder cancer: Cell-free DNA, epigenetic modifications and non-coding rnas. Int J Mol Sci 2022; 23.