6th edition
Abstract book LIVE & VIRTUAL MEETING 14 & 15 October 2021
All abstracts are available online • Browse published posters • Contact authors directly
ABSTRACTS OF THE GLOBAL CONGRESS ON BLADDER CANCER, 6TH EDITION LIVE & VIRTUAL MEETING, 14 & 15 OCTOBER 2021
You can find an overview of all accepted abstracts on https://www.morressier.com/event/ bladdr2021/61373f8eeef6420012dc9d4c.
ORGANISING COMMITTEE OF THE CONGRESS Ananya Choudhury, Christie NHS Foundation Trust, Manchester, UK Daan De Maeseneer, University Hospital, Ghent & AZ Sint-Lucas, Bruges, Belgium Maria De Santis, Charité Medical University Hospital, Berlin, Germany Valérie Fonteyne, University Hospital, Ghent, Belgium Badrinath Konety, University of Minnesota, Minneapolis, USA Thierry Roumeguère, Erasme Hospital & Jules Bordet Institute, ULB, Brussels, Belgium
COPYRIGHT
This abstract book and the individual abstracts published in it are protected under copyright by e-HIMS.
PUBLISHED BY: e-HIMS bv Leopoldplein 39 B1 2500 Lier, Belgium T: +32 3 491 87 46
SPONSORED BY: The abstract book is sponsored by
For appropriate referencing to the abstracts, please use: ISBN 9789462210219
Except as outlined here below, no part of this abstract book may be copied, distributed, modified, published, reproduced, stored, transmitted, created derivative works from, or sold or licensed in any medium to anyone, without prior written permission of the Publisher. Copy, modification or use of any content of the abstract book for any commercial purpose without the authorisation of the Publisher is a violation of copyright. Any copying or redistribution for commercial purposes or for compensation of any kind requires prior written permission from the Publisher. Any unapproved use may result in actions being taken by e-HIMS to require removal of material concerned from display/distribution and possible legal action. To obtain permission for the reproduction of (parts of ) this work, email to info@mirrorsofmedicine.org.
Photocopying Single photocopies of single abstracts may be made for personal use as allowed by national copyright laws. Permission of the Publisher and payment of a fee is required for all other photocopying, including multiple or systematic copying, copying for promotional purposes, resale, etc. Notice No responsibility is assumed by the Publisher for any injury and/or damage to persons or property as a matter of products liability, negligence or otherwise, or from any use or operation of any methods, products, instructions or ideas contained in this abstract book.
OPDIVO® demonstrated an ORR of 20.7%2 CheckMate 275 – ORR (33.7-month minimum follow-up)2
OPDIVO® (nivolumab) as monotherapy is indicated for the treatment of locally advanced unresectable or metastatic urothelial carcinoma in adults after failure of prior platinum-containing therapy.1
20.7% ORR (56/270)
95% CI: 16.1–26.1 PR: 14.1% (n=38) CR: 6.7% (n=18) ORR by PD-L1 Expression Status
Adapted from Galsky MD, et al. Clin Cancer Res 2020.
≥1% (124/270) 25.8% ORR
<1% (146/270) 16.4% ORR
(95% CI: 18.4–34.4)
(95% CI: 10.8–23.5)
• 20.7% of patients (56/270) experienced stable disease Primary analysis (median follow-up of 11.5 months)1
OPDIVO® demonstrated efficacy in urothelial carcinoma patients after failure of prior platinum-containing therapy 1-3 In the Phase II CheckMate 275 study (33.7-month minimum follow-up),* OPDIVO® demonstrated:
• ORR in ITT population: 20.0% (95% CI: 15.4–25.3), (PR=17.0%, CR=3.0%) • ORR in PD-L1 ≥1%: 25% (95% CI: 17.7–33.6) • ORR in PD-L1 <1%: 16% (95% CI: 10.3–22.7)
OPDIVO® has a safety profile in urothelial carcinoma consistent with that established across its use across other tumour types1-3 In CheckMate 275 (33.7-month follow-up):2 • Grade 3/4 TRAEs occurred in 24.8% of patients (n=67)
20.7% ORR 20.7% objective response rate (95% CI: 16.1–26.1, n=56/270)2
– Three patients (1%) died from TRAEs†
8.6
• No new safety signals were identified
months 8.6 months median OS (95% CI: 6.1–11.3)2
* Phase II, single-arm study of OPDIVO® in 270 patients with metastatic or unresectable urothelial carcinoma who have progressed or recurred following treatment with a platinum agent. Patients received OPDIVO® 3 mg/kg intravenously every 2 weeks, with treatment continuing until progression, unacceptable toxicity or other protocol-defined reasons. The primary endpoint was objective response rate per blinded independent review committee (BIRC).1-3
Consistent safety profile with that established across tumour types for OPDIVO® monotherapy1-3
This material has been produced to ensure compliance with the EFPIA and local Belgian regulations. The indication is according to European Commission licensing. Prescribing information may vary depending on local approval in each country or region. Therefore, before prescribing any product, always refer to local materials such as the prescribing information and/or the Summary of Product Characteristics (SPC).
† One instance each of pneumonitis, respiratory failure and circulatory collapse.2 AE – adverse event; CI – confidence interval; CR – complete response; ITT – intention-to-treat; ORR – objective response rate; OS – overall survival; PD-L1 – programmed death-ligand 1; PR – partial response; TRAE – treatment-related adverse event References: 1. OPDIVO® Summary of Product Characteristics. Available at: https://www.ema.europa.eu/en/documents/product-information/opdivo-epar-product-information_en.pdf [Accessed September 2021]. 2. Galsky MD, Saci A, Szabo PM et al. Nivolumab in Patients with Advanced Platinum-resistant Urothelial Carcinoma: Efficacy, Safety, and Biomarker Analyses with Extended Follow-up from CheckMate 275. Clin Cancer Res 2020; published online. DOI:10.1158/1078-0432.CCR-19-4162. 3. Sharma P, Retz M, Siefker-Radtke A et al. Nivolumab in metastatic urothelial carcinoma after platinum therapy (CheckMate 275): a multicentre, single-arm, phase 2 trial. Lancet Oncol 2017;18(3):312-322. © 2021 Bristol-Myers Squibb Company. All rights reserved. OPDIVO® and the related logos are trademarks of Bristol-Myers Squibb Company. 1506-BE-2100024 Date of preparation: September 2021
For Belgian essential information, please see following pages. 4
5
6
7
1
2
Evert Baten, Associatie Urologie Hageland Frank Van der Aa, Hans Goethuys, Koen Slabbaert, Ingrid Arijs, Koenraad van Renterghem
Emma Corke, Southmead Hospital NHS trust Helena Burden, Kate Warren
Introduction & Objectives: To determine whether omitting antimicrobial prophylaxis (AMP) in TURB is safe in patients undergoing TURB without an indwelling pre-operative catheter/ nephrostomy/DJ and a negative pre-operative urinary culture.
Introduction & Objectives: In this study we investigated the recurrence rate of NMIBC (Non Muscle Invasive Bladder Cancer) following Hyperthermic Mitomycin C (MMC) therapy (COMBAT). Currently there are no guidelines about standard regimes.
Materials & Methods: A multi-centered randomized controlled trial (RCT) from 17-09-2017 until 31-12-2019 in 5 hospitals. Patients with a pre-operative indwelling catheter/DJ-stent or nephrostomy and a positive pre-operative urinary culture (> 104 uropathogens/mL) were excluded. Post-operative fever was defined as body temperature ≥ 38.3°C. A non-inferiority design with a 6% noninferiority margin and null hypothesis (H0) that the infection risk is at least 6% higher in the experimental (E) than in the control (C) group; H0: C (AMP-group) − E (no AMP-group) ≥ Δ (6% noninferiority margin). A multivariable, logistic regression was performed for AMP and post-TURB fever with covariates: tumor size and (clot-) retention. The R Project® for statistical computing was used for statistical analysis and a p value of 0.05 was considered as statistically significant.
Materials & Methods: We retrospectively reviewed data on 56 patients who were offered Hyperthermic MMC therapy for NMIBC between 2018 and 2021. Patients with high grade NMIBC were treated for 1 year following a BCG style SWOG regime, intermediate NMIBC patients were treated with 6 weeks induction. We investigated recurrence rates following treatment.
A randomized trial regarding antimicrobial prophylaxis (AMP) in transurethral resection of bladder tumor (TURB)
3
Recurrence rate following hyperthermic mitomycin C treatment (COMBAT system), with BCG style regime
A multicenter Dutch cohort of patients treated with definitive concurrent chemoradiotherapy (CRT) for muscle invasive bladder carcinoma: results of the past 10 years Ben-Max de Ruiter, Academic Medical Center Introduction & Objectives: Concurrent chemoradiotherapy (CRT) is increasingly applied in clinical practice as a definitive treatment for patients with localized muscle invasive bladder carcinoma (MIBC). This multicenter retrospective study aimed to assess the oncological outcomes in a contemporary cohort of non-metastatic MIBC patients treated with CRT. Materials & Methods: Patients with non-metastatic MIBC (cT2-4aN0M0) who received CRT with curative intent between January 2010 and April 2020 in three centers were retrospectively identified. The primary endpoint of the study was 2-year locoregional disease-free survival (LDFS), which was defined as time from date of diagnosis to first documented recurrence in the bladder, non-muscle invasive (NMIBC) or MIBC, or pelvis, as assessed by cystoscopy and/or imaging. Secondary endpoints were disease-specific survival (DSS), overall survival (OS) and complete response at 3 months. The Kaplan-Meier method was used to estimate survival outcomes. Multivariable Cox regression analysis was used to identify associating variables with survival outcomes.
Results: Of the 56 patients, 10 were removed from the trial due to side effects. 3 patients developed new urinary symptoms, 5 had previous urinary symptoms that worsened and 2 developed a rash. 46 patients were therefore reviewed in total. 34 (74%) had high risk NMIBC and 12 (26%) had intermediate NMIBC, as defined by EAU. Only 3 patients had the full maintenance course of MMC, see below for the flowchart indicating the dropout rates. The overall recurrence rate was 47.8%. In intermediate disease this dropped to 25% and in high risk disease it was 55.9%. In patients who were BCG naïve, the recurrence rate was 31% and in patients who had been previously given BCG treatment, the recurrence rate was 56.7%. In patients with no CIS in the sample, the recurrence rate was 33.3%, but in patients with CIS, the recurrence rate rose to 68.4%. The follow up range was 2-29 months (average recurrence was at 7 months).
Results: 459 Patients were included and 202/459 (44.1%) received AMP vs 257/459 (55.9%) without AMP. Fever occurred in 6/202 (2.9%; 95% CI [1.2%-6.6%]) patients with AMP vs 8/257 (3.1%; 95% CI [1.5%-6.1%]) without AMP (p=0.44). Multivariable, logistic regression showed no significant harm in omitting AMP when controlled for (clot-)retention and tumor size (p=0.85) and an adjusted risk difference in developing post-TURB fever of 0.0016; 95% CI [-0.029; 0.032].
Results: The study population included 240 patients with a median age of 74 years. 31 (13%) patients underwent neoadjuvant chemotherapy and 19 patients (8%) underwent pelvic lymph node dissection, prior to CRT. Median follow up was 27 months (IQR 11- 44). LDFS probability (Figure 1) for 1, 2 and 5 years(y) after diagnosis were 0.89 (95% CI 0.85 – 0.93), 0.76 (95% CI 0.70- 0.82) and 0.56 (95% CI 0.47-0.66), respectively. The 5y local recurrence rate for NMIBC for MIBC and for regional lymph node metastasis after CRT was 11%, 7%, and 8%, respectively. DSS probability for 1, 2 and 5y after diagnosis were 0.94 (95% CI 0.91 – 0.97), 0.83 (95% CI 0.0.78- 0.89) and 0.70 (95% CI 0.62-0.79), respectively. OS probability for 1, 2 and 5 y after diagnosis were 0.88 (95% CI 0.84 - 0.93), 0.74 (95% CI 0.69 – 0.81) and 0.50 (95% 0.42 – 0.59), respectively. Complete response after CRT was seen in 210 cases (87.5%). On multivariable analysis T2 versus T3-4 tumor stage was significantly associated with a better DSS (HR 1.01. 95% CI 1-1.05, p 0.024). Nine out of 14 (64%) patients with MIBC recurrences underwent salvage cystectomy.
Conclusion: Hyperthermic MMC has shown to have benefit in decreasing recurrence rates in bladder cancer patients. BCG naïvity improved recurrence rates, as did the lack of CIS in the initial sample. In general, the more courses of maintenance doses the patient had, the lower the recurrence rates. Our data is in line with previous data for recurrence rates, however we had a higher drop-out rate in the induction period than previously has been recorded, this may be due to our BCG style regime, and therefore consideration will be given to alternative regimes in our centre.
Conclusion: Our data suggest the safety of omitting AMP in patients undergoing TURB without an indwelling, pre-operative catheter/nephrostomy/DJ and a negative pre-operative urinary culture.
Conclusion: In this multicenter retrospective cohort study we showed that 2 years after CRT the LDFS, including non-muscle invasive recurrence, is 76% for non-metastatic MIBC patients. DSS 5 years after CRT for non-metastatic MIBC patients is 70%.
8
9
4
5
Adjuvant radiotherapy after radical cystectomy for patients with muscle invasive bladder cancer: results of a phase 2 trial
Bladder cancer rapid autopsy program provides a critical resource for the biological study of advanced bladder cancer
Verghote Flor, Ghent University Hospital Piet Dirix, Charles Van Praet, Charlien Berghen, Maarten Albersen,Sara Junius, Nick Liefhooghe, Marc Brosens, Gert De Meerleer, Piet Ost, Geert Villeirs, Sofie Verbeke, Daan De Maeseneer, Elhaseen Elamin, Kathia De Man, Karel Decaestecker, Valérie Fonteyne
Jose Garcia, University of Washington Funda Vakar-Lopez, Martine P Roudier, Petros Grivas, Evan Yu, John K. Lee, Eva Corey, Bruce Montgomery, Andrew Hsieh, Jonathan L. Wright, Hung-Ming Lam Introduction & Objectives: Rapid autopsies are unique in their ability to provide adequate tissue specimens sampled across multiple anatomic locations, allowing biological studies in the metastatic disease. We have established a bladder cancer rapid autopsy program to acquire bladder cancer metastases, and build patient-derived xenograft (PDX) and organoid models for biological and preclinical studies.
Introduction & Objectives: The majority of patients with non-metastatic (M0) muscle-invasive bladder cancer (MIBC) are treated with neo-adjuvant chemotherapy followed by radical cystectomy (RC) and extended lymph node dissection. Despite this aggressive treatment the outcome is poor and ultimately 30% of the patients with ≥ pathological (p)T3 tumors develop a pelvic recurrence, which is rarely salvageable and is often associated with debilitating sequels. In the past, adjuvant external beam radiotherapy (EBRT) has shown to increase disease free survival, but excessive toxicity hampered enthusiasm for further implementation. In this phase 2 trial the place of adjuvant EBRT after RC for high-risk MIBC is re-investigated.
Materials & Methods: Patients with metastatic bladder cancer were consented and rapid autopsies were performed 2-6 hours after death to allow acquisition of normal and metastatic specimens. Frozen and formalin-fixed, paraffin embedded specimens were deposited into University of Washington GU biorepository. Pathological evaluation of each specimen was performed. Additionally, fresh tumors were implanted subcutaneously into SCID mice to establish PDXs. Once PDXs were established, PDXs were dissociated for companion organoid culture. Clinical history and treatment information was documented for each participant.
Materials & Methods: In this Belgian multicentric prospective phase 2 trial, seventy-two patients were included and received treatment. Eligible patients were patients with MIBC, treated with RC and presenting with ≥1 of the following characteristics: pT3 stage + presence of lymphovascular invasion on pathological examination, pT4 stage, <10 lymph nodes removed, positive lymph nodes, positive surgical margins. Prior to EBRT, a post-operative 18F-FDG-PET-CT was performed to rule out distant metastases. A median dose of 50 Gy in 25 fractions was administered, using Volumetric-Modulated Arc Therapy (VMAT), to the pelvic lymph node regions with inclusion of the cystectomy bed in case of a positive surgical margin. A simultaneous integrated boost up to 70 Gy was delivered to PET-CT positive lymph nodes. The primary endpoint was acute grade ≥3 gastro-intestinal (GI) toxicity and in case of a neobladder reconstruction grade 4 urinary toxicity, within 12 weeks after EBRT. Secondary endpoints were late toxicity, local control, disease free survival and overall survival (OS). Toxicity was scored using the Radiation Therapy Oncology Group (RTOG) criteria and the Common Terminology Criteria for Adverse Events (CTCEA) v4.0.
Results: We have performed 20 bladder cancer rapid autopsies to date, and have acquired 243 metastatic and 97 normal specimens. The tumor histologies of the tumors include urothelial cell carcinoma (7/20), squamous cell carcinoma (5/20), plasmacytoid variant of urothelial carcinoma (6/10) and neuroendocrine carcinoma (2/10). The cohort represented one treatment naïve patient (1/20), and patients treated with BCG only (2/20), chemotherapy (4/20), immune checkpoint inhibitors (5/20), and chemotherapy with immune checkpoint inhibitors (8/20). The most common sites of metastases were liver (14/20), lymph node (13/20), followed by lung (11/20), intestine (6/20), adrenal gland (3/20), and omentum (2/20). More than half of the patients with liver metastasis 10/14) had extensive disease that allowed acquisition of multiple tumor foci. We have successfully developed PDX and organoid models from two independent metastases (omentum – CoCaB 14.1 and liver –CoCaB 14.2) from the same patient, representing the first PDX developed from bladder cancer metastasis.
Results: The median follow up was 15 months (1-72 months). Five (7%) patients had acute grade ≥3 GI toxicity. The 2-year probability of developing grade ≥3 and grade ≥2 GI toxicity was 18% and 79%, respectively. Of the seventeen neobladder patients 18% and 11% reported acute and late grade 2 urinary toxicity, respectively. Grade ≥3 urinary toxicity was not reported. The 2-year local recurrence-free survival and clinical relapse-free survival (CRFS) rates were 88% ±4% and 44% ±7, respectively. The mean CRFS time was 31 months (95% CI: 22-39 months). The 2-year OS rate was 52% ±7%, with a mean OS time of 39 months (95% CI: 31-48 months).
Conclusion: Rapid autopsies allow multiple and simultaneous collection of metastatic disease tissue specimens. The availability of metastatic bladder cancer specimens, PDXs and organoids represent a valuable resource for molecular and preclinical study to advance the much needed therapeutic discovery for metastatic bladder cancer. The accrual is ongoing and molecular analyses of these specimens are in progress.
6
7
Immunotherapy in advanced urothelial cancer: first steps, multidisciplinary approach
A randomized, double-blind, placebocontrolled, phase 3 trial of infigratinib as adjuvant therapy in patients with invasive urothelial carcinoma harboring susceptible FGFR3 genetic alterations: PROOF 302
Lorena García, Hospital Universitario Nuestra Señora de Candelaria Carrion Valencia Almudena, Adrián Amador Robayna, Cristina Gomez de Segura, Marta Jiménez Navarro, Sara González Nieto, Lara Torres León, Raquel Clemente Graffigna, Jonathan Rodríguez Talavera
Petros Grivas, University of Washington Shilpa Gupta, Sumanta K. Pal, Diederik M. Somford, Srikala, Joaquim Bellmunt Molins, Guru Sonpavde, Mark Fleming, Seth P. Lerner, Yohann Loriot, Sijian Ge, Corina Andresen, Florian Roghmann, Maria De Santis, Siamak Daneshmand
Introduction & Objectives: Immunotherapy has become a therapeutic strategy that has changed the natural history of several tumors. Data on the expression of PDL-1 among other immunological biomarkers, have allowed the development of modern immunotherapeutics in the treatment of urothelial cancer for those patients whose disease progresses after chemotherapy treatment or are not Fit for it.
Introduction & Objectives: Radical surgery is the mainstay of treatment for urothelial bladder cancer (UBC) and invasive upper tract urothelial cancer (UTUC) but, even when surgery is combined with cisplatin-based (neo)adjuvant chemotherapy, recurrence rates are high. Many patients are ineligible for, do not respond to cisplatin-based chemotherapy, or refuse this treatment because of toxicity concerns. Fibroblast growth factor receptor 3 (FGFR3) genetic alterations occur in 60% of UTUC and 21–38% of muscleinvasive UBC and are candidates for targeted therapy. Infigratinib (BGJ398) is an ATP-competitive FGFR1–3 selective oral tyrosine kinase inhibitor that has shown clinical activity and tolerability in patients with advanced urothelial carcinoma (UC) harboring susceptible FGFR3 alterations. PROOF 302 was designed to investigate the efficacy and safety of adjuvant infigratinib versus placebo in patients with high-risk, muscle-invasive UC and susceptible FGFR3 alterations.
Materials & Methods: We present our series of 15 patients treated with immunotherapy for urothelial cancer. Review of the characteristics of the sample, toxicity and overall survival. Results: 15 patients with a mean age of 69 years, in a range between 55 and 85 years, have been treated. 14 males and 1 female. 14 of the cases have their origin in the bladder and one in the renal pelvis. Most are ECOG 0-1. Out of the 14 patients with bladder cancer, 12 with stage IV: 7 underwent cystectomy. 4 were treated with immunotherapy for being unfit for chemotherapy regimens and 10 after disease progression. The patient with urothelial cancer of the upper urinary tract was treated with chemotherapy and immunotherapy was indicated after progression. 6 of the patients have died. The average overall survival of these patients was 2.5 months. 10 months for nivolumab, 5 months for pembrolizumab, and 1.5 months for atezolizumab. The immunotherapy associated toxicity profile is related to the immune system. Most had fatigue, a diabetic debut?? and skin reactions were also observed. Three serious adverse reactions have been detected that required discontinuing the treatment. Pulmonary embolism with Pembrolizumab, a colitis that required admission and severe cholangitis. Immunotherapy toxicity requires multidisciplinary management, supported by endocrinology, dermatology, pulmonology, digestive, etc.
Materials & Methods: PROOF 302 is a randomized, doubleblind, placebo-controlled, phase 3 trial (NCT04197986). Eligible patients have high-risk, muscle-invasive UTUC or UBC (85% and 15%, respectively) with susceptible FGFR3 alterations (activating mutations, gene fusions, or rearrangements by FoundationOne® CDx test), and are ≤120 days following nephroureterectomy, distal ureterectomy, or cystectomy. The protocol was amended to include patients with carcinoma in situ, and those ineligible for or refusing cisplatin-based chemotherapy. Pathologic disease stage is ≥ypT2 and/or yN+ M0 after cisplatin-based neoadjuvant chemotherapy. For patients who are ineligible for, or have refused, cisplatin-based therapy, pathologic disease stage is: UTUC (≥pT2 pN0–2 M0 [post-lymphadenectomy or no lymphadenectomy]); UBC (≥pT3 or pN+). Patients are randomized 1:1 to receive oral infigratinib 125 mg or placebo, QD on days 1–21 every 28 days for up to 52 weeks (13 cycles) or until recurrence, unacceptable toxicity, or death. Primary endpoint: centrally reviewed disease-free survival (DFS), analyzed via stratified log-rank test, and Cox model (hazard ratio [HR]). Secondary endpoints: investigator-reviewed DFS; metastasisfree survival; overall survival; safety/tolerability. Exploratory endpoints: quality of life; pharmacokinetics, determination of the prevalence of genomic alterations and correlation with features of UC; genomic and proteomic assessment of tumor tissue and cell-free DNA samples from baseline and at recurrence to identify the predictive/prognostic value of biomarkers. Based on the HR of 0.5 for infigratinib vs. placebo, an interim analysis for adaptation and futility will be conducted after ~35 centrally confirmed DFS events. The initial sample size of 218 patients may be increased up to 328 patients based on an adaptive design. The power to detect a difference in DFS is 80%.
Conclusion: The majority of toxicities observed are Grade I (fatigue). Treatment has been discontinued in 3 of the cases due to severe toxicity (colitis, cholangitis and PE). The immunotherapy toxicity profile requires multidisciplinary management (dermatology, endocrinology, etc.). In our experience, the overall survival is 2.5 months on median. Underway Clinical trials working hand in hand with translational research allow us to continue defining the role of immunotherapy in this type of tumors.
Conclusion: Using newer techniques such as VMAT, toxicity after adjuvant EBRT is acceptable. Preliminary survival data are encouraging and underline the importance of further evaluating the place of adjuvant EBRT in patients with high-risk and poor prognostic MIBC characteristics.
Results: PROOF 302 was initiated in March 2020 and is open internationally and expected to end in 2024.
10
11
8
9
Perioperative enfortumab vedotin plus pembrolizumab versus chemotherapy in cisplatin-eligible patients with muscle invasive bladder cancer (MIBC): randomized phase 3 KEYNOTE-B15/EV-304 study
10
Platin-based neoadjuvant chemotherapy for muscle invasive bladder cancer
Prognostic value of pretreatment neutrophil-to-lymphocyte ratio and status PD-L1 for pembrolizumab in second-line in metastatic urothelial carcinoma in a Portuguese center – real world data
Luísa Leal-Costa, Hospital Beatriz Ângelo Diana Silva, Pedro Simões, Carlota Sofia Vieira Baptista, Madalena Machete, João Moreira-Pinto, João Paulo Baptista Godinho, Ana Faria, José Alberto Teixeira, José Luís Passos Coelho
Christopher J Hoimes, Duke University Jens Bedke, Yohann Loriot, Hiroyuki Nishiyama, Xiao Fang, Ritesh Kataria, Blanca Homet Moreno, Matthew Galsky
Lúcia Moreira Gil, Centro Hospitalar de Lisboa Central Ana Filipa Reis, Ana Sofia Chantre Spencer, Diana Simão, Mariana Sardinha, Chiara Rodriguez, Ricardo Jorge Martins Da Luz
Introduction & Objectives: Neoadjuvant chemotherapy (NAC) followed by radical cystectomy (RC) and pelvic lymph node dissection is standard treatment for non-metastatic muscleinvasive bladder cancer (MIBC). Gemcitabine-cisplatin (GC) has similar efficacy to the MVAC regimen with lower toxicity in advanced BC and both regimens are considered adequate options for NAC. We report the experience of our centre with NAC for MIBC.
Introduction & Objectives: Although neoadjuvant cisplatinbased chemotherapy followed by radical cystectomy and pelvic lymph node dissection (RC+PLND) is standard of care for patients with MIBC, patients experience disease recurrence or progression. The PD-1 inhibitor pembrolizumab has been effective for cisplatin-ineligible and platinum-resistant/refractory metastatic urothelial carcinoma (mUC) and Bacillus Calmette-Guerin– unresponsive high-risk non–muscle-invasive bladder cancer. Enfortumab vedotin (EV) is a Nectin-4–directed antibody-drug conjugate comprising a fully human, monoclonal antibody and the microtubule-disrupting agent monomethyl auristatin E, which has been effective in patients with mUC who previously received a PD-1/PD-L1 inhibitor. Results of KEYNOTE-869/EV-103, a phase 1/2 study (NCT03288545), showed that EV + pembrolizumab had encouraging antitumor activity and acceptable safety as first-line treatment for cisplatin-ineligible patients with metastatic urothelial cancer, suggesting that combination therapy might be promising (Rosenberg JE et al. J Clin Oncol. 2020;38[15 suppl]:5044).
Introduction & Objectives: According to the KEYNOTE-045 Phase III trial, pembrolizumab is approved for the treatment of patients with locally advanced or metastatic urothelial carcinoma (mUC) that progressed after the first-line with platinum-based chemotherapy with benefit in OS, PFS and ORR, regardless of PD-L1 expression. However, in clinical practice, pembrolizumab is effective in a limited number of patients with mUC and the role of PD-L1 expression in second-line therapy is uncertain. Therefore, the development of clinical biomarkers with prognostic value is essential to personalize the choice of treatments and improve outcomes. The aim of this retrospective study is to evaluate the prognostic and predictive value of neutrophil/lymphocyte ratio (NLR) and PD-L1 expression as clinical biomarkers in the patients with metastatic urothelial carcinoma treated with pembrolizumab in second-line.
Materials & Methods: We conducted an observational, retrospective, single-centre cohort study. All patients (pts) with non-metastatic MIBC (≥cT2) after initial transurethral resection of the bladder tumour (TURBT), treated with GC between 2012 and 2020 were included. The primary objective was to evaluate pathological complete response (pCR) rates after NAC, defined as ypT0N0 or ypTisN0. We also assessed disease free survival (DFS) and overall survival (OS) as secondary objectives. Results: Thirty-nine pts with MIBC were treated with NAC followed by RC between 2012 and 2020. The median age was 66yo (range, 46-79), 31 (80%) were males. At the pre-NAC staging, 36 tumours were T2 (92%), 2 T3 (5%) and 1 T4 (3%). Thirty-four pts received the full 4 cycles of NAC; 11 pts (28%) had to delay at least 1 cycle of NAC due to toxicity and 4 pts (10%) required dose reductions. Eleven pts (28%) had grade 3 or 4 toxicities, mainly hematological (73%). The pCR rate was 28% (11 pts). With a median follow-up of 57 months, 17 pts (44%) had tumour recurrence, 1 pt after pCR, and 14 pts (36%) died, none of those with pCR. The association between pCR and DFS (median 22 months if residual disease versus not reached if pCR, log rank p=0.017) and between pCR and OS (median not reached for both groups, log rank p=0.01) were confirmed.
Materials & Methods: KEYNOTE-B15/EV-304 (NCT04700124) is a randomized, open-label, phase 3 study to evaluate the efficacy and safety of perioperative EV + pembrolizumab versus neoadjuvant chemotherapy using gemcitabine/cisplatin in cisplatin-eligible patients with MIBC. Approximately 784 patients will be randomly assigned 1:1 to receive either 4 cycles of neoadjuvant EV + pembrolizumab followed by 5 cycles of adjuvant EV + 13 cycles of adjuvant pembrolizumab after RC+PLND or 4 cycles of neoadjuvant cisplatin-based chemotherapy followed by observation after RC+PLND. Neoadjuvant and adjuvant pembrolizumab 200 mg + EV 1.25 mg/kg will be administered intravenously every 3 weeks (Q3W), and neoadjuvant chemotherapy will consist of gemcitabine 1000 mg/m2+ cisplatin 70 mg/m2 Q3W. Randomization will be stratified by centrally determined (pathology or imaging) initial T and N stage (T2N0 or T3/T4aN0 or T1-T4aN1), PD-L1 combined positive score (CPS ≥10 or CPS <10), and geographic region (United States or Europe or most of world). Patients must have histologically confirmed urothelial cancer/MIBC (clinical stage T2T4aN0M0 or T1-T4aN1M0) with predominant (≥50%) urothelial histology, have nonmetastatic disease (≥N2 disease and/or M1 excluded) confirmed by blinded independent central review (BICR), have ECOG PS 0 or 1, and not have previously received systemic therapy for MIBC. Imaging (CT or MRI) will be performed ≤6 weeks before cystectomy and 6 weeks after cystectomy. After postcystectomy imaging, additional imaging will be performed Q12W up to the end of year 2 (week 96) and at discontinuation. In year 3 and beyond, imaging will be performed Q24W. Primary end points are pathologic complete response and event-free survival by BICR. Secondary end points are overall survival, disease-free survival, pathologic downstaging, and safety and tolerability.
Materials & Methods: We analyzed retrospectively 16 patients with metastatic urothelial carcinoma refractory to platinumbased chemotherapy who received second-line pembrolizumab between December 2017 and March 2021. Pretreatment NLR was calculated by dividing the number of neutrophils by the number of lymphocytes in peripheral blood, before the first administration of pembrolizumab and the PD-L1 expression was evaluated in the tumor specimen (CPS). Progression-free survival (PFS) and overall survival (OS) were compared by the Kaplan-Meier method and the Cox proportional hazard model. Results: Overall 80% are male, with a median age of 70 (range: 55-82) years. Patients with NLR ≤ 5 before starting treatment with pembrolizumab had a median PFS of 13 months (7.71-14.57) and those with NLR > 5 was 8 months (4.07-10.35 ) (p < 0.001). In the multivariate analysis, the presence of bone metastases (HR: 3.01, 95% CI: 1.73-5.61, p = 0.021) and NLR> 5 (HR: 2.97, 95% CI: 2, 699.02, p<0.001) were independent predictors for shorter PFS and lower ORR. When the same analysis was performed for OS, it was not statistically significant, however, NLR ≤ 5 pretreatment with pembrolizumab and PD-L1 CPS > 10 were associated with better OS (p < 0.027) and ORR (p < 0.001).
Conclusion: In our single institution cohort, the observed pCR rate (28%) was lower than that reported in the phase III GETUG/ AFU V05 VESPER trial comparing dose-dense MVAC with GC (42% vs 36%). However, we also documented a positive association between pCR and both DFS and OS. While less toxic than MVAC, the full 12-week NAC with GC was only tolerated without treatment delays or dose reductions in 23 of the 39 pts.
Conclusion: These data suggest that pretreatment NLR can estimate the ORR, PFS and OS of patients with mUC treated with second-line pembrolizumab and that NLR ≤ 5 is a potential biomarker predicting response to second-line pembrolizumab as well as PD -L1 CPS > 10 and it may represent accessible tools in clinical practice to identify appropriate patients for second-line treatment with pembrolizumab. Prospective studies and clinical trials are needed to validate NLR as predictive biomarker of response to checkpoint inhibitors in mUC.
Results: The KEYNOTE-B15/EV-304 study is enrolling in Africa, Asia, Australia, Europe, and North America. Conclusion: Results of KEYNOTE-B15/EV-304 will provide further clarity on the efficacy of perioperative EV + pembrolizumab in cisplatin-eligible patients with MIBC.
12
13
11
12
The evolution of intravesical therapy for bladder cancer
Open (ORC) versus intracorporeal robotic-assisted radical cystectomy (iRARC) for bladder cancer: a high volume specialist centre experience
Cecile Pham, Northern Beaches Hospital Jordan Cohen
Mutie Raslan, University of Leeds
Introduction & Objectives: Intravesical therapy is currently used to prevent disease recurrence following transurethral resection of bladder tumours for superficial non-muscle invasive bladder cancer (NMIBC). We review the history and development of these agents.
13
Table 1. iRARC
ORC
Overall survival of patients receiving cisplatin and carboplatin for primary metastatic urothelial carcinoma of the bladder: a contemporary Dutch nationwide cohort study
Gender Male
61 (74%)
100 (76.3%)
Mean age (43-85)
69
70
Mean BMI (16-47)
27
28
38/82 (46.3%)
60/131 (45.8%)
MIBC (%)
Anke Richters, The Netherlands Comprehensive Cancer Organisation Introduction & Objectives: Radical cystectomy is the gold standard treatment for aggressive bladder cancer. Traditionally, this has been carried out as an open procedure, however, there is a growing body of evidence to suggest that intracorporeal robotic approach may improve perioperative outcomes. The aim of this study was to examine early- and medium-term outcomes for patients undergoing iRARC and ORC at a single specialist centre.
Materials & Methods: A systematic literature search using MedLine, Embase, and secondary historical sources was performed to identify the key historical medical advances in intravesical therapy. Results: Mitomycin-C was first conceived as an antibiotic derived from streptomyces species but was later found to have antineoplastic properties. This discovery by T. Hata with S. Wakagi in Japan in the late 1950’s led to the first intravesical injections. Thiotepa, an alkylating agent and organophosphate, was first synthesised in 1952 after the mutagenic effects of mustard gas were discovered in World War I. J. Bateman of the USA trialled its injections into cavities for breast and ovarian cancer, leading to intravesical use in 1961 by H.C. Jones and J. Swinney. However, the use of thiotepa was superseded by mitomycin-C after K. Shida and his team published their positive findings in 1967. J.D. Coe and J.E. Feldman (1965), and Bloomberg (1975) in the USA demonstrated the effect of Bacillus Calmette-Guerin (BCG) toxin on guinea pig and dog bladders, respectively. Following their success, A. Morales of Canada trialled intravesical BCG in humans in 1976. This revolutionised the treatment of high risk NMIBC with one of the earliest forms of immunotherapy, which remains the most efficacious therapy to date.
Materials & Methods: Patients undergoing curative resection for muscle-invasive (MIBC) or high-risk non muscle invasive bladder cancer (HR-NMIBC) at a high-volume tertiary centre in the United Kingdom, were identified from a prospectively maintained database. Outcomes of interest included perioperative complications, and overall survival. Cumulative survival rates were calculated using the Kaplan Meir method and Logrank test was used to compare survival.
Table 2. Peri-operative and medium-term surgical outcome comparison: Introduction & Objectives: Cisplatin is preferred to carboplatin when treating metastatic urothelial carcinoma of the bladder (mUCB), despite its greater toxicity. Randomised studies underpinning this have been performed in non-contemporary populations with limitations in sample sizes and analyses, affecting their validity in current clinical practice. The objective of this study is to estimate overall survival (OS) and assess the benefit of cisplatin-based regimens over carboplatin-based regimens in a contemporary cohort of patients with mUCB.
P-value Clavien-Dindo complications >=3 (%)
5/82 (6.1%)
19/131 (14.5%)
0.055
Blood transfusion rate (%)
3/82 (3.7%)
21/131 (16.0%)
<0.05
Median hospital stay
8 days (3-50)
10 days (3-80)
0.067
5/82 (6%)
11/131 (8.4%)
0.53
17
14
0.83
5/82 (6%)
6/131 (4.6%)
0.62
Positive surgical margin Mean LN yield Uretero-ileal anastomotic stricture
Materials & Methods: A nationwide retrospective cohort study of patients diagnosed with de novo mUCB in the Netherlands between 2016 and 2019 who underwent first-line treatment with cisplatin- or carboplatin-based chemotherapy, based on data from the Netherlands Cancer Registry. A propensity model for receiving cisplatin-based chemotherapy based on age, sex, age-adjusted Charlson Comorbidity Index, renal function, performance status, serum haemoglobin, and the presence of visceral and bone metastases was used to produce inverse probability weights (IPW) per patient. Unadjusted and IPWadjusted Kaplan–Meier OS curves of both chemotherapy groups were compared by restricted mean survival times (RMST).
Results: Overall, 213 patients were identiQed and included in the study (iRARC: 82 and ORC: 131). Patient and disease demographics between the two groups were comparable (Table 1). Between the two groups, iRARC group had lower risk of high-grade complications (6.1% vs 14.5%), lower blood transfusion rates (3.7% vs 16%) and shorter median hospital stay (8 vs 10 days). There was no statistical difference between the groups for positive margins, lymph node (LN) yield and uretero-ileal stricture rates (Table 2). Thirty-day mortality for iRARC and open cystectomy was 2.4% vs 3%. The overall 1-year survival for the iRARC and ORC group was 86.7% and 80.9% respectively (P=0.83, see figure).
Conclusion: The rapid global development of intravesical therapy since the 1950’s has created agents that remain standard of care today. While new therapeutic agents are being trialled, the importance of these discoveries and their stories should be reflected upon as we have the privilege to deliver these treatments today.
Results: Of the 1041 patients with mUCB, 359 received either cisplatin (n = 170; 47%) or carboplatin (n = 189; 53%) first line. The cisplatin group was younger, had fewer comorbidities, and had better performance status and renal function. The median OS in the cisplatin and carboplatin groups was 13.1 and 11.5 months, respectively. After IPW-adjustment, prognostic factors were balanced between the two chemotherapy groups (standardised differences <0.1) and differences in RMST were <2.0 months and not statistically significant up to 24 months.
Conclusion: This single centre experience provides constructive data to suggest iRARC leads to favourable short-term outcomes compared to ORC. Of note, there were lower rates of high-grade complications, requirement for postoperative blood transfusion and shorter hospital stay. There was no significant difference in 1-year overall survival or development of uretero-ileal anastomotic stricture.
14
Conclusion: After accounting for all known prognostic factors, we found no significant survival benefit for cisplatin over carboplatin as a first-line chemotherapy in mUCB.
15
14
15
Specific optical spectral tumor markers of bladder cancer
Preoperative neutrophil-tolymphocyte ratio predicting outcome in muscle invasive bladder cancer undergoing radical cystectomy
Gilmanova Rita Flaridovna, Federal State Budgetary Educational Institution of Higher Education "Bashkir State Medical University" of the Ministry of Healthcare of the Russian Federation, Ufa
Diana Simão, Centro Hospitalar de Lisboa Central Mariana Sardinha, Ana Filipa Reis, Ana Sofia Chantre Spencer, Lúcia Moreira Gil, Chiara Rodriguez, Ricardo Jorge Martins Da Luz
Introduction & Objectives: A fundamental factor in the classification of bladder cancer is the involvement of the muscle layer in the pathological process. Despite the progress achieved in the morphological diagnosis of bladder cancer, the issues of scientific support for the creation of new high-precision and effective methods for early diagnosis are an urgent problem of modern oncourology. Purposes of the study: determination of optical markers of bladder cancer using Raman spectroscopy.
Introduction & Objectives: Radical cystectomy (RC) is the standard treatment for patients with muscle-invasive bladder cancer (MIBC) and no evidence of metastasis. Despite surgical treatment and improvements in neoadjuvant chemotherapy, a significant proportion of patients develop disease recurrence. Recent studies report neutrophil-to-lymphocyte ratio (NLR) as an useful biomarker of systemic inflammatory response in several cancer types. NLR has been proposed as a prognostic factor for patients with MIBC following RC. The aim of our study was to evaluate the clinical usefulness of preoperative NLR as a prognostic marker in patients treated with RC for MIBC.
Materials & Methods: We studied the material obtained from 85 patients diagnosed with bladder cancer (male –65, female – 20), who were examined and treated at the oncology department of the BSMU Clinic. Tumor tissues of the urinary bladder removed during TUR of the bladder and radical cystoprostatvesiculectomy were examined. Tumor tissue sample: without sample preparation. The study of the samples was carried out on a Horiba Scientific apparatus. Configuration: wavelength 785 nm, grating 1200 gr/mm, filter 100%, confocal aperture 300 μm. Integration time 50 s. Spectral data analysis was carried out using Spectragryph software.
Materials & Methods: A retrospective analysis was performed at our institution between January 2013 and January 2021. Baseline clinical, demographic and histopathological features and related outcomes are reported. We established a NLR cut-off value of 3 based on previous publications. Disease-free survival (DFS) and overall survival (OS) were estimated by Kaplan-Meier method, and the log-rank test was used to determine significance. Multivariate analysis was performed based on these survival endpoints using a Cox regression. Statistics were evaluated with IBM™ SPSS software, version 23.
Results: Recurrence after 3 months was detected in 2 patients (primary histology peaks corresponded to T1G3. Secondary histology - T2G3). Peaks: high peaks 564 cm-1, 1450 cm-1, 1650 cm-1, 850 cm-1, 2930 cm-1. Recurrence after 6 months - in 5 patients (2 patients T1G3: 1 patient - T1G3, 2 patients - transfer to T2G3, after 12 months in both of them no recurrence was not verified, 3 patients - T2G3: 2 patients completed radical cystectomy and 1 evisceration). Peaks: Patient 2 - transfer to T2G3: initially high peaks at 564 cm-1, 865 cm-1, 1450 cm-1, 1650 cm-1, 850 cm-1, 2935 cm-1 (primary histology matched T2G3 peaks). 3 patients - T2G3 - 2 radical cystectomy and 1 evisceration: 565 cm1, 655 cm-1, 850 cm-1, 1420 cm-1, 1450 cm-1, 1650 cm-1, 2290 cm-1, 2870 cm -1, 2930 cm-1, 3120 cm-1, 3254 cm-1, 3150 cm-1 (initially histologically T2G3, MRI revealed no germination, primary histology had peaks T3-T4G3). Recurrence after 12 months - 3 patients (2 patients initially T2G3, 1 patient with primary histology T1G3 – recurrence corresponded to multifocal lesion - the patient completed radical cystectomy). Patient T1G3: 560 cm-1, 1450 cm1, 1650 cm-1, 2930 cm-1, 3055 cm-1, 3120 cm-1 (peaks from T34G3).
Results: 120 patients were included, of which 103 men (86%) and 17 women (14%). Median age at diagnosis was 70 years (range: 40-86). 55 (46%) patients were active smokers, 37 (31%) former smokers and 28 (23%) never smoked. Median NLR was 2.6. No association was found between NLR and age (p=0.12), gender (p=0.38), smoking status (p=0.45) or comorbidities assessed by Charlson Index (p=0.42). Statistically significant associations were found between NLR > 3 and advanced pathological tumour stages (p=0.02) and lymphovascular invasion (p=0.01). Survival analysis showed that NLR > 3 was associated with worse DFS (29.6 vs 61.2 months, p=0.001) and OS (36.3 vs 66.0 months, p=0.002). In multivariate analysis, a NLR > 3 was independently associated with both DFS (HR 2.18; 95% CI 1.25-3.82, p=0.006) and OS (HR 2.22; 95% CI 1.24–3.97, p=0.007).
Conclusion: Raman spectroscopy can be used in the early diagnosis of bladder cancer, to confirm the fact of tumor invasion, in the algorithm for treating patients.
Conclusion: MIBC is associated with high morbidity and mortality. Consequently, there is a need to identify predictors of survival. Our results suggest that patients with preoperative NLR > 3 undergoing RC for MIBC have worse outcomes, both DFS and OS. As an easy achievable and not expensive biomarker, NLR can be useful and needs to be validated in risk stratification models.
16
17
SGNTUC-019: phase 2 basket study of tucatinib and trastuzumab in previously treated solid tumors with HER2 alterations: urothelial cancer cohort (trial in progress)
Analysing cause of death during follow-up for non-muscle invasive bladder cancer: is there a role for watchful waiting? Yuhao Zhang, Stepping Hill Hospital Mahmoud Nosseir, James Dyer
Evan Yu, University of Washington Jorge Ramos, Sherry Tan, Matthew Galsky
Introduction & Objectives: Most patients with bladder cancer have non muscle-invasive disease (NMIBC), requiring multiple invasive procedures during follow-up. Surveillance guidelines do not adjust for increasing age or frailty. We aim to evaluate the influence of frailty on the natural history of NMIBC.
Introduction & Objectives: Tucatinib, a highly selective HER2directed tyrosine kinase inhibitor approved in multiple regions for HER2+ metastatic breast cancer, is being investigated as a novel therapy for patients with metastatic colorectal cancer, gastric cancer, and other GI tumors. In xenograft models of HER2+ and HER2-mutated tumors, dual targeting of HER2 with tucatinib and trastuzumab showed superior activity to either alone (Kulukian 2020; Peterson 2020). Despite development of several new therapies for metastatic urothelial cancer, response durations generally remain short and the majority of patients succumb to the disease, highlighting the need for additional therapeutic approaches. Given 20-30% of metastatic urothelial cancers have molecular alterations of the ErbB family, tucatinib + trastuzumab warrants further evaluation. SGNTUC-019 (NCT04579380) is a multi-cohort, open-label, international Phase 2 study evaluating tucatinib + trastuzumab in patients with previously treated solid tumors displaying HER2+ or HER2-mutated solid tumors, including a cohort with locally advanced or metastatic urothelial cancer (UC).
Materials & Methods: Patients who died between 1/1/20171/1/2020 with bladder cancer and underwent a flexible cystoscopy within two years were identified via clinical coding. Notes were reviewed to establish relevant clinical parameters and follow-up procedures. Frailty was assessed using the 9-point Rockwood Clinical Frailty Scale (CFS): 1-3 no frailty, 4 vulnerable, 5-9 mild/severe frailty. The Charlson co-morbidity index and American Society of Anesthesiologists (ASA) score were similarly categorised. Results: 69 patients were identified (mean age 78) with a median overall survival (OS) of 32.5 months. 36, 20 and 13 patients had frailty scores of 1-3, 4, and 5-9, respectively. There was no difference in the proportion of higher risk NMIBC between groups (table 1). Increasing frailty was associated with reduced OS (58.5, 28.5, 13 months; p <0.05) but not recurrence-free survival (24, 16, 27 months; p =0.98) or progress-free survival (144, n/a, n/a months; p =0.58) (figure 1). There was no clear difference in the number procedures per year of follow-up between groups (table 2). Progression or disease-specific mortality was the same across all groups (table 2). Similar results were obtained for the Charlson index and ASA scores. There were no frail patients with low or intermediate NMIBC who had a clinically significant progression prior to death from other causes (table 3).
Materials & Methods: Eligible patients must have HER2+ or HER2-mutated locally advanced or metastatic solid tumors, with progression on or after the last systemic therapy for advanced disease. Additionally, an ECOG PS ≤1, adequate hepatic, hematological, renal, and cardiac function, and no prior exposure to HER2-directed therapy in the UC cohort. For eligibility, HER2 alterations can be demonstrated by HER2 overexpression/ amplification in tumor tissue by prior IHC/ISH, or by HER2 amplification/mutation in prior or pre‑screening NGS assay of ctDNA or prior tissue NGS assay. The HER2 overexpression/ amplification UC cohort will enroll 12 response-evaluable patients per RECIST 1.1. If ≥2 responses are observed, the cohort will be expanded to 30. Patients with HER2-mutated UC will be enrolled into a cohort of 30 patients with HER2-mutated solid tumors (except breast and non-squamous non‑small cell lung cancers). The primary objective is antitumor activity, with confirmed ORR as the primary endpoint, and disease control rate, duration of response, PFS, and OS as secondary endpoints. Patients will receive tucatinib 300 mg orally twice daily and trastuzumab 8 mg/kg intravenous on Cycle 1 Day 1 and 6 mg/kg q21 days from Cycle 2 Day 1. Disease assessments per RECIST 1.1 will occur q6 weeks for 24 weeks, then q12 weeks. Trough concentrations of tucatinib will be evaluated in Cycles 2-6, with a peak concentration sampled in Cycle 3. Quality of life will be evaluated q2 cycle using EQ-5D-5L.
Conclusion: There are frail patients with NMIBC who have limited life expectancy. For such patients with lower risk NMIBC, protocoldriven cystoscopic surveillance may not improve survival and watchful waiting may be more appropriate. Further investigation is required to determine the feasibility of this approach.
Results: Sites are currently enrolling within the US, EU, and Asia.
16
17
18
19
Zongtai Zheng, Shanghai Tenth People’s Hospital, Tongji University School of Medicine Xudong Yao
Zongtai Zheng, Shanghai Tenth People’s Hospital, Tongji University School of Medicine Xudong Yao
Introduction & Objectives: Muscle-invasive bladder cancer (MIBC) is highly heterogeneous with distinct molecular subtypes. We aimed to investigate the heterogeneity of different molecular subtypes from a tumor microenvironment perspective and develop a model to predict the molecular subtype, prognosis and immunotherapy response.
Introduction & Objectives: The Ki67 expression is associated with the malignant characteristics of bladder cancer (BCa) and can only be postoperatively detected by immunohistochemistry. We aimed to develop and validate Magnetic Resonance Imaging (MRI)-based radiomics signatures to preoperatively predict the Ki67 expression status and survival outcome in BCa.
Materials & Methods: MIBC patients in The Cancer Genome Atlas (TCGA) and IMvigor210 were classified into luminal and basal subtypes according to the UNC classification. The proportions of tumor-infiltrating immune cells (TIICs) were evaluated through The Cell Type Identification by Estimating Relative Subsets of RNA Transcripts algorithm. Immune-related genes that were differently expressed between luminal and basal subtypes and associated with prognosis were selected to develop the immune prognostic signature (IPS). Nomogram based on the IPS was developed. Functional enrichment analysis (GSEA) was performed on the IPS. In addition, the RNA-sequence data of ten MIBC samples in our center were utilized to validate hub genes expression between molecular subtypes.
Materials & Methods: We retrospectively collected 179 BCa patients (41 low Ki67 expression, 138 high Ki67 expression) with preoperative MRI. Patients were randomly allocated into training and validation sets based on a 7:3 ratio. Axial T2-weighted images (T2WI) and dynamic contrast-enhancement (DCE) were used for radiomics features extraction. The synthetic minority oversampling technique (SMOTE) was used to balance the low Ki67 expression group. After feature filtering and feature selection, four radiomic signatures were developed in the training set and assessed in the validation set. The predictive performance of radiomics signatures was assessed by the area under the curve (AUC) and accuracy. The decision curve analyses and calibration curve and were used to investigate the clinical usefulness and calibration of the radiomics signature, respectively. Univariate and multivariate Cox regression analyses were used to obtain independent predictors for Ki67 expression status prediction. Kaplan-Meier test and Cox analyses were performed to investigate the prognostic value of radiomics-predicted Ki67 expression status.
Development and validation of a molecular subtype-associated immune prognostic signature for muscle invasive bladder cancer
Figure 1: Kaplan-Meier analyses comparing Rockwood clinical frailty scale (CFS), Charlson co-morbidity index (Ch), and American Society of Anesthesiologists (ASA) physical status score categories with respect to overall survival, recurrence-free survival, and progression-free survival. Survival time is shown in months. Hashed marks represent censored data. P-values are shown for each analysis.
≤3
CFS 4
≥5
≤5
Charlson 6-7
≥8
≤2
ASA 3
≥4
37
19
13
21
30
18
21
31
9
Mean age, years
76.2
78.7
82.3
71.6
79.4
83.4
76.1
79.4
79.4
Median age, years Age standard deviation, years Male sex, %
78.1
80.0
84.3
70.5
80.2
84.1
78.0
81.8
78.8
9.08
7.62
8.81
6.99
9.03
5.55
8.67
8.84
9.21
78.4
73.7
76.9
71.4
80
77.8
75.9
77.4
77.8
Higher risk, % (p-value)
59.5
47.4
69.2
57.1
56.7
61.1
48.3
61.3
77.8
ref
(0.411)
(0.742)
ref
(1)
(1)
ref
(0.437)
(0.148)
Number, n
CFS Charlson The total study population ASA (N Table 1: Demographics and characteristics of the study population. ≤3way of Rockwood 4 ≥59-point≤5 6 - 7 scale≥8(CFS), Charlson ≤2 3 ≥4 =69) was categorised by clinical frailty co-morbidity index, and Amercian Society of Anesthesiologists (ASA) risk” NMIBC 58 25 13 59 Score. 38.5“Higher20 60 is defined 30 as EAU 21 OS, years “high” or “very high” risk [13].(<0.01 Comparisons were made between (“ref”) <0.01 the healthiest reference <0.01 categories against the others; p-values were calculated using Fisher’s exact test. 24 16 27 26 21 25 26 24 16 RFS, years 0.98
PDS, years
144
n/a
0.76 n/a
144
0.58
n/a
0.53 n/a
n/a
0.39
144
n/a
Table 2: Median overall survival (OS), recurrence-free survival (RFS), and progression-free survival (PFS) are shown in years. The rates of surveillance and non-surveillance flexible cystoscopies are shown per year. The proportions of bladder cancer (“BCa”) deaths and/or progressions (“prog”) are shown. Comparisons were made between the healthiest reference (“ref”) categories against the others. Survival analysis performed using Kaplan-Meier for OS, RFS, and PFS with associated p-values shown. P-values were calculated using Student’s T-test for continuous and Fisher’s exact test for categorical variables.
Results: Half of TIICs were differently distributed between luminal and basal subtypes. The IPS was highly associated with molecular subtypes, critical immune checkpoint genes expression, prognoses and immunotherapy response. Several validation data sets (GSE32894, GSE31684, GSE13507 and GSE48277) and meta-analysis further validated the prognostic value of IPS. GSEA revealed that some oncogenic pathways were coenriched in the high risk group. An IPS-based nomogram was constructed for prognostic predictions with good calibration and clinical usefulness. The expression of hub genes was significantly different between luminal and basal subtypes in MIBC samples from our center.
Results: A 38-feature-based support vector machine model achieved the best predictive performance in the SMOTE-balanced training (AUC, 0.978; accuracy, 91.19%) and validation sets (AUC, 0.951; accuracy, 84.34%) with a good calibration performance and clinical usefulness. Multivariate logistic regression showed that radiomics signature was an independent predictor for Ki67 expression prediction. Immunohistochemistry-based high Ki67 expression and radiomics-predicted high Ki67 expression were significantly associated with poor disease-free survival (P=0.004, 0.026, respectively).
Conclusion: Dysregulation of TIICs contributed to the heterogeneity between luminal and basal subtypes. The IPS can facilitate molecular subtyping, prognostic evaluation and personalized immunotherapy.
Conclusion: A radiomics signature could predict the Ki67 expression status and the prognoses of the BCa patients, thereby may aid in clinical decision-making.
0.65
Surveillance, n/years
1.73
0.310
1.08
2.00
1.32
1.08
1.78
1.16
1.47
ref
<0.01
0.105
ref
0.206
0.112
ref
0.128
0.563
Nonsurveillance, n/years BCa death, %
0.536
0.404
0.261
0.664
0.370
0.325
0.430
0.525
0.240
ref
0.323
0.102
ref
0.0263
0.0278
ref
0.432
0.307
18.9
5.26
15.4
28.6
10.0
5.6
20.7
9.68
11.1
ref
0.243
1
ref
0.136
0.0985
ref
0.437
0.148
BCa death/Prog, %
Table 1: Demographics and characteristics of the study population. The total study population (N =69) was categorised by way of Rockwood 9-point clinical frailty scale (CFS), Charlson co-morbidity index, and Amercian Society of Anesthesiologists (ASA) Score. “Higher risk” NMIBC is defined as EAU “high” or “very high” risk [13]. Comparisons were made between the healthiest reference (“ref”) categories against the others; p-values were calculated using Fisher’s exact test.
Magnetic resonance imaging-based radiomics signature for preoperative prediction of Ki67 expression and outcome in bladder cancer
29.7
15.8
15.4
33.3
20.0
16.7
20.7
25.8
22.2
ref
0.3377
0.4686
ref
0.3382
0.2898
ref
0.7634
1
2: Median Bladder Bladder Cancer Recurrence Table overallCancer survivalDeath/Progression (OS), recurrence-free survival (RFS), and progression-free survival (PFS) are shown years. The ratesHigher of surveillance and non-surveillance LowerinRisk Risk Lower Risk flexible cystoscopies Higher Risk are shown per year. The proportions of bladder cancer (“BCa”) deaths and/or progressions (“prog”) are 10/21 (47.6%) 11/16 (68.8%) 14/21 (66.7%) CFS ≤3 2/16 (12.5%) shown. Comparisons were made between the healthiest reference (“ref”) categories against the 3/19 (15.8%) 2/13 (15.4%) 9/19 (47.4%) CFS ≥4 1/13 (7.69%) others. Survival analysis performed using Kaplan-Meier for OS, RFS, and PFS with associated p-values shown. P-values were calculated using Student’s T-test for continuous and Fisher’s exact test for Table 3: Two-by-two categorical variables.matrices showing the proportion of patients who died/progressed of bladder cancer as well as those who recurred. Non-frail (CFS ≤3) and frail (CFS ≥3) patients were 18 compared with lower (low/intermediate) vs higher (high/very high) risk NMIBC as defined by EAU [13].
Table 3: Two-by-two matrices showing the proportion of patients who died/ progressed of bladder cancer as well as those who recurred. Non-frail (CFS ≤3) and frail (CFS ≥3) patients were compared with lower (low/intermediate) vs higher (high/very high) risk NMIBC as defined by EAU [13].
19
Case reports 20
21
Metabolic-associated signature and hub genes associated with immune microenvironment and prognosis in bladder cancer
Multiparametric MRI-based radiomics signature for preoperative estimation of basal and luminal features in bladder cancer
Zongtai Zheng, Shanghai Tenth People’s Hospital, Tongji University School of Medicine Xudong Yao
Zongtai Zheng, Shanghai Tenth People’s Hospital, Tongji University School of Medicine Xudong Yao
Introduction & Objectives: The relationship between metabolism and immune microenvironment remains to be studied in bladder cancer (BCa). We aimed to construct a metabolic-associated signature for prognostic prediction and investigate its relationship with the immune microenvironment in BCa.
Introduction & Objectives: We aimed to develop a quantitative biomarker referred to as basal-luminal score to assess the luminal and basal features in bladder cancer (BCa). Multiparametric MRI (mpMRI)-based radiomics signatures were also developed to preoperatively predict the basal-luminal score. Materials & Methods: Gene Set Variation Analysis was performed to evaluate the basal-luminal score using RNA expression data of gene sets for basal and luminal features. Kaplan-Meier curves with the log-rank test, univariate/multivariate Cox regression analysis and meta-analysis were used to investigate the prognostic value of the basal-luminal score. Gene Set Enrichment Analysis and western blot were used to explore the biology behavior of the basal-luminal score. 111 BCa patients with RNA-sequence data and preoperative mpMRI were collected in our center to construct mpMRI-based radiomics signatures for the prediction of the basal-luminal score.
Materials & Methods: The RNA expression of metabolism associated genes was obtained from a combined data set including The Cancer Genome Atlas, GSE48075, and GSE13507 to divide BCa patients into different clusters. A metabolic-associated signature was constructed using the differentially expressed genes between clusters in the combined data set and validated in the IMvigor210 trial and our center. The composition of tumorinfiltrating immune cells (TIIC) was assessed using a single-sample gene set mutation analysis (ssGSVA). Thiosulfate transferase (TST) and S100 calcium-binding protein A16 (S100A16) were identified by hub genes screening. The loss of function study further investigated the effects of TST and S100A16 on the biological function and immune microenvironment of BCa cells.
Results: The basal-luminal score was highly associated with the molecular subtypes of BCa. Patients with high basal-luminal scores had poorer prognoses than patients with low basalluminal scores in several public data sets, including The Cancer Genome Atlas, IMvigor210, GSE13507, GSE31684, GSE32894 and GSE48277. Hallmarks of multiple oncogenic pathways were significantly enriched in the high basal-luminal group. The support vector machine based on 18 radiomics features achieved the optimal performance in the prediction of the basal-luminal score in both the training (accuracy: 85.7%) and validation sets (accuracy: 82.4%).
Results: BCa patients in Cluster A or high-risk group were associated with advanced clinicopathological features and poor survival outcomes. The percentage of complete response/ partial response and the IC50 values of cisplatin and gemcitabine were significantly lower in high-risk patients than in low-risk patients. ssGSVA revealed that the composition of TIICs including macrophages and regulatory T cells between risk groups. TST and S100A16 were significantly associated with clinicopathological features and prognosis. Down-regulation of TST promoted BCa cell invasion, migration, and EMT, which are inhibited by downregulation of S100A16. CD8+T cells, neutrophils, and dendritic cells had higher infiltration in the TST low-group and the S100A16 high-group. Furthermore, loss of function TST and S100A16 significantly affected the expression of PD-L1 and CD47.
Conclusion: The basal-luminal score is associated with molecular subtypes and prognosis in BCa. Biological functional analysis revealed the oncogenic role of the basal-luminal score. The mpMRI-based radiomics signature may be useful for preoperatively predicting the basal and luminal features in BCa.
Conclusion: The metabolic-associated signature can stratify BCa patients into distinct risk groups with different immunotherapeutic susceptibility and survival outcomes, may contribute to effective individualized treatment. Dysexpression of TST or S100A16 contributes to the tumor progression and dysregulation of immune microenvironment in BCa.
1
2
Bladder involvement due to extraurothelial lineage tumors: report of three cases
Bladder necrosis after immediate postoperative mitomycin-C Lorena García, Hospital Universitario Nuestra Señora de Candelaria Carrion Valencia Almudena, Sergio Fumero-Arteaga, Marta Jiménez Navarro, Sara González Nieto, Lara Torres León, Raquel Clemente Graffigna, Jesús Monllor Gisbert, Jonathan Rodríguez Talavera, Jesús Monllor Gisbert
Raquel Clemente Graffigna, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife Carrion Valencia Almudena, Lorena García, Marta Jiménez Navarro, Sara González Nieto, Lara Torres León, Adrián Amador Robayna, Jonathan Rodríguez Talavera, Jesús Monllor Gisbert
Introduction: The early intravesical Instillation of mitomycin C (MMC) is accepted as safe adjuvant therapy after TURBT by nonmuscle- invasive bladder cancer if there is no perforation and it has been shown to reduce recurrence.
Introduction: Bladder cancer is the 4th most common tumor, however, secondary bladder tumors are rare, involving less than 1% of all tumors in this organ. We present three cases of bladder involvement due to extraurothelial lineage tumors.
Case report: We report a 69-year-old male patient, formersmoker with a history of alcoholic cirrhosis with adequate functional reserve. On follow-up in urology consultations for recurrent bladder neoplasia (low-grade Ta) since 2012 with predominance at the anterior bladder wall. The last transurethral resection was in July 2020 with low-grade Ta urotelial neoplasia and received immediate postoperative MMC. After control cystoscopy, it was included in the surgical waiting list due to suspicion of tumor recurrence and symptoms of dysuria with a fatty “supernatant” in the urine. During the surgical procedure, a 3-4 cm cottony, overgrowing lesion is seen on the anterior side. After compressing it, lipid drops come out, showing fat necrosis. The pathological anatomy confirms the presence of connective tissue with extensive areas of necrosis. Conservative treatment was performed with bladder catheterization and antibiotic therapy, and a cystographic control was carried out which confirmed the persistence of the defect in the anterior bladder wall. The bladder catheter was kept for a further 3 months and, given the persistence of a leak in the imaging tests, it was proposed to repair the fistula vs partial cystectomy.
Case report: Case 1: 34 years old male patient, who presented episodes of acute urine retention (AUR), macroscopic hematuria and pain in the right iliac fossa in October 2020. Magnetic resonance (MRI) revealed a mass in right seminal vesicle. Biopsy was diagnostic of squamous carcinoma. TAP CT revealed involvement mesorectal fascia, iliac chains, bladder wall and left distal ureter. Multidisciplinary committee decides start treatment with neoadjuvant chemotherapy and radiotherapy, achieving a partial response to treatment. Therefore, radical cystoprostatectomy and rectal resection were performed in July 2021. Case 2: 46 years old male patient, underwent subtotal gastrectomy for carcinoid tumor in december 2018, that in control PET-CT shows recurrence in form of a tumor mass in seminal vesicles. Asymptomatic, from urologic point of view. An extension study through CT TAP and pelvic MRI was performed, which confirmed the mass in seminal vesicles, with bladder and anterior rectal wall infiltration. Biopsy was diagnostic of neuroendocrine tumor. Radical cystoprostatectomy and rectal resection were performed in december 2019. Nonetheless, he presents disease progression and he is in treatment with Irinotecan.
Conclusion: The most common side effects reported about de MMC are irritative symptoms (10%) and allergic skin reactions (3%). Rare complications such as necrosis of the bladder wall or perforation are rarely mentioned in the literature, but we must take them into account in patients with suggestive symptoms and signs in order to opt for an adequate definitive treatment.
Case 3: 54 years old male patient, diagnosed of intestinal origin GIST located in left iliac fossa, with left kidney involvement, therefore was performed nephroureterectomy in march 2020. Patient has presented macroscopic hematuria. Despite Imatinib treatment, control MRI revealed disease progression as well as bladder wall involvement, that was confirmed by cystoscopy. Currently, we're waiting to pathologic diagnosis, to assess the attitude towards bladder lesion. Conclusion: Metastasis bladder are rare and usually, it occur as a manifestation of advanced disease and for dissemination from adjoining organs (prostate, female genital tract, colon). Most patients with secondary bladder involvement present symptoms, like hematuria and AUR. In the same way, it's common that there is involvement of other organs at the time of diagnosis. Definitive diagnosis and management of secondary bladder tumors is often complicated. It's important rule out the primary origin of the bladder lesions and and confirm the diagnosis of metastasis by radiological and pathological study. In the presence of histology other than urothelial, we must rule out the metastatic origin of the lesion.
20
21
3
4
Urachal remnant, risk of malignancy a case report
Pembrolizumab: complete pathological remission in metastatic bladder cancer - a case report
Lorena García, Hospital Universitario Nuestra Señora de Candelaria Carrion Valencia Almudena, Jonathan Rodríguez Talavera, Marta Jiménez Navarro, Sara González Nieto, Lara Torres León, Raquel Clemente Graffigna, Nuria Orribo Morales, Jesús Monllor Gisbert
Sara González Nieto, Hospital Universitario Nuestra Señora de Candelaria Carrion Valencia Almudena, Lorena García García, Marta Jiménez Navarro, Lara Torres León, Raquel Clemente Graffigna, Jonathan Rodríguez Talavera, Jesús Monllor Gisbert
Introduction: The urachus is an embryologic remnant which is formed from the obliteration of the allantois. Urachal abnormalities are caused when defective obliteration of the urachus happens. They are an infrequent condition.
Introduction: Inhibition of programmed death receptor-1 (PD1)/PD-L1 checkpoint has demonstrated significant benefit in patients with unresectable and metastatic Bladder Cancer for first-line treatment in cisplatin-unfit patients in case of positive PD-L1 status. Data have been presented with encouraging results, showing that using the PD-1 inhibitor pembrolizumab reported a complete pathological remission (pT0) in 42% and pathological response (< pT2) in 54% of patients.
Case report: We present the case of a 38-year-old male patient, with no previous medical history, evaluated in the urology consultation due to dysuria and the presence of whitish exudate after urination for years of evolution, without any other symptom. The physical examination is normal, with negative urethral exudate and negative urine culture. A cystoscopy is requested: a dome image suspicious of urachal pathology is observed. A CT scan was performed that showed a 19x24x45 cm tubular structure with a cystic appearance, compliant with a urachal diverticulum. After these findings, the case was discussed in a clinical session and a systematic search of the literature was carried out to determine the attitude to follow. There is no clear indication in the literature about the management of urchal pathology, being able to opt for surgery o conservative management. Given the persistent symptoms and despite the low risk of urachal cancer (incidence of 0.18 per 100,000), we performed a laparoscopic resection The histological analysis report a mucinous cystic tumor with low potential for malignancy. Urooncological committee decided very close monitoring, with imaging test.
Case report: A 72-year-old male presented with acute bilateral obstructive uropathy, haematuria and abdominal pain. A CTUrogram was performed, manifesting left ureteral rupture and bladder wall enlargement. Bilateral nephrostomy and a bladderprostate transurethral resection (TURBT + TURP) were performed, showing bladder tumour with prostate and left ureteral invasion. Tissue analysis exhibited high grade urothelial carcinoma PD-L1 positive. Following the surgery, disease was updated with ThoraxAbdomen-Pelvis CT-scan, observing retroperitoneal lymph node invasion. After urinary diversion with bilateral nephrostomy, patient persisted with GFR< 30 ml/min, so after discussing the case in a multidisciplinary team, it was decided to start a treatment with Pembrolizumab. Following 8 Pembrolizumab-cycles ThoraxAbdomen-Pelvis CT-scan was repeated, showing response, so our multidisciplinary team proposed radical cystectomy + lymphadenectomy and Bricker urinary diversion. Surgical piece was analyzed, exhibiting absence of tumour (pT0 N0). During the next 17-months follow-up, Thorax-Abdomen-Pelvis CT-scan have not shown tumour relapse or metastasis.
Conclusion: Despite the low risk of urachal cancer in adults, we must consider the possibility of resection or close control of it, given the possibility of malignancy.
Conclusion: Immunotherapy is a promising therapy in the treatment of urothelial cancer for patients unfit for chemotherapy, showing pathological remission in 30-40% of patients, therefore it is a key-point for those who are not able to benefit from first line therapies.
5
6
Endoscopic management of upper urinary tract tumor
Metastatic invasive lobular carcinoma mimicking plasmacytoid urothelial carcinoma of the bladder
Marta Jiménez Navarro, Marta Jiménez Navarro Sergio Fumero-Arteaga, Lorena García, Sara González Nieto, Lara Torres León, Raquel Clemente Graffigna, Jonathan Rodríguez Talavera, Jesus Monllor Gisbert
George Naveen Weeratunga, Capital and Coast District Health Board David Valsenti, Peter Bethwaite
Introduction: Urothelial carcinomas represent the sixth most common tumors in developed countries and 5-10% of cases are located at upper urinary tract. At diagnosis, more than half of patients with upper urinary tract urothelial carcinoma (UTUC) present an invasive tumor. Patients with low risk tumors (solitary tumor, size <2cm, low grade biopsy and negative high grade cytology) and select patients with chronic kidney disease or solitary kidney, kidney sparing management is a primary treatment option.
Introduction: Plasmacytoid urothelial carcinoma (PUC) is rare and is known to mimic invasive lobular carcinoma of the breast and signet cell gastric adenocarcinoma. PUC has a very poor prognosis, with high cancer mortality, despite being chemosensitive, largely due to its high recurrence rate. Metastatic lesions presenting as bladder tumours, are also rare and has poor prognosis when presenting as a widespread metastatic disease process. Case report: A 69-year-old woman with a history of metastatic lobular carcinoma of the breast was noted to have new unilateral hydronephrosis with associated luminal thickening of the proximal right ureter. She originally had a previously wide local excision thirteen years prior, subsequently re-excision then requiring salvage mastectomy due to positive margins. She underwent appropriate neoadjuvant chemotherapy and anastrozole. She then developed evidence of gastric involvement of lobular carcinoma. She was then placed on exemestane which showed radiological and biochemical regression. The finding of hydronephrosis was identified on surveillance imaging with no haematuria.
We present 3 UTUC cases with endoscopic treatment: biopsy with NGage® basket 1.7 + Holmium laser (HoL) + base biopsy with Piranha® forceps and retrograde instillation of mitomycin C (MMC). Case report: Case 1: 80 year old woman patient, with solitary kidney (right nephroureterectomy 5 years ago due to high grade UTUC pT2N0M0) and chronic kidney disease. URO-TC performed yearly, showed a lesion in upper pyelocaliceal cavity, suggestive of neoformative process. Endoscopic exam revealed normal bladder and 25mm exofitic tumor in upper pyelocaliceal cavity. Anatomopathology reported urothelial carcinoma pTaG3. In order to decrease risk recurrence, retrograde instillation with MMC during 6 sessions was performed. 6 months later, endoscopic control showed 5mm papillary lesion on the previous tumor area and resection with HoL was made. After 6 additional instillations with MMC, endoscopic control shows no recurrence with negative cytology. Other cycle of 3 instillations with MMC was made, and nowadays endoscopic revision is expected to perform.
Given the clinical findings the patient came forwards for ureteroscopy. Upon entering the bladder, a small raised erythematous lesion was identified just below the right ureteric orifice and was biopsy. Ureteroscopy of the affected ureter was abandoned due to excessive tightness of the ureter. Ureteric washings of the renal pelvis were taken, and a retrograde pyelogram was performed. A 6 Fr stent was placed. Despite the initial histology suggestion of PUC, clinical correlation and evaluation of the ureteric washing sample was in keeping with metastatic lobular. Evaluation of the ureteric washings revealed mammaglobin positive, and oestrogen receptor and progesterone receptor positive.
Case 2: 54 year old male patient, with solitary kidney (right nephroureterectomy 1 year before because high grade UTUC pT2N0M0). Successive control with URO-TC evidence 9mm lesion in left renal pelvis. Endoscopic procedure revealed 20mm papillary lesion. Pathological report showed urothelial carcinoma pTaG3, and local treatment with cycle of 6 MMC instillations was carried out. Endoscopic control at 6 months showed no lesions. During 2 years subsequent instillations with MMC each 6 months and endoscopic control has carried out and has no shown relapse.
Given the asymptomatic nature of metastatic disease, the patient was kept under observation. Unfortunately, the patient presented acutely with contralateral hydronephrosis, due to extrinsic compression from a metastatic peritoneal deposit. Retrograde decompression with a JJ stent was performed and subsequent palliative paclitaxel and then Palbociclib were trailed.
Case 3: 72 year old woman patient, with chronic kidney disease due to mellitus diabetes, presented with left renal colic and haematuria. URO-TC showed a 12mm solid lesion in left pelvis kidney. Cystoscopy ruled out bladder tumor. Endoscopic examination showed papillary tumor in pelvis kidney. Anatomopathological result: urothelial carcinoma pTaG3. To complete treatment, retrograde instillation with MMC during 6 sessions was made. 6 months after first surgery, endoscopic control shows no relapse. At present, she is being treated with MMC maintenance schedule and subsequent endoscopic control will be performed.
Conclusion: PUC is a rare variant malignancy, and clinicians should be aware of its mimicry of metastatic malignancy. The need for clear communication between surgical and pathology teams is also highlighted for accurate reporting. Positive staining of mammaglobin and hormone receptors in a bladder tumour with initial appearance of plasmacytoid urothelial carcinoma should raise suspicion for metastatic lobular carcinoma of the breast.
Conclusion: Kidney sparing treatment for UTUC by endoscopic management is not standardized. In patients with low risk UTUC, solitary kidney and chronic kidney disease can be a safe option as long as close follow-up is carried out.
Figure 1,2 Urachal remnant
22
23
7
8
Management of recurrent urothelial disease in non-muscle invasive urothelial cancer of a solitary kidney after pyelo-ileoconduit
Carcinomatous lymphangitis associated with urothelial carcinoma Lara Torres León, Hospital Universitario Nuestra Señora de Candelaria Adrián Amador Robayna, Lorena García, Marta Jiménez Navarro, Sara González Nieto, Raquel Clemente Graffigna, Omar Halawa González, Jonathan Rodríguez Talavera, Jesús Monllor Gisbert
Melanie Schouten, KU Leuven Carl Van Haute
Introduction: Cutaneous metastasis associated with urothelial carcinomas are extremely rare with an incidence of 1% to 2%, mostly secondary to iatrogenic tumor cell implantation or hematogenous spread. They are usually diagnosed in advanced stages of the disease and are associated with poor survival (median 2-year survival of 14.2%).
Introduction: Despite the low progression and mortality rates of non-muscle-invasive bladder cancer these cancers can demonstrate high recurrence rates and have the potential to progress to panurothelial disease. Case report: A 61-year old male presented with a history of recurrent non-muscle-invasive bladder cancer. Since the initial diagnosis of low-grade Ta transitional-cell carcinoma in 2014 he had four recurrences after treatment with transurethral resection and adjuvant bacillus Calmette-Guérin and mitomycine C till 2015. Last pathology report showed high-grade pT1. In 2015 an early radical cystoprostatectomy was performed with ileal orthotopic neobladder urinary diversion, resection revealed a Ta/Tis partially low-grade but predominantly high-grade disease with negative lymph nodes. Eight months later a single lesion of 3cm in the right renal pelvis was diagnosed without evidence of lymphadenopathy or metastatic disease. He underwent a nephro-ureterectomy on the right side, final pathology showed a low-grade Ta urothelialcell carcinoma with negative section margins. In 2019 detection of local recurrence in the cranial aspect of the neobladder with involvement of both distal ureters and a metachrone lesion in the proximal left lumbal ureter. A resection of the neobladder, residual right ureter and complete left ureter and reconstruction with pyelo-ileoconduit was performed. Pathology report revealed high grade urothelialcell carcinoma from the right ureter expanding into the neobladder, with a positive section margin of the proximal left ureter. The patient developed tumor recurrences in his solitary kidney. He declines left side nefrectomy since it necessitates dialysis. Decision was made to treat him endoscopically. Since then he had 6 recurrences all treated endoscopic with biopsy and lasering. Pathology samples has always shown Ta or T1. Since December 2020 he underwent adjuvant intracavitary chemotherapy with Epirubicine but continues to develop recurrences.
Case report: We present the case of a 65-year-old male exsmoker diagnosed with high-grade T1 bladder carcinoma in 2008 who was treated with bladder TUR plus induction treatment with endovesical instillations of BCG. During the clinical followup, in view of the pathological result of a cytological study (C4 suspicious of malignancy), endoscopic bladder exploration under anesthesia, standardized multiple mapping biopsy and prostate biopsy were indicated with a diagnosis of prostatic infiltration by high histological grade papillary urothelial carcinoma in the prostate samples, in addition to a focus of prostate adenocarcinoma Gleason 3 + 3. The case was presented in uroconcologic committee proposing radical cystectomy and ileal conduit (Bricker) plus ileoobturator lymphadenectomy performed in September 2014. The anatomopathological study showed high-grade infiltrating urothelial carcinoma of the prostate and prostatic urethra (pT4a N0 M0). Subsequently, intensity-modulated radiation therapy (IMRT) was started on the prostate bed (60 Gy to 2Gy/day) as adjuvant treatment. After 4 years of follow-up the patient reported the appearance of a lesion compatible with Zoon's balanitis in the penis. Circumcision and biopsy of the glans was decided with an anatomopathological diagnosis of intraepithelial carcinoma with presence of carcinomatous lymphangitis (metastatic cutaneous form). MRI showed multiple external iliac and femoral lymphadenopathies and a lesion in the midline of the corpus spongiosum, of uncertain nature (Image 1). Before the radiological findings a cold endoscopic biopsy of the urethral lesion was performed, which was found to be compatible with urothelial carcinoma in situ. An extension study was performed by CT finding mediastinal and iliac lymph node metastases and multiple liver metastases not present in the previous study performed two months earlier (PT4 N3M1).
Multiple recurrence of urothelialcell carcinoma of the upper urinary tract after extended excision of the urothelial surface. Currently nephron sparing endoscopic restrain on 3-monthly intervals with adjuvant intracavitary Epirubicine. Intracavitary chemotherapy is impeded by rapid washout. Conclusion: A radical approach with extended excision in order to further decrease the urothelial surface and reduce the risk of recurrence might be necessary in some patients with non-muscle-invasive bladder cancer and upper tract urothelial carcinoma but can not prevent all recurrence.
First line systemic treatment was started according to CDDPGemcitabine scheme, after 3 cycles rapid progression of liver and bone disease was observed and second line treatment with Atezolizumab was tried for 3 months until significant deterioration of the patient and death. Conclusion: High risk urothelial carcinoma is a serious entity with a high recurrence rate that can exceed 80%. It is important strict follow-up by periodic endoscopic study (urethrocystoscopies) and not only with urine cytology studies as was carried out in this case. Early diagnosis of progression is imperative for right management and prognosis of the disease.
24
25
Notes
Notes
26
27
PinPoint Case Platform
Every week a new case! Join 3000 colleagues and see what they would do... and what the experts say! Stay up-to-date about clinical advances in: Prostate cancer Bladder cancer Renal cell carcinoma Lower urinary tract dysfunction Breast cancer COVID-19 Chronic pain (in development)
Patient cases right into your mailbox! Weekly case challenges Easy & fast to consume Compare with peers and experts Individual pathways with QuickScan & dashboard Accredited course per topic
Why not join your colleagues...it’s free!
ppcp.mirrorsmed.org