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

1
Q

Introduction

A

Upper Tract Urothelial Cancer (UTUC) is a rare but serious disease which presents distinct clinical and treatment challenges. While it shares similarities with bladder urothelial cancer, differences in their pathogenesis, biology, and clinical attributes have been identified. UTUC, particularly of the renal pelvis, carries a higher 5-year mortality rate (>50%) compared to bladder cancer (<25%). Additionally, the risk of renal functional loss adds to patient morbidity, requiring specialized approaches for disease assessment, clinical staging, and management. A standardized approach, tailored to patient risks, is essential to optimize care, minimize toxicity, improve cancer control, and enhance survival rates. Dissemination of this knowledge, particularly in relation to this rare, clinically complex disease, can inform better patient care and promote referral to experienced, multidisciplinary teams in more complex cases.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Which of the following cancers has a higher 5-year mortality rate?
a) Bladder urothelial cancer
b) Upper Tract Urothelial Cancer of the renal pelvis
c) Both have similar mortality rates

A

b) Upper Tract Urothelial Cancer of the renal pelvis. UTUC has a >50% mortality rate, which is higher than the <25% rate for bladder cancer.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

What additional risk does UTUC pose beyond cancer itself?
a) Risk of renal functional loss
b) Risk of bladder dysfunction
c) Risk of prostate enlargement

A

a) Risk of renal functional loss. UTUC, especially of the renal pelvis, may result in renal functional loss, adding to patient morbidity.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

Why is a standardized, risk-stratified approach considered optimal in managing UTUC?

A

A standardized, risk-stratified approach to managing UTUC is considered optimal as it helps to customize the treatment and management strategy according to the individual patient’s risk factors. This approach aims to optimize patient care by balancing the need for aggressive treatment with the potential for toxicity, aiming to improve both cancer control and patient survival. Furthermore, it can help reduce under- and over-treatment, thereby minimizing the risk of treatment-associated morbidity.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

How might UTUC treatment and management be optimized in more challenging cases?

A

In more challenging cases, optimization of UTUC treatment and management might be achieved through referral to experienced, multidisciplinary teams. These teams can bring a diverse set of skills and perspectives to patient care, resulting in more comprehensive and specialized management. In complex diseases like UTUC, multidisciplinary teams can offer better informed and coordinated care, improving patient outcomes. The sharing and dissemination of knowledge among these teams also play a crucial role in such optimization.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Background

A

Upper Tract Urothelial Cancer (UTUC) refers to urothelial tumors originating from the ureter, calyces, or renal pelvis, anatomically distinct from the bladder and urethra. UTUC, though related to lower tract urothelial cancer, is less common, affecting 5-10% of all patients with urothelial carcinoma. The exact incidence rate is difficult to determine but estimates suggest around 7,000 cases per year in the U.S. Knowledge of patient demographics, staging distribution and causative factors is critical for UTUC management. SEER data shows 25% of cases present as localized disease, over 50% as regionally advanced cancers, and nearly 20% as distant disease at diagnosis. Peak incidence is in adults over 70 years, and it’s thrice as common in men than women in western countries. Risk factors include occupational exposure, geographic location, Balkan endemic nephropathy associated with aristolochic acid ingestion, chronic upper tract inflammation, and hereditary factors such as Lynch and Lynch-like syndromes.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

What percentage of UTUC cases present as localized disease at the time of diagnosis?
a) 10%
b) 25%
c) 50%

A

b) 25%. According to SEER data, approximately 25% of UTUC cases present as localized disease at diagnosis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Which demographic has the highest incidence of UTUC?
a) Women over 70 years
b) Men over 70 years
c) Men under 70 years

A

b) Men over 70 years. UTUC’s peak incidence is in adults aged over 70 years and is three times more common in men than women in western countries.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What are some risk factors for developing UTUC?

A

Risk factors for developing UTUC include occupational exposure (potentially to carcinogens), geographic location (it’s notably prevalent in certain regions), Balkan endemic nephropathy (associated with the ingestion of aristolochic acid from certain herbs), chronic inflammation of the upper urinary tract, and hereditary factors such as Lynch and Lynch-like syndromes.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

How does the incidence of UTUC compare to that of testicular cancer?

A

The incidence of UTUC is slightly lower than that of testicular cancer. It’s estimated that there are around 7,000 cases of UTUC per year in the U.S., whereas testicular cancer has an annual incidence of between 8,000 – 10,000 cases.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

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)

A

In diagnosing and evaluating suspected Upper Tract Urothelial Cancer (UTUC), a cystoscopy and cross-sectional imaging of the upper tract with contrast, including delayed images of the collecting system and ureter, are recommended due to the risk of concurrent lower tract urothelial cancer. CT urography is advised if no contraindications exist, as it demonstrates high sensitivity (92%) and specificity (95%). When contrast-enhanced CT is not feasible, such as in chronic kidney disease (CKD) or allergy to iodinated contrast medium, clinicians can utilize MR urography, although its sensitivity is lower (63-74%). Retrograde pyelography, in conjunction with non-contrast axial imaging, or renal ultrasound may be used when both multiphasic CT and MR urography are contraindicated, even though ultrasound has not been specifically evaluated for diagnosing UTUC and its routine use is not recommended.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

For patients with suspected UTUC, which diagnostic tool is initially recommended?
a) Cystoscopy and cross-sectional imaging of the upper tract with contrast
b) Retrograde pyelography
c) Renal ultrasound

A

a) Cystoscopy and cross-sectional imaging of the upper tract with contrast. This is recommended due to the risk of concurrent lower tract urothelial cancer.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

If a patient has contraindications to contrast-enhanced CT, what alternative imaging method could be used?
a) MR urography
b) Renal ultrasound
c) None of the above

A

a) MR urography. For patients with contraindications to contrast-enhanced CT, MR urography may be utilized.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Why is a cystoscopy an essential component of the evaluation for patients with suspected UTUC?

A

A cystoscopy is an essential part of the evaluation for patients with suspected UTUC due to the risk of concurrent lower tract urothelial cancer. This means that patients suspected of having UTUC could also have cancer in the bladder or urethra, necessitating a comprehensive examination of the lower urinary tract through cystoscopy.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

How does the sensitivity and specificity of CT urography compare to MR urography in the evaluation of UTUC?

A

CT urography has a higher sensitivity and specificity in the evaluation of UTUC compared to MR urography. The pooled sensitivity of CT urography is approximately 92%, with a specificity of 95%. In contrast, MR urography has a lower sensitivity ranging from 63% to 74%, but retains a high specificity of 96% to 97%. The difference in sensitivity means that CT urography is likely more effective in detecting UTUC when it is present.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

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)

A

For patients with suspected UTUC, a diagnostic ureteroscopy and biopsy of any identified lesion, along with cytologic washing from the upper tract system being inspected, are recommended. The procedure should be standardized and carefully document all key features of UTUC, including tumor size, number, location, focality, and appearance. It is vital to distinguish between diagnostic and therapeutic endoscopic procedures: diagnostic procedures provide crucial clinical information, whereas therapeutic procedures involve more extensive operations aimed at curative treatment. The choice of approach (retrograde ureteroscopy versus antegrade percutaneous nephroscopy and/or ureteroscopy) depends on tumor characteristics and patient factors. Urine cytology can assist in identifying carcinoma in the upper tracts, and urine fluorescence in situ hybridization (FISH) testing also offers high diagnostic accuracy for identifying UTUC. However, the use of FISH is not yet fully established due to its high sensitivity and low specificity compared to voided cytology.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

Which of the following should be performed on patients with suspected UTUC for diagnostic purposes?
a) Retrograde ureteroscopy
b) Antegrade percutaneous nephroscopy
c) Either retrograde ureteroscopy or antegrade percutaneous nephroscopy, depending on factors such as tumor location, configuration, size, and patient factors

A

c) Either retrograde ureteroscopy or antegrade percutaneous nephroscopy, depending on factors such as tumor location, configuration, size, and patient factors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

What diagnostic test can be used to assist in identifying carcinoma in the upper tracts?
a) Urine cytology
b) Urine fluorescence in situ hybridization (FISH) testing
c) Both urine cytology and urine fluorescence in situ hybridization (FISH) testing

A

c) Both urine cytology and urine fluorescence in situ hybridization (FISH) testing

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

What key features should be documented during a ureteroscopic evaluation in patients with suspected UTUC?

A

Key features that should be documented during a ureteroscopic evaluation in patients with suspected UTUC include tumor size, number, location, focality, and appearance. These factors are crucial in guiding further diagnostic testing, informing therapeutic interventions, and providing points of comparison for subsequent ureteroscopic surveillance.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What are some of the considerations when choosing between retrograde ureteroscopy and antegrade percutaneous nephroscopy for diagnosing UTUC?

A

The choice between retrograde ureteroscopy and antegrade percutaneous nephroscopy for diagnosing UTUC is influenced by several factors, including the location, configuration, and size of the tumor, as well as patient-specific factors such as previous surgical history (e.g., prior cystectomy). There is a lack of comprehensive data comparing the effectiveness of these two techniques across all clinical situations, making individualized decision-making based on these factors important.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

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)

A

The guideline recommends that patients who have concomitant lower tract tumors (in the bladder or urethra) discovered during ureteroscopy should receive appropriate, guideline-directed management for these tumors in the same surgical setting. This could include biopsy, resection, or ablation as clinically indicated. Genomic studies indicate a clonal similarity between upper and lower tract tumors, suggesting potential downstream or upstream tumor implantation. The sequencing of procedures (managing bladder before or after ureteroscopy) is scenario-dependent and lacks consensus. Managing the bladder first may optimize visualization, avoid back-pressure or back-washing into the upper tract, and confirm bladder hemostasis. Addressing the upper tract first could be preferred in cases of bulky tumor involvement where complete resection is not possible or when risk assessment is the priority.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

When lower tract tumors are discovered during ureteroscopy for suspected UTUC, what is the recommended approach?
a) Ignore them until the upper tract is fully examined
b) Manage them in the same surgical setting
c) Schedule a separate surgery for lower tract tumors

A

b) Manage them in the same surgical setting

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

The sequencing of procedures (managing bladder before or after ureteroscopy) when lower tract tumors are discovered during ureteroscopy is:
a) Always manage bladder first
b) Always manage bladder last
c) Lacks consensus and heavily scenario-dependent

A

c) Lacks consensus and heavily scenario-dependent

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

What are the considerations for managing bladder tumors discovered at the same time as UTUC during ureteroscopy?

A

Several factors are considered when managing bladder tumors discovered concurrently with UTUC during ureteroscopy. The sequence of procedures, whether to address bladder or upper tract first, lacks consensus and heavily depends on the clinical scenario. Arguments for managing the bladder first include optimizing visualization within the bladder, preventing back-pressure or back-washing into the upper tract, and confirming bladder hemostasis. However, addressing the upper tract first might be preferred in cases of bulky bladder tumor involvement where complete resection isn’t feasible or in cases of bulky upper tract disease where risk assessment is the priority. Seeding of tumors from bladder to upper tract or vice versa has been raised as a concern, and some recommend using ureteral access sheaths to mitigate this, but the benefits of this approach need further study.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

What insights do genomic studies provide about concomitant upper and lower tract tumors?

A

Genomic studies have shown clonal similarity between upper and lower tract tumors, which indicates a potential downstream or upstream tumor implantation mechanism. This finding underscores the interconnected nature of these tumors and justifies treating them in the same surgical setting when they are discovered concurrently. It’s important to note, however, that while bladder tumor pathology often reflects that of upper tract tumors, it is not reliable enough to avoid separate upper tract endoscopy and biopsy when feasible.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

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)

A

The guideline recommends minimizing the risk of ureteral injury in patients with existing ureteral strictures or difficult access to the upper tract by using gentle dilation techniques, such as temporary stenting (pre-stenting), and limiting the use of aggressive dilation access techniques like ureteral access sheaths. This is crucial because perforation or disruption of the urothelium in patients with upper tract urothelial carcinoma (UTUC) can risk tumor seeding outside the urinary tract. Pre-stenting can decrease the risk of iatrogenic injury and allow for a safer and more successful procedure. If perforation or injury events occur, they should be immediately documented and the procedure should be stopped as soon as safely possible, with additional steps taken to limit complications.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

What is recommended to minimize the risk of ureteral injury in cases of existing ureteral strictures or difficult access to the upper tract?
a) Use of aggressive dilation access techniques like ureteral access sheaths
b) Use of gentle dilation techniques such as temporary stenting (pre-stenting)
c) Use of non-dilation techniques only

A

b) Use of gentle dilation techniques such as temporary stenting (pre-stenting)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

If perforation or injury events occur during the procedure, what should be done?
a) Continue the procedure ignoring the injury
b) Document the injury event immediately and cease the procedure as soon as safely possible
c) Ignore the injury until the end of the procedure

A

b) Document the injury event immediately and cease the procedure as soon as safely possible

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

What is the significance of gentle dilation techniques such as pre-stenting in patients with UTUC and difficult ureteral access?

A

In patients with upper tract urothelial carcinoma (UTUC) who have existing ureteral strictures or difficulty accessing the upper tract, gentle dilation techniques such as temporary stenting or pre-stenting can decrease the risk of iatrogenic injury. This is critical as perforation or disruption of the urothelium in patients with UTUC can risk seeding the tumor outside the urinary tract. Pre-stenting provides an opportunity for a safer and more successful procedure.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

What measures should be taken in case of recognized perforation or injury events during the procedure?

A

If a perforation or injury event is recognized during the procedure, it should be immediately documented, and the procedure should be stopped as soon as it is safe to do so. Additional measures to limit complications should be undertaken, such as stenting, bladder decompression with urethral catheter drainage to limit reflux, and placement of a nephrostomy tube in cases of a completely obstructive ureteral tumor and evidence of contrast extravasation. These measures are important to manage the injury and prevent further complications.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

Table 3: Standardized Upper Tract Endoscopy Suggested Reporting Elements

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

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)

A

The guideline suggests that in cases where ureteroscopy can’t be performed safely or isn’t possible, attempts at selective upper tract washing or barbotage for cytology can be made, and pyeloureterography could be performed in cases where high-quality imaging such as CT or MR urography cannot be obtained. The findings from these procedures may provide useful and objective information for risk stratification when endoscopic examination of the involved upper tract isn’t feasible. This guidance is considered a conditional recommendation due to limited data from endoscopy, biopsy, and imaging, but is supported by the evidence associated with Statement 2.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

When is it recommended to attempt selective upper tract washing or barbotage for cytology?
a) When ureteroscopy can be performed safely and easily
b) When ureteroscopy cannot be safely performed or is not possible
c) When ureteroscopy can be performed, but imaging like CT or MR urography is not possible

A

b) When ureteroscopy cannot be safely performed or is not possible

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

When is pyeloureterography recommended?
a) When high-quality imaging such as CT or MR urography is readily available
b) When ureteroscopy is possible
c) When high-quality imaging such as CT or MR urography cannot be obtained

A

c) When high-quality imaging such as CT or MR urography cannot be obtained

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

How can selective upper tract washing or barbotage for cytology and pyeloureterography be useful in managing UTUC?

A

In cases where ureteroscopy can’t be performed safely or isn’t possible, attempts at selective upper tract washing or barbotage for cytology can provide useful, objective information for risk stratification. Furthermore, pyeloureterography can be beneficial in scenarios where high-quality imaging such as CT or MR urography is not obtainable. These methods provide an alternative approach to gather critical information about the patient’s condition when traditional methods are not viable.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

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)

A

The guideline advises against performing ureteroscopic inspection of a radiographically and clinically normal contralateral upper tract at the time of ureteroscopy for suspected upper tract urothelial carcinoma (UTUC). Indications for upper urinary tract ureteroscopy or percutaneous endoscopy include lateralizing hematuria, suspicious selective cytology, and radiographic presence of a mass or urothelial thickening. These endoscopic procedures carry risks for patient injury and potential for tumor seeding in the presence of urothelial cancer. Unnecessary upper tract endoscopy in the setting of a completely normal contralateral upper urinary tract, without clinical indication or as a “screening” procedure, puts patients at undue risk and is not recommended.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

Under what circumstances is it suggested to perform ureteroscopic inspection of the contralateral upper tract?
a) For screening purposes
b) When there are findings such as lateralizing hematuria, suspicious selective cytology, and radiographic presence of a mass or urothelial thickening
c) When the contralateral upper tract appears normal in radiographic and clinical evaluations

A

b) When there are findings such as lateralizing hematuria, suspicious selective cytology, and radiographic presence of a mass or urothelial thickening

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

What are the potential risks associated with performing ureteroscopic inspection of a radiographically and clinically normal contralateral upper tract?
a) There are no significant risks associated with this procedure
b) Potential risks include patient injury and the possibility of tumor seeding in the presence of urothelial cancer
c) The only risk is patient discomfort during the procedure

A

b) Potential risks include patient injury and the possibility of tumor seeding in the presence of urothelial cancer

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

Why is it recommended not to perform ureteroscopic inspection of a radiographically and clinically normal contralateral upper tract in suspected UTUC?

A

Performing a ureteroscopic inspection of a radiographically and clinically normal contralateral upper tract is not recommended because it unnecessarily exposes the patient to risks associated with the procedure. These risks include potential injury to the patient and the possibility of tumor seeding in the presence of urothelial cancer. The guideline suggests that this procedure should only be performed when there are specific indications such as lateralizing hematuria, suspicious selective cytology, and radiographic presence of a mass or urothelial thickening.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

Under what circumstances should ureteroscopy or percutaneous endoscopy of the upper urinary tract be performed?

A

Ureteroscopy or percutaneous endoscopy of the upper urinary tract should be performed when there are specific clinical indications. These indications include lateralizing hematuria, suspicious selective cytology, and the radiographic presence of a mass or urothelial thickening. The decision to perform these procedures should be guided by the need to diagnose and treat suspected upper tract urothelial carcinoma, balanced against the potential risks associated with the procedures.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

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)

A

Clinicians should obtain a personal and family history from patients with suspected or diagnosed upper tract urothelial carcinoma (UTUC) to identify known hereditary risk factors for familial diseases associated with Lynch Syndrome (LS), including colorectal, ovarian, endometrial, gastric, biliary, small bowel, pancreatic, prostate, skin, and brain cancer. If such risks are identified, referral for genetic counseling should be offered. LS is common among UTUC patients, accounting for 7-20% of U.S. cases. It’s an autosomal-dominant multi-organ cancer syndrome affecting approximately 1 in 280 individuals in the U.S. LS is associated with several cancers, with patients recommended to undergo routine screening due to increased life-long risk for developing associated malignancies. For UTUC patients with familial risk factors, clinical suspicion, or interest in further testing for hereditary syndromes, clinicians can perform initial screening tests and should offer referral for genetic counseling and, if indicated, genetic testing.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

What is the estimated percentage of UTUC cases in the U.S. associated with Lynch Syndrome?
a) 1-3%
b) 7-20%
c) 25-30%

A

b) 7-20%

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

Lynch Syndrome is associated with which of the following cancers?
a) Colorectal, ovarian, and endometrial
b) Gastric, biliary, and small bowel
c) Pancreatic, prostate, skin, and brain
d) All of the above

A

d) All of the above

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

How should clinicians approach patients with suspected or diagnosed UTUC in the context of potential Lynch Syndrome?

A

Clinicians should obtain a detailed personal and family history from patients with suspected or diagnosed UTUC to identify known hereditary risk factors for familial diseases associated with Lynch Syndrome. If such risk factors are identified, clinicians should offer referral for genetic counseling and, if indicated, genetic testing. Routine evaluation should include specific questions about Lynch Syndrome associated cancers to help identify at-risk patients and their family members. In UTUC specifically, Lynch Syndrome may increase the possibility of contralateral upper tract involvement, which should be considered when developing a treatment plan.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

Why is it important to recognize Lynch Syndrome as a potential risk factor in patients with UTUC?

A

It’s important to recognize Lynch Syndrome as a potential risk factor in patients with UTUC as it’s common among such patients, accounting for an estimated 7-20% of U.S. cases. Moreover, Lynch Syndrome is an autosomal-dominant multi-organ cancer syndrome that increases the lifetime risk for developing associated malignancies. Therefore, recognition of this syndrome can help guide clinical decision-making, including routine screening, development of treatment plans, and referral for genetic counseling and testing. It’s particularly important to consider when LS might increase the risk of contralateral upper tract involvement in UTUC. Also, future therapeutic options are likely to target LS-associated cancers, further emphasizing the importance of aligning clinical guidelines with genetic risk factors for the preventative and therapeutic management of UTUC in patients with LS.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

Table 4: Clinical Screening Criteria for LS (also referred to as hereditary non-polyposis colorectal cancer [HNPCC])

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

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)

A

Universal histologic testing of upper tract urothelial carcinoma (UTUC) should be performed, using additional studies like immunohistochemical (IHC) or microsatellite instability (MSI) testing. This helps identify patients with high probability of Lynch-related cancers, who should then be referred for genetic counseling and germline testing. Lynch Syndrome (LS) results from inherited germline mutations in DNA damage response genes responsible for mismatch repair (MMR), specifically MLH1, MSH2, MSH6, PMS2, or EPCAM. Histologic studies like IHC can indicate loss of these specific MMR proteins or evaluate for MSI status as a standard means to assess for the possibility of LS association. Recommendations in other LS-related cancers strongly endorse universal MMR and MSI testing. The panel recommends genetic testing to all patients with UTUC due to the higher identified prevalence of LS association in UTUC relative to colorectal cancer.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

Why should universal histologic testing of UTUC be performed?
a) To identify patients with high probability of Lynch-related cancers
b) To evaluate for the presence of mismatch repair proteins
c) To assess microsatellite instability
d) All of the above

A

d) All of the above

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

Which genetic mutations are commonly associated with Lynch Syndrome?
a) BRCA1 and BRCA2
b) TP53 and PTEN
c) MLH1, MSH2, MSH6, PMS2, or EPCAM
d) FLT3 and IDH1

A

Which genetic mutations are commonly associated with Lynch Syndrome?
a) BRCA1 and BRCA2
b) TP53 and PTEN
c) MLH1, MSH2, MSH6, PMS2, or EPCAM
d) FLT3 and IDH1

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

What is the importance of universal histologic testing of UTUC and what additional studies should be performed?

A

Universal histologic testing of UTUC is crucial as it helps to identify patients with a high probability of Lynch-related cancers. It’s the first-line means to detect Lynch Syndrome-associated features in tumor samples, thus providing clinically significant information for patient counseling, management, and screening for family members. Additional studies that should be performed include immunohistochemical (IHC) testing and microsatellite instability (MSI) testing. IHC testing can preliminarily identify the altered proteins associated with Lynch Syndrome, while MSI testing can assess the genome stability. These tests serve as a standard means to assess for the possibility of LS association, and suspicious findings with these tests require further confirmatory testing.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

Why is genetic testing recommended for all patients with UTUC?

A

Genetic testing is recommended for all patients with UTUC due to the higher identified prevalence of Lynch Syndrome association in UTUC relative to colorectal cancer. Lynch Syndrome results from inherited germline mutations in mismatch repair (MMR) genes, specifically MLH1, MSH2, MSH6, PMS2, or EPCAM. Alterations in these genes can lead to the accumulation of DNA errors and increase the potential for cancer development. Therefore, genetic testing can help to identify patients who have a high probability of Lynch-related cancers, leading to more effective patient counseling, management strategies, and familial screening. Furthermore, recommendations and guidelines in other LS-related cancers strongly endorse universal MMR and MSI testing.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

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)

A

At the time of identifying upper tract urothelial carcinoma (UTUC), a standardized assessment documenting endoscopic (focality, location, appearance, size) and radiographic (invasion, obstruction, lymphadenopathy) features should be performed to facilitate clinical staging and risk assessment. Such tumor features identified by endoscopic and radiologic assessment are strongly associated with disease risk, hence vital for proper risk stratification, treatment decision-making, and treatment response assessment. Key features to document include sites of involvement, number of tumors or presence of multifocality, and tumor appearance. Quality of visualization and possible impacts on accuracy should be documented. Radiographic characterization of tumor features is also important for clinical staging, including details of tumor characteristics suggesting invasive features, obstruction of the urinary tract, locoregional progression, and presence of metastatic disease.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

Why should a standardized assessment be performed at the time of identified UTUC?
a) To facilitate clinical staging and risk assessment
b) To ensure proper risk stratification
c) To guide treatment decision-making
d) All of the above

A

d) All of the above

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

What key features should be documented during the endoscopic examination of UTUC?
a) Sites of involvement
b) Number of tumors or presence of multifocality
c) Tumor appearance
d) All of the above

A

d) All of the above

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

Why is it important to document the endoscopic and radiographic features of a UTUC?

A

The endoscopic and radiographic features of a UTUC are strongly associated with disease risk, which is crucial for proper risk stratification, guiding treatment decisions, and assessing the response to treatment. By standardizing the assessment and documentation of these features, clinicians can better communicate and evaluate the objective clinical findings, enhancing patient management and treatment outcomes.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

What is the importance of radiographic characterization of tumor features in UTUC?

A

Radiographic characterization of tumor features is crucial for clinical staging of UTUC. It provides details about tumor characteristics suggesting invasive features, obstruction of the urinary tract, locoregional progression, and the presence of metastatic disease. This information assists in determining the extent of the disease, helping to inform the appropriate treatment strategy and assess the potential treatment response. Moreover, these radiographic features can aid in the monitoring of disease progression and response to therapy over time.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

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)

A

After standardized assessment, clinicians should categorize patients into “low-“ or “high” risk for invasive disease (pT2 or greater) based on endoscopic, cytologic, pathologic, and radiographic findings. Further stratification into favorable and unfavorable risk groups should be based on standard identified features. The factors assisting sub-stratification include biopsy, cytology, fluorescence in situ hybridization (FISH), and CT imaging. Biopsy has a positive predictive value (PPV) of 60% and negative predictive value (NPV) of 77% for high-stage (HS) disease. FISH may have value as an adjunct test where tissue sampling is challenging and cytology is indeterminate. The sensitivity and specificity of specific CT findings for identifying high-grade (HG) disease varies; heterogenous texture (versus homogeneous) has a sensitivity of 70% and specificity of 100%. Moreover, retrograde pyelograms, MRI, and features such as tumor appearance, growth characteristics, and lower tract involvement also contribute to risk stratification. However, the panel advises that studies like endoluminal US and MRI should only be used as supplements to current standards of care.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

Which factors can aid in sub-stratification within the high-risk and low-risk categories for UTUC?
a) Biopsy
b) Cytology
c) FISH
d) CT Imaging
e) All of the above

A

e) All of the above

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

Which CT finding has the best combination of sensitivity and specificity for identifying high-grade UTUC?
a) Hydroureteronephrosis
b) Heterogeneous texture
c) Local invasion
d) Pathologically enlarged lymph nodes

A

b) Heterogeneous texture

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

How does cytology contribute to the risk stratification of UTUC?

A

Selective ipsilateral upper tract cytology provides supplemental histologic data to tumor biopsies. If high-grade cytology is found in the setting of low-grade biopsy findings, it indicates the likely presence of higher-risk features, such as a high-grade tumor, that were missed on biopsy sampling. Obtaining selective cytology after tumor biopsy can improve the yield of cells for cytologic analysis, providing more accurate and detailed information about the tumor.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

How can imaging modalities like CT and MRI be used in the risk stratification of UTUC?

A

Specific CT findings, such as the presence of heterogeneous texture, can be indicative of high-grade disease in UTUC. It’s important to note that the sensitivity and specificity of CT findings for identifying high-stage UTUC vary widely. MRI can provide some soft tissue details in patients who cannot receive contrast, offering some advantages by identifying features of fat invasion with diffusion-weighted imaging associated with very advanced, T3 disease. However, MRI can falsely overestimate tumor stage due to surrounding tissue effects that may mimic tumor invasion. At present, the guidelines recommend such studies only as supplements to current standards of care.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

Table 5: Presurgical Clinical Risk Categories

A
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

GUIDELINE STATEMENT 11
Patients with UTUC should be assessed prior to surgery for the risk of post-NU CKD or dialysis. (Expert Opinion)

A

The 11th guideline emphasizes the assessment of patients with Upper Tract Urothelial Carcinoma (UTUC) for risk of Chronic Kidney Disease (CKD) or dialysis following nephroureterectomy (NU). This process is crucial for operative decision-making and systemic therapy administration. An estimated glomerular filtration rate (eGFR) should be obtained before surgery. If hydronephrosis or renal atrophy is present, these may affect accurate preoperative renal function estimation. Hence, renal decompression may be needed. Considerations for postoperative renal function, especially for patients needing perioperative systemic treatment, must be taken into account. Regular monitoring and management of CKD risks, such as Diabetes Mellitus (DM) and hypertension, are also crucial.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

What tests are recommended to assess renal function before surgery in UTUC patients?
a) Serum creatinine
b) Differential renal scan
c) CT volumetric studies
d) All of the above

A

d) All of the above. All these tests are recommended to assess the renal function before surgery.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

What factors could potentially cause a false under-estimate or overestimate of renal function in UTUC patients prior to surgery?
a) Hydronephrosis
b) Renal atrophy
c) Both a and b
d) None of the above

A

c) Both a and b. Hydronephrosis may falsely under-estimate, and renal atrophy may overestimate the preoperative renal function.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

Why is it crucial to assess patients with UTUC for the risk of post-NU CKD or dialysis prior to surgery?

A

Assessing UTUC patients for the risk of post-NU CKD or dialysis is crucial because the operative approach and administration of systemic therapy are decided based on the patient’s baseline renal function and their estimated post-operative glomerular filtration rate (eGFR). Further, since the postoperative renal function can diminish due to the loss of a renal unit in NU, this can exacerbate CKD and affect a patient’s eligibility to receive adjuvant chemotherapy. Thus, an accurate preoperative assessment helps in planning the treatment strategy and managing the risks effectively.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

Explain the importance of optimizing glycemic and blood pressure control in the management of postoperative renal function in UTUC patients.

A

Optimizing glycemic and blood pressure control is essential in managing postoperative renal function in UTUC patients because it helps reduce the degree of renal dysfunction in the perioperative period. Particularly in patients with diabetes mellitus, maintaining good glycemic control can prevent the risk of acute kidney injury. Similarly, effective blood pressure control can mitigate the risk of hypertensive nephropathy. These measures, along with smoking cessation and minimizing the risk of acute kidney injury, can significantly contribute to preserving renal function and managing CKD in the postoperative period.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

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)

A

The 12th guideline underlines the importance of informing patients about the short- and long-term risks associated with diagnostic and therapeutic options for Upper Tract Urothelial Carcinoma (UTUC). These risks include the need for endoscopic follow-up, possible complications like significant strictures, surgical treatment toxicities, and side effects from neoadjuvant and adjuvant therapies. Urothelial recurrences are common in UTUC management, necessitating long-term surveillance and possible additional treatments. Ablative therapies, while providing local control and durable long-term kidney-sparing outcomes, come with added endoscopic surveillance requirements and associated risks such as stricture and infection. Treatment options such as chemoablative treatment with reverse thermo-hydrogel preparation of mitomycin carry specific risks and require appropriate patient counseling.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

Why is long-term surveillance necessary in the management of UTUC?
a) To monitor the effectiveness of the therapy
b) Due to the common occurrence of urothelial recurrences
c) To check for possible complications from therapies
d) All of the above

A

d) All of the above. Long-term surveillance is necessary to monitor the effectiveness of the therapy, due to the common occurrence of urothelial recurrences, and to check for possible complications from therapies.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

What are the risks associated with ablative therapies in UTUC treatment?
a) Need for additional endoscopic surveillance
b) Risk of significant stricture formation
c) Risk of infection
d) All of the above

A

d) All of the above. Ablative therapies for UTUC can lead to a need for additional endoscopic surveillance, risk of significant stricture formation, and the risk of infection.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

What is the importance of informing patients about the short- and long-term risks associated with diagnostic and therapeutic options for UTUC?

A

Informing patients about the short- and long-term risks associated with diagnostic and therapeutic options for UTUC is a crucial part of care as it enables the patient to understand the potential consequences and complications of the treatment. It allows the patients to make informed decisions about their treatment options, prepare for possible long-term surveillance and additional treatments, and manage their expectations. Additionally, this information helps patients to adhere to the treatment plan and encourages them to promptly report any side effects or complications, which contributes to better overall management of UTUC.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

What are the specific risks associated with the use of chemoablative treatment with reverse thermo-hydrogel preparation of mitomycin for pyelocaliceal instillation for low-grade tumors?

A

The specific risks associated with the use of chemoablative treatment with reverse thermo-hydrogel preparation of mitomycin for pyelocaliceal instillation for low-grade tumors include a high risk of ureteral obstruction (over 44%), potential bone marrow suppression, and embryo-fetal toxicity. These risks necessitate specific patient counseling and careful monitoring of the patient’s condition during the treatment.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

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)

A

The 13th guideline states that tumor ablation should be the initial management option for patients with low-risk (LR) favorable Upper Tract Urothelial Carcinoma (UTUC) due to low rates of metastatic progression. Endoscopic management is recommended as the first-line treatment when feasible. Observational studies have shown comparable cancer-specific survival (CSS) and improved renal functional outcomes with endoscopic ablation compared to nephroureterectomy (NU). However, there is strong case-selection bias, and outcomes should be interpreted within this context. There have been instances where complete endoscopic ablation may not be feasible, and chemoablation can serve as an alternative. The benefits of chemoablation, however, need to be balanced against the risk of potential ureteral stricture. Whenever possible, complete endoscopic ablation is preferred over chemoablation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

Why is tumor ablation the initial management option for patients with low-risk UTUC?
a) It has lower rates of metastatic progression
b) It preserves kidney function
c) It has a lower risk of ureteral stricture
d) Both a and b

A

d) Both a and b. Tumor ablation is the initial management option because it is associated with lower rates of metastatic progression and helps preserve kidney function.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

When is chemoablation considered in the management of UTUC?
a) When complete endoscopic ablation is not feasible
b) When tumor is at high risk
c) In all patients with UTUC
d) When tumor is low grade

A

a) When complete endoscopic ablation is not feasible. Chemoablation is considered when specific tumor (location and focality) and patient factors (age, comorbidities, baseline renal function, procedural risk) make complete endoscopic ablation unfeasible.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

How does the effectiveness of endoscopic management compare with nephroureterectomy (NU) in treating patients with low-risk UTUC?

A

Observational studies have found endoscopic management to have similar cancer-specific survival (CSS) outcomes as nephroureterectomy (NU) in patients with low-risk UTUC. Additionally, endoscopic management has been shown to lead to improved renal functional outcomes compared to NU. However, these findings must be interpreted in the context of strong case-selection bias inherent in retrospective studies. Therefore, while the endoscopic approach can be advantageous, especially in terms of preserving renal function, the decision between endoscopic management and NU must be made on a case-by-case basis.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

What are some potential drawbacks of using chemoablation in the management of UTUC, and when should it be considered?

A

The primary drawback of using chemoablation is the potential risk of ureteral stricture. However, it can be considered in situations where complete endoscopic ablation is not feasible due to specific tumor characteristics (such as location and focality) or patient factors (like age, comorbidities, baseline renal function, and procedural risk). Despite these drawbacks, chemoablation can be a beneficial treatment alternative in certain clinical scenarios. But it’s important to note that chemoablation should not be used as a substitute for complete endoscopic ablation when the latter is feasible.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

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)

A

The 14th guideline proposes that tumor ablation may be the initial management option offered to patients with low-risk (LR) unfavorable UTUC and select patients with high-risk (HR) favorable disease with low-volume tumors or those who cannot undergo radical nephroureterectomy (RNU). There’s no high-quality evidence comparing endoscopic management versus RNU for these patients. Nonetheless, comparable cancer-specific survival and improved renal functional outcomes have been reported for endoscopic management compared to RNU. Certain considerations need to be factored, such as tumor size. Tumors less than 1.5 cm may be optimal for endoscopic ablation due to a lower risk of invasive disease. However, larger tumors (≥ 1.5 cm) could also be considered for ablation based on provider’s experience and assessment for kidney sparing surgery. For LR unfavorable disease showing progression in tumor size, focality, or grade, further endoscopic-assisted attempts are discouraged, and definitive resection via segmental ureterectomy (SU) or NU is recommended.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

Tumor ablation may be considered the initial management option for which types of UTUC patients?
a) Patients with LR unfavorable UTUC
b) Select patients with HR favorable disease with low-volume tumors
c) Patients who cannot undergo RNU
d) All of the above

A

d) All of the above. Tumor ablation can be considered for patients with LR unfavorable UTUC, select patients with HR favorable disease with low-volume tumors, and patients who cannot undergo RNU.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

When might larger tumors (≥ 1.5 cm) be considered for ablation?
a) When the tumor is at a low-risk stage
b) When the provider’s experience supports it and there is a need for kidney sparing surgery
c) When the tumor is high-grade
d) In all cases

A

b) When the provider’s experience supports it and there is a need for kidney sparing surgery. Larger tumors may be considered for ablation based on provider’s experience and assessment of the need for kidney sparing surgery.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

What are the considerations when deciding the initial management for LR unfavorable or HR favorable UTUC?

A

Several factors need to be considered when deciding the initial management for LR unfavorable or HR favorable UTUC. These include the size of the tumor, the volume of the tumor, and the patient’s ability to undergo radical nephroureterectomy (RNU). Tumors less than 1.5 cm in size may be more optimal for endoscopic ablation due to a lower risk of invasive disease. However, larger tumors (greater or equal to 1.5 cm) may also be considered for ablation based on the provider’s experience and the need for kidney-sparing surgery. For patients with LR unfavorable disease showing progression in size, focality, or grade, further endoscopic attempts are discouraged, and definitive resection via segmental ureterectomy (SU) or NU is recommended.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

What should be done if there is progression in tumor size, focality, or grade in patients with LR unfavorable disease?

A

For patients with low-risk unfavorable disease who demonstrate progression in tumor size, focality, or grade, further endoscopic-assisted attempts are discouraged. Instead, consideration should be given to definitive resection via segmental ureterectomy (SU) or radical nephroureterectomy (NU). This decision must be made based on the individual patient’s circumstances, the extent of disease progression, and the potential benefits and risks of the different surgical approaches.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

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)

A

Guideline 15 emphasizes that tumor ablation in UTUC can be accomplished via either a retrograde or antegrade percutaneous approach, and it’s crucial to repeat endoscopic evaluation within three months. Retrograde approaches, including ureteroscopy with pyeloscopy, are common, while percutaneous techniques are typically reserved for larger tumors, those difficult to access in a retrograde fashion, or in patients who have undergone prior radical cystectomy or urinary diversion. The energy source for ablation can vary, including thulium laser, holmium laser, Neodymium (Nd:YAG), electrocautery devices, or chemoablation. Using a ureteral access sheath during ureteroscopic ablation can provide advantages, such as allowing repeated scope passage and managing irrigation solutions. Because UTUC has a high chance of recurrence and residual disease after the first ablation, repeat endoscopic evaluation should take place within three months, with a 30-day window on either side of this period. Clinicians may opt for shorter intervals for more challenging cases.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

When is a percutaneous approach to tumor ablation generally reserved?
a) For smaller tumors
b) For tumors that are easy to access in a retrograde fashion
c) For patients who have undergone prior radical cystectomy or urinary diversion
d) For tumors with low recurrence risk

A

c) For patients who have undergone prior radical cystectomy or urinary diversion. Percutaneous techniques are typically reserved for larger tumors, those difficult to access in a retrograde fashion, or in patients who have undergone prior radical cystectomy or urinary diversion.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
85
Q

What is one advantage of using a ureteral access sheath during ureteroscopic ablation?
a) Increases the risk of intravesical recurrence
b) Allows for repeated scope passage up and down the ureter
c) Allows for excess pelvicalyceal hydrostatic pressure from irrigation solutions
d) Reduces the need for repeat endoscopic evaluation

A

b) Allows for repeated scope passage up and down the ureter. The use of a ureteral access sheath allows for repeated scope passage up and down the ureter and provides a means of fluid egress from the upper tract to avoid excess pelvicalyceal hydrostatic pressure from irrigation solutions.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
86
Q

What are the considerations for the choice of approach and energy source for tumor ablation in UTUC?

A

The choice of approach for tumor ablation in UTUC depends on the size and accessibility of the tumor and the patient’s past surgical history. Retrograde approaches, including ureteroscopy with pyeloscopy, are commonly used, while percutaneous techniques are usually reserved for larger tumors, those difficult to access in a retrograde fashion, or in patients who have undergone prior radical cystectomy or urinary diversion. The choice of energy source for ablation can vary based on the availability of instrumentation and tumor characteristics. These can include the thulium laser, holmium laser, Neodymium (Nd:YAG), and electrocautery devices. Additionally, chemoablation can be employed either through retrograde ureteral catheter instillation or percutaneous access with fluoroscopic imaging guidance.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

Why is it recommended to perform a repeat endoscopic evaluation within three months of initial treatment?

A

The recommendation to perform a repeat endoscopic evaluation within three months of the initial treatment is due to the propensity of UTUC to recur and for residual disease to remain after the first ablation. Optimal timing of follow-up endoscopic evaluation has not been definitively established, but several factors such as tumor size, visualization, access, treatment efficacy, etc., could influence this decision. A 30-day window on either side of the three-month period is justified to allow for timely identification of recurrences. Especially for more challenging cases or when incomplete treatment is a possibility, clinicians may opt for a more conservative approach with shorter interval endoscopic diagnostic and therapeutic procedures.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

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.

A

Guideline 16 advocates for the instillation of adjuvant pelvicalyceal chemotherapy or intravesical chemotherapy following the ablation of UTUC tumors, provided there is no bladder or upper tract perforation. This recommendation is aimed at decreasing the risk of urothelial cancer recurrence. The principle of immediate instillation of chemotherapy after endoscopic tumor ablation for UTUC comes from extrapolation of data related to urothelial carcinoma of the lower tract. Despite limited compelling evidence, some studies show a trend towards improved urothelial recurrence-free survival with the use of chemotherapy post-ablation. Technical aspects are still not fully established, but the guideline considers three approaches to be acceptable: 1) antegrade perfusion by nephrostomy tube, 2) retrograde perfusion via ureteral catheter, and 3) bladder instillation by transurethral catheter with reflux via a double J ureteral stent. While bacillus Calmette-Guerin (BCG) is the primary topical therapy for UTUC, other agents have been described including mitomycin c, gemcitabine, docetaxel, epirubicin, adriamycin, thiotepa, and BCG with interferon.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
89
Q

After UTUC tumor ablation, why might clinicians instill adjuvant pelvicalyceal or intravesical chemotherapy?
a) To facilitate wound healing
b) To decrease the risk of urothelial cancer recurrence
c) To reduce the risk of post-operative infection
d) To manage post-operative pain

A

b) To decrease the risk of urothelial cancer recurrence. The purpose of instilling adjuvant pelvicalyceal or intravesical chemotherapy after UTUC tumor ablation is to reduce the risk of urothelial cancer recurrence.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
90
Q

What are the three approaches to chemotherapy instillation considered acceptable by the Panel following UTUC tumor ablation?
a) Antegrade perfusion by nephrostomy tube, retrograde perfusion via ureteral catheter, and bladder instillation by transurethral catheter with reflux via a double J ureteral stent
b) Oral chemotherapy, retrograde perfusion via ureteral catheter, and bladder instillation by transurethral catheter with reflux via a double J ureteral stent
c) Antegrade perfusion by nephrostomy tube, oral chemotherapy, and bladder instillation by transurethral catheter with reflux via a double J ureteral stent
d) Antegrade perfusion by nephrostomy tube, retrograde perfusion via ureteral catheter, and intravenous chemotherapy

A

a) Antegrade perfusion by nephrostomy tube, retrograde perfusion via ureteral catheter, and bladder instillation by transurethral catheter with reflux via a double J ureteral stent. These are the three approaches considered acceptable for the administration of chemotherapy following UTUC tumor ablation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
91
Q

What is the purpose of instilling adjuvant pelvicalyceal or intravesical chemotherapy following UTUC tumor ablation and what evidence supports its use?

A

The purpose of instilling adjuvant pelvicalyceal or intravesical chemotherapy following UTUC tumor ablation is to decrease the risk of urothelial cancer recurrence. The recommendation is largely based on extrapolation of data from the management of urothelial carcinoma of the lower tract, where an immediate instillation of intravesical chemotherapy after transurethral resection has been shown to reduce the rate of intravesical tumor recurrence. Some small studies specifically investigating UTUC have shown a trend towards improved urothelial recurrence-free survival with the use of chemotherapy post-ablation.

92
Q

Describe the three approaches to chemotherapy instillation considered acceptable by the Panel following UTUC tumor ablation.

A

The three approaches to chemotherapy instillation considered acceptable by the Panel following UTUC tumor ablation are: 1) Antegrade perfusion by nephrostomy tube, where the chemotherapy is instilled directly into the kidney via a tube; 2) Retrograde perfusion via a ureteral catheter, where the chemotherapy is instilled upwards through the ureter into the kidney; 3) Bladder instillation by transurethral catheter with reflux via a double J ureteral stent, where the chemotherapy is instilled into the bladder and then allowed to reflux back into the kidneys via a stent.

93
Q

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)

A

Guideline 17 suggests that pelvicalyceal therapy with Bacillus Calmette-Guerin (BCG) can be offered to patients with high-risk but favorable Upper Tract Urothelial Carcinoma (UTUC) after complete tumor ablation, or to patients with upper tract carcinoma in situ (CIS). This is particularly beneficial for patients with imperative indications such as solitary kidney status, bilateral UTUC, or the risk of progression to end-stage renal disease. However, there is limited literature specifically addressing the treatment of such UTUC with topical therapy like BCG. The outcomes of studies vary widely, with complete response rates ranging from 41% to 100%, and adverse effects reported in 0% to 92% of patients. Systematic reviews indicate that the treatment of UTUC Ta/T1 disease with BCG yields a cancer-specific survival and overall survival rate of 94% and 71% respectively. Furthermore, there seems to be no significant differences in outcomes when other therapeutic agents are used.

94
Q

Why might pelvicalyceal therapy with BCG be offered to patients with high-risk UTUC?
a) To promote rapid healing post-ablation
b) To prevent post-ablation infection
c) To manage symptoms of UTUC
d) To reduce the risk of UTUC recurrence post-ablation

A

d) To reduce the risk of UTUC recurrence post-ablation. Bacillus Calmette-Guerin (BCG) therapy may be offered to patients with high-risk but favorable UTUC after complete tumor ablation to reduce the risk of UTUC recurrence.

95
Q

Why is BCG therapy considered for patients with high-risk but favorable UTUC and what are the reported outcomes?

A

BCG therapy is considered for patients with high-risk but favorable UTUC following complete tumor ablation to reduce the risk of UTUC recurrence. This therapy is particularly beneficial for patients with solitary kidney status, bilateral UTUC, or the risk of progression to end-stage renal disease. However, outcomes from the studies are highly variable, with complete response rates ranging from 41% to 100%. Adverse effects have been reported in 0% to 92% of patients, including complications such as cystitis, fever, sepsis, renal tuberculosis, ureteral stricture, and pericarditis.

96
Q

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)

A

Guideline 18 suggests that when tumor ablation is not feasible or evidence of risk group progression is identified in patients with low-risk Upper Tract Urothelial Carcinoma (UTUC), surgical resection of all involved sites, either by radical nephroureterectomy (RNU) or segmental resection of the ureter, should be offered. This is due to the potential risk of cancer progression from curable to an incurable form of UTUC. Any signs of a change in tumor growth pattern toward a more aggressive subtype should lead to a reconsideration of the management strategy. Studies have shown mixed results when comparing endoscopic management versus RNU. Despite these disparities, clear communication with patients is essential to manage their expectations and make a shared decision about their treatment plan.

97
Q

What should be done when tumor ablation is not feasible or evidence of risk group progression is identified in patients with low-risk UTUC?
a) Continue with conservative strategies and closely monitor the patient
b) Initiate systemic chemotherapy immediately
c) Perform surgical resection of all involved sites
d) Initiate radiation therapy

A

c) Perform surgical resection of all involved sites. If tumor ablation is not feasible or if there’s evidence of progression from low-risk UTUC, surgical resection of all involved sites should be considered to avoid the risk of cancer progression to an incurable state.

98
Q

What is a potential risk if conservative strategies for kidney preservation fail in UTUC?
a) Risk of kidney failure
b) Risk of progression to incurable UTUC
c) Risk of metastasis to the brain
d) Risk of kidney stone formation

A

b) Risk of progression to incurable UTUC. If conservative strategies for kidney preservation in UTUC fail, there’s a potential risk of cancer progression from a curable to an incurable form of UTUC.

99
Q

What should be done if signs of a change in tumor growth pattern toward a more aggressive subtype are observed in a patient with low-grade UTUC?

A

If a more aggressive tumor growth pattern is observed in a patient with low-grade UTUC, the management strategy should be reassessed. This could prompt a consideration for a more definitive treatment with extirpative surgical resection, either by radical nephroureterectomy (RNU) or segmental resection of the ureter. The observed findings and their clinical significance suggesting a shift in disease risk should be thoroughly discussed with the patient, and a decision for the change in strategy should be made collaboratively.

100
Q

What were the reported cancer-specific survival and overall survival rates following the treatment of UTUC Ta/T1 disease with BCG, according to the meta-analysis conducted by Foerster et al.?
a) 70% and 90% respectively
b) 85% and 75% respectively
c) 94% and 71% respectively
d) 100% and 80% respectively

A

c) 94% and 71% respectively. According to the meta-analysis by Foerster et al., the cancer-specific survival and overall survival rates following the treatment of UTUC Ta/T1 disease with BCG were 94% and 71% respectively.

101
Q

What were the reported cancer-specific survival and overall survival rates following the treatment of UTUC Ta/T1 disease with BCG, according to the meta-analysis conducted by Foerster et al.?
a) 70% and 90% respectively
b) 85% and 75% respectively
c) 94% and 71% respectively
d) 100% and 80% respectively

A

c) 94% and 71% respectively. According to the meta-analysis by Foerster et al., the cancer-specific survival and overall survival rates following the treatment of UTUC Ta/T1 disease with BCG were 94% and 71% respectively.

102
Q

How have studies compared endoscopic management versus radical nephroureterectomy (RNU) in different risk groups of UTUC?

A

Studies comparing endoscopic management versus RNU in different risk groups of UTUC have shown mixed results. For instance, one study found that endoscopic management was associated with an increased risk of any local, intravesical, or distant recurrence, while another study found endoscopic management associated with improved intravesical recurrence-free survival. Yet another study found endoscopic management associated with an increased risk of local recurrence but no difference in the risk of intravesical recurrence-free survival. Regarding cancer-specific survival or all-cause mortality, some studies have found endoscopic management to be associated with worse outcomes. These mixed findings underscore the importance of vigilance in follow-up and recognizing clinical signs indicating thresholds for recommending altering care.

103
Q

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)

A

Guideline 19 suggests that clinicians may offer watchful waiting or surveillance alone to selected patients with Upper Tract Urothelial Carcinoma (UTUC) who have significant comorbidities, competing risks of mortality, or at significant risk of End-Stage Renal Disease (ESRD) with any intervention resulting in dialysis. These are patients for whom the risk of treatment-related complications may outweigh the potential benefits. Rates of complications following radical nephroureterectomy (RNU) range from 15% to 50%, including a 30-day mortality risk of 1%. Discussions should involve treatment-related risks, including perioperative mortality, leading to a shared decision for active surveillance or watchful waiting/expectant management. However, patients and families should be prepared for potential disease-related events like bleeding, obstruction, infection, and pain. Studies comparing non-surgical management versus surgery for UTUC have generally found worse overall survival (OS) associated with non-surgical management, likely due to the compromised medical condition of these patients.

104
Q

Who could be considered for watchful waiting or surveillance alone in the context of UTUC?
a) Patients with no comorbidities and low-risk UTUC
b) Patients with significant comorbidities, competing risks of mortality, or at significant risk of End-Stage Renal Disease (ESRD) with any intervention resulting in dialysis
c) Young patients with high-risk UTUC
d) All UTUC patients regardless of risk group

A

b) Patients with significant comorbidities, competing risks of mortality, or at significant risk of End-Stage Renal Disease (ESRD) with any intervention resulting in dialysis.

105
Q

What are some of the potential complications of radical nephroureterectomy (RNU) in patients with UTUC?
a) Severe allergic reactions
b) Risk of viral infections
c) Bleeding, obstruction, infection, and pain
d) Sudden hearing loss

A

c) Bleeding, obstruction, infection, and pain.

106
Q

What were the reported cancer-specific survival and overall survival rates following the treatment of UTUC Ta/T1 disease with BCG, according to the meta-analysis conducted by Foerster et al.?
a) 70% and 90% respectively
b) 85% and 75% respectively
c) 94% and 71% respectively
d) 100% and 80% respectively

A

c) 94% and 71% respectively. According to the meta-analysis by Foerster et al., the cancer-specific survival and overall survival rates following the treatment of UTUC Ta/T1 disease with BCG were 94% and 71% respectively.

107
Q

What were the reported cancer-specific survival and overall survival rates following the treatment of UTUC Ta/T1 disease with BCG, according to the meta-analysis conducted by Foerster et al.?
a) 70% and 90% respectively
b) 85% and 75% respectively
c) 94% and 71% respectively
d) 100% and 80% respectively

A

c) 94% and 71% respectively. According to the meta-analysis by Foerster et al., the cancer-specific survival and overall survival rates following the treatment of UTUC Ta/T1 disease with BCG were 94% and 71% respectively.

108
Q

What are the outcomes of non-surgical management versus surgical treatment for UTUC based on large database studies?

A

Studies utilizing large databases have shown that non-surgical management is generally associated with worse overall survival (OS) compared to surgical treatment for UTUC. For example, an analysis of the Surveillance, Epidemiology, and End Results (SEER) database found that non-surgical management was associated with worse OS and 3-year cancer-specific survival (CSS). Similarly, a study using the National Cancer Database found non-surgical management to be associated with worse OS. These results, however, likely reflect the compromised medical condition of the patients who opt for non-surgical management.

109
Q

GUIDELINE STATEMENT 20
Clinicians should recommend RNU or SU for surgically eligible patients with HR UTUC. (Strong Recommendation; Evidence Level: Grade B)

A

Guideline 20 suggests that clinicians should recommend radical nephroureterectomy (RNU) or segmental ureterectomy (SU) for surgically eligible patients with high-risk UTUC. RNU with complete bladder cuff excision (BCE) and lymphadenectomy is the standard of care for patients with high-risk UTUC. Oncological principles should be maintained whether an open, robotic, or laparoscopic approach is used. Minimally invasive approaches are favored for most patients as they offer shorter lengths of stay and fewer complications. However, open surgical approaches might be preferred for large, bulky UTUC with clinical evidence for direct invasion to adjacent structures.

Complete BCE is important, as studies have shown worse outcomes for patients who did not receive it. Distal ureterectomy or SU are reasonable alternatives to RNU for well-selected patients. The best candidates for distal ureterectomy have ureteral tumors in the lower third of the ureter and a mobile bladder with sufficient capacity for reimplantation. Those suitable for SU have small, unifocal tumors (typically 1 cm or smaller) isolated to a short segment of the proximal or mid-ureter.

110
Q

What is the standard of care for patients with high-risk UTUC?
a) Palliative care only
b) Radical nephroureterectomy (RNU) with complete bladder cuff excision (BCE) and lymphadenectomy
c) Chemotherapy only
d) Radiation therapy only

A

b) Radical nephroureterectomy (RNU) with complete bladder cuff excision (BCE) and lymphadenectomy.

111
Q

What are the potential benefits of minimally invasive approaches for RNU?
a) Longer hospital stay
b) Higher recurrence rates
c) Shorter length of stay and fewer complications
d) Longer recovery times

A

c) Shorter length of stay and fewer complications.

112
Q

What are the basic principles to be maintained during a radical nephroureterectomy (RNU)?

A

The basic principles of an RNU include complete excision of the ipsilateral upper tract urothelium, including the intramural portion of the ureter and ureteral orifice with negative margins. It’s also crucial to avoid urinary spillage by, for example, early low ligation of the ureter, to minimize the risk of seeding urothelial cancer outside the urinary tract. The RNU specimen should be removed en bloc whenever technically feasible. These principles must be adhered to regardless of whether an open, robotic, or laparoscopic approach is used.

113
Q

Which patients may be suitable for segmental ureterectomy (SU) as an alternative to RNU?

A

The most suitable patients for SU are those with small, unifocal tumors (typically 1 cm or smaller) that are isolated to a short segment of the proximal or mid-ureter. The requirement is that the resection involves 2 cm or less of ureteral length to allow for primary ureteroureterostomy. Longer sections of ureteral involvement and resection may require more complex reconstruction techniques when kidney sparing is desired.

114
Q

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)

A

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)

115
Q

Which approach is preferred for surgically eligible patients with high-risk and unfavorable low-risk cancers that are endoscopically confirmed as confined to the lower ureter in a functional renal unit?
a) Endoscopic assisted tumor ablation
b) Distal ureterectomy and ureteral reimplantation
c) Watchful waiting or surveillance
d) Topical therapies such as BCG

A

b) Distal ureterectomy and ureteral reimplantation

116
Q

What are the potential drawbacks of endoscopic assisted tumor ablation for tumors confined to the lower ureter?
a) Requires multiple additional procedures
b) Does not preserve kidney function
c) Carries a high risk for upper tract tumor recurrence
d) Both a) and c)

A

d) Both a) and c)

117
Q

Why is distal ureterectomy and ureteral reimplantation the preferred treatment for localized cancers in the lower ureter?

A

Distal ureterectomy and ureteral reimplantation is preferred because it offers definitive curative management for tumors confined to the lower ureter while preserving kidney function. It is ideal for patients with localized cancers in this location who have an increased risk of disease recurrence and progression.

118
Q

How can carcinoma in situ (CIS) limited to the region within the ureteral orifice be managed?

A

Carcinoma in situ (CIS) that is limited to the region within the ureteral orifice can be managed with topical therapies such as BCG. Additionally, transurethral resection of the transmural portion of the ureter can be used for very distal tumors, when the tumor is limited to the region inside the ureteral orifice and not beyond the bladder wall, thus it can be anatomically managed as bladder cancer.

119
Q

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)

A

Guideline 22 advises that when performing nephroureterectomy (NU) or distal ureterectomy, the entire distal ureter, including the intramural ureteral tunnel and ureteral orifice, should be excised. Moreover, the urinary tract should be closed in a watertight manner to avoid urinary extravasation, facilitate more rapid catheter removal, and allow for instillation of intravesical adjuvant chemotherapy in the perioperative setting. Various surgical approaches have been described for managing the ureteral orifice during these procedures, but there are insufficient data to recommend one over the other. Retrospective observational studies have shown that formal bladder cuff excision (BCE) is associated with improved survival rates compared to no BCE. No randomized controlled trial has compared the different surgical techniques for managing the distal ureter and ureteral orifice during NU or distal ureterectomy for upper tract urothelial carcinoma (UTUC).

120
Q

What is the recommendation for managing the ureteral orifice during nephroureterectomy or distal ureterectomy?
a) Partial excision of the distal ureter
b) Excision of the entire distal ureter, including the intramural ureteral tunnel and ureteral orifice
c) Excision of only the ureteral orifice
d) No specific recommendation due to insufficient data

A

b) Excision of the entire distal ureter, including the intramural ureteral tunnel and ureteral orifice

121
Q

What is the benefit of closing the urinary tract in a watertight fashion during nephroureterectomy or distal ureterectomy?
a) It minimizes the risk of urinary extravasation
b) It facilitates more rapid catheter removal
c) It permits instillation of intravesical adjuvant chemotherapy in the perioperative setting
d) All of the above

A

d) All of the above

122
Q

What were the reported cancer-specific survival and overall survival rates following the treatment of UTUC Ta/T1 disease with BCG, according to the meta-analysis conducted by Foerster et al.?
a) 70% and 90% respectively
b) 85% and 75% respectively
c) 94% and 71% respectively
d) 100% and 80% respectively

A

c) 94% and 71% respectively. According to the meta-analysis by Foerster et al., the cancer-specific survival and overall survival rates following the treatment of UTUC Ta/T1 disease with BCG were 94% and 71% respectively.

123
Q

d) All of the above

A
124
Q

What have retrospective observational studies suggested about the benefits of formal bladder cuff excision (BCE) during nephroureterectomy or distal ureterectomy?

A

Retrospective observational studies have shown that formal bladder cuff excision (BCE) is associated with improved 5-year overall survival rates compared to no BCE in patients undergoing these procedures for upper tract urothelial carcinoma.

125
Q

Why is the urinary tract recommended to be closed in a watertight fashion during nephroureterectomy or distal ureterectomy?

A

Closing the urinary tract in a watertight fashion during these procedures is recommended to avoid urinary extravasation, facilitate more rapid catheter removal, and permit the instillation of intravesical adjuvant chemotherapy in the perioperative setting.

126
Q

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)

A

Guideline 23 recommends administering a single dose of perioperative intravesical chemotherapy to patients undergoing radical nephroureterectomy (RNU) or segmental ureterectomy (SU), including distal ureterectomy, for upper tract urothelial carcinoma (UTUC). This is done to reduce the risk of bladder recurrence. Two prospective randomized controlled trials (RCTs) have shown that a single instillation of intravesical chemotherapy around the time of NU reduces the risk of subsequent bladder recurrence of urothelial carcinoma. Although there’s little data to support one intravesical chemotherapeutic over another, many clinicians have started using gemcitabine due to compelling evidence supporting its use and concerns about potential chemical peritonitis from extravesical extravasation of mitomycin-C.

127
Q

Why is a single dose of perioperative intravesical chemotherapy recommended in patients undergoing RNU or SU for UTUC?
a) To increase survival rates
b) To reduce the risk of bladder recurrence
c) To minimize complications during surgery
d) To reduce the risk of kidney disease

A

b) To reduce the risk of bladder recurrence

128
Q

Which agent is increasingly being favored by many clinicians for intravesical chemotherapy?
a) Mitomycin-C
b) Gemcitabine
c) Cisplatin
d) Doxorubicin

A

b) Gemcitabine

129
Q

What evidence supports the use of a single dose of intravesical chemotherapy around the time of RNU to reduce the risk of subsequent bladder recurrence?

A

The evidence comes from two prospective randomized controlled trials (RCTs). The ODMIT-C trial by O’Brien et al. showed that 17% of patients who received a single post-operative intravesical dose of mitomycin-C developed a bladder recurrence in the first year, compared to 27% in the standard treatment arm. Another phase II trial by Ito et al. showed similar results with a single intravesical instillation of pirarubicin.

130
Q

What has led to a shift towards the use of gemcitabine over mitomycin-C for intravesical chemotherapy?

A

There are compelling data supporting the use of a single dose of intravesical gemcitabine at the time of trans urethral resection of bladder tumor to reduce the rate of intravesical recurrences. Additionally, there are concerns about potential chemical peritonitis if there is extravesical extravasation of mitomycin-C. These factors have led many clinicians to prefer gemcitabine over mitomycin-C.

131
Q

GUIDELINE STATEMENT 24
For patients with LR UTUC, clinicians may perform LND at time of NU or ureterectomy. (Conditional Recommendation; Evidence Level: Grade C)

A

Guideline 24 suggests that clinicians may perform a lymph node dissection (LND) at the time of nephroureterectomy (NU) or ureterectomy in patients with low-risk (LR) upper tract urothelial carcinoma (UTUC). However, the evidence supporting this approach is limited. No randomized controlled trials (RCTs) have compared LND versus no LND concerning oncologic outcomes. Two recent systematic reviews of observational studies also showed no significant differences in oncologic outcomes whether LND was performed or not. Given these findings, the benefit of LND among patients with LR UTUC remains unclear. Thus, the decision to perform LND may be left to the clinician’s discretion based on clinically or radiographically suspicious regional lymphadenopathy or other intraoperative findings suggesting more advanced disease.

132
Q

According to Guideline 24, when should clinicians consider performing a lymph node dissection (LND) in patients with low-risk UTUC?
a) In all cases of nephroureterectomy (NU) or ureterectomy
b) Only when there is a high likelihood of disease progression
c) When there are clinically or radiographically suspicious regional lymphadenopathy or other intraoperative findings suggesting more advanced disease
d) LND should not be performed in cases of low-risk UTUC

A

c) When there are clinically or radiographically suspicious regional lymphadenopathy or other intraoperative findings suggesting more advanced disease

133
Q

What is the current state of evidence concerning the impact of LND on oncologic outcomes in patients with low-risk UTUC?
a) There is strong evidence from randomized controlled trials suggesting significant benefits of LND
b) Observational studies show significant improvements in oncologic outcomes with LND
c) The evidence is limited, with no randomized controlled trials available, and observational studies showing no significant differences in oncologic outcomes
d) There is strong evidence suggesting LND has a detrimental effect on oncologic outcomes

A

c) The evidence is limited, with no randomized controlled trials available, and observational studies showing no significant differences in oncologic outcomes

134
Q

What does Guideline 24 recommend regarding lymph node dissection (LND) in patients with low-risk upper tract urothelial carcinoma (UTUC)?

A

Guideline 24 suggests that for patients with low-risk UTUC, clinicians may perform lymph node dissection (LND) at the time of nephroureterectomy (NU) or ureterectomy. However, the decision to perform LND should be based on clinically or radiographically suspicious regional lymphadenopathy or other intraoperative findings suggesting more advanced disease.

135
Q

What is the evidence supporting lymph node dissection (LND) in patients with low-risk upper tract urothelial carcinoma (UTUC)?

A

The evidence supporting LND in patients with low-risk UTUC is limited. No randomized controlled trials have compared LND versus no LND in terms of oncologic outcomes. Also, two recent systematic reviews of observational studies did not find statistically significant differences in oncologic outcomes between patients who had LND and those who did not. Therefore, the benefit of LND among patients with low-risk UTUC is unclear.

136
Q

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)

A

Guideline 25 advises that for patients with high-risk (HR) upper tract urothelial carcinoma (UTUC), clinicians should perform a lymph node dissection (LND) at the time of nephroureterectomy (NU) or ureterectomy. Although there are no randomized controlled trials (RCTs) that evaluate the effect of LND on oncologic outcomes, the panel conducted a re-analysis of some studies described in the systematic review. The results indicate an association between LND and better recurrence-free survival (RFS). Two additional recent cohort studies also found LND was associated with improved overall survival (OS) and cancer-specific survival (CSS), especially in patients with T3 and T4 tumors. However, there’s no consensus yet on the appropriate template to yield maximal oncologic outcomes and prognostic information.

137
Q

FIGURE 1: REANALYSIS OF RECURRENCE-FREE SURVIVAL FROM CHAN 2020 SYSTEMATIC REVIEW

A
138
Q

What is the recommendation for patients with high-risk UTUC regarding LND?
a) LND should not be performed at the time of NU or ureterectomy
b) LND should be performed at the time of NU or ureterectomy
c) LND may be performed at the time of NU or ureterectomy
d) LND should only be performed if there are additional risk factors present

A

b) LND should be performed at the time of NU or ureterectomy

139
Q

Based on the findings in the re-analysis of some studies, what was the association between LND and oncologic outcomes in high-risk UTUC patients?
a) LND was associated with worse oncologic outcomes
b) LND was not associated with any significant changes in oncologic outcomes
c) LND was associated with better oncologic outcomes, specifically better recurrence-free survival
d) There was no clear association between LND and oncologic outcomes

A

c) LND was associated with better oncologic outcomes, specifically better recurrence-free survival

140
Q

What is the recommendation given in Guideline 25 for patients with high-risk upper tract urothelial carcinoma (UTUC) regarding lymph node dissection (LND)?

A

Guideline 25 strongly recommends that for patients with high-risk UTUC, clinicians should perform lymph node dissection (LND) at the time of nephroureterectomy (NU) or ureterectomy.

141
Q

What is the current state of evidence supporting the use of lymph node dissection (LND) in patients with high-risk upper tract urothelial carcinoma (UTUC)?

A

Despite the lack of randomized controlled trials (RCTs), recent systematic reviews and a re-analysis conducted by the panel suggest a potential benefit of LND in patients with high-risk UTUC. The re-analysis revealed that LND was associated with better recurrence-free survival (RFS). Moreover, additional cohort studies found LND to be associated with improved overall survival (OS) and cancer-specific survival (CSS), especially in patients with T3 and T4 tumors.

142
Q

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)

A

Guideline 26 recommends offering cisplatin-based neoadjuvant chemotherapy (NAC) to patients undergoing radical nephroureterectomy (RNU) or ureterectomy with high-risk (HR) upper tract urothelial carcinoma (UTUC). This is particularly important for patients whose post-operative estimated glomerular filtration rate (eGFR) is expected to be less than 60 mL/min/1.73m2 or those with other medical comorbidities that would prevent platinum-based chemotherapy in the post-operative setting. This strong recommendation is supported by evidence from several meta-analyses and two recent NAC trials, which show improved pathologic outcomes, cancer-specific survival (CSS), and overall survival (OS) with this approach. The guideline highlights that alternatives to cisplatin-based chemotherapy, such as immune checkpoint inhibitors, carboplatin, antibody drug conjugates, targeted FGFR therapies, are not recommended in the neoadjuvant setting outside of clinical trials.

143
Q

Why is neoadjuvant chemotherapy (NAC) recommended before radical nephroureterectomy (RNU) or ureterectomy in high-risk (HR) upper tract urothelial carcinoma (UTUC) patients?
a) NAC can be better tolerated in the neoadjuvant setting, allowing for more complete courses and enabling patients to proceed to the surgical intervention
b) NAC can only be administered before surgical intervention
c) NAC has no effect on the outcome of surgery
d) NAC increases the risk of surgical complications

A

a) NAC can be better tolerated in the neoadjuvant setting, allowing for more complete courses and enabling patients to proceed to the surgical intervention

144
Q

What is the expected effect of cisplatin-based neoadjuvant chemotherapy (NAC) on high-risk (HR) upper tract urothelial carcinoma (UTUC) patients?
a) It has no significant effect on survival outcomes
b) It leads to improved pathologic outcomes, cancer-specific survival (CSS), and overall survival (OS)
c) It worsens cancer-specific survival (CSS) and overall survival (OS)
d) It can cause progression of the disease

A

b) It leads to improved pathologic outcomes, cancer-specific survival (CSS), and overall survival (OS)

145
Q

What is the recommendation given in Guideline 26 for high-risk (HR) upper tract urothelial carcinoma (UTUC) patients regarding neoadjuvant chemotherapy (NAC)?

A

Guideline 26 strongly recommends that clinicians should offer cisplatin-based neoadjuvant chemotherapy (NAC) to high-risk (HR) upper tract urothelial carcinoma (UTUC) patients undergoing radical nephroureterectomy (RNU) or ureterectomy. This recommendation is particularly crucial for patients whose post-operative estimated glomerular filtration rate (eGFR) is expected to be less than 60 mL/min/1.73m2 or for those with other medical comorbidities that would preclude the use of platinum-based chemotherapy in the post-operative setting.

146
Q

What were the reported cancer-specific survival and overall survival rates following the treatment of UTUC Ta/T1 disease with BCG, according to the meta-analysis conducted by Foerster et al.?
a) 70% and 90% respectively
b) 85% and 75% respectively
c) 94% and 71% respectively
d) 100% and 80% respectively

A

c) 94% and 71% respectively. According to the meta-analysis by Foerster et al., the cancer-specific survival and overall survival rates following the treatment of UTUC Ta/T1 disease with BCG were 94% and 71% respectively.

147
Q

What is the current state of evidence supporting the use of cisplatin-based neoadjuvant chemotherapy (NAC) in high-risk (HR) upper tract urothelial carcinoma (UTUC) patients?

A

There are several meta-analyses and recent neoadjuvant chemotherapy trials supporting the use of cisplatin-based NAC for high-risk UTUC patients. These studies have shown that NAC improves pathologic outcomes, cancer-specific survival (CSS), and overall survival (OS) in these patients. Two recently completed NAC trials of cisplatin-based chemotherapy prior to RNU strongly support the use of this approach. Additionally, it’s important to note that alternative treatments to cisplatin-based chemotherapy are not recommended in the neoadjuvant setting outside of clinical trials.

148
Q

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)

A

Guideline 27 recommends offering 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. This strong recommendation is based on the results from the randomized phase III POUT trial, which showed that adjuvant platinum-based chemotherapy improved disease-free survival (DFS) compared with observation. Patients in the adjuvant chemotherapy arm had a significantly lower risk of metastases or death. However, the trial also showed that outcomes for patients with lymph node involvement and those treated with carboplatin chemotherapy were worse than those without positive nodes or treated with cisplatin chemotherapy. Therefore, carboplatin remains a reasonable choice for HR cisplatin-ineligible patients post-RNU if neoadjuvant chemotherapy was not given.

149
Q

Why is adjuvant chemotherapy recommended for advanced pathological stage UTUC patients after RNU or ureterectomy who have not received neoadjuvant platinum-based therapy?
a) It has no effect on the survival outcomes.
b) It leads to improved disease-free survival (DFS) and lower risk of metastases or death.
c) It worsens disease-free survival (DFS) and increases the risk of metastases.
d) It can cause progression of the disease.

A

b) It leads to improved disease-free survival (DFS) and lower risk of metastases or death.

150
Q

What is the role of carboplatin in the treatment of high-risk UTUC patients post-RNU who have not received neoadjuvant chemotherapy?
a) It is not recommended at all.
b) It is the first-line treatment.
c) It is a reasonable choice for cisplatin-ineligible patients.
d) It is only used in combination with other chemotherapy drugs.

A

c) It is a reasonable choice for cisplatin-ineligible patients.

151
Q

What is the recommendation given in Guideline 27 for advanced pathological stage UTUC patients regarding adjuvant chemotherapy?

A

Guideline 27 strongly recommends that clinicians should offer platinum-based adjuvant chemotherapy to advanced pathological stage (pT2–T4 pN0–N3 M0 or pTany N1–3 M0) UTUC patients after radical nephroureterectomy (RNU) or ureterectomy who have not received neoadjuvant platinum-based therapy. The chemotherapy can improve disease-free survival (DFS) and reduce the risk of metastases or death.

152
Q

What is the current state of evidence supporting the use of platinum-based adjuvant chemotherapy in advanced pathological stage UTUC patients who have not received neoadjuvant platinum-based therapy?

A

The strong recommendation for platinum-based adjuvant chemotherapy in advanced pathological stage UTUC patients is supported by results from the randomized phase III POUT trial. This trial found that adjuvant platinum-based chemotherapy improved disease-free survival (DFS) and lowered the risk of metastases or death compared to observation. However, it also showed that outcomes were worse for patients with lymph node involvement and those treated with carboplatin chemotherapy, suggesting that carboplatin remains a reasonable choice for cisplatin-ineligible patients if neoadjuvant chemotherapy was not given.

153
Q

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)

A

Guideline 28 suggests that adjuvant nivolumab therapy can be offered to patients who have either received neoadjuvant platinum-based chemotherapy or are ineligible for or refuse perioperative cisplatin. This recommendation is based on the results of two RCTs - IMvigor 010 and CheckMate 274, which involved patients with high-risk non-metastatic urothelial carcinoma, including a subset with upper tract urothelial carcinoma (UTUC). The IMvigor 010 trial, with adjuvant atezolizumab, did not meet its primary endpoint of improved disease-free survival (DFS) compared to observation. In contrast, the CheckMate 274 study demonstrated that adjuvant nivolumab did improve DFS compared to placebo. The approval of adjuvant nivolumab for UTUC and urothelial carcinoma of the bladder is based on these overall findings, despite the lack of a distinct difference in DFS for renal pelvic cancers or ureter in either arm within a UTUC subgroup. However, the small sample size limits the statistical power of this subgroup analysis. Nivolumab was well tolerated, with toxicities similar to other checkpoint inhibitor studies. Adjuvant platinum-based chemotherapy is still recommended over adjuvant nivolumab for eligible patients who did not receive neoadjuvant chemotherapy, while nivolumab might be more suitable for those with contraindications to platinum-based chemotherapy, high-risk pathology after neoadjuvant chemotherapy, or those who refuse standard adjuvant chemotherapy.

154
Q

What are the conditions under which adjuvant nivolumab therapy is recommended for UTUC patients, according to Guideline 28?
a) Only for patients with HR pathology after NAC.
b) Only for patients with contraindications to platinum-based chemotherapy.
c) For patients who have received neoadjuvant platinum-based chemotherapy, are ineligible for or refuse perioperative cisplatin, or who refuse standard forms of adjuvant chemotherapy after appropriate counseling.
d) For all UTUC patients, regardless of their previous treatment history.

A

c) For patients who have received neoadjuvant platinum-based chemotherapy, are ineligible for or refuse perioperative cisplatin, or who refuse standard forms of adjuvant chemotherapy after appropriate counseling.

155
Q

What were the outcomes of the CheckMate 274 study concerning adjuvant nivolumab?
a) Adjuvant nivolumab did not meet its primary endpoints of improved DFS compared to placebo.
b) Adjuvant nivolumab improved DFS compared to placebo in the intention to treat population.
c) Adjuvant nivolumab led to worse DFS compared to placebo.
d) The CheckMate 274 study did not include nivolumab in its protocol.

A

b) Adjuvant nivolumab improved DFS compared to placebo in the intention to treat population.

156
Q

How did the IMvigor 010 trial and the CheckMate 274 study compare in terms of outcomes related to adjuvant therapy in UTUC patients?

A

The IMvigor 010 trial and the CheckMate 274 study yielded different results. The IMvigor 010 trial, which used adjuvant atezolizumab, did not meet its primary endpoint of improved disease-free survival (DFS) compared to observation. On the other hand, the CheckMate 274 study, which used adjuvant nivolumab, did meet its co-primary endpoints and showed improved DFS compared to placebo.

157
Q

In what cases might the use of adjuvant nivolumab be considered over adjuvant platinum-based chemotherapy for UTUC patients?

A

Adjuvant nivolumab might be considered over adjuvant platinum-based chemotherapy for UTUC patients in the following cases: 1) patients with contraindications to platinum-based chemotherapy, such as poor renal function, poor performance status, sensorineural hearing loss, neuropathy or congestive heart failure, or allergy, 2) patients with high-risk pathology after receiving neoadjuvant chemotherapy, 3) patients who refuse standard forms of adjuvant chemotherapy after they have been properly counseled.

158
Q

What type of drug is Nivolumab?

A

Nivolumab is an immune checkpoint inhibitor.

159
Q

What specific receptor does Nivolumab target?

A

Nivolumab targets the programmed cell death protein 1 (PD-1) receptor on T cells.

160
Q

How does the interaction between PD-1 and PD-L1 influence immune response?

A

The interaction between PD-1 on T cells and PD-L1 on normal and cancerous cells sends an “off” signal that prevents T cells from attacking these cells.

161
Q

How does Nivolumab enhance the immune system’s response to cancer?

A

By blocking the PD-1 receptor, Nivolumab prevents the interaction between PD-1 and PD-L1, effectively removing the “off” signal. This allows T cells to recognize and attack cancer cells.

162
Q

What types of cancers can Nivolumab be used to treat?

A

Nivolumab can be used to treat non-small cell lung cancer, renal cell carcinoma, melanoma, Hodgkin lymphoma, head and neck cancer, urothelial carcinoma, colorectal cancer, and hepatocellular carcinoma, among others.

163
Q

GUIDELINE STATEMENT 29
In patients with metastatic (M+) UTUC, RNU or ureterectomy should not be offered as initial therapy. (Expert Opinion)

A

Guideline 29 advises against offering radical nephroureterectomy (RNU) or ureterectomy as initial therapy in patients with metastatic upper tract urothelial carcinoma (UTUC). There is no clear evidence supporting upfront surgery without chemotherapy for metastatic UTUC, as oncologic outcomes are primarily determined by the response to systemic therapy. Surgical treatment does not have demonstrable therapeutic efficacy for cytoreduction or as a stand-alone modality in this setting. Additionally, the potential harms, such as delays or an inability to receive systemic therapy due to surgical consequences, can significantly and negatively impact oncologic outcomes and overall survival. Instead, systemic therapy and alternative approaches, such as radiotherapy with or without chemotherapy, should be considered for inoperable or symptomatic patients with metastatic UTUC. The retrospective studies that suggest a clinical benefit from surgery to the primary site in metastatic UTUC patients typically involve those who have already received first-line chemotherapy or lack well-documented use of perioperative chemotherapy. This limits the interpretation and applicability of these studies due to strong selection biases and significant weaknesses in the data sets.

164
Q

According to Guideline 29, why is surgery not recommended as initial therapy for patients with metastatic UTUC?
a) There is clear evidence against the effectiveness of surgery for metastatic UTUC.
b) Surgery has a demonstrated therapeutic efficacy for cytoreduction in metastatic UTUC.
c) Surgery can delay or prevent the receipt of systemic therapy, which can negatively impact oncologic outcomes and overall survival.
d) All of the above.

A

c) Surgery can delay or prevent the receipt of systemic therapy, which can negatively impact oncologic outcomes and overall survival.

165
Q

What alternative approaches to surgery does Guideline 29 suggest for inoperable or symptomatic patients with metastatic UTUC?
a) Systemic therapy and radiotherapy, potentially with chemotherapy.
b) Increased frequency of medical examinations.
c) Strict lifestyle modifications, including diet and exercise.
d) No alternative approaches are suggested.

A

a) Systemic therapy and radiotherapy, potentially with chemotherapy.

166
Q

Why does Guideline 29 advise against offering RNU or ureterectomy as initial therapy for patients with metastatic UTUC?

A

Guideline 29 advises against offering RNU or ureterectomy as initial therapy in patients with metastatic UTUC because there is no clear evidence supporting upfront surgery without chemotherapy in these patients. Oncologic outcomes are primarily determined by the response to systemic therapy. Surgical treatment does not have demonstrable therapeutic efficacy for cytoreduction or as a single modality in this setting. Furthermore, surgery can potentially delay or prevent the receipt of systemic therapy, which can significantly and negatively impact oncologic outcomes and overall survival.

167
Q

What alternative treatment approaches are suggested for inoperable or symptomatic patients with metastatic UTUC?

A

For inoperable or symptomatic patients with metastatic UTUC, the guideline suggests considering systemic therapy and alternative approaches, such as radiotherapy with or without chemotherapy.

168
Q

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)

A

Guideline 30 recommends that patients with clinical, regional node-positive (cN1-3, M0) upper tract urothelial carcinoma (UTUC) should first be treated with systemic therapy. In cases where the patients have a partial or complete response to the systemic therapy, consolidative radical nephroureterectomy (RNU) or ureterectomy with lymph-node dissection may be performed. Patients with clinically suspicious lymph nodes are considered to have high-risk unfavorable disease with likely locally advanced or metastatic disease. In patients whose disease is converted to a surgically resectable state following a response to systemic therapy, surgical treatment should be offered if they are medically suitable. Comparative data supports this approach, with three reviews of up to six studies indicating that neoadjuvant chemotherapy (NAC) improves oncologic outcomes compared to nephroureterectomy alone.

169
Q

What is the initial recommended treatment for patients with clinical, regional node-positive (cN1-3, M0) UTUC according to Guideline 30?
a) Immediate surgery.
b) Systemic therapy.
c) Radiotherapy.
d) Watchful waiting.

A

b) Systemic therapy.

170
Q

What were the reported cancer-specific survival and overall survival rates following the treatment of UTUC Ta/T1 disease with BCG, according to the meta-analysis conducted by Foerster et al.?
a) 70% and 90% respectively
b) 85% and 75% respectively
c) 94% and 71% respectively
d) 100% and 80% respectively

A

c) 94% and 71% respectively. According to the meta-analysis by Foerster et al., the cancer-specific survival and overall survival rates following the treatment of UTUC Ta/T1 disease with BCG were 94% and 71% respectively.

171
Q

b) Systemic therapy.

A
172
Q

When is surgery recommended for patients with clinical, regional node-positive (cN1-3, M0) UTUC after initial systemic therapy?
a) Only in patients who have a complete response to the systemic therapy.
b) In patients who have either a partial or complete response to the systemic therapy.
c) In patients who do not respond to the systemic therapy.
d) Surgery is not recommended in these patients.

A

b) In patients who have either a partial or complete response to the systemic therapy.

173
Q

How does Guideline 30 recommend treating patients with clinical, regional node-positive (cN1-3, M0) UTUC?

A

Guideline 30 recommends treating patients with clinical, regional node-positive (cN1-3, M0) UTUC initially with systemic therapy. If these patients respond partially or completely to the systemic therapy, consolidative radical nephroureterectomy or ureterectomy with lymph-node dissection can be performed.

174
Q

What outcomes are improved by neoadjuvant chemotherapy according to the data supporting Guideline 30?

A

The data supporting Guideline 30, derived from a pooled analysis of various studies, indicates that neoadjuvant chemotherapy is associated with significantly better overall survival and cancer-specific survival compared to nephroureterectomy alone. In the subgroup of patients with locally advanced tumors (≥cT3 or cN+), the findings for overall survival were similar. Additionally, a review found neoadjuvant chemotherapy associated with improved recurrence-free survival compared to nephroureterectomy.

175
Q

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)

A

Guideline 31 recommends that patients with unresectable upper tract urothelial carcinoma (UTUC) or those who are ineligible for or refuse surgical treatment should be offered a clinical trial or best supportive care, including palliative management. Unresectable disease may be due to significant medical comorbidities, or other factors such as refusal of surgical treatment or possession of a solitary kidney. Treatment options should be determined with a multidisciplinary approach, focusing on achieving realistic care goals like local control for functional preservation (for instance, renal function) and palliation (such as bleeding, infection). Treatment plans should be explained clearly to patients, and their understanding and acceptance of the goals and expectations should be documented. Clinical trials should be sought and offered to eligible patients where available. Multimodal approaches may involve a combination of endoscopic management, systemic treatment options, and in rare cases, radiation, angioembolization, or percutaneous ablation for palliation of bleeding.

176
Q

What are the recommended options for managing patients with unresectable UTUC or those who are ineligible or refuse surgery?
a) Immediate surgical intervention.
b) A clinical trial or best supportive care including palliative management.
c) Only radiation treatment.
d) No treatment.

A

b) A clinical trial or best supportive care including palliative management.

177
Q

What is the focus when formulating alternative care options for patients with unresectable UTUC or those who are ineligible or refuse surgery?
a) Only prolonging life regardless of quality.
b) Achieving realistic care goals such as local control for functional preservation and palliation.
c) Undertaking the most expensive treatment options.
d) Performing unnecessary procedures.

A

b) Achieving realistic care goals such as local control for functional preservation and palliation.

178
Q

What does Guideline 31 recommend for patients with unresectable UTUC or those who are ineligible or refuse surgery?

A

Guideline 31 recommends that patients with unresectable UTUC or those who are ineligible or refuse surgery should be offered a clinical trial or best supportive care, including palliative management. Multimodal approaches may involve a combination of endoscopic management and systemic treatment options. In rare cases, radiation, angioembolization, or percutaneous ablation can be offered for palliation of bleeding.

179
Q

What are the key factors that need to be considered when formulating alternative care options for patients with unresectable UTUC or those who are ineligible or refuse surgery?

A

When formulating alternative care options for patients with unresectable UTUC or those who are ineligible or refuse surgery, a multi-disciplinary approach should be taken. The focus should be on achieving realistic care goals such as local control for functional preservation (like renal function) and palliation (for example, bleeding, infection). It’s crucial to provide appropriate patient counseling to explain goals and expectations, and document these discussions. If available, clinical trials should be discussed and offered to eligible patients.

180
Q

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)

A

Guideline 32 advises that low-risk patients treated with kidney-sparing treatments should undergo a follow-up cystoscopy and upper tract endoscopy within one to three months to confirm successful treatment. Upon confirmation, these patients should continue cystoscopic surveillance of the bladder at least every six to nine months for the first two years, and then at least annually afterwards. 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. The follow-up evaluation schedule aims to balance the morbidity and cost of follow-up with the risk of disease recurrence. Clinicians may elect to increase the intensity of surveillance according to their assessment of an individual patient’s risk and shared decision-making.

181
Q

What is the recommended follow-up protocol for low-risk patients managed with kidney sparing treatment for UTUC?
a) They should undergo a follow-up cystoscopy and upper tract endoscopy within one to three months, followed by continued cystoscopic surveillance of the bladder at least every six to nine months for the first two years and then at least annually thereafter.
b) They should undergo a follow-up cystoscopy and upper tract endoscopy only when they show symptoms.
c) They should undergo a follow-up cystoscopy and upper tract endoscopy within six months, followed by annual cystoscopic surveillance of the bladder.
d) They should undergo a follow-up cystoscopy and upper tract endoscopy within one year, followed by cystoscopic surveillance of the bladder every three years.

A

a) They should undergo a follow-up cystoscopy and upper tract endoscopy within one to three months, followed by continued cystoscopic surveillance of the bladder at least every six to nine months for the first two years and then at least annually thereafter.

182
Q

What should be the focus of follow-up evaluation schedule for low-risk patients treated with kidney sparing treatment?
a) It should aim to maximize the intensity of surveillance.
b) It should aim to balance the morbidity and cost of follow-up with the risk of disease recurrence.
c) It should aim to minimize the cost of follow-up.
d) It should aim to maximize the morbidity of follow-up.

A

b) It should aim to balance the morbidity and cost of follow-up with the risk of disease recurrence.

183
Q

What is the recommended surveillance protocol after kidney sparing treatment for low-risk UTUC patients according to Guideline 32?

A

According to Guideline 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.

184
Q

What factors should clinicians consider when deciding the intensity of surveillance for low-risk patients treated with kidney sparing treatment for UTUC?

A

When deciding the intensity of surveillance for low-risk patients treated with kidney sparing treatment for UTUC, clinicians should balance the morbidity and cost of follow-up with the risk of disease recurrence. They can elect to increase the intensity of surveillance above the minimum recommendations according to their assessment of an individual patient’s risk and shared decision-making with the patient.

185
Q

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)

A

Guideline 33 suggests that 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. If there’s no evidence of disease, these patients 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. Risk-adapted surveillance reflecting high recurrence risk within this patient population should incorporate cross-sectional imaging of the abdomen, pelvis, and chest to evaluate sites of metastasis.

186
Q

What is the recommended follow-up protocol for high-risk patients managed with kidney sparing treatment for UTUC?
a) They should undergo a follow-up cystoscopy and upper tract endoscopy within six months, followed by cystoscopic surveillance of the bladder every three years.
b) They should undergo a follow-up cystoscopy and upper tract endoscopy within one to three months, followed by 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.
c) They should undergo a follow-up cystoscopy and upper tract endoscopy only when they show symptoms.
d) They should undergo a follow-up cystoscopy and upper tract endoscopy within one year, followed by cystoscopic surveillance of the bladder every three to six months for two years.

A

b) They should undergo a follow-up cystoscopy and upper tract endoscopy within one to three months, followed by 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.

187
Q

What factors should clinicians consider when deciding the intensity of surveillance for high-risk patients treated with kidney sparing treatment for UTUC?
a) It should aim to maximize the intensity of surveillance.
b) It should aim to balance the morbidity and cost of follow-up with the risk of disease recurrence and metastasis.
c) It should aim to minimize the cost of follow-up.
d) It should aim to maximize the morbidity of follow-up.

A

b) It should aim to balance the morbidity and cost of follow-up with the risk of disease recurrence and metastasis.

188
Q

What is the recommended surveillance protocol after kidney sparing treatment for high-risk UTUC patients according to Guideline 33?

A

According to Guideline 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. If there’s no evidence of disease, these patients 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.

189
Q

How should clinicians tailor the surveillance regimen for high-risk patients managed with kidney sparing treatment for UTUC?

A

Clinicians should tailor the surveillance regimen for high-risk patients managed with kidney sparing treatment for UTUC based on disease risk and treatment modalities received. The regimen includes cystoscopic surveillance starting within three months after treatment, continuing every 3-6 months for 3 years, and then every 6-12 months through year 5. Follow-up ureteroscopy should be performed at least once within three to six months of endoscopic therapy and subsequently at the discretion of the clinician. Upper tract imaging with CT urogram and basic metabolic panel should be performed every 3-6 months for 3 years, then every 6-12 months for 2 years, and annually thereafter. Chest imaging with chest X-ray or CT of the chest is recommended every 6-12 months to evaluate for intrathoracic metastasis up to 5 years following the last diagnosis/treatment.

190
Q

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)

A

Guideline 34 suggests that if a patient with a history of UTUC develops urothelial recurrence in the bladder or urethra, or shows positive cytology, there might be a possibility of recurrent disease in the upper tracts. In such cases, an evaluation of the upper tracts should be carried out. This evaluation can be done through cross-sectional imaging or retrograde pyelography with or without selective upper tract cytology. If these tests suggest the presence of upper tract involvement, then further endoscopic evaluation is warranted.

191
Q

What does Guideline 34 suggest clinicians should do if a patient with a history of UTUC shows positive cytology or develops urothelial recurrence in the lower urinary tract?
a) Ignore it, as it is likely not related to the UTUC.
b) Only monitor the lower urinary tract, as the upper tracts are likely not involved.
c) Conduct an evaluation of the upper tracts, as there may be a recurrence of the disease there.
d) Conduct a complete re-evaluation of the patient’s medical history, as it might be an unrelated condition.

A

c) Conduct an evaluation of the upper tracts, as there may be a recurrence of the disease there.

192
Q

What were the reported cancer-specific survival and overall survival rates following the treatment of UTUC Ta/T1 disease with BCG, according to the meta-analysis conducted by Foerster et al.?
a) 70% and 90% respectively
b) 85% and 75% respectively
c) 94% and 71% respectively
d) 100% and 80% respectively

A

c) 94% and 71% respectively. According to the meta-analysis by Foerster et al., the cancer-specific survival and overall survival rates following the treatment of UTUC Ta/T1 disease with BCG were 94% and 71% respectively.

193
Q

c) Conduct an evaluation of the upper tracts, as there may be a recurrence of the disease there.

A
194
Q

If the evaluation of the upper tracts suggests the presence of involvement, what is the next step according to Guideline 34?
a) Discontinue treatment.
b) Wait and observe without further action.
c) Conduct further endoscopic evaluation.
d) Immediately proceed with aggressive treatment.

A

c) Conduct further endoscopic evaluation.

195
Q

What is the recommended action according to Guideline 34 if a patient with a history of UTUC develops urothelial recurrence in the bladder or urethra, or shows positive cytology?

A

According to Guideline 34, if a patient with a history of UTUC develops urothelial recurrence in the bladder or urethra, or shows positive cytology, it’s recommended to conduct an evaluation of the upper tracts, as there may be a recurrence of the disease there.

196
Q

What kind of evaluation is suggested by Guideline 34 if a patient with a history of UTUC shows positive cytology or develops urothelial recurrence in the bladder or urethra?

A

Guideline 34 suggests conducting an evaluation of the upper tracts in such cases. This evaluation could be carried out through cross-sectional imaging or retrograde pyelography with or without selective upper tract cytology. If these tests suggest the presence of upper tract involvement, then a further endoscopic evaluation is warranted.

197
Q

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)

A

Guideline 35 recommends post-radical nephroureterectomy (NU) surveillance for patients with <pT2 N0/M0 disease. After surgery, cystoscopy and cytology should be performed within three months, with the frequency of repetition based on pathological grade. For Low-Grade (LG), tests should be repeated every six to nine months for the first two years and annually thereafter. For High-Grade (HG), the tests should be repeated every three to six months for the first three years and annually thereafter. Cross-sectional imaging of the abdomen and pelvis should be done within six months after surgery and then at least annually for a minimum of five years due to metastasis risk and estimated 5% probability for contralateral disease. Surveillance after five years without recurrence should be encouraged based on shared decision-making between patient and clinician. The studies reviewed indicate a significant risk of intravesical recurrence and low to intermediate risk of regional or distant metastases depending on factors such as tumor grade, lymphovascular invasion (LVI), and tumor multifocality.

198
Q

According to Guideline 35, how frequently should patients with High-Grade (HG) disease undergo cystoscopy and cytology within the first three years after NU?
a) Every three to six months
b) Every six to nine months
c) Annually
d) Every two years

A

a) Every three to six months

199
Q

What is the risk of contralateral disease after NU, as noted in Guideline 35?
a) Approximately 50%
b) Approximately 5%
c) Approximately 25%
d) Approximately 75%

A

b) Approximately 5%

200
Q

According to Guideline 35, how should surveillance be carried out after NU in patients with <pT2 N0/M0 disease?

A

According to Guideline 35, after NU, cystoscopy and cytology should be performed within three months. For Low-Grade (LG) disease, these tests should be repeated every six to nine months for the first two years and then annually. For High-Grade (HG) disease, tests should be performed every three to six months for the first three years, then annually. Cross-sectional imaging of the abdomen and pelvis should be done within six months after surgery and annually for a minimum of five years due to metastasis risk and a 5% probability for contralateral disease.

201
Q

What are the key findings from the studies referenced in Guideline 35 about recurrence rates after NU?

A

The studies indicate that the rate of intravesical recurrence after NU is approximately 29% with a median time to recurrence of 6-12 months. The overall risk of recurrence to the contralateral upper tract is 2.2% (with a range of 0% to 4.6%). Local recurrence rates are around 24% with a median time to recurrence of 7 months. Also, most local recurrences were identified within the first two years. These findings highlight the need for regular attention to monitoring for intravesical recurrence through regular cystoscopic surveillance.

202
Q

TABLE 6: SURVEILLANCE AFTER COMPLETE TREATMENT

A
203
Q

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)

A

Guideline 36 provides post-operative surveillance recommendations for patients who have undergone nephroureterectomy (NU) for >pT2 Nx/0 disease. Surveillance includes cystoscopy with cytology, cross-sectional imaging of the abdomen and pelvis, and chest imaging. The frequency varies based on the years passed since surgery. Beyond five years without recurrence, annual surveillance may be continued based on shared/informed decision-making.

A systematic review and meta-analysis of studies highlighted the need for such surveillance due to the significant risk of intravesical recurrence and retroperitoneal or distant metastasis. A range of risk factors associated with recurrences were identified, including male sex, prior bladder cancer, positive preoperative urinary cytology, multifocality, necrosis, positive surgical margins, etc.

In addition to monitoring for cancer recurrence or metastasis, patients should also be observed for any health complications arising from the NU procedure. Periodic laboratory assessments, including serum creatinine level, eGFR, and urinalysis, are recommended. Referrals to nephrology should be considered in patients who develop progressive renal insufficiency or proteinuria.

204
Q

According to Guideline 36, how frequently should clinicians perform surveillance cystoscopy with cytology for the first 3 years in patients who have undergone NU for >pT2 Nx/0 disease?
a) Annually
b) Every three to six months
c) Every six months
d) Every nine months

A

b) Every three to six months

205
Q

Which of the following factors was identified as a risk for intravesical recurrence after nephroureterectomy?
a) Female sex
b) Negative preoperative urinary cytology
c) Absence of necrosis
d) Positive surgical margins

A

d) Positive surgical margins

206
Q

What are the key elements of post-operative surveillance for patients who have undergone NU for >pT2 Nx/0 disease as per Guideline 36?

A

The key elements of post-operative surveillance as per Guideline 36 include performing cystoscopy with cytology every three to six months for the first three years, then annually thereafter. Additionally, 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 in year three, and annually up to year five. Chest imaging should be performed every 6-12 months for the first 5 years. Beyond five years without recurrence, annual surveillance may be continued based on shared/informed decision-making.

207
Q

What are some of the risk factors for intravesical recurrence after NU as indicated in Guideline 36?

A

Some of the risk factors for intravesical recurrence after NU include male sex, previous bladder cancer, preoperative chronic kidney disease, positive preoperative urinary cytology, ureteral tumor site, multifocality, invasive pathologic T-stage, presence of necrosis, laparoscopic approach, extravesical bladder cuff removal, and positive surgical margins.

208
Q

GUIDELINE STATEMENT 37
For patients with reduced or deteriorating renal function following NU or other intervention, clinicians should consider referral to nephrology. (Expert Opinion)

A

Guideline 37 emphasizes the importance of postoperative renal function monitoring in patients who have undergone nephroureterectomy (NU) or other interventions. If patients display reduced or deteriorating renal function, clinicians are recommended to consider a referral to nephrology. This is particularly crucial for patients with an estimated Glomerular Filtration Rate (eGFR) less than 45 mL/min/1.73m^2, confirmed proteinuria, diabetics with preexisting Chronic Kidney Disease (CKD), or if eGFR is projected to be less than 30 mL/min/1.73m^2 post-intervention.

Long-term renal dysfunction can heighten the risk of osteoporosis, anemia, metabolic and cardiovascular disease, hospitalization, and death. However, early detection of progressing renal dysfunction or proteinuria can provide opportunities for medical interventions to slow CKD progression and reduce cardiovascular risks. The Modification of Diet in Renal Disease and CKD – Epidemiology Collaboration (CKDEPI) equations are commonly used formulas for monitoring eGFR.

209
Q

According to Guideline 37, referral to nephrology should be considered for patients with:
a) eGFR more than 45 mL/min/1.73m^2
b) eGFR less than 45 mL/min/1.73m^2
c) Confirmed absence of proteinuria
d) No preexisting CKD

A

b) eGFR less than 45 mL/min/1.73m^2

210
Q

What were the reported cancer-specific survival and overall survival rates following the treatment of UTUC Ta/T1 disease with BCG, according to the meta-analysis conducted by Foerster et al.?
a) 70% and 90% respectively
b) 85% and 75% respectively
c) 94% and 71% respectively
d) 100% and 80% respectively

A

c) 94% and 71% respectively. According to the meta-analysis by Foerster et al., the cancer-specific survival and overall survival rates following the treatment of UTUC Ta/T1 disease with BCG were 94% and 71% respectively.

211
Q

Which of the following formulas are commonly used for monitoring eGFR?
a) CKDEPI and Kidney Disease Outcomes Quality Initiative (KDOQI) equations
b) Modification of Diet in Renal Disease and CKD – Epidemiology Collaboration (CKDEPI) equations
c) KDOQI and Renal Disease Outcomes and Practice Patterns Study (DOPPS) equations
d) DOPPS and Modification of Diet in Renal Disease equations

A
212
Q

Which of the following formulas are commonly used for monitoring eGFR?
a) CKDEPI and Kidney Disease Outcomes Quality Initiative (KDOQI) equations
b) Modification of Diet in Renal Disease and CKD – Epidemiology Collaboration (CKDEPI) equations
c) KDOQI and Renal Disease Outcomes and Practice Patterns Study (DOPPS) equations
d) DOPPS and Modification of Diet in Renal Disease equations

A

b) Modification of Diet in Renal Disease and CKD – Epidemiology Collaboration (CKDEPI) equations

213
Q

What were the reported cancer-specific survival and overall survival rates following the treatment of UTUC Ta/T1 disease with BCG, according to the meta-analysis conducted by Foerster et al.?
a) 70% and 90% respectively
b) 85% and 75% respectively
c) 94% and 71% respectively
d) 100% and 80% respectively

A

c) 94% and 71% respectively. According to the meta-analysis by Foerster et al., the cancer-specific survival and overall survival rates following the treatment of UTUC Ta/T1 disease with BCG were 94% and 71% respectively.

214
Q

When should a referral to nephrology be considered according to Guideline 37?

A

According to Guideline 37, a referral to nephrology should be considered for patients with reduced or deteriorating renal function following nephroureterectomy (NU) or other interventions. Specifically, a nephrology referral should be considered for patients with an eGFR less than 45 mL/min/1.73m^2, confirmed proteinuria, diabetics with preexisting CKD, or when eGFR is expected to be less than 30 mL/min/1.73m^2 after intervention.

215
Q

What are some of the potential long-term risks of renal dysfunction?

A

Long-term renal dysfunction can increase the risk of various health conditions, including osteoporosis, anemia, metabolic and cardiovascular disease. It can also increase the likelihood of hospitalization and death.

216
Q

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)

A

Guideline 38 advises clinicians to discuss disease-related stressors and risk factors with patients who have urothelial cancer. Clinicians should also encourage these patients to adopt healthy lifestyle habits, such as quitting smoking, exercising regularly, and maintaining a healthy diet, to promote long-term health benefits and enhance their quality of life.

Certain risk factors like smoking are linked to advanced disease stage, recurrence, and worse cancer-specific mortality (CSM) among patients with Upper Tract Urothelial Carcinoma (UTUC), especially for current smokers. UTUC is also associated with metabolic syndrome and obesity. Obesity has been found to negatively impact disease-specific outcomes among patients undergoing Radical Nephroureterectomy (RNU).

As such, clinicians should emphasize the adoption of healthy lifestyle habits to UTUC patients to foster long-term health benefits and improve their quality of life. Clinicians should also collaborate with patients and their primary care providers to optimally manage comorbidities throughout the UTUC treatment and surveillance process, ensuring a better quality of life during survivorship.

217
Q

Which of the following risk factors is associated with advanced disease stage, recurrence, and worse CSM among patients with UTUC?
a) Physical inactivity
b) Smoking
c) Low-carb diet
d) Absence of comorbidities

A

b) Smoking

218
Q

UTUC is also associated with which of the following conditions?
a) Metabolic syndrome and obesity
b) Hypertension and diabetes
c) Chronic respiratory diseases
d) Autoimmune disorders

A

a) Metabolic syndrome and obesity

219
Q

What are some lifestyle habits that clinicians should encourage UTUC patients to adopt, according to Guideline 38?

A

According to Guideline 38, clinicians should encourage UTUC patients to adopt healthy lifestyle habits such as quitting smoking, regularly exercising, and maintaining a healthy diet. These habits are associated with long-term health benefits and improved quality of life.

220
Q

What role does obesity play in the context of UTUC?

A

Obesity is associated with UTUC and it has been found to negatively impact disease-specific outcomes among patients undergoing Radical Nephroureterectomy (RNU). Therefore, maintaining a healthy weight can be a crucial aspect of managing UTUC.

221
Q

What recent research finding has highlighted potential therapeutic targets for UTUC?
a) High prevalence of activating FGFR3 mutations in UTUC
b) A higher rate of obesity in UTUC patients compared to bladder cancer patients
c) A significant decrease in kidney function following UTUC diagnosis
d) The frequent occurrence of UTUC in conjunction with hypertension

A

a) High prevalence of activating FGFR3 mutations in UTUC

222
Q

Why is a multidisciplinary approach crucial in the management of UTUC patients?
a) It allows for more extensive tissue sampling
b) It ensures all potential treatments are considered
c) It provides optimal care by integrating various fields of expertise
d) It reduces the need for invasive diagnostic methods

A

c) It provides optimal care by integrating various fields of expertise

223
Q

How can genomic markers and urinary biomarkers potentially improve the management of UTUC?

A

Genomic markers and urinary biomarkers have the potential to improve UTUC management by providing less invasive methods of grading and staging tumors, as well as identifying potential treatment pathways. Urinary biomarkers may also be beneficial for refining post-treatment follow-up procedures and enabling better-informed decisions about endoscopic surveillance.

224
Q

What recent advancements have been made in the visualization and treatment of upper urinary tract tumors?

A

Advancements in flexible digital endoscopes have improved the visualization and accessibility of upper urinary tract tumors. In terms of treatment, the development of new therapies such as reverse thermo-hydrogel preparation of mitomycin offers a new method of treating low-risk tumors. Energy devices like the thulium:YAG laser have also been approved for thermal ablative procedures, and photodynamic treatments are currently being tested in Phase III clinical trials.

225
Q

What were the reported cancer-specific survival and overall survival rates following the treatment of UTUC Ta/T1 disease with BCG, according to the meta-analysis conducted by Foerster et al.?
a) 70% and 90% respectively
b) 85% and 75% respectively
c) 94% and 71% respectively
d) 100% and 80% respectively

A

c) 94% and 71% respectively. According to the meta-analysis by Foerster et al., the cancer-specific survival and overall survival rates following the treatment of UTUC Ta/T1 disease with BCG were 94% and 71% respectively.

226
Q

What are some of the reasons clinicians may consider watchful waiting or surveillance alone for patients with UTUC?

A

Clinicians may consider watchful waiting or surveillance alone for patients with UTUC who have significant comorbidities, competing risks of mortality, or are at significant risk of End-Stage Renal Disease (ESRD) with any intervention resulting in dialysis. These patients have a higher risk of severe treatment-related complications, which could outweigh the potential benefits of treatment. Also, if the patient has a very limited life expectancy, interventions might be limited to palliation or awaiting symptomatic progression.

227
Q

What are the cancer-specific survival and overall survival rates reported for the treatment of UTUC Ta/T1 disease with BCG according to the meta-analysis by Foerster et al.?

A

The meta-analysis conducted by Foerster et al. reported cancer-specific survival and overall survival rates of 94% and 71% respectively for the treatment of UTUC Ta/T1 disease with BCG. These figures suggest that BCG therapy can be effective in managing high-risk but favorable UTUC.