Rashid Sayyid, Zizo Al-Daqqaq, Shideh Abedi, Alexandra Gleave, Rui Bernardino, Raj Tiwari, Jessica Grace Cockburn, Ur Metser, Alejandro Berlin, Neil E. Fleshner
{"title":"PSMA PET/CT-DETECTED MESORECTAL LYMPH NODE METASTASES: TREATMENT OUTCOMES AND CLINICAL IMPLICATIONS","authors":"Rashid Sayyid, Zizo Al-Daqqaq, Shideh Abedi, Alexandra Gleave, Rui Bernardino, Raj Tiwari, Jessica Grace Cockburn, Ur Metser, Alejandro Berlin, Neil E. Fleshner","doi":"10.1016/j.urolonc.2024.12.065","DOIUrl":"10.1016/j.urolonc.2024.12.065","url":null,"abstract":"<div><h3>Introduction</h3><div>PSMA-PET/CT-detected mesorectal lymph node (MLN) metastases are present in 11% of biochemically recurrent prostate cancer patients in the post-radical prostatectomy or radiotherapy settings. The biologic significance and clinical implications of these lesions remains unknown. Whether these novel findings represent classical ‘stage migration’ or advanced stage disease associated with adverse clinical outcomes remains unknown. The study objectives were thus to compare the oncologic outcomes of biochemically recurrent patients with MLN metastases to those with other ‘classical’ sites of metastases, using the primary outcome of time to development of castration-resistant disease, and evaluate clinical responses to treatment.</div></div><div><h3>Methods</h3><div>This is a retrospective analysis of biochemically recurrent prostate cancer patients (post-radical prostatectomy and/or radiotherapy) with a positive <sup>18</sup>F-DCFPyL-PSMA-PET/CT that was performed at a tertiary center between December 2018 and February 2021. The study patients were prospectively enrolled in the PSMA-PET for Recurrent Prostate Cancer (PREP) provincial registry.</div><div>We evaluated the association between site of most advanced disease (mesorectal-only versus other sites) and development of castration resistance using a multivariable Cox model, adjusted for PSA level at PSMA-PET/CT and biopsy Grade Group. The site of most advanced disease was defined in the following ascending order: prostatic fossa-only residual/recurrent disease, MLN metastases, pelvic lymph node disease (cN1), non-regional nodal disease (cM1a), bone metastases (cM1b), and visceral organ metastases (cM1c).</div><div>The secondary study objectives were to evaluate oncologic outcomes (prostate-specific antigen [PSA] decrease by 50% [PSA50] and freedom from androgen deprivation therapy [ADT]) in patients with MLN-only metastases treated with metastasis-directed therapy (MDT).</div><div>Survival analysis with Kaplan Meier curves was performed for time-to-event outcomes.</div></div><div><h3>Results</h3><div>The cohort included 301 patients with PSMA PET-positive disease, of whom 71 had mesorectal nodal disease (44 had mesorectal-only disease). At a median follow-up of 36 months, patients with mesorectal nodal-only disease had equivalent rates of castration resistance development, compared to patients with prostatic fossa-limited disease (HR=0.99, p=0.88). Sixteen patients with mesorectal nodal disease underwent metastasis-directed therapy. A PSA50 response was observed in 5/10 patients with mesorectal nodal-only disease and 2/6 for those with additional pelvic nodal disease. All patients without a PSA50 response were started on hormones, with no castrate-resistance development to date. The median hormone-free survival was 28 months.</div></div><div><h3>Conclusions</h3><div>Biochemically recurrent patients with mesorectal nodal-only disease on PSMA-PET/CT have identical rates o","PeriodicalId":23408,"journal":{"name":"Urologic Oncology-seminars and Original Investigations","volume":"43 3","pages":"Page 26"},"PeriodicalIF":2.4,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143509962","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Soki Kashima, Ruchi Gupta, Vivien Moritz, Katherine Sadak, Julia Walker, Adebowale Adeniran, Peter Humphrey, Daniel Dinulescu, Doug Palmer, Scott Hammond, Marcus Bosenberg, Michael Hurwitz, Patrick Kenney, David A. Braun
{"title":"DEVELOPMENT OF EX VIVO PATIENT-DERIVED MODELS TO UNCOVER THE TUMOR-IMMUNE MICROENVIRONMENT IN RENAL CELL CARCINOMA","authors":"Soki Kashima, Ruchi Gupta, Vivien Moritz, Katherine Sadak, Julia Walker, Adebowale Adeniran, Peter Humphrey, Daniel Dinulescu, Doug Palmer, Scott Hammond, Marcus Bosenberg, Michael Hurwitz, Patrick Kenney, David A. Braun","doi":"10.1016/j.urolonc.2024.12.057","DOIUrl":"10.1016/j.urolonc.2024.12.057","url":null,"abstract":"<div><h3>Introduction</h3><div>With the rise of immune checkpoint inhibitors (ICIs) as the primary treatment option for metastatic renal cell carcinoma (RCC), investigating the role of T cells within the tumor microenvironment (TME) is a critical component of understanding both treatment response and resistance. Prior efforts, including single-cell transcriptomic approaches, have provided an important landscape of T cell transcriptional phenotypes. However, these immuno-profiling efforts require validation through functional interrogation of the TME to facilitate the development of novel immunomodulatory therapies. Thus, we established a patient-derived tumor model (PDTM) system to directly assess the effect of inhibitory immune interactions on T cell function and anti-tumor activity in the RCC TME. In this initial proof-of-concept study, we evaluated T cell activation in the RCC TME using the PDTM system.</div></div><div><h3>Methods</h3><div>Fresh tumor samples were obtained from surgical resections of RCC at Yale-New Haven Hospital. The tumor was minced to ∼1-3 mm³ pieces and suspended in an air-liquid interface system, consisting of tumor fragments embedded in a collagen matrix on an insert with a semi-permeable membrane, exposed to culture media. The tumor fragment and matrix suspension were carefully pipetted onto the Millicell insert, which served as the top layer. The PDTM setup includes an inner dish containing the bottom gel layer and the tissue-containing top layer. To complete the assembly, a low-dose T cell stimulant, 1.5 ml of DMEM media with or without 500 nM of anti-PD-1 monoclonal antibody (aPD1mAb) was added to the outer dish surrounding the insert.</div></div><div><h3>Results</h3><div>We successfully optimized PDTM experimental workflows for culture, dissociation, and analysis using immunohistochemistry (IHC), flow cytometry (FCM), and enzyme-linked immunosorbent assays (ELISA). Hematoxylin and eosin (H&E) staining and IHC showed that the TME cellular architecture and immune cell composition was broadly preserved during the three-day experimental period. Using FCM to analyze the dissociated tumor samples, we identified well-preserved CA9+ tumor cells, CD4+ and CD8+ T cell populations, CD4+CD25+ regulatory T cells, CD56+ natural killer cells, CD20+ B cells, and CD14+CD11b+ myeloid subsets including monocytes and CD163-/+macrophages. Among the T cells, we detected PD1+, LAG3+, TIM3+, and TIGIT+ cells. We tested the effect of the anti-PD-1 antibody on PDTM, and importantly, found that treatment of the PDTM with aPD1mAb resulted in more activated CD8 T cells and higher IFN-γ production than the control samples.</div></div><div><h3>Conclusions</h3><div>Through optimization of assays evaluating T cell cytokine production, we were able to assess multiple axes of T cell function in the RCC TME. This study revealed that our PDTM system preserves the RCC TME for functional interrogation. Furthermore, our system for assessing T cell ph","PeriodicalId":23408,"journal":{"name":"Urologic Oncology-seminars and Original Investigations","volume":"43 3","pages":"Pages 22-23"},"PeriodicalIF":2.4,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143509993","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Aaron Isaac Ahdoot, Margaret Meagher, Dhruv Puri, Giacomo Musso, Kit Yuen, Julian Cortes, Cesare Saitta, Dattatraya Patil, Hajime Tanaka, Melis Guer, Masaki Kobayashi, Shohei Fukuda, Alberto Briganti, Andrea Salonia, Umberto Capitanio, Alessandro Larcher, Francesco Montorsi, Yasuhisa Fujii, Viraj Master, Ithaar Derweesh
{"title":"UTILITY OF C-REACTIVE PROTEIN LEVELS IN IMPROVING RISK STRATIFICATION IN METASTATIC RENAL CELL CARCINOMA","authors":"Aaron Isaac Ahdoot, Margaret Meagher, Dhruv Puri, Giacomo Musso, Kit Yuen, Julian Cortes, Cesare Saitta, Dattatraya Patil, Hajime Tanaka, Melis Guer, Masaki Kobayashi, Shohei Fukuda, Alberto Briganti, Andrea Salonia, Umberto Capitanio, Alessandro Larcher, Francesco Montorsi, Yasuhisa Fujii, Viraj Master, Ithaar Derweesh","doi":"10.1016/j.urolonc.2024.12.052","DOIUrl":"10.1016/j.urolonc.2024.12.052","url":null,"abstract":"<div><h3>Introduction</h3><div>The International Metastatic Renal Cell Carcinoma Database Consortium (IMDC) serves as the main prognostic model for metastatic Renal Cell Carcinoma (mRCC). C-reactive Protein (CRP), an acute phase inflammatory marker, is associated with outcomes in mRCC. We sought to evaluate utility of addition of CRP to the current IMDC model and determine whether such additions may improve predictive capability of this model.</div></div><div><h3>Methods</h3><div>We conducted a retrospective analysis utilizing the International Marker Consortium for Renal Cancer registry. Inclusion criteria were mRCC at presentation or mRCC progression/recurrence. Main outcome was overall survival (OS)/all-cause mortality (ACM). Cox proportional hazard multivariable analysis (MVA) was conducted to elucidate independent predictors for ACM including preoperative CRP and current IMDC strata. Kaplan-Meier analysis (KMA) was conducted to evaluate for survival outcomes stratified by IMDC category, which were further subdivided based on normal or elevated preoperative CRP and used to propose new inclusion criteria for each IMDC category. To compare predictive capability of the proposed IMDC stratification to the current stratification, receiver operating characteristic/area under the curve (ROC/AUC) analysis was conducted.</div></div><div><h3>Results</h3><div>515 patients met inclusion criteria (median follow up 33 months), which were stratified into 156 favorable, 345 intermediate, and 14 poor-risk. IMDC strata were subdivided based on preoperative CRP. MVA for ACM revealed elevated CRP (CRP ≥ 5 ng/mL, HR=2.1, p=0.01) and worsening IMDC status (HR=2.2-2.5; p-value=0.006-0.03) associated with ACM. KMA comparing IMDC favorable/intermediate/poor-risk revealed 3-year OS of 60%/42%/50%, p=0.001; further subdivision by CRP revealed alignment between favorable/elevated-CRP with intermediate/normal-CRP and intermediate/elevated-CRP with poor-risk. Proposed realignment of new favorable (current favorable/normal-CRP), new intermediate (current favorable/elevated-CRP and current intermediate/normal-CRP) and new poor-risk (current intermediate/elevated-CRP and poor-risk) revealed 3-year OS of 53%/50%/40%, p=0.001 with improved predictive capability of the CRP-augmented model compared to the current IMDC (ROC/AUC of 0.677 vs. 0.649, respectively).</div></div><div><h3>Conclusions</h3><div>Addition of CRP to the IMDC criteria can improve patient stratification and thereby more accurately stratify patients based on their probability of survival. Validation of our findings is requisite.</div></div>","PeriodicalId":23408,"journal":{"name":"Urologic Oncology-seminars and Original Investigations","volume":"43 3","pages":"Pages 20-21"},"PeriodicalIF":2.4,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143510048","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Kirill E. Medvedev, Anna Savelyeva, John Lafin, Liwei Jia, Feng Wang, Yanfeng Zhang, Jane Smitham, Christina Jamieson, Amir Horowitz, John Sfakianos, Lin Xu, Krinio Giannikou, James Amatruda, Aditya Bagrodia
{"title":"GENOMIC AND TRANSCRIPTOMIC ABERRATIONS DIFFERENTIATING TERATOMAS AT METASTATIC SITES FROM PRIMARY GERM CELL TUMORS OF TESTIS","authors":"Kirill E. Medvedev, Anna Savelyeva, John Lafin, Liwei Jia, Feng Wang, Yanfeng Zhang, Jane Smitham, Christina Jamieson, Amir Horowitz, John Sfakianos, Lin Xu, Krinio Giannikou, James Amatruda, Aditya Bagrodia","doi":"10.1016/j.urolonc.2024.12.086","DOIUrl":"10.1016/j.urolonc.2024.12.086","url":null,"abstract":"<div><h3>Introduction</h3><div>Testicular germ cell tumors (TGCTs) are the most common malignancy occurring in young men between the ages of 20-40 years and include various histologically diverse cancer subtypes. Amongst all TGCTs, mature teratomas present the most advanced differentiation state and can lead to secondary malignant transformation into non-TGCT cancer types (such as sarcoma and adenocarcinoma) in various organs and late relapse with worse clinical outcome. Currently, there are no effective systemic therapies in the teratoma cases of unresectability, incomplete resection, or multifocal metastases and teratoma patients often demonstrate resistance to chemotherapy. Both primary and metastatic teratomas share several morphological similarities. However, they inhabit distinct microenvironments, potentially driven by different molecular alterations and transcriptomic vulnerabilities, which are currently limitedly studied.</div></div><div><h3>Methods</h3><div>We performed whole exome sequencing in 16 primary TGCTs and 17 matched post-chemotherapy pure metastatic teratomas collected from retroperitoneal lymph nodes undergoing orchiectomy. Most primary tumors (n=11) had mixed histological components, seven of them had dominant embryonal carcinoma component (>50%), two were 99-100% teratomas, one was a pure testicular seminoma. Recurrent cases were treated with one cycle of bleomycin, etoposide or cisplatin chemotherapy followed by surgical resection of residual tumor. Tumors removed from two different metastatic sites were analyzed for two patients. Tumor samples were compared to matching normal samples (blood) to distinguish germline and somatic genetic alterations. Bulk RNA-sequencing was performed in 13 residual teratomas removed from lymph nodes after chemotherapy (metastatic teratomas) and 12 primary testicular teratomas. Key findings were validated at spatial level by 10X Visium transcriptomics (Illumina platform) in one mixed TGCT sample, predominantly enriched in teratoma (95%).</div></div><div><h3>Results</h3><div>A total of 583 single nucleotide variants (SNVs) were identified across all 33 tumors analyzed, with the majority being missense alterations (82%). The most frequently mutated genes were associated with RTK/RAS signaling, with KRAS gain-of-function alterations being the predominant oncogenic event. Metastatic teratomas had median tumor mutation burden (TMB) of 0.65 and matched primary tumors had a lower median TMB of 0.47. GISTIC analysis revealed recurrent chromosomal 3p11.1 gain in metastatic site teratoma comparison to two primary TGCT datasets (internal and The Cancer Genome Atlas/TCGA cohort) (p-value = 0.0001). Five loci including chr10q26.3, 16p11.2, 19p13.2, 19q12 and 22q12 showed recurrent loss events in metastatic teratomas compared to primary tumors. In addition, gene set enrichment analysis of bulk RNA-sequencing data revealed significantly enriched, clinically relevant pathways e.g., collagen network forma","PeriodicalId":23408,"journal":{"name":"Urologic Oncology-seminars and Original Investigations","volume":"43 3","pages":"Page 34"},"PeriodicalIF":2.4,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143510086","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Michael Forster, Maryna Gozhenko, Rebekka Kraemer, Marc Höppner, Tim A. Steiert, Xiaoli Yi, Nicole Braun, Anja Tanck, Mona Elhadidi, Reza Mehrazin, Pamela Pinzani, Stefania Gelmini, Philipp Nuhn, Andre Franke Andre Franke, Ahmed Eraky
{"title":"REPEATABILITY OF CANCER MUTATION DETECTION IN URINE LIQUID BIOPSY: MORE SAMPLES ARE BETTER","authors":"Michael Forster, Maryna Gozhenko, Rebekka Kraemer, Marc Höppner, Tim A. Steiert, Xiaoli Yi, Nicole Braun, Anja Tanck, Mona Elhadidi, Reza Mehrazin, Pamela Pinzani, Stefania Gelmini, Philipp Nuhn, Andre Franke Andre Franke, Ahmed Eraky","doi":"10.1016/j.urolonc.2024.12.042","DOIUrl":"10.1016/j.urolonc.2024.12.042","url":null,"abstract":"<div><h3>Introduction</h3><div>Single urine samples are the standard for cytology examination of urothelial tumors. The detection of urothelial cancer mutations in urine is an emerging research field, but there is currently no evidence-based standard for the timing or frequency of sampling. Our primary objective was to investigate the repeatability of urothelial cancer mutations in urine samples of bladder cancer patients. Our secondary objective was to refine a panel of bladder cancer-associated genes for future diagnostic research.</div></div><div><h3>Methods</h3><div>Urine samples from 39 bladder cancer patients were sequenced using a 127-gene panel. The Wilcoxon signed rank sum test was used to compare one urine sample versus multiple samples from the same patient with respect to mutation presence. For panel refinement, genes were considered if they were mutated in at least three patients of our cohort and confirmed by literature.</div></div><div><h3>Results</h3><div>Somatic mutation presence in a single urine sample is incomplete compared to several samples (V=120, p=.0005843). Our refined set of 32 genes enabled bladder cancer- associated mutation detection in all 39 patients (100%). The mutational landscape was congruent with literature.</div></div><div><h3>Conclusions</h3><div>More than one urine sample is needed to discover the mutational landscape in most bladder cancer patients, speculatively due to physiological or circadian reasons. Our refined set of 32 genes is probably sufficient for bladder cancer-associated mutation detection, even in a single sample. Urine-based mutation detection is promising for bladder cancer diagnostics and management, especially when multiple urine samples are used.</div></div>","PeriodicalId":23408,"journal":{"name":"Urologic Oncology-seminars and Original Investigations","volume":"43 3","pages":"Pages 16-17"},"PeriodicalIF":2.4,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143510145","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Madison Krischak, David Elliott, Zach Landis-Lewis, Patrick Lewicki, Gretchen Piatt, Todd Morgan, Geoffrey Barnes, Alex Bryant, Megan Caram, Phoebe Tsao, Molly Harrod, Ted Skolarus, Anne Sales, Kristian Stensland
{"title":"ARE VETERANS WITH PROSTATE CANCER SCIENTIFICALLY UNDERSERVED? DESCRIBING ACCESS TO PROSTATE CANCER CLINICAL TRIALS IN THE VA","authors":"Madison Krischak, David Elliott, Zach Landis-Lewis, Patrick Lewicki, Gretchen Piatt, Todd Morgan, Geoffrey Barnes, Alex Bryant, Megan Caram, Phoebe Tsao, Molly Harrod, Ted Skolarus, Anne Sales, Kristian Stensland","doi":"10.1016/j.urolonc.2024.12.028","DOIUrl":"10.1016/j.urolonc.2024.12.028","url":null,"abstract":"<div><h3>Introduction</h3><div>Clinical trials advance science and provide innovative care that is sometimes the only treatment option for people with cancer. Some groups do not have access to clinical trials, excluding them from the benefits of trial participation, and could be considered “scientifically underserved”. Specific to Veterans with prostate cancer, it is unclear how frequently and where prostate cancer clinical trials are offered within the VA, and who may be scientifically underserved. Describing these gaps could identify opportunities for improving Veteran access to cancer clinical trials, ensuring Veterans are afforded the benefits of clinical trial enrollment. To understand gaps in access, we assessed VA site inclusion in prostate cancer clinical trials registered on ClinicalTrials.gov.</div></div><div><h3>Methods</h3><div>In March 2024, using a Python script, we identified all phase 2-3 prostate cancer clinical trial records registered on ClinicalTrials.gov after January 1, 2007, and extracted trial site information. A custom algorithm was used to identify VA sites recorded on ClinicalTrials.gov. We identified the number of unique trials hosted at each VA grouped by trial phase and overall trial status. We manually identified the total number of VA hospitals from the VA directory (va.gov/directory), grouping hospitals by unique address and manually removing duplicates. We then described the proportion of VA sites hosting at least 1 clinical trial, and the proportion of trials with at least 1 VA site. We further identified geographic variation in trial availability by describing the proportion of VA facilities in each census division with at least 1 prostate cancer trial.</div></div><div><h3>Results</h3><div>Of 1,884 registered phase 2 or 3 prostate cancer clinical trials, 118 (6%) included at least one VA site. Of 353 phase 3 trials, 55 (16%) included at least 1 VA site. Of 159 identified VA facilities, 66 (42%) have had at least one phase 2 or 3 prostate cancer clinical trial, while 58 (36%) have had at least one phase 3 trial. As of March 2024, there were 53 VA sites (33%) with a currently active phase 3 prostate cancer clinical trial, 60 (38%) with an active phase 2 or 3 prostate cancer clinical trial, and 8 sites (5%) with >10 currently active prostate cancer clinical trials. Availability of prostate cancer trials at VA sites within census divisions ranged from 3 of 10 VA facilities in the New England census division to 10 of 19 VA facilities in the Pacific census division.</div></div><div><h3>Conclusions</h3><div>Veterans treated at VA facilities have access to only 6% of prostate cancer clinical trials. Further, less than half of VA hospitals are sites for at least one prostate cancer clinical trial, and only a third have had a phase 3 prostate cancer clinical trial. There was some geographic variation in VA trial availability, with a higher proportion of Pacific division VA facilities offering trials than New England o","PeriodicalId":23408,"journal":{"name":"Urologic Oncology-seminars and Original Investigations","volume":"43 3","pages":"Page 11"},"PeriodicalIF":2.4,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143509708","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
John Pfail, Alain Kaldany, Rachel Passarelli, Melinda Fu, Benjamin Lichtbroun, Kevin Chua, Vignesh Packiam, David Golombos, Thomas Jang, Saum Ghodoussipour
{"title":"A COMPARATIVE ANALYSIS OF COMPLICATION RATES IN URINARY DIVERSIONS: EXPLORING THE IMPACT OF CONTEMPORARY SURGICAL TRENDS","authors":"John Pfail, Alain Kaldany, Rachel Passarelli, Melinda Fu, Benjamin Lichtbroun, Kevin Chua, Vignesh Packiam, David Golombos, Thomas Jang, Saum Ghodoussipour","doi":"10.1016/j.urolonc.2024.12.030","DOIUrl":"10.1016/j.urolonc.2024.12.030","url":null,"abstract":"<div><h3>Introduction</h3><div>Historically, outcomes have been similar among patients undergoing continent and incontinent diversion at time of radical cystectomy, with no difference in complication rates. Given the decreasing rates of continent diversion, we sought to examine postoperative complication rates based on type of diversion in a contemporary cohort.</div></div><div><h3>Methods</h3><div>Data was extracted from the National Surgical Quality Improvement Program (NSQIP) database including all patients from 2019-2021 who underwent radical cystectomy. Patients were stratified based on diversion type. Statistical endpoints included thirty-day complications, length of stay (LOS), and readmissions. Optimal RC outcome was defined as absence of any postoperative complication, reoperation, prolonged LOS (75th percentile, 8 days) with no readmission. Multivariable analyses with Bonferroni correction were performed to assess the association between urinary diversion and postoperative outcomes in patients undergoing RC.</div></div><div><h3>Results</h3><div>A total of 4375 patients were identified, including 3780 (86.4%) who underwent incontinent diversion and 595 (13.6%) who underwent continent diversion. Compared to patients with continent diversion, those with incontinent diversion were more likely to be older, female, have higher ASA, worse renal function, robotic/laparoscopic approach, history of radiation or pelvic surgery, and higher stage (Table 1). On multivariable analysis, after Bonferroni adjustment, patients with continent diversion had increased odds of high grade complications (OR 1.58; 99% CI [1.15-2.15]) and readmission (OR 1.7 [1.28-2.27]) as well as lower odds of an optimal outcome (OR 0.74 [0.58-0.95]) compared to incontinent diversion (Figure 1).</div></div><div><h3>Conclusions</h3><div>In a contemporary cohort of patients undergoing radical cystectomy, odds of adverse postoperative outcomes were increased among those undergoing continent diversion compared to incontinent diversion, despite more favorable baseline characteristics in this cohort.</div></div>","PeriodicalId":23408,"journal":{"name":"Urologic Oncology-seminars and Original Investigations","volume":"43 3","pages":"Pages 11-12"},"PeriodicalIF":2.4,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143509710","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Randie E. White, Joshua Linscott, Connor Pelletier, Evelyn James, Erin Santos, Jeffrey M. Howard, Stephen T. Ryan, Matthew H. Hayn, Jesse D. Sammon
{"title":"OPTIMIZING A POST-OPERATIVE PATHWAY IN RADICAL CYSTECTOMY PATIENTS REDUCES INFECTIOUS COMPLICATIONS","authors":"Randie E. White, Joshua Linscott, Connor Pelletier, Evelyn James, Erin Santos, Jeffrey M. Howard, Stephen T. Ryan, Matthew H. Hayn, Jesse D. Sammon","doi":"10.1016/j.urolonc.2024.12.046","DOIUrl":"10.1016/j.urolonc.2024.12.046","url":null,"abstract":"<div><h3>Introduction</h3><div>Radical Cystectomy (RC) is a morbid procedure with a known high risk of complications and readmissions. Our institution is the only academic center in a rural state and provides the majority of cystectomy care, leading to a robust bladder cancer program. Longitudinally, a series of peri-operative changes to our RC care pathway have been made with the aim of improving outcomes. After the introduction of ERAS in 2020, we did not observe a change in outcomes which prompted a change in our perioperative pathway. Furthermore, genitourinary (GU) infections represented the largest body of post-operative complications amongst our complications. We describe the introduction of a post-operative discharge pathway (PODP), incorporated in 2022, aimed to reduce low-grade complications and associated readmissions.</div></div><div><h3>Methods</h3><div>Data was extracted from a prospectively maintained database identifying 323 RC pts from 2015-2024. Complications were graded and classified by the Memorial Sloan Kettering Cancer Center system. A statewide health information exchange allows for record sharing across institutions. This allowed for capture of complete 90d follow-up. All 90d readmissions, complications, and GU infections after surgery were recorded and analyzed.</div><div>An antibiogram specific to our RC pts was previously created, leading to prophylaxis (ppx) with low dose Levaquin or Bactrim starting POD#4 until stent removal at 2 weeks. This began April 1, 2022, as part of a larger PODP that included increased post-operative education, a post-discharge day 2 and 5 phone call with an oncology RN, and oral 30d DVT ppx post-discharge. Chi-square and multivariable logistic regression analyses were performed to assess the association between antibiotic ppx and 30 and 90d readmissions due to GU infection.</div></div><div><h3>Results</h3><div>90d follow-up was available for all 323 pts. 462 complications were recorded pre-PODP. 75.4% and 24.6% occurred at 30d and 90d, respectively. Infectious (27.3%), gastrointestinal (17.7%), and genitourinary (12.6%) causes were the most common, predominantly due to GU infections, ileus, and renal failure (Table 1). The majority were grade 2 (35.5%, Table 1<em>).</em> 30d and 90d mortality was 2.7% and 5.0%, respectively<em>.</em> Post-PODP implementation, 93 complications were recorded. 66.7% and 33.3% occurred at 30d and 90d, respectively. Gastrointestinal (33.3%), infectious (19.4%), and genitourinary (16.1%) causes were the most common (Table 1). The majority were grade 1 (23.4%, Table 1).</div><div>After PODP implementation, 53/56 pts received antibiotic ppx. The 30d risk of GU infections decreased following antibiotic ppx from 84/172 (48.0%) to 3/24 (12.5%), (p=0.001, Table 2). Of 66 pts readmitted at 30d pre-ppx, 41/66 (62.1%) were due to a GU infection vs 1/10 (10%) post-ppx (p=0.002, Table 2).</div></div><div><h3>Conclusions</h3><div>Here we show complete capture of follow-up for","PeriodicalId":23408,"journal":{"name":"Urologic Oncology-seminars and Original Investigations","volume":"43 3","pages":"Page 18"},"PeriodicalIF":2.4,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143510149","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Optimal imaging techniques across the spectrum of testicular cancer","authors":"Kshitij Pandit, Dhruv Puri, Kit Yuen, Nuphat Yodkhunnatham, Margaret Meagher, Aditya Bagrodia","doi":"10.1016/j.urolonc.2024.05.023","DOIUrl":"10.1016/j.urolonc.2024.05.023","url":null,"abstract":"<div><div>Over the years, several imaging techniques<span> have been used in the diagnosis and management of testicular cancer. We compartmentalize disease stages into preorchiectomy, stage 1, initial stage 2 and 3 and postchemotherapy stage 2 and 3. We then elaborate on various imaging modalities that are relevant to each of these stages. We also describe evolving imaging tools that have shown promise. We attempt to provide a comprehensive review of these techniques over the spectrum of testicular cancer.</span></div></div>","PeriodicalId":23408,"journal":{"name":"Urologic Oncology-seminars and Original Investigations","volume":"43 3","pages":"Pages 150-155"},"PeriodicalIF":2.4,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141499063","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jacob L. Roberts M.D. , Luke Luchen Wang M.D. , Brent Rose M.D. , Tyler M. Seibert M.D., Ph.D. , Lisa Madlensky Ph.D. , Sarah M. Nielsen , Amir Salmasi M.D. , A. Karim Kader M.D. , Christopher J. Kane M.D. , E. David Crawford M.D. , Juan Javier-Desloges M.D., M.S. , Rana R. McKay M.D. , Aditya Bagrodia M.D.
{"title":"Germline genetic testing for prostate cancer: Ordering trends in the era of expanded hereditary cancer screening recommendations","authors":"Jacob L. Roberts M.D. , Luke Luchen Wang M.D. , Brent Rose M.D. , Tyler M. Seibert M.D., Ph.D. , Lisa Madlensky Ph.D. , Sarah M. Nielsen , Amir Salmasi M.D. , A. Karim Kader M.D. , Christopher J. Kane M.D. , E. David Crawford M.D. , Juan Javier-Desloges M.D., M.S. , Rana R. McKay M.D. , Aditya Bagrodia M.D.","doi":"10.1016/j.urolonc.2024.10.010","DOIUrl":"10.1016/j.urolonc.2024.10.010","url":null,"abstract":"<div><h3>Purpose</h3><div>The availability of targeted therapies for advanced prostate cancer led to the expansion of national guidelines recommending germline genetic testing. The aim of this study was to describe recent trends in germline test ordering patterns for patients with prostate cancer.</div></div><div><h3>Materials and Methods</h3><div>A retrospective cohort analysis of patients with prostate cancer who underwent germline testing through a single commercial laboratory (Invitae Corporation) between 2015–2020 was performed. Ordering trends between provider medical specialties were compared. Our primary hypothesis was that the proportion of tests ordered by urologists would increase over time.</div></div><div><h3>Results</h3><div>In total, 17,256 prostate cancer patients underwent germline genetic testing; 14,400 patients had an ordering provider with an associated medical specialty and were included in the final comparison cohort. Total prostate cancer patients undergoing germline testing increased quarterly from 21 in Q2 of 2015 to 1,509 in Q3 of 2020. The proportion of tests ordered by urologists increased from 0% in Q2 2015 to 8.3% in Q3 2020 (<em>P</em> < 0.001). Compared to medical genetics, medical oncology, and other specialties, urology ordered more tests for patients under 70 years old (66% vs 51%–55%, <em>P</em> <0.004) and for patients who reported negative family history (25% vs 12%–20%, <em>P</em> = 0.012).</div></div><div><h3>Conclusions</h3><div>As awareness and indications for germline testing continue to expand, aggregate ordering volume is increasing, and urologists are becoming more involved in facilitating testing. This highlights the continued importance of educating urologists on the indications for and implications of germline genetic testing, as well as providing tools to support implementation.</div></div>","PeriodicalId":23408,"journal":{"name":"Urologic Oncology-seminars and Original Investigations","volume":"43 3","pages":"Pages 195.e21-195.e27"},"PeriodicalIF":2.4,"publicationDate":"2025-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142558882","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}