1
|
de Vos II, Rosenstand C, Hogenhout R, van den Bergh RCN, Remmers S, Roobol MJ, ERSPC Rotterdam Study Group. Reducing Overtreatment of Prostate Cancer Patients: Revisiting the European Association of Urology Pretreatment Risk Group Classification Using Long-term Follow-up Data from the European Randomized Study of Screening for Prostate Cancer Rotterdam. Eur Urol Oncol 2025; 8:747-754. [PMID: 39603883 DOI: 10.1016/j.euo.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/04/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND AND OBJECTIVE Tailored treatment for prostate cancer (PCa) requires accurate risk stratification. This study examines the effectiveness of the European Association of Urology (EAU) classification in predicting long-term PCa-specific mortality (PCSM) and assesses whether an alternative system can improve the identification of patients with low-risk disease. METHODS This study included two cohorts of patients with localized PCa: one with screen-detected PCa (n = 1563; S-cohort) and the other with clinically detected PCa (n = 755; C-cohort), all from a population-based, randomized screening study, who underwent primary radical prostatectomy or radiation monotherapy. Patients were stratified according to the traditional EAU risk classification and an alternative risk classification where low-risk disease is adjusted according to contemporary active surveillance (AS) eligibility criteria. The 15-yr time-dependent area under the curve (AUC) and the cumulative incidence of PCSM at 15 yr after diagnosis were assessed for each risk classification and cohort. KEY FINDINGS AND LIMITATIONS With a median follow-up of 20 yr in the S-cohort and 12 yr in the C-cohort, the EAU classification demonstrated 15-yr AUCs of 0.76 (95% confidence interval [CI]: 0.71-0.80) and 0.72 (95% CI: 0.65-0.79), respectively, for predicting PCSM. The alternative classification showed a 15-yr AUC of 0.74 (95% CI: 0.69-0.79) in the S-cohort and 0.75 (95% CI: 0.68-0.81) in the C-cohort. The alternative classification identified 45% more men having a low risk in the S-cohort and 83% more in the C-cohort than the EAU classification, with no statistically significant increase in the 15-yr PCSM incidence (S-cohort subhazard ratio: 1.33 [95% CI: 0.66-2.68]; C-cohort subhazard ratio: 0.99 [95% CI: 0.29-3.38]). CONCLUSIONS AND CLINICAL IMPLICATIONS The EAU classification predicts PCSM accurately, but an alternative classification, adjusted for AS eligibility, identifies substantially more men as having a low risk. This could enhance AS acceptance and utilization in clinical practice, reducing overtreatment. PATIENT SUMMARY This study shows that while a commonly used pretreatment risk classification for prostate cancer predict disease prognosis accurately, an alternative system based on active surveillance eligibility criteria identifies many more men as having a low risk. Adopting this classification could enhance the acceptance and use of active surveillance, reducing unnecessary treatments.
Collapse
Affiliation(s)
- Ivo I de Vos
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands.
| | - Charlotte Rosenstand
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Renée Hogenhout
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Roderick C N van den Bergh
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Sebastiaan Remmers
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Monique J Roobol
- Department of Urology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | |
Collapse
|
2
|
Soeterik TFW, Heetman JG, Hermsen R, Wever L, Lavalaye J, Vinken M, Bahler CD, Yong C, Tann M, Kesch C, Seifert R, Telli T, Chiu PKF, Wu KK, Zattoni F, Evangelista L, Bettella S, Ceci F, Barone A, Miszczyk M, Matsukawa A, Rajwa P, Marra G, Briganti A, Montorsi F, Scheltema MJ, van Basten JPA, van Melick HHE, van den Bergh RCN, Gandaglia G, European Association of Urology Young Academic Urologists Prostate Cancer Working Party. The Added Value of Prostate-specific Membrane Antigen Positron Emission Tomography/Computed Tomography to Magnetic Resonance Imaging for Local Staging of Prostate Cancer in Patients Undergoing Radical Prostatectomy. Eur Urol Oncol 2025; 8:731-738. [PMID: 39613565 DOI: 10.1016/j.euo.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/21/2024] [Accepted: 11/08/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND AND OBJECTIVE The role of prostate-specific membrane antigen (PSMA)-based positron emission tomography (PET)/computed tomography (CT) in addition to magnetic resonance imaging (MRI) for local staging of prostate cancer (PC) has been poorly addressed so far. Our aim was to assess the diagnostic accuracy of PSMA PET/CT and MRI, alone and combined, for detection of extraprostatic extension (EPE) and seminal vesicle invasion (SVI) in PC. METHODS We conducted a multicenter retrospective study evaluating patients undergoing PSMA PET/CT and MRI before radical prostatectomy. The sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV), and area under the receiver operating characteristic curve (AUC) for detection of EPE and SVI were calculated for MRI and PSMA PET/CT alone and combined. KEY FINDINGS AND LIMITATIONS We included 550 patients, of whom 2%, had low-risk, 43% had intermediate-risk, and 55% had high-risk PC. Overall, 52% of patients had EPE and 21% had SVI at histopathology. Patient-based comparison of MRI versus PSMA PET/CT for detection of EPE revealed sensitivity of 60% versus 41% (p < 0.001), specificity of 77% versus 83% (p = 0.075), PPV of 75% versus 73% (p = 0.6), NPV of 64% versus 56% (p < 0.001), and AUC of 69% versus 62% (p = 0.01). Combining the modalities increased the sensitivity (73%; p < 0.001) and NPV (69%; p < 0.001) and decreased the specificity (67%; p < 0.001) and PPV (71%; p = 0.01) over MRI alone. Patient-based comparison of MRI versus PSMA PET/CT for detection of SVI revealed sensitivity of 36% versus 44% (p = 0.2), specificity of 96% versus 96% (p > 0.99), PPV of 71% versus 75% (p = 0.6), NPV of 85% versus 87% (p = 0.2), and AUC of 66% versus 70% (p = 0.2). Combining the modalities increased the sensitivity (60%; p < 0.001), NPV (90%; p < 0.001), and AUC (76%; p < 0.001) and decreased the specificity (92%; p < 0.001) over MRI alone. Limitations include the retrospective nature of the study, selection of higher-risk cases for PSMA PET/CT, and lack of central review. CONCLUSIONS AND CLINICAL IMPLICATIONS PSMA PET/CT has lower sensitivity for EPE detection in comparison to MRI. However, addition of PSMA PET information to MRI improved the sensitivity for EPE and SVI detection. Thus, the two modalities should be combined to guide treatment selection. PATIENT SUMMARY Combining MRI (magnetic resonance imaging) scans with another type of imaging called PSMA PET/CT (prostate-specific membrane antigen positron emission tomography/computed tomography) for patients with prostate cancer leads to better identification of cancer growth outside the prostate in comparison to MRI alone. This could potentially improve the choice of prostate cancer treatment.
Collapse
Affiliation(s)
- Timo F W Soeterik
- Department of Urology, St. Antonius Hospital, Nieuwegein/Utrecht, The Netherlands; Department of Radiation Oncology, University Medical Center Utrecht, Utrecht, The Netherlands.
| | - Joris G Heetman
- Department of Urology, St. Antonius Hospital, Nieuwegein/Utrecht, The Netherlands
| | - Rick Hermsen
- Department of Nuclear Medicine, Canisius Wilhelmina Hospital, Nijmegen, The Netherlands
| | - Lieke Wever
- Department of Urology, St. Antonius Hospital, Nieuwegein/Utrecht, The Netherlands
| | - Jules Lavalaye
- Department of Nuclear Medicine, St. Antonius Hospital, Nieuwegein/Utrecht, The Netherlands
| | - Maarten Vinken
- Department of Nuclear Medicine, Canisius Wilhelmina Hospital, Nijmegen, The Netherlands
| | - Clinton D Bahler
- Department of Urology, Indiana University Medical Center, Indianapolis, IN, USA
| | - Courtney Yong
- Department of Urology, Indiana University Medical Center, Indianapolis, IN, USA
| | - Mark Tann
- Department of Radiology and Imaging Sciences, Indiana University Medical Center, Indianapolis, IN, USA
| | - Claudia Kesch
- Department of Urology, University Hospital Essen, Essen German Cancer Consortium (DKTK) University Hospital Essen, Essen, Germany
| | - Robert Seifert
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | - Tugce Telli
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | - Peter Ka-Fung Chiu
- S.H. Ho Urology Centre, Department of Surgery, The Chinese University of Hong Kong, Hong Kong, China
| | - Kwan Kit Wu
- Department of Nuclear Medicine and PET, Hong Kong Sanatorium and Hospital, Hong Kong, China
| | - Fabio Zattoni
- Department of Surgery, Oncology, and Gastroenterology - Urology Clinic, University of Padua, Padua, Italy; Department of Medicine - DIMED, University of Padua, Padua, Italy
| | - Laura Evangelista
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy; Division of Nuclear Medicine, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Sara Bettella
- Department of Surgery, Oncology, and Gastroenterology - Urology Clinic, University of Padua, Padua, Italy
| | - Francesco Ceci
- Division of Nuclear Medicine and Theranostics, IRCCS European Institute of Oncology, Milan, Italy
| | - Antonio Barone
- Division of Nuclear Medicine and Theranostics, IRCCS European Institute of Oncology, Milan, Italy
| | - Marcin Miszczyk
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Collegium Medicum, WSB University, Dąbrowa Górnicza, Poland
| | - Akihiro Matsukawa
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Department of Urology, The Jikei University School of Medicine, Tokyo, Japan
| | - Pawel Rajwa
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Second Department of Urology, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Giancarlo Marra
- Department of Urology, University Hospital S. Giovanni Battista, Azienda Ospedaliero Universitaria Città della Salute e della Scienza di Torino, Turin, Italy
| | - Alberto Briganti
- Division of Oncology/Unit of Urology, Soldera Prostate Cancer Laboratory, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Francesco Montorsi
- Division of Oncology/Unit of Urology, Soldera Prostate Cancer Laboratory, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | - Matthijs J Scheltema
- Department of Urology, St. Antonius Hospital, Nieuwegein/Utrecht, The Netherlands
| | | | - Harm H E van Melick
- Department of Urology, St. Antonius Hospital, Nieuwegein/Utrecht, The Netherlands
| | | | - Giorgio Gandaglia
- Division of Oncology/Unit of Urology, Soldera Prostate Cancer Laboratory, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy
| | | |
Collapse
|
3
|
Mazzone E, Thomson A, Chen DC, Cannoletta D, Quarta L, Pellegrino A, Gandaglia G, Moon D, Eapen R, Lawrentschuk N, Montorsi F, Siva S, Hofman MS, Chiti A, Briganti A, Perera ML, Murphy DG. The Role of Prostate-specific Membrane Antigen Positron Emission Tomography for Assessment of Local Recurrence and Distant Metastases in Patients with Biochemical Recurrence of Prostate Cancer After Definitive Treatment: A Systematic Review and Meta-analysis. Eur Urol 2025:S0302-2838(25)00285-4. [PMID: 40393864 DOI: 10.1016/j.eururo.2025.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 04/21/2025] [Accepted: 05/06/2025] [Indexed: 05/22/2025]
Abstract
BACKGROUND AND OBJECTIVE Positron emission tomography (PET) with prostate-specific membrane antigen (PSMA) radioligands has an established role in the assessment of recurrence of prostate cancer (PC) after primary treatment. However, an updated understanding of its diagnostic utility is warranted, particularly in the context of emerging targeted and systemic treatment options for recurrent PC. Our aim was to evaluate the diagnostic performance of PSMA PET for staging and detection of local or metastatic disease in patients with biochemical recurrence (BCR) following definitive treatment for PC. METHODS We conducted a systematic review (Web of Science/MEDLINE, Cochrane Library, and EMBASE from inception to April 25, 2024) and meta-analysis. Eligible retrospective and prospective studies had extractable data on PSMA PET for patients with BCR after radical prostatectomy (RP) or radiation therapy (RT). Risk of bias was assessed using the QUADAS-2 tool. Random-effects models were used to evaluate PSMA PET positivity rates across clinical subgroups stratified by PSA, primary treatment, PSMA PET radioligand, and anatomic lesion sites. KEY FINDINGS AND LIMITATIONS A total of 43 studies (8119 patients) were included. PSMA PET positivity rates varied substantially among patients with BCR after primary definitive treatment, with significant study heterogeneity (τ2 = 0.6; p < 0.001), which was mitigated in part after stratification by PSA category. Positivity rates were significantly higher after RT (92%) than after RP (60%; p < 0.001). PSMA PET positivity by anatomic location was 23% for local recurrence, 32% for pelvic nodes, 14% for extrapelvic nodes, 16% for bone metastases, and just 1% for visceral metastases. The positivity rate increased with PSA, from 48% at PSA 0.2-0.5 ng/ml to >90% at PSA >2 ng/ml. Gleason score (GS) at RP did not significantly influence the positivity rate (50% for GS ≤7 vs 62% for GS 8-10; p = 0.08). Heterogeneity limits the generalisability of our findings. CONCLUSIONS AND CLINICAL IMPLICATIONS We observed substantial variability in PSMA PET positivity rates in BCR because of significant study heterogeneity, mostly related to differences in treatment type, PSA, and anatomic sites. In post-RP BCR, approximately half of patients undergoing PSMA PET had positive findings, even at low PSA (0.2-0.5 ng/ml). In the post-RT setting, PSMA PET use was generally aligned with the Phoenix criterion for BCR, with most studies performing PSMA PET at PSA ≥2 ng/ml. Further research is needed to refine PSA thresholds for PSMA PET, particularly in the post-RT setting, and to assess its role in guiding salvage treatment decisions.
Collapse
Affiliation(s)
- Elio Mazzone
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Australia; Division of Oncology/Unit of Urology, Gianfranco Soldera Prostate Cancer Laboratory, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Alice Thomson
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - David C Chen
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Donato Cannoletta
- Division of Oncology/Unit of Urology, Gianfranco Soldera Prostate Cancer Laboratory, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Leonardo Quarta
- Division of Oncology/Unit of Urology, Gianfranco Soldera Prostate Cancer Laboratory, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Antony Pellegrino
- Division of Oncology/Unit of Urology, Gianfranco Soldera Prostate Cancer Laboratory, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giorgio Gandaglia
- Division of Oncology/Unit of Urology, Gianfranco Soldera Prostate Cancer Laboratory, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Daniel Moon
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia; Royal Melbourne Clinical School, University of Melbourne, Melbourne, Australia
| | - Renu Eapen
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Nathan Lawrentschuk
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia; Department of Urology, Royal Melbourne Hospital, Melbourne, Australia
| | - Francesco Montorsi
- Division of Oncology/Unit of Urology, Gianfranco Soldera Prostate Cancer Laboratory, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Shankar Siva
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia; Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Michael S Hofman
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia; Prostate Cancer Theranostics and Imaging Centre of Excellence, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Arturo Chiti
- Vita-Salute San Raffaele University, Milan, Italy; Department of Nuclear Medicine, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alberto Briganti
- Division of Oncology/Unit of Urology, Gianfranco Soldera Prostate Cancer Laboratory, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Marlon L Perera
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Australia; Department of Urology, Austin Hospital, Heidelberg, Australia
| | - Declan G Murphy
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| |
Collapse
|
4
|
Sciorio C, Giannella R, Romano L, Mirto BF, Di Girolamo A, Ruffo A, Romeo G, Esposito F, Crocetto F, Napolitano L, Balsamo R, Trama F, Bottone F, Quattrone C, Imperatore V, Spirito L. Clinical Predictors and Risk Factors of Gleason Score Upgrade: A Retrospective Cohort Analysis. Diagnostics (Basel) 2025; 15:1238. [PMID: 40428230 PMCID: PMC12110525 DOI: 10.3390/diagnostics15101238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/25/2025] [Accepted: 04/29/2025] [Indexed: 05/29/2025] Open
Abstract
Background: In prostate cancer (PCa) patients, discrepancies between biopsy-assigned Gleason Scores and those determined from surgical specimens are frequently reported. This phenomenon, known as Gleason score upgrade (GSU), can have significant clinical implications. This work aims to understand the factors contributing to GSU for refining prostate cancer management strategies. Methods: Data from 779 patients diagnosed with histologically confirmed PCa who underwent robot-assisted radical prostatectomy at a single tertiary care institution between January 2005 and December 2020 were examined. Results: In the univariable setting, 5-alpha reductase inhibitor (5-ARI) use was associated with a higher percentage of upgrading (42.3% vs. 30.4% among non-users; p = 0.03942). A more advanced pathological T stage (p = 0.01114) and lymph node positivity (p < 0.00001) correlated significantly with GSU. In the logistic regression model, advanced pathological stage increased the odds more than twofold (OR = 2.807, p = 0.00135). 5-ARI use was associated with notably higher odds of upgrading (OR = 3.809, p = 0.00004). Younger age slightly increased the likelihood of GSU (OR = 0.951 per year increase in age, p = 0.01101). Conclusions: Younger age, advanced pathological stage, and the use of 5-alpha reductase inhibitors were identified as significant predictors of GSU.
Collapse
Affiliation(s)
| | | | - Lorenzo Romano
- Department of Woman, Child and General and Specialized Surgery, Università degli Studi della Campania “Luigi Vanvitelli”, 81100 Napoli, Italy; (L.R.); (C.Q.); (L.S.)
| | - Benito Fabio Mirto
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples “Federico II”, 80131 Naples, Italy;
| | | | - Antonio Ruffo
- Dipartimento di Medicina e di Scienze della Salute “Vincenzo Tiberio”, UNIMOL, 86039 Termoli, Italy;
| | - Giuseppe Romeo
- UOC Urologia, AORN “A. Cardarelli”, 80131 Napoli, Italy; (R.G.); (G.R.)
| | - Fabio Esposito
- Urology Unit, Casa di Cura “Nostra Signora di Lourdes”, 80040 Napoli, Italy;
| | - Felice Crocetto
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples “Federico II”, 80131 Naples, Italy;
| | - Luigi Napolitano
- Azienda Sanitaria Locale (ASL) Salerno, Via Vernieri, 84125 Salerno, Italy;
| | | | - Francesco Trama
- UOC Urologia, Asl Napoli 2 Nord, PO “Santa Maria delle Grazie”, 80078 Pozzuoli, Italy;
| | - Francesco Bottone
- UOC Urologia, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy;
| | - Carmelo Quattrone
- Department of Woman, Child and General and Specialized Surgery, Università degli Studi della Campania “Luigi Vanvitelli”, 81100 Napoli, Italy; (L.R.); (C.Q.); (L.S.)
| | | | - Lorenzo Spirito
- Department of Woman, Child and General and Specialized Surgery, Università degli Studi della Campania “Luigi Vanvitelli”, 81100 Napoli, Italy; (L.R.); (C.Q.); (L.S.)
| |
Collapse
|
5
|
Zaurito P, Stabile A, Montorsi F, Briganti A, Gandaglia G. The prognostic role of prostate MRI in prostate cancer patients. Curr Opin Urol 2025:00042307-990000000-00241. [PMID: 40269557 DOI: 10.1097/mou.0000000000001288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
PURPOSE OF REVIEW Multiparametric MRI (mpMRI) has been included in the diagnostic pathway of prostate cancer (PCa). However, the role of this imaging modality in predicting clinical outcomes after diagnosis has been poorly addressed so far. This review aims to summarize the most relevant updates on the prognostic role of mpMRI. RECENT FINDINGS Baseline mpMRI features help to predict adverse pathology at radical prostatectomy (RP) and grade reclassification during active surveillance. Parameters derived at prostate mpMRI such as PI-RADS score 4-5, the maximum diameter of the index lesion and the presence of extracapsular invasion/seminal vesicle invasion are among the strongest predictors of biochemical recurrence (BCR) for men treated with RP. mpMRI-based predictive models can achieve similar accuracy for BCR prediction when compared with validated models that relied on final pathology. Moreover, the use of mpMRI findings to predict disease recurrence after radiotherapy or focal therapy seems to optimize patient's risk stratification after treatment, ruling out disease recurrence. SUMMARY Clinicians should account for prostate mpMRI findings when predicting clinical outcomes in patients diagnosed with PCa.
Collapse
Affiliation(s)
- Paolo Zaurito
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele
- Vita-Salute San Raffaele University, Milan, Italy
| | - Armando Stabile
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele
| | - Francesco Montorsi
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele
- Vita-Salute San Raffaele University, Milan, Italy
| | - Alberto Briganti
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele
- Vita-Salute San Raffaele University, Milan, Italy
| | - Giorgio Gandaglia
- Division of Experimental Oncology/Unit of Urology, Urological Research Institute, IRCCS Ospedale San Raffaele
- Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
6
|
Spirito L, Sciorio C, Romano L, Di Girolamo A, Ruffo A, Romeo G, Crocetto F, Napolitano L, Stizzo M, Bottone F, Quattrone C, Imperatore V. Impact of Nerve-Sparing Techniques on Prostate-Specific Antigen Persistence Following Robot-Assisted Radical Prostatectomy: A Multivariable Analysis of Clinical and Pathological Predictors. Diagnostics (Basel) 2025; 15:987. [PMID: 40310337 PMCID: PMC12025793 DOI: 10.3390/diagnostics15080987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 03/02/2025] [Accepted: 04/09/2025] [Indexed: 05/02/2025] Open
Abstract
Background/Objectives: Prostate-specific antigen (PSA) persistence, defined as a postoperative PSA level ≥ 0.1 ng/mL measured within 4-8 weeks after radical prostatectomy (RP), predicts biochemical recurrence (BCR) and adverse oncological outcomes. The influence of nerve-sparing (NS) surgical techniques on PSA persistence remains debated, especially among patients with high-risk pathological features. This study aimed to evaluate the impact of NS techniques on PSA persistence following robot-assisted radical prostatectomy (RARP), considering tumor characteristics, surgical parameters, and patient-specific factors. Methods: A retrospective cohort analysis was performed on 779 patients who underwent RARP at a single institution between January 2002 and December 2015. The inclusion criteria consisted of histologically confirmed prostate cancer with available preoperative and postoperative data, including PSA measurements taken 4-8 weeks after surgery. PSA persistence served as the primary outcome. Statistical analyses included descriptive statistics, univariate and multivariable logistic regression models to identify predictors of PSA persistence, and Spearman's correlation along with the Kruskal-Wallis H test to evaluate associations. Results: Of the 779 patients included, 55% underwent NS surgery (51% unilateral, 49% bilateral). The mean preoperative PSA was 11.85 ng/mL (SD: 7.63), while the mean postoperative PSA was 0.70 ng/mL (SD: 4.42). An elevated postoperative PSA was associated with a larger tumor size (r = 0.1285, p < 0.001), advanced pathological stages (χ2 = 45.10, p = 3.79 × 10-9), and higher Gleason scores (χ2 = 24.74, p = 1.57 × 10-4). NS surgery correlated with a lower postoperative PSA (mean: 0.20 ng/mL) compared to non-NS procedures (mean: 0.65 ng/mL), with slight differences between unilateral (mean: 0.30 ng/mL) and bilateral (mean: 0.35 ng/mL) NS approaches. Multivariable regression analysis identified advanced pathological stage (coefficient = 1.16, p = 0.04) as an independent predictor of PSA persistence, while NS techniques had no significant independent effect (coefficient = -0.01, p = 0.99). Conclusions: Nerve-sparing surgical techniques do not independently predict PSA persistence after RARP when adjusting for tumor-related factors and confounders. Advanced pathological stage, particularly stage pT3b, primarily determines PSA persistence. These findings highlight the necessity of personalized surgical planning informed by preoperative imaging and patient-centered decision making to optimize oncological and functional outcomes.
Collapse
Affiliation(s)
- Lorenzo Spirito
- Unit of Urology, Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy; (L.R.); (M.S.); (C.Q.)
| | | | - Lorenzo Romano
- Unit of Urology, Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy; (L.R.); (M.S.); (C.Q.)
| | - Antonio Di Girolamo
- AORN Moscati di Avellino, UOC di Urologia, 83100 Avellino, Italy; (A.D.G.); (V.I.)
| | - Antonio Ruffo
- Dipartimento di Medicina e di Scienze della Salute “Vincenzo Tiberio”, 86100 Campobasso, Italy;
| | | | - Felice Crocetto
- Department of Neurosciences, Science of Reproduction and Odontostomatology, University of Naples Federico II, 80131 Naples, Italy;
| | - Luigi Napolitano
- Azienda Sanitaria Locale (ASL) Salerno, via Vernieri, 84125 Salerno, Italy;
| | - Marco Stizzo
- Unit of Urology, Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy; (L.R.); (M.S.); (C.Q.)
| | - Francesco Bottone
- DAI Medico Chirurgico di Alta Specialità, UOC Urologia Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Napoli, Italy;
| | - Carmelo Quattrone
- Unit of Urology, Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 80131 Naples, Italy; (L.R.); (M.S.); (C.Q.)
| | - Vittorio Imperatore
- AORN Moscati di Avellino, UOC di Urologia, 83100 Avellino, Italy; (A.D.G.); (V.I.)
| |
Collapse
|
7
|
Li T, Graham PL, Cao B, Nalavenkata S, Patel MI, Kim L. Accuracy of MRI in detecting seminal vesicle invasion in prostate cancer: a systematic review and meta-analysis. BJU Int 2025; 135 Suppl 3:17-28. [PMID: 39436642 DOI: 10.1111/bju.16547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
OBJECTIVE To determine the diagnostic test accuracy of multiparametric magnetic resonance imaging (mpMRI) in detecting seminal vesicle invasion (SVI). METHODS The Medical Literature Analysis and Retrieval System Online (MEDLINE), PubMed, the Excerpta Medica dataBASE (EMBASE) and Cochrane databases were search up to May 2023. We included studies that investigated the accuracy of mpMRI in detecting SVI when compared to radical prostatectomy specimens as the reference standard. Data extraction was performed by two independent reviewers to construct 2 × 2 tables, as well as patient and study characteristics. The methodological quality of the included studies was assessed with the Quality of Assessment of Diagnostic Accuracy Studies-2 tool. Sensitivity and specificity were pooled and presented graphically with summary receiver operator characteristic (SROC) plots. RESULTS A total of 27 articles with 4862 patients were included for analysis. The summary sensitivity and specificity were 0.57 (95% confidence interval [CI] 0.45-0.68) and 0.95 (95% CI 0.92-0.99), respectively. Meta-regression indicated that there was no evidence that coil strength (P = 0.079), coil type (P = 0.589), year of publication (P = 0.503) or use of the Prostate Imaging-Reporting and Data System (P = 0.873) significantly influenced these results. The summary diagnostic odds ratio was 28.3 (95% CI 15.0-48.8) and the area under the curve for the SROC curve was 0.87. The I2 statistic was a modest 11.9%. In general, methodological quality was good. CONCLUSION The use of mpMRI in detecting SVI has excellent specificity but poor sensitivity. Both endorectal coils and magnetic field strength do not significantly impact the accuracy of MRI. These findings suggest that mpMRI cannot reliably rule out SVI in patients with prostate cancer.
Collapse
Affiliation(s)
- Thomas Li
- Westmead Hospital, Westmead, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - Petra L Graham
- Macquarie University, Sydney, New South Wales, Australia
| | - Brooke Cao
- Westmead Hospital, Westmead, New South Wales, Australia
| | - Sunny Nalavenkata
- Westmead Hospital, Westmead, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - Manish I Patel
- Westmead Hospital, Westmead, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| | - Lawrence Kim
- Westmead Hospital, Westmead, New South Wales, Australia
- University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
8
|
Lin H, Yao F, Yi X, Yuan Y, Xu J, Chen L, Wang H, Zhuang Y, Lin Q, Xue Y, Yang Y, Pan Z. Prediction of adverse pathology in prostate cancer using a multimodal deep learning approach based on [ 18F]PSMA-1007 PET/CT and multiparametric MRI. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07134-0. [PMID: 39969539 DOI: 10.1007/s00259-025-07134-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 02/01/2025] [Indexed: 02/20/2025]
Abstract
PURPOSE Accurate prediction of adverse pathology (AP) in prostate cancer (PCa) patients is crucial for formulating effective treatment strategies. This study aims to develop and evaluate a multimodal deep learning model based on [18F]PSMA-1007 PET/CT and multiparametric MRI (mpMRI) to predict the presence of AP, and investigate whether the model that integrates [18F]PSMA-1007 PET/CT and mpMRI outperforms the individual PET/CT or mpMRI models in predicting AP. METHODS 341 PCa patients who underwent radical prostatectomy (RP) with mpMRI and PET/CT scans were retrospectively analyzed. We generated deep learning signature from mpMRI and PET/CT with a multimodal deep learning model (MPC) based on convolutional neural networks and transformer, which was subsequently incorporated with clinical characteristics to construct an integrated model (MPCC). These models were compared with clinical models and single mpMRI or PET/CT models. RESULTS The MPCC model showed the best performance in predicting AP (AUC, 0.955 [95% CI: 0.932-0.975]), which is higher than MPC model (AUC, 0.930 [95% CI: 0.901-0.955]). The performance of the MPC model is better than that of single PET/CT (AUC, 0.813 [95% CI: 0.780-0.845]) or mpMRI (AUC, 0.865 [95% CI: 0.829-0.901]). Additionally, MPCC model is also effective in predicting single adverse pathological features. CONCLUSION The deep learning model that integrates mpMRI and [18F]PSMA-1007 PET/CT enhances the predictive capabilities for the presence of AP in PCa patients. This improvement aids physicians in making informed preoperative decisions, ultimately enhancing patient prognosis.
Collapse
Affiliation(s)
- Heng Lin
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Fei Yao
- The Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xinwen Yi
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, 315300, China
| | - Yaping Yuan
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Jian Xu
- The Department of Nuclear Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Lixuan Chen
- The Department of Nuclear Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Hongyan Wang
- The Department of Nuclear Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yuandi Zhuang
- The Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Qi Lin
- The Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yingnan Xue
- The Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Yunjun Yang
- The Department of Nuclear Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Zhifang Pan
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
9
|
Ah-Thiane L, Campion L, Allouache N, Meyer E, Pommier P, Mesgouez-Nebout N, Serre AA, Créhange G, Guimas V, Rio E, Sargos P, Ladoire S, Mahier Ait Oukhatar C, Supiot S. Combination of Abiraterone Acetate, Prostate Bed Radiotherapy, and Luteinizing Hormone-releasing Hormone Agonists in Biochemically Relapsing Patients After Prostatectomy (CARLHA): A Phase 2 Clinical Trial. Eur Urol Oncol 2025; 8:38-46. [PMID: 38734543 DOI: 10.1016/j.euo.2024.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/26/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND The relevance of next-generation hormone therapies and circulating tumor cells (CTCs) are not elucidated in biochemical recurrence after prostatectomy. OBJECTIVE To evaluate the combination of abiraterone acetate plus prednisone (AAP), prostate bed radiotherapy (PBRT), and goserelin in biochemically relapsing men after prostatectomy, and to investigate the utility of CTCs. DESIGN, SETTING, AND PARTICIPANTS In this single-arm multicenter phase 2 trial, 46 biochemically relapsing men were enrolled between December 2012 and January 2019. The median follow-up was 47 mo. INTERVENTION All patients received AAP 1000 mg daily (but 750 mg during PBRT), salvage PBRT, and goserelin. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The primary outcome was 3-yr biochemical recurrence-free survival (bRFS) when prostate-specific antigen (PSA) levels were ≥0.2 ng/ml. The secondary outcomes included alternative bRFS (alt-bRFS) when PSA levels were ≥0.5 ng/ml and safety assessment. CTC count was assessed. RESULTS AND LIMITATIONS The 3-yr bRFS and alt-bRFS were 81.5% (95% confidence interval or CI [66.4-90.3%]) and 95.6% (95% CI [83.5-98.9%]), respectively. The most common acute radiotherapy-related adverse effect (AE; all grades was pollakiuria (41.3%). The most common late AE (all grades) was urinary incontinence (15.2%). Grade 3-4 acute or late radiotherapy-related AEs were scarce. Most frequent AEs nonrelated to radiotherapy were hot flashes (76%), hypertension (63%), and hepatic cytolysis (50%, of which 20% were of grades 3-4). Of the patients, 11% had a CTC count of ≥5, which was correlated with poorer bRFS (p = 0.042) and alt-bRFS (p = 0.008). The association between CTC count and higher rates of relapse was independent of the baseline PSA level and PSA doubling time (p = 0.42 and p = 0.09, respectively). This study was nonrandomized with a limited number of patients, and few clinical events were reported. CONCLUSIONS Adding AAP to salvage radiation therapy and goserelin resulted in high bRFS and alt-bRFS. AEs remained manageable, although a close liver surveillance is advised. CTC count appears as a promising biomarker for prognosis and predicting response to treatment. PATIENT SUMMARY Our study was a phase 2 clinical trial that exhibited the efficacy and tolerance of a novel androgen-receptor targeting agent (abiraterone acetate plus prednisone) in patients with prostate cancer who experienced rising prostate-specific antigen after radical prostatectomy, in combination with prostate bed radiotherapy. The results also indicated the feasibility and potential value of circulating tumor cell detection, which constitutes a possible advance in managing prostate cancers.
Collapse
Affiliation(s)
- Loic Ah-Thiane
- Department of Radiotherapy, ICO Rene Gauducheau, St-Herblain, France
| | - Loic Campion
- Department of Biostatistics, ICO Rene Gauducheau, St-Herblain, France
| | - Nedjla Allouache
- Department of Radiotherapy, Francois Baclesse Center, Caen, France
| | - Emmanuel Meyer
- Department of Radiotherapy, Francois Baclesse Center, Caen, France
| | - Pascal Pommier
- Department of Radiotherapy, Leon Berard Center, Lyon, France
| | | | | | - Gilles Créhange
- Department of Radiotherapy, Georges Francois Leclerc Center, Dijon, France
| | - Valentine Guimas
- Department of Radiotherapy, ICO Rene Gauducheau, St-Herblain, France
| | - Emmanuel Rio
- Department of Radiotherapy, ICO Rene Gauducheau, St-Herblain, France
| | - Paul Sargos
- Department of Radiotherapy, Bergonie Institute, Bordeaux, France
| | - Sylvain Ladoire
- Department of Radiotherapy, Georges Francois Leclerc Center, Dijon, France
| | | | - Stéphane Supiot
- Department of Radiotherapy, ICO Rene Gauducheau, St-Herblain, France; Inserm UMR1232, CNRS ERL 6001, Nantes University, Nantes, France.
| |
Collapse
|
10
|
Tao H, Wu F, Li R, Du X, Zhu Y, Dong L, Pan J, Dong B, Xue W. Efficacy and Predictive Factors Analysis of Androgen Deprivation Plus Novel Hormone Therapy as Neoadjuvant Treatment for High-Risk Prostate Cancer. Prostate 2025; 85:198-206. [PMID: 39488849 DOI: 10.1002/pros.24817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 09/24/2024] [Accepted: 10/18/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND This investigation explored the clinical features, pathological outcomes, and biochemical recurrence (BCR) duration among high-risk prostate cancer (HRPC) patients who have undergone neoadjuvant therapy (NAT) in combination with radical prostatectomy (RP) and pelvic lymph node dissection (PLND). Additionally, we identified prognostic indicators that discern pathological complete response (pCR) or minimal residual disease (MRD) and BCR. METHODS In total, we examined 76 HRPC patients, who received NAT with either androgen deprivation therapy (ADT) plus apalutamide or ADT plus abiraterone, with subsequent RP and PLND. We conducted a genetic evaluation of patients receiving neoadjuvant apalutamide. Additionally, patient pathological outcomes, circulating prostate-specific antigen (PSA) response rates, and BCR duration were analyzed. Lastly, we employed uni- and multivariate analyses to screen for prognostic factors that govern pCR or MRD and BCR duration. RESULTS Patient median age and median PSA at presentation were 69 years (IQR: 66-73), and 47.6 ng/mL (IQR: 24.1-105.75), respectively. We observed marked changes in pCR or MRD rates between the two cohorts. In particular, the ADT plus apalutamide cohort (51.5%) exhibited enhanced rates relative to the ADT plus abiraterone cohort (25.6%) (p = 0.03). The median BCR duration was substantially prolonged among neoadjuvant apalutamide cohort relative to the neoadjuvant abiraterone cohort (261 days vs. 76 days, p = 0.04). Using multivariate analysis, we revealed that the postintervention pre-RP PSA content (≤ 0.1 ng/mL vs. > 0.1 ng/mL) remained a substantial stand-alone indicator of pCR or MRD (odds ratio: 10.712, 95% CI: 2.725-42.105, p < 0.001). Furthermore, supplemental analyses revealed that the ADT plus apalutamide cohort exhibited an augmented serum response rate, which, in turn, reduced the post-intervention pre-RP PSA content. Based on our genetic profiling of the neoadjuvant apalutamide cohort demonstrated high-frequency deleterious changes in the AR axis (30.3%), followed by TP53 mutations (15.15%). Patients with defective AR axis experienced a remarkably shorter median BCR duration relative to patients with other or no genetic alterations (52.5 days vs. 286 and 336 days, respectively, p < 0.0001). Furthermore, using multivariate analysis, we demonstrated that achieving pCR or MRD (hazard ratio [HR]: 0.170, 95% CI: 0.061-0.477, p < 0.001) and presence of defective AR signaling (HR: 11.193, 95% CI: 3.499-35.806, p < 0.001) were strong stand-alone indicators of BCR. CONCLUSIONS Herein, we demonstrated the superior performance of ADT plus apalutamide in achieving pCR or MRD and in extending BCR duration among HRPC patients. Post-intervention pre-RP PSA content as well as genetic shifts, especially in the AR axis, are critical indicators of patient pathological and clinical outcomes. These findings highlight the significance of genetic testing and PSA content monitoring in treating HRPC patients.
Collapse
Affiliation(s)
- Hanyang Tao
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fan Wu
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rui Li
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinxing Du
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yinjie Zhu
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liang Dong
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiahua Pan
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Baijun Dong
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Xue
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Jung G, Song B, Ahn H, Hwang SI, Lee HJ, Huh KY, Song SH, Lee S, Byun SS, Hong SK. Oncological outcomes after radical prostatectomy of localized prostate cancer: stratified by magnetic resonance imaging and risk classification. Prostate Int 2024; 12:224-230. [PMID: 39735202 PMCID: PMC11681324 DOI: 10.1016/j.prnil.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 12/31/2024] Open
Abstract
Background We investigated whether combining T2-weighted magnetic resonance imaging (MRI) findings and clinical risk categories improves upon established prognostic indicators of oncological outcomes in prostate cancer. Methods Patients who underwent radical prostatectomy, but not preoperative hormone therapy, radiotherapy, or chemotherapy, for localized prostate cancer at Seoul National University Bundang Hospital from October 2007 to April 2016 were included. MRIs were classified according to the Prostate Imaging-Reporting and Data System (PI-RADS). Patients were divided into the following five groups: 1, no focal suspicious lesion; 2, organ-confined suspicious lesion PI-RADS ≤3; 3, organ-confined suspicious lesion PI-RADS 4 or 5; 4, suspicious lesion with extraprostatic extension (EPE), no seminal vesicle invasion (SVI); 5, suspicious lesion with EPE and SVI. Risk classified according to the National Comprehensive Cancer Network (NCCN) and MRI findings were combined to analyze survival curves for biochemical recurrence (BCR)-free and metastasis-free survival. The area under a time-dependent receiver operating characteristic was analyzed for event prediction after 5 years. Results We analyzed 1,290 patients. In multivariate Cox regression models, PI-RADS ≥4 (hazard ratio [HR] 2.33, P < 0.001), EPE (HR 1.46, P = 0.027), SVI (HR 5.03, P < 0.001) and NCCN high-risk (HR 2.33, 95% CI 1.66-3.26, P < 0.001) were associated with BCR. For metastasis, EPE (HR 2.33, P = 0.047), SVI (HR 13.08, P < 0.001) and NCCN high-risk (HR 2.78, P = 0.026) were independent risk factors. Depending on MRI group, BCR-free survival significantly decreased in NCCN intermediate-risk (P = 0.001) and high-risk (P < 0.001) groups, and metastasis-free survival decreased in the intermediate-risk group (P = 0.39) and significantly decreased in the high-risk (P < 0.001) group. Adding MRI group to NCCN risk classification significantly improved the predictive accuracy for BCR in comparison with NCCN risk classification alone (P = 0.042), but not for metastasis (P = 0.012). Conclusion Combining prostate MRI with NCCN risk classification improves the prediction value of BCR following radical prostatectomy for localized prostate cancer.
Collapse
Affiliation(s)
- Gyoohwan Jung
- Department of Urology, Hanyang University College of Medicine, Seoul, Korea
| | - Byeongdo Song
- Department of Urology, Hanyang University Guri Hospital, Guri, Kyunggi-Do, Korea
| | - Hyungwoo Ahn
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Sung Il Hwang
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Hak Jong Lee
- Department of Radiology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Ki Young Huh
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Korea
| | - Sang Hun Song
- Department of Urology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Sangchul Lee
- Department of Urology, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Urology, Seoul National University College of Medicine, Seoul, Korea
| | - Seok-Soo Byun
- Department of Urology, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Medical Device Development, Seoul National University College of Medicine, Seoul, Korea
| | - Sung Kyu Hong
- Department of Urology, Seoul National University Bundang Hospital, Seongnam, Korea
- Department of Urology, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
12
|
Akpinar C, Kuru Oz D, Oktar A, Ozsoy F, Ozden E, Haliloglu N, Ibis MA, Suer E, Baltaci S. Preoperative multiparametric magnetic resonance imaging based risk stratification system for predicting biochemical recurrence after radical prostatectomy. Surg Oncol 2024; 57:102150. [PMID: 39348786 DOI: 10.1016/j.suronc.2024.102150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/22/2024] [Accepted: 09/25/2024] [Indexed: 10/02/2024]
Abstract
BACKGROUND Multiparametric magnetic resonance imaging (mpMRI) is used as a current marker in preoperative staging and surgical decision-making, but current evidence on predicting post-surgical oncological outcomes based on preoperative mpMRI findings is limited. In this study We aimed to develop a risk classification based on mpMRI and mpMRI-derived biopsy findings to predict early biochemical recurrence (BCR) after radical prostatectomy. METHODS Between January 2017 and January 2023, the data of 289 patients who underwent mpMRI, transrectal ultrasound-guided cognitive and fusion targeted biopsies, and subsequent radical prostatectomy (RP) with or without pelvic lymph node dissection in a single center were retrospectively re-evaluated. BCR was defined as a prostate specific-antigen (PSA) ≥ 0.2 ng/mL at least twice after RP. Multivariate logistic regression models tested the predictors of BCR. The regression tree analysis stratified patients into risk groups based on preoperative mpMRI characteristics. Receiver operating characteristic (ROC)-derived area under the curve (AUC) estimates were used to test the accuracy of the regression tree-derived risk stratification tool. RESULTS BCR was detected in 47 patients (16.2 %) at a median follow-up of 24 months. In mpMRI based multivariate analyses, the maximum diameter of the index lesion (HR 1.081, 95%Cl 1.015-1.151, p = 0.015) the presence of PI-RADS 5 lesions (HR 2.604, 95%Cl 1.043-6.493, p = 0.04), ≥iT3a stage (HR 2.403, 95%Cl 1.013-5.714, p = 0.046) and ISUP grade ≥4 on biopsy (HR 2.440, 95%Cl 1.123-5.301, p = 0.024) were independent predictors of BCR. In regression tree analysis, patients were stratified into three risk groups: maximum diameter of index lesion, biopsy ISUP grade, and clinical stage on mpMRI. The regression tree-derived risk stratification model had moderate-good accuracy in predicting early BCR (AUC 77 %) CONCLUSION: Straightforward mpMRI and mpMRI-derived biopsy-based risk stratification for BCR prediction provide an additional clinical predictive model to the currently available pathological risk tools.
Collapse
Affiliation(s)
- Cagri Akpinar
- Department of Urology, Ankara Etlik City Hospital, Ankara, Turkey.
| | - Digdem Kuru Oz
- Department of Radiology, School of Medicine, Ankara University, Ankara, Turkey.
| | - Alkan Oktar
- Department of Urology, School of Medicine, Ankara University, Ankara, Turkey.
| | - Furkan Ozsoy
- Department of Urology, School of Medicine, Ankara University, Ankara, Turkey.
| | - Eriz Ozden
- Department of Radiology, School of Medicine, Ankara University, Ankara, Turkey.
| | - Nuray Haliloglu
- Department of Radiology, School of Medicine, Ankara University, Ankara, Turkey.
| | - Muhammed Arif Ibis
- Department of Urology, School of Medicine, Ankara University, Ankara, Turkey.
| | - Evren Suer
- Department of Urology, School of Medicine, Ankara University, Ankara, Turkey.
| | - Sumer Baltaci
- Department of Urology, School of Medicine, Ankara University, Ankara, Turkey.
| |
Collapse
|
13
|
Schrader A, Netzer N, Hielscher T, Görtz M, Zhang KS, Schütz V, Stenzinger A, Hohenfellner M, Schlemmer HP, Bonekamp D. Prostate cancer risk assessment and avoidance of prostate biopsies using fully automatic deep learning in prostate MRI: comparison to PI-RADS and integration with clinical data in nomograms. Eur Radiol 2024; 34:7909-7920. [PMID: 38955845 PMCID: PMC11557625 DOI: 10.1007/s00330-024-10818-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 04/15/2024] [Accepted: 04/21/2024] [Indexed: 07/04/2024]
Abstract
OBJECTIVES Risk calculators (RCs) improve patient selection for prostate biopsy with clinical/demographic information, recently with prostate MRI using the prostate imaging reporting and data system (PI-RADS). Fully-automated deep learning (DL) analyzes MRI data independently, and has been shown to be on par with clinical radiologists, but has yet to be incorporated into RCs. The goal of this study is to re-assess the diagnostic quality of RCs, the impact of replacing PI-RADS with DL predictions, and potential performance gains by adding DL besides PI-RADS. MATERIAL AND METHODS One thousand six hundred twenty-seven consecutive examinations from 2014 to 2021 were included in this retrospective single-center study, including 517 exams withheld for RC testing. Board-certified radiologists assessed PI-RADS during clinical routine, then systematic and MRI/Ultrasound-fusion biopsies provided histopathological ground truth for significant prostate cancer (sPC). nnUNet-based DL ensembles were trained on biparametric MRI predicting the presence of sPC lesions (UNet-probability) and a PI-RADS-analogous five-point scale (UNet-Likert). Previously published RCs were validated as is; with PI-RADS substituted by UNet-Likert (UNet-Likert-substituted RC); and with both UNet-probability and PI-RADS (UNet-probability-extended RC). Together with a newly fitted RC using clinical data, PI-RADS and UNet-probability, existing RCs were compared by receiver-operating characteristics, calibration, and decision-curve analysis. RESULTS Diagnostic performance remained stable for UNet-Likert-substituted RCs. DL contained complementary diagnostic information to PI-RADS. The newly-fitted RC spared 49% [252/517] of biopsies while maintaining the negative predictive value (94%), compared to PI-RADS ≥ 4 cut-off which spared 37% [190/517] (p < 0.001). CONCLUSIONS Incorporating DL as an independent diagnostic marker for RCs can improve patient stratification before biopsy, as there is complementary information in DL features and clinical PI-RADS assessment. CLINICAL RELEVANCE STATEMENT For patients with positive prostate screening results, a comprehensive diagnostic workup, including prostate MRI, DL analysis, and individual classification using nomograms can identify patients with minimal prostate cancer risk, as they benefit less from the more invasive biopsy procedure. KEY POINTS The current MRI-based nomograms result in many negative prostate biopsies. The addition of DL to nomograms with clinical data and PI-RADS improves patient stratification before biopsy. Fully automatic DL can be substituted for PI-RADS without sacrificing the quality of nomogram predictions. Prostate nomograms show cancer detection ability comparable to previous validation studies while being suitable for the addition of DL analysis.
Collapse
Affiliation(s)
- Adrian Schrader
- Division of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg University Medical School, Heidelberg, Germany
| | - Nils Netzer
- Division of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Heidelberg University Medical School, Heidelberg, Germany
| | - Thomas Hielscher
- Division of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Magdalena Görtz
- Department of Urology, University of Heidelberg Medical Center, Heidelberg, Germany
- Junior Clinical Cooperation Unit 'Multiparametric Methods for Early Detection of Prostate Cancer', German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kevin Sun Zhang
- Division of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Viktoria Schütz
- Department of Urology, University of Heidelberg Medical Center, Heidelberg, Germany
| | - Albrecht Stenzinger
- Institute of Pathology, University of Heidelberg Medical Center, Heidelberg, Germany
| | - Markus Hohenfellner
- Department of Urology, University of Heidelberg Medical Center, Heidelberg, Germany
| | - Heinz-Peter Schlemmer
- Division of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
| | - David Bonekamp
- Division of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Heidelberg University Medical School, Heidelberg, Germany.
- National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany.
| |
Collapse
|
14
|
Stabile A, Giannarini G, Radtke JP, Crestani A, Albers P, Briganti A. PI-QUAL version 2: the urologist's perspective. Eur Radiol 2024; 34:7063-7064. [PMID: 38904759 DOI: 10.1007/s00330-024-10844-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 05/31/2024] [Accepted: 06/03/2024] [Indexed: 06/22/2024]
Affiliation(s)
- Armando Stabile
- Department of Urology and Division of Experimental Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gianluca Giannarini
- Urology Unit, Santa Maria della Misericordia University Hospital, Udine, Italy.
| | - Jan Philipp Radtke
- Department of Urology, University Hospital, Medical Faculty, Heinrich-Heine University Duesseldorf, Duesseldorf, Germany
- Department of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Alessandro Crestani
- Urology Unit, Santa Maria della Misericordia University Hospital, Udine, Italy
| | - Peter Albers
- Department of Urology, University Hospital, Medical Faculty, Heinrich-Heine University Duesseldorf, Duesseldorf, Germany
- Division of Personalized Early Detection of Prostate Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Alberto Briganti
- Department of Urology and Division of Experimental Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
15
|
Morelli L, Paganelli C, Marvaso G, Parrella G, Annunziata S, Vicini MG, Zaffaroni M, Pepa M, Summers PE, De Cobelli O, Petralia G, Jereczek-Fossa BA, Baroni G. Addressing intra- and inter-institution variability of a radiomic framework based on Apparent Diffusion Coefficient in prostate cancer. Med Phys 2024; 51:8096-8107. [PMID: 39172115 DOI: 10.1002/mp.17355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 06/27/2024] [Accepted: 08/05/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Prostate cancer (PCa) is a highly heterogeneous disease, making tailored treatment approaches challenging. Magnetic resonance imaging (MRI), notably diffusion-weighted imaging (DWI) and the derived Apparent Diffusion Coefficient (ADC) maps, plays a crucial role in PCa characterization. In this context, radiomics is a very promising approach able to disclose insights from MRI data. However, the sensitivity of radiomic features to MRI settings, encompassing DWI protocols and multicenter variations, requires the development of robust and generalizable models. PURPOSE To develop a comprehensive radiomics framework for noninvasive PCa characterization using ADC maps, focusing on identifying reliable imaging biomarkers against intra- and inter-institution variations. MATERIALS AND METHODS Two patient cohorts, including an internal cohort (118 PCa patients) used for both training (75%) and hold-out testing (25%), and an external cohort (50 PCa patients) for independent testing, were employed in the study. DWI images were acquired with three different DWI protocols on two different MRI scanners: two DWI protocols acquired on a 1.5-T scanner for the internal cohort, and one DWI protocol acquired on a 3-T scanner for the external cohort. One hundred and seven radiomics features (i.e., shape, first order, texture) were extracted from ADC maps of the whole prostate gland. To address variations in DWI protocols and multicenter variability, a dedicated pipeline, including two-way ANOVA, sequential-feature-selection (SFS), and ComBat features harmonization was implemented. Mann-Whitney U-tests (α = 0.05) were performed to find statistically significant features dividing patients with different tumor characteristics in terms of Gleason score (GS) and T-stage. Support-Vector-Machine models were then developed to predict GS and T-stage, and the performance was assessed through the area under the curve (AUC) of receiver-operating-characteristic curves. RESULTS Downstream of ANOVA, two subsets of 38 and 41 features stable against DWI protocol were identified for GS and T-stage, respectively. Among these, SFS revealed the most predictive features, yielding an AUC of 0.75 (GS) and 0.70 (T-stage) in the hold-out test. Employing ComBat harmonization improved the external-test performance of the GS model, raising AUC from 0.72 to 0.78. CONCLUSION By incorporating stable features with a harmonization procedure and validating the model on an external dataset, model robustness, and generalizability were assessed, highlighting the potential of ADC and radiomics for PCa characterization.
Collapse
Affiliation(s)
- Letizia Morelli
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, Milan, Italy
| | - Chiara Paganelli
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, Milan, Italy
| | - Giulia Marvaso
- Department of Radiation Oncology, European Institute of Oncology (IEO), Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Giovanni Parrella
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, Milan, Italy
| | - Simone Annunziata
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, Milan, Italy
| | - Maria Giulia Vicini
- Department of Radiation Oncology, European Institute of Oncology (IEO), Milan, Italy
| | - Mattia Zaffaroni
- Department of Radiation Oncology, European Institute of Oncology (IEO), Milan, Italy
| | - Matteo Pepa
- Department of Radiation Oncology, European Institute of Oncology (IEO), Milan, Italy
| | | | - Ottavio De Cobelli
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Urology, European Institute of Oncology (IEO), Milan, Italy
| | - Giuseppe Petralia
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- Department of Radiology, European Institute of Oncology (IEO), Milan, Italy
| | - Barbara Alicja Jereczek-Fossa
- Department of Radiation Oncology, European Institute of Oncology (IEO), Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Guido Baroni
- Department of Electronics, Information and Bioengineering (DEIB), Politecnico di Milano, Milan, Italy
| |
Collapse
|
16
|
Westhofen T, Buchner A, Lennartz S, Rodler S, Eismann L, Aydogdu C, Askari-Motlagh D, Berg E, Feyerabend E, Kazmierczak P, Jokisch F, Becker A, Stief CG, Kretschmer A. Optimizing risk stratification for intermediate-risk prostate cancer - the prognostic value of baseline health-related quality of life. World J Urol 2024; 42:585. [PMID: 39427278 PMCID: PMC11491415 DOI: 10.1007/s00345-024-05298-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 09/24/2024] [Indexed: 10/22/2024] Open
Abstract
OBJECTIVE To investigate the prognostic value of baseline health-related quality of life (HRQOL) for patients with intermediate-risk localized prostate cancer (IR-PCa) undergoing radical prostatectomy (RP). METHODS 4780 patients with IR-PCa according to NCCN risk stratification were identified from a prospectively maintained database. All patients were treated with RP and had prospectively assessed baseline HRQOL. Main outcomes were oncologic endpoints metastasis-free survival (MFS); biochemical recurrence free survival (BRFS) and overall survival (OS). Multivariable Cox regression models assessed prognostic significance of baseline global health status (GHS) on survival outcomes. Harrell's discrimination C-index was applied to calculate the predictive accuracy of the model. Decision curve analysis (DCA) tested the clinical net benefit associated with adding the GHS domain to our multivariable model (p < 0.05). RESULTS Median follow-up was 51 months. Multivariable analysis confirmed baseline GHS as an independent predictor for increased MFS (HR 0.976, 95%CI 0.96-0.99; p < 0.001), increased BRFS (HR 0.993, 95%CI 0.99-1.00; p = 0.027) and increased OS (HR 0.969, 95%CI 0.95-0.99; p = 0.002), indicating a relative risk reduction of 2.4% for MFS, 0.7% for BRFS and 3.1% for OS per 1-point increase of baseline GHS. Baseline HRQOL improved discrimination in predicting MFS, BRFS and OS. DCA revealed a net benefit over all threshold probabilities. CONCLUSIONS We found baseline HRQOL to substantially improve risk stratification for the heterogeneous cohort of IR-PCa. Baseline HRQOL accurately predicts increased MFS, BRFS and OS. Our findings therefore support the role of preoperative HRQOL as an adjunct to established prognosticators for IR-PCa, potentially facilitating guidance of therapy.
Collapse
Affiliation(s)
- Thilo Westhofen
- Department of Urology, Ludwig-Maximilians-University of Munich, Marchioninistrasse 15, 81377, Munich, Germany.
| | - Alexander Buchner
- Department of Urology, Ludwig-Maximilians-University of Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Simon Lennartz
- Institute for Diagnostic and Interventional Radiology, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
| | - Severin Rodler
- Department of Urology, Ludwig-Maximilians-University of Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Lennert Eismann
- Department of Urology, Ludwig-Maximilians-University of Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Can Aydogdu
- Department of Urology, Ludwig-Maximilians-University of Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Darjusch Askari-Motlagh
- Department of Urology, Ludwig-Maximilians-University of Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Elena Berg
- Department of Urology, Ludwig-Maximilians-University of Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Enya Feyerabend
- Department of Urology, Ludwig-Maximilians-University of Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Philipp Kazmierczak
- Institute for Diagnostic and Interventional Radiology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Friedrich Jokisch
- Department of Urology, Ludwig-Maximilians-University of Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Armin Becker
- Department of Urology, Ludwig-Maximilians-University of Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Christian G Stief
- Department of Urology, Ludwig-Maximilians-University of Munich, Marchioninistrasse 15, 81377, Munich, Germany
| | - Alexander Kretschmer
- Department of Urology, Ludwig-Maximilians-University of Munich, Marchioninistrasse 15, 81377, Munich, Germany
| |
Collapse
|
17
|
Yang DD, Lee LK, Tsui JMG, Leeman JE, McClure HM, Sudhyadhom A, Guthier CV, Taplin ME, Trinh QD, Mouw KW, Martin NE, Orio PF, Nguyen PL, D'Amico AV, Shin KY, Lee KN, King MT. AI-derived Tumor Volume from Multiparametric MRI and Outcomes in Localized Prostate Cancer. Radiology 2024; 313:e240041. [PMID: 39470422 DOI: 10.1148/radiol.240041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/30/2024]
Abstract
Background An artificial intelligence (AI)-based method for measuring intraprostatic tumor volume based on data from MRI may provide prognostic information. Purpose To evaluate whether the total volume of intraprostatic tumor from AI-generated segmentations (VAI) provides independent prognostic information in patients with localized prostate cancer treated with radiation therapy (RT) or radical prostatectomy (RP). Materials and Methods For this retrospective, single-center study (January 2021 to August 2023), patients with cT1-3N0M0 prostate cancer who underwent MRI and were treated with RT or RP were identified. Patients who underwent RT were randomly divided into cross-validation and test RT groups. An AI segmentation algorithm was trained to delineate Prostate Imaging Reporting and Data System (PI-RADS) 3-5 lesions in the cross-validation RT group before providing segmentations for the test RT and RP groups. Cox regression models were used to evaluate the association between VAI and time to metastasis and adjusted for clinical and radiologic factors for combined RT (ie, cross-validation RT and test RT) and RP groups. Areas under the receiver operating characteristic curve (AUCs) were calculated for VAI and National Comprehensive Cancer Network (NCCN) risk categorization for prediction of 5-year metastasis (RP group) and 7-year metastasis (combined RT group). Results Overall, 732 patients were included (combined RT group, 438 patients; RP group, 294 patients). Median ages were 68 years (IQR, 62-73 years) and 61 years (IQR, 56-66 years) for the combined RT group and the RP group, respectively. VAI was associated with metastasis in the combined RT group (median follow-up, 6.9 years; adjusted hazard ratio [AHR], 1.09 per milliliter increase; 95% CI: 1.04, 1.15; P = .001) and the RP group (median follow-up, 5.5 years; AHR, 1.22; 95% CI: 1.08, 1.39; P = .001). AUCs for 7-year metastasis for the combined RT group for VAI and NCCN risk category were 0.84 (95% CI: 0.74, 0.94) and 0.74 (95% CI: 0.80, 0.98), respectively (P = .02). Five-year AUCs for the RP group for VAI and NCCN risk category were 0.89 (95% CI: 0.80, 0.98) and 0.79 (95% CI: 0.64, 0.94), respectively (P = .25). Conclusion The volume of AI-segmented lesions was an independent, prognostic factor for localized prostate cancer. © RSNA, 2024 Supplemental material is available for this article.
Collapse
Affiliation(s)
- David D Yang
- From the Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, 75 Francis St, Boston, MA 02115 (D.D.Y., J.E.L., A.S., C.V.G., K.W.M., N.E.M., P.F.O., P.L.N., A.V.D., K.Y.S., K.N.L., M.T.K.); Departments of Radiology (L.K.L.) and Urology (Q.D.T.), Brigham and Women's Hospital, Boston, Mass; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Ma (H.M.M., M.E.T.); and Department of Radiation Oncology, McGill University, Montreal, Canada (J.M.G.T.)
| | - Leslie K Lee
- From the Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, 75 Francis St, Boston, MA 02115 (D.D.Y., J.E.L., A.S., C.V.G., K.W.M., N.E.M., P.F.O., P.L.N., A.V.D., K.Y.S., K.N.L., M.T.K.); Departments of Radiology (L.K.L.) and Urology (Q.D.T.), Brigham and Women's Hospital, Boston, Mass; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Ma (H.M.M., M.E.T.); and Department of Radiation Oncology, McGill University, Montreal, Canada (J.M.G.T.)
| | - James M G Tsui
- From the Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, 75 Francis St, Boston, MA 02115 (D.D.Y., J.E.L., A.S., C.V.G., K.W.M., N.E.M., P.F.O., P.L.N., A.V.D., K.Y.S., K.N.L., M.T.K.); Departments of Radiology (L.K.L.) and Urology (Q.D.T.), Brigham and Women's Hospital, Boston, Mass; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Ma (H.M.M., M.E.T.); and Department of Radiation Oncology, McGill University, Montreal, Canada (J.M.G.T.)
| | - Jonathan E Leeman
- From the Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, 75 Francis St, Boston, MA 02115 (D.D.Y., J.E.L., A.S., C.V.G., K.W.M., N.E.M., P.F.O., P.L.N., A.V.D., K.Y.S., K.N.L., M.T.K.); Departments of Radiology (L.K.L.) and Urology (Q.D.T.), Brigham and Women's Hospital, Boston, Mass; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Ma (H.M.M., M.E.T.); and Department of Radiation Oncology, McGill University, Montreal, Canada (J.M.G.T.)
| | - Heather M McClure
- From the Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, 75 Francis St, Boston, MA 02115 (D.D.Y., J.E.L., A.S., C.V.G., K.W.M., N.E.M., P.F.O., P.L.N., A.V.D., K.Y.S., K.N.L., M.T.K.); Departments of Radiology (L.K.L.) and Urology (Q.D.T.), Brigham and Women's Hospital, Boston, Mass; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Ma (H.M.M., M.E.T.); and Department of Radiation Oncology, McGill University, Montreal, Canada (J.M.G.T.)
| | - Atchar Sudhyadhom
- From the Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, 75 Francis St, Boston, MA 02115 (D.D.Y., J.E.L., A.S., C.V.G., K.W.M., N.E.M., P.F.O., P.L.N., A.V.D., K.Y.S., K.N.L., M.T.K.); Departments of Radiology (L.K.L.) and Urology (Q.D.T.), Brigham and Women's Hospital, Boston, Mass; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Ma (H.M.M., M.E.T.); and Department of Radiation Oncology, McGill University, Montreal, Canada (J.M.G.T.)
| | - Christian V Guthier
- From the Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, 75 Francis St, Boston, MA 02115 (D.D.Y., J.E.L., A.S., C.V.G., K.W.M., N.E.M., P.F.O., P.L.N., A.V.D., K.Y.S., K.N.L., M.T.K.); Departments of Radiology (L.K.L.) and Urology (Q.D.T.), Brigham and Women's Hospital, Boston, Mass; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Ma (H.M.M., M.E.T.); and Department of Radiation Oncology, McGill University, Montreal, Canada (J.M.G.T.)
| | - Mary-Ellen Taplin
- From the Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, 75 Francis St, Boston, MA 02115 (D.D.Y., J.E.L., A.S., C.V.G., K.W.M., N.E.M., P.F.O., P.L.N., A.V.D., K.Y.S., K.N.L., M.T.K.); Departments of Radiology (L.K.L.) and Urology (Q.D.T.), Brigham and Women's Hospital, Boston, Mass; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Ma (H.M.M., M.E.T.); and Department of Radiation Oncology, McGill University, Montreal, Canada (J.M.G.T.)
| | - Quoc-Dien Trinh
- From the Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, 75 Francis St, Boston, MA 02115 (D.D.Y., J.E.L., A.S., C.V.G., K.W.M., N.E.M., P.F.O., P.L.N., A.V.D., K.Y.S., K.N.L., M.T.K.); Departments of Radiology (L.K.L.) and Urology (Q.D.T.), Brigham and Women's Hospital, Boston, Mass; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Ma (H.M.M., M.E.T.); and Department of Radiation Oncology, McGill University, Montreal, Canada (J.M.G.T.)
| | - Kent W Mouw
- From the Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, 75 Francis St, Boston, MA 02115 (D.D.Y., J.E.L., A.S., C.V.G., K.W.M., N.E.M., P.F.O., P.L.N., A.V.D., K.Y.S., K.N.L., M.T.K.); Departments of Radiology (L.K.L.) and Urology (Q.D.T.), Brigham and Women's Hospital, Boston, Mass; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Ma (H.M.M., M.E.T.); and Department of Radiation Oncology, McGill University, Montreal, Canada (J.M.G.T.)
| | - Neil E Martin
- From the Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, 75 Francis St, Boston, MA 02115 (D.D.Y., J.E.L., A.S., C.V.G., K.W.M., N.E.M., P.F.O., P.L.N., A.V.D., K.Y.S., K.N.L., M.T.K.); Departments of Radiology (L.K.L.) and Urology (Q.D.T.), Brigham and Women's Hospital, Boston, Mass; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Ma (H.M.M., M.E.T.); and Department of Radiation Oncology, McGill University, Montreal, Canada (J.M.G.T.)
| | - Peter F Orio
- From the Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, 75 Francis St, Boston, MA 02115 (D.D.Y., J.E.L., A.S., C.V.G., K.W.M., N.E.M., P.F.O., P.L.N., A.V.D., K.Y.S., K.N.L., M.T.K.); Departments of Radiology (L.K.L.) and Urology (Q.D.T.), Brigham and Women's Hospital, Boston, Mass; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Ma (H.M.M., M.E.T.); and Department of Radiation Oncology, McGill University, Montreal, Canada (J.M.G.T.)
| | - Paul L Nguyen
- From the Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, 75 Francis St, Boston, MA 02115 (D.D.Y., J.E.L., A.S., C.V.G., K.W.M., N.E.M., P.F.O., P.L.N., A.V.D., K.Y.S., K.N.L., M.T.K.); Departments of Radiology (L.K.L.) and Urology (Q.D.T.), Brigham and Women's Hospital, Boston, Mass; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Ma (H.M.M., M.E.T.); and Department of Radiation Oncology, McGill University, Montreal, Canada (J.M.G.T.)
| | - Anthony V D'Amico
- From the Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, 75 Francis St, Boston, MA 02115 (D.D.Y., J.E.L., A.S., C.V.G., K.W.M., N.E.M., P.F.O., P.L.N., A.V.D., K.Y.S., K.N.L., M.T.K.); Departments of Radiology (L.K.L.) and Urology (Q.D.T.), Brigham and Women's Hospital, Boston, Mass; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Ma (H.M.M., M.E.T.); and Department of Radiation Oncology, McGill University, Montreal, Canada (J.M.G.T.)
| | - Kee-Young Shin
- From the Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, 75 Francis St, Boston, MA 02115 (D.D.Y., J.E.L., A.S., C.V.G., K.W.M., N.E.M., P.F.O., P.L.N., A.V.D., K.Y.S., K.N.L., M.T.K.); Departments of Radiology (L.K.L.) and Urology (Q.D.T.), Brigham and Women's Hospital, Boston, Mass; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Ma (H.M.M., M.E.T.); and Department of Radiation Oncology, McGill University, Montreal, Canada (J.M.G.T.)
| | - Katie N Lee
- From the Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, 75 Francis St, Boston, MA 02115 (D.D.Y., J.E.L., A.S., C.V.G., K.W.M., N.E.M., P.F.O., P.L.N., A.V.D., K.Y.S., K.N.L., M.T.K.); Departments of Radiology (L.K.L.) and Urology (Q.D.T.), Brigham and Women's Hospital, Boston, Mass; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Ma (H.M.M., M.E.T.); and Department of Radiation Oncology, McGill University, Montreal, Canada (J.M.G.T.)
| | - Martin T King
- From the Department of Radiation Oncology, Brigham and Women's Hospital/Dana-Farber Cancer Institute, 75 Francis St, Boston, MA 02115 (D.D.Y., J.E.L., A.S., C.V.G., K.W.M., N.E.M., P.F.O., P.L.N., A.V.D., K.Y.S., K.N.L., M.T.K.); Departments of Radiology (L.K.L.) and Urology (Q.D.T.), Brigham and Women's Hospital, Boston, Mass; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Ma (H.M.M., M.E.T.); and Department of Radiation Oncology, McGill University, Montreal, Canada (J.M.G.T.)
| |
Collapse
|
18
|
Xu H, Xing Z, Wang J, Lv Z, Deng P, Hong Y, Li Y. Development and External Validation of Nomograms for Predicting Disease-Free Survival and Overall Survival in Patients with cT1-ccRCC After Partial Nephrectomy: A Multicenter Retrospective Study. Ann Surg Oncol 2024; 31:5827-5838. [PMID: 38971957 DOI: 10.1245/s10434-024-15718-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/19/2024] [Indexed: 07/08/2024]
Abstract
BACKGROUND To develop a novel nomogram for predicting 2-year and 5-year disease-free survival (DFS) and overall survival (OS) in patients with cT1-clear cell renal cell carcinoma (ccRCC) undergoing partial nephrectomy (PN). METHODS A retrospective study was conducted across five urological centers, including 940 patients who underwent PN for cT1N0M0-ccRCC. Four centers were randomly selected to constitute the training group, while the remaining center served as the testing group. We employed the LASSO and multivariate Cox regression to develop new nomograms. The 1,000 bootstrap-corrected c-index, net reclassification improvement (NRI) and receiver operating characteristic curve were employed to compare the predictive abilities of new nomograms with the widely used UUIS and SSIGN models. Finally, the novel nomograms underwent external validation. RESULTS The training group included 714 patients, while the testing group consisted of 226 patients. The bootstrap-corrected c-indexes for the DFS and OS model were 0.870 and 0.902, respectively. In the training cohort, the AUC for the DFS and OS models at 2 years and 5 years were 0.953, 0.902, 0.988, and 0.911, respectively. These values were also assessed in the testing cohort. The predictive capabilities of the new nomograms surpassed those of the UUIS and SSIGN models (NRI > 0). Decision curve analysis demonstrated that the novel nomograms provide greater net benefits compared to the UUIS and SSIGN models. CONCLUSIONS Our novel nomograms demonstrated strong predictive ability for forecasting oncological outcomes in cT1-ccRCC patients after PN. These user-friendly nomograms are simple and convenient for clinical application, providing tangible clinical benefits.
Collapse
Affiliation(s)
- Haozhe Xu
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhuo Xing
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jie Wang
- Department of Oncology, Hunan Cancer Hospital, Changsha, Hunan, China
| | - Zhengtong Lv
- Department of Urology, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Piye Deng
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yulong Hong
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuan Li
- Department of Urology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
19
|
Sun R, A J, Yu H, Wang Y, He M, Tan L, Cheng H, Zhang J, Wang Y, Sun X, Lyu M, Qu M, Huang L, Li Z, Zhang W, Ma K, Dong Z, Ge W, Zhang Y, Ding X, Yang B, Hou J, Xu C, Wang L, Zhu Y, Guo T, Gao X, Yang C. Proteomic landscape profiling of primary prostate cancer reveals a 16-protein panel for prognosis prediction. Cell Rep Med 2024; 5:101679. [PMID: 39168102 PMCID: PMC11384950 DOI: 10.1016/j.xcrm.2024.101679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 06/13/2024] [Accepted: 07/16/2024] [Indexed: 08/23/2024]
Abstract
Prostate cancer (PCa) is the most common malignant tumor in men. Currently, there are few prognosis indicators for predicting PCa outcomes and guiding treatments. Here, we perform comprehensive proteomic profiling of 918 tissue specimens from 306 Chinese patients with PCa using data-independent acquisition mass spectrometry (DIA-MS). We identify over 10,000 proteins and define three molecular subtypes of PCa with significant clinical and proteomic differences. We develop a 16-protein panel that effectively predicts biochemical recurrence (BCR) for patients with PCa, which is validated in six published datasets and one additional 99-biopsy-sample cohort by targeted proteomics. Interestingly, this 16-protein panel effectively predicts BCR across different International Society of Urological Pathology (ISUP) grades and pathological stages and outperforms the D'Amico risk classification system in BCR prediction. Furthermore, double knockout of NUDT5 and SEPTIN8, two components from the 16-protein panel, significantly suppresses the PCa cells to proliferate, invade, and migrate, suggesting the combination of NUDT5 and SEPTIN8 may provide new approaches for PCa treatment.
Collapse
Affiliation(s)
- Rui Sun
- School of Medicine, Westlake University, Hangzhou, Zhejiang Province, China; Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province 310030, China; Research Center for Industries of the Future, Westlake University, 600 Dunyu Road, Hangzhou, Zhejiang 310030, China; Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Jun A
- School of Medicine, Westlake University, Hangzhou, Zhejiang Province, China; Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province 310030, China; Research Center for Industries of the Future, Westlake University, 600 Dunyu Road, Hangzhou, Zhejiang 310030, China; Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Haolan Yu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China; CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yan Wang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China; Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Miaoxia He
- Department of Pathology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Lingling Tan
- Westlake Omics (Hangzhou) Biotechnology Co., Ltd., Hangzhou 310024, China
| | - Honghan Cheng
- School of Medicine, Westlake University, Hangzhou, Zhejiang Province, China; Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province 310030, China; Research Center for Industries of the Future, Westlake University, 600 Dunyu Road, Hangzhou, Zhejiang 310030, China; Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Jili Zhang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yingrui Wang
- School of Medicine, Westlake University, Hangzhou, Zhejiang Province, China; Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province 310030, China; Research Center for Industries of the Future, Westlake University, 600 Dunyu Road, Hangzhou, Zhejiang 310030, China; Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Xiaochen Sun
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China; CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Mengge Lyu
- School of Medicine, Westlake University, Hangzhou, Zhejiang Province, China; Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province 310030, China; Research Center for Industries of the Future, Westlake University, 600 Dunyu Road, Hangzhou, Zhejiang 310030, China; Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Min Qu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Lingling Huang
- School of Medicine, Westlake University, Hangzhou, Zhejiang Province, China; Westlake Omics (Hangzhou) Biotechnology Co., Ltd., Hangzhou 310024, China
| | - Zijian Li
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Wenhui Zhang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Kunpeng Ma
- School of Medicine, Westlake University, Hangzhou, Zhejiang Province, China; Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province 310030, China; Research Center for Industries of the Future, Westlake University, 600 Dunyu Road, Hangzhou, Zhejiang 310030, China; Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Zhenyang Dong
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Weigang Ge
- Westlake Omics (Hangzhou) Biotechnology Co., Ltd., Hangzhou 310024, China
| | - Yun Zhang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xuan Ding
- Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province 310030, China; Research Center for Industries of the Future, Westlake University, 600 Dunyu Road, Hangzhou, Zhejiang 310030, China; Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Bo Yang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jianguo Hou
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Chuanliang Xu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China; Shanghai Key Laboratory of Cell Engineering, Shanghai 200433, China
| | - Linhui Wang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China; Shanghai Key Laboratory of Cell Engineering, Shanghai 200433, China
| | - Yi Zhu
- School of Medicine, Westlake University, Hangzhou, Zhejiang Province, China; Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province 310030, China; Research Center for Industries of the Future, Westlake University, 600 Dunyu Road, Hangzhou, Zhejiang 310030, China; Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China
| | - Tiannan Guo
- School of Medicine, Westlake University, Hangzhou, Zhejiang Province, China; Westlake Center for Intelligent Proteomics, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang Province 310030, China; Research Center for Industries of the Future, Westlake University, 600 Dunyu Road, Hangzhou, Zhejiang 310030, China; Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, China.
| | - Xu Gao
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China; Shanghai Key Laboratory of Cell Engineering, Shanghai 200433, China.
| | - Chenghua Yang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai, China; Shanghai Key Laboratory of Cell Engineering, Shanghai 200433, China.
| |
Collapse
|
20
|
Li T, Xu M, Yang S, Wang G, Liu Y, Liu K, Zhao K, Su X. Development and validation of [18 F]-PSMA-1007 PET-based radiomics model to predict biochemical recurrence-free survival following radical prostatectomy. Eur J Nucl Med Mol Imaging 2024; 51:2806-2818. [PMID: 38691111 DOI: 10.1007/s00259-024-06734-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/23/2024] [Indexed: 05/03/2024]
Abstract
PURPOSE Biochemical recurrence (BCR) following radical prostatectomy (RP) is a significant concern for patients with prostate cancer. Reliable prediction models are needed to identify patients at risk for BCR and facilitate appropriate management. This study aimed to develop and validate a clinical-radiomics model based on preoperative [18 F]PSMA-1007 PET for predicting BCR-free survival (BRFS) in patients who underwent RP for prostate cancer. MATERIALS AND METHODS A total of 236 patients with histologically confirmed prostate cancer who underwent RP were retrospectively analyzed. All patients had a preoperative [18 F]PSMA-1007 PET/CT scan. Radiomics features were extracted from the primary tumor region on PET images. A radiomics signature was developed using the least absolute shrinkage and selection operator (LASSO) Cox regression model. The performance of the radiomics signature in predicting BRFS was assessed using Harrell's concordance index (C-index). The clinical-radiomics nomogram was constructed using the radiomics signature and clinical features. The model was externally validated in an independent cohort of 98 patients. RESULTS The radiomics signature comprised three features and demonstrated a C-index of 0.76 (95% CI: 0.60-0.91) in the training cohort and 0.71 (95% CI: 0.63-0.79) in the validation cohort. The radiomics signature remained an independent predictor of BRFS in multivariable analysis (HR: 2.48, 95% CI: 1.47-4.17, p < 0.001). The clinical-radiomics nomogram significantly improved the prediction performance (C-index: 0.81, 95% CI: 0.66-0.95, p = 0.007) in the training cohort and (C-index: 0.78 95% CI: 0.63-0.89, p < 0.001) in the validation cohort. CONCLUSION We developed and validated a novel [18 F]PSMA-1007 PET-based clinical-radiomics model that can predict BRFS following RP in prostate cancer patients. This model may be useful in identifying patients with a higher risk of BCR, thus enabling personalized risk stratification and tailored management strategies.
Collapse
Affiliation(s)
- Tiancheng Li
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, People's Republic of China
| | - Mimi Xu
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, People's Republic of China
| | - Shuye Yang
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, People's Republic of China
| | - Guolin Wang
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, People's Republic of China
| | - Yinuo Liu
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, People's Republic of China
| | - Kaifeng Liu
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, People's Republic of China
| | - Kui Zhao
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, People's Republic of China
| | - Xinhui Su
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou, 310003, People's Republic of China.
| |
Collapse
|
21
|
Calleris G, Filleron T, Kesch C, Roubaud G, Pradère B, Cabarrou B, Malavaud B, Roupret M, Mourey L, Ploussard G. Surgery with or Without Darolutamide in High-risk and/or Locally Advanced Prostate Cancer: The SUGAR (CCAFU-PR2) Phase 2 Trial Rationale and Protocol. Eur Urol Oncol 2024; 7:494-500. [PMID: 37806843 DOI: 10.1016/j.euo.2023.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 09/25/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND High-risk prostate cancer (PCa) patients frequently experience recurrence and progression after radical prostatectomy (RP). Neoadjuvant androgen deprivation therapy (ADT) has not demonstrated a clear oncological benefit and is not currently recommended. OBJECTIVE The SUGAR trial is the first phase 2, randomised, controlled, multicentre, noncommercial, open-label study investigating single-agent perioperative darolutamide compared with the standard of care (ie, upfront RP, without neoadjuvant ADT). DESIGN, SETTING, AND PARTICIPANTS SUGAR aims to randomise 240 men affected by nonmetastatic PCa, with the major eligibility criteria being International Society of Urological Pathology grade group ≥4, seminal vesicle invasion at magnetic resonance imaging and/or clinically node-positive disease. Patients in the experimental arm will undergo neoadjuvant darolutamide monotherapy, RP, and adjuvant darolutamide, completing 9 mo of treatment. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The primary endpoint is noncurable recurrence-free survival, an innovative and clinically meaningful measure; the secondary endpoints encompass safety; recurrence-free, metastasis-free, and overall survival; pathological response; and quality of life. A predictive biomarker analysis will also be performed. RESULTS AND LIMITATIONS Initial data suggest that intensified neoadjuvant treatment with androgen receptor signalling inhibitors (ARSIs) is associated with a sustained pathological response and may improve outcomes, via tumour downstaging and micrometastasis eradication. ARSI monotherapy could further enhance tolerability. CONCLUSIONS SUGAR will provide efficacy and safety information on perioperative darolutamide monotherapy compared with upfront RP, in a contemporary high-risk PCa population undergoing surgery. PATIENT SUMMARY The on-going SUGAR clinical trial evaluates 9 mo of darolutamide treatment in addition to radical prostatectomy, in men affected by prostate cancer with specific high-risk characteristics. It investigates whether this hormonal treatment can lower the rates of noncurable recurrences, maintaining a favourable tolerability profile.
Collapse
Affiliation(s)
- Giorgio Calleris
- Department of Urology UROSUD, La Croix du Sud Hospital, Toulouse, France; Department of Urology, San Giovanni Battista Hospital, Città della Salute e della Scienza and University of Turin, Turin, Italy
| | - Thomas Filleron
- Biostatistics & Health Data Science Unit, Institut Claudius Regaud, Institut Universitaire du Cancer Toulouse-Oncopole, Toulouse, France
| | - Claudia Kesch
- Department of Urology UROSUD, La Croix du Sud Hospital, Toulouse, France; Department of Urology and West German Cancer Center, University Hospital Essen, Essen, Germany
| | | | - Benjamin Pradère
- Department of Urology UROSUD, La Croix du Sud Hospital, Toulouse, France
| | - Bastien Cabarrou
- Biostatistics & Health Data Science Unit, Institut Claudius Regaud, Institut Universitaire du Cancer Toulouse-Oncopole, Toulouse, France
| | - Bernard Malavaud
- Surgery Department, Institut Claudius Regaud, Institut Universitaire du Cancer Toulouse-Oncopole, Toulouse, France
| | - Morgan Roupret
- GRC 5 Predictive Onco-Urology Research Group and Urology Department, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris, Sorbonne University, Paris, France
| | - Loic Mourey
- Oncology Department, Institut Claudius Regaud, Institut Universitaire du Cancer Toulouse-Oncopole, Toulouse, France
| | - Guillaume Ploussard
- Department of Urology UROSUD, La Croix du Sud Hospital, Toulouse, France; Surgery Department, Institut Claudius Regaud, Institut Universitaire du Cancer Toulouse-Oncopole, Toulouse, France.
| |
Collapse
|
22
|
Mazzone E, Gandaglia G, Robesti D, Rajwa P, Gomez Rivas J, Ibáñez L, Soeterik TFW, Bianchi L, Afferi L, Kesch C, Darr C, Guo H, Zhuang J, Zattoni F, Fendler WP, Amparore D, Huebner NA, Giesen A, Joniau S, Schiavina R, Brunocilla E, Mattei A, Dal Moro F, Moreno Sierra J, Porpiglia F, Picchio M, Chiti A, van den Bergh R, Shariat SF, Montorsi F, Briganti A. Which Patients with Prostate Cancer and Lymph Node Uptake at Preoperative Prostate-specific Membrane Antigen Positron Emission Tomography/Computerized Tomography Scan Are at a Higher Risk of Prostate-specific Antigen Persistence After Radical Prostatectomy? Identifying Indicators of Systemic Disease by Integrating Clinical, Magnetic Resonance Imaging, and Functional Imaging Parameters. Eur Urol Oncol 2024; 7:231-240. [PMID: 37689506 DOI: 10.1016/j.euo.2023.08.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/11/2023] [Accepted: 08/23/2023] [Indexed: 09/11/2023]
Abstract
BACKGROUND The role of local therapies including radical prostatectomy (RP) in prostate cancer (PCa) patients with clinical lymphadenopathies on prostate-specific membrane antigen (PSMA) positron emission tomography/computerized tomography (PET/CT) has scarcely been explored. Limited data are available to identify men who would benefit from RP; on the contrary, those more likely to benefit already have systemic disease. OBJECTIVE We aimed to assess the predictors of prostate-specific antigen (PSA) persistence in surgically managed PCa patients with lymphadenopathies on a PSMA PET/CT scan by integrating clinical, magnetic resonance imaging (MRI), and PSMA PET/CT parameters. DESIGN, SETTING, AND PARTICIPANTS We identified 519 patients treated with RP and extended lymph node dissection, and who received preoperative PSMA PET between 2017 and 2022 in nine referral centers. Among them, we selected 88 patients with nodal uptake at preoperative PSMA PET (miTxN1M0). OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The outcome was PSA persistence, defined as a PSA value of ≥0.1 ng/ml at the first measurement after surgery. Multivariable logistic regression models tested the predictors of PSA persistence. Covariates consisted of biopsy International Society of Urological Pathology (ISUP) grade group, clinical stage at MRI, and number of positive spots at a PET/CT scan. A regression tree analysis stratified patients into risk groups based on preoperative characteristics. RESULTS AND LIMITATIONS Overall, lymph node invasion (LNI) was detected in 63 patients (72%) and 32 (36%) experienced PSA persistence after RP. At multivariable analyses, having more than two lymph nodal positive findings at PSMA PET, seminal vesicle invasion (SVI) at MRI, and ISUP grade group >3 at biopsy were independent predictors of PSA persistence (all p < 0.05). At the regression tree analysis, patients were stratified in four risk groups according to biopsy ISUP grade, number of positive findings at PET/CT, and clinical stage at MRI. The model depicted good discrimination at internal validation (area under the curve 78%). CONCLUSIONS One out of three miN1M0 patients showed PSA persistence after surgery. Patients with ISUP grade 2-3, as well as patients with organ-confined disease at MRI and a single or two positive nodal findings at PET are those in whom RP may achieve the best oncological outcomes in the context of a multimodal approach. Conversely, patients with a high ISUP grade and extracapsular extension or SVI or more than two spots at PSMA PET should be considered as potentially affected by systemic disease upfront. PATIENT SUMMARY Our novel and straightforward risk classification integrates currently available preoperative risk tools and should, therefore, assist physician in preoperative counseling of men candidates for radical treatment for prostate cancer with positive lymph node uptake at prostate-specific membrane antigen positron emission tomography.
Collapse
Affiliation(s)
- Elio Mazzone
- Unit of Urology/Division of Oncology, Gianfranco Soldera Prostate Cancer Laboratory, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy.
| | - Giorgio Gandaglia
- Unit of Urology/Division of Oncology, Gianfranco Soldera Prostate Cancer Laboratory, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Daniele Robesti
- Unit of Urology/Division of Oncology, Gianfranco Soldera Prostate Cancer Laboratory, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Pawel Rajwa
- Department of Urology, Medical University of Vienna, Vienna, Austria; Department of Urology, Medical University of Silesia, Zabrze, Poland
| | - Juan Gomez Rivas
- Department of Urology, Hospital Clinico San Carlos, Madrid, Spain
| | - Laura Ibáñez
- Department of Urology, Hospital Clinico San Carlos, Madrid, Spain
| | - Timo F W Soeterik
- Department of Urology, St Antonius Hospital, Utrecht, The Netherlands; Department of Radiation Oncology, University Medical Center Utrecht, The Netherlands
| | - Lorenzo Bianchi
- Division of Urology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Luca Afferi
- Department of Urology, Luzerner Kantonsspital, Luzern, Switzerland
| | - Claudia Kesch
- Department of Urology, West German Cancer Center, University of Duisburg, Essen, Germany; German Cancer Consortium, University Hospital Essen, Essen, Germany
| | - Christopher Darr
- Department of Urology, West German Cancer Center, University of Duisburg, Essen, Germany; German Cancer Consortium, University Hospital Essen, Essen, Germany
| | - Hongqian Guo
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Jiangsu, China
| | - Junlong Zhuang
- Department of Urology, Drum Tower Hospital, Medical School of Nanjing University, Institute of Urology, Nanjing University, Jiangsu, China
| | - Fabio Zattoni
- Department Surgery, Oncology and Gastroenterology, Urologic Unit, University of Padova, Padua, Italy
| | - Wolfgang P Fendler
- German Cancer Consortium, University Hospital Essen, Essen, Germany; Department of Nuclear Medicine, University of Duisburg, Essen, Germany
| | - Daniele Amparore
- Department of Oncology, Division of Urology, San Luigi Gonzaga Hospital, Turin, Italy
| | - Nicolai A Huebner
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Alexander Giesen
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - Steven Joniau
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - Riccardo Schiavina
- Division of Urology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Eugenio Brunocilla
- Division of Urology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Agostino Mattei
- Department of Urology, Luzerner Kantonsspital, Luzern, Switzerland
| | - Fabrizio Dal Moro
- Department Surgery, Oncology and Gastroenterology, Urologic Unit, University of Padova, Padua, Italy
| | | | - Francesco Porpiglia
- Department of Oncology, Division of Urology, San Luigi Gonzaga Hospital, Turin, Italy
| | - Maria Picchio
- Vita-Salute San Raffaele University, Milan, Italy; Department of Nuclear Medicine, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Arturo Chiti
- Vita-Salute San Raffaele University, Milan, Italy; Department of Nuclear Medicine, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Shahrokh F Shariat
- Department of Urology, Medical University of Vienna, Vienna, Austria; Department of Urology, Weill Cornell Medical College, New York, NY, USA; Department of Urology, Second Faculty of Medicine, Charles University, Prague, Czech Republic; Department of Urology, University of Texas Southwestern, Dallas, TX, USA; Division of Urology, Department of Special Surgery, The University of Jordan, Amman, Jordan
| | - Francesco Montorsi
- Unit of Urology/Division of Oncology, Gianfranco Soldera Prostate Cancer Laboratory, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Alberto Briganti
- Unit of Urology/Division of Oncology, Gianfranco Soldera Prostate Cancer Laboratory, IRCCS San Raffaele Scientific Institute, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
23
|
Zhong Q, Sun R, Aref AT, Noor Z, Anees A, Zhu Y, Lucas N, Poulos RC, Lyu M, Zhu T, Chen GB, Wang Y, Ding X, Rutishauser D, Rupp NJ, Rueschoff JH, Poyet C, Hermanns T, Fankhauser C, Rodríguez Martínez M, Shao W, Buljan M, Neumann JF, Beyer A, Hains PG, Reddel RR, Robinson PJ, Aebersold R, Guo T, Wild PJ. Proteomic-based stratification of intermediate-risk prostate cancer patients. Life Sci Alliance 2024; 7:e202302146. [PMID: 38052461 PMCID: PMC10698198 DOI: 10.26508/lsa.202302146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 12/07/2023] Open
Abstract
Gleason grading is an important prognostic indicator for prostate adenocarcinoma and is crucial for patient treatment decisions. However, intermediate-risk patients diagnosed in the Gleason grade group (GG) 2 and GG3 can harbour either aggressive or non-aggressive disease, resulting in under- or overtreatment of a significant number of patients. Here, we performed proteomic, differential expression, machine learning, and survival analyses for 1,348 matched tumour and benign sample runs from 278 patients. Three proteins (F5, TMEM126B, and EARS2) were identified as candidate biomarkers in patients with biochemical recurrence. Multivariate Cox regression yielded 18 proteins, from which a risk score was constructed to dichotomize prostate cancer patients into low- and high-risk groups. This 18-protein signature is prognostic for the risk of biochemical recurrence and completely independent of the intermediate GG. Our results suggest that markers generated by computational proteomic profiling have the potential for clinical applications including integration into prostate cancer management.
Collapse
Affiliation(s)
- Qing Zhong
- ProCan, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Rui Sun
- iMarker Lab, Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| | - Adel T Aref
- ProCan, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Zainab Noor
- ProCan, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Asim Anees
- ProCan, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Yi Zhu
- iMarker Lab, Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| | - Natasha Lucas
- ProCan, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Rebecca C Poulos
- ProCan, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Mengge Lyu
- iMarker Lab, Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| | - Tiansheng Zhu
- iMarker Lab, Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| | - Guo-Bo Chen
- Urology & Nephrology Center, Department of Urology, Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Yingrui Wang
- iMarker Lab, Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| | - Xuan Ding
- iMarker Lab, Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| | - Dorothea Rutishauser
- Department of Pathology and Molecular Pathology, University Hospital Zürich, Zürich, Switzerland
| | - Niels J Rupp
- Department of Pathology and Molecular Pathology, University Hospital Zürich, Zürich, Switzerland
| | - Jan H Rueschoff
- Department of Pathology and Molecular Pathology, University Hospital Zürich, Zürich, Switzerland
| | - Cédric Poyet
- Department of Urology, University Hospital Zürich, Zürich, Switzerland
| | - Thomas Hermanns
- Department of Urology, University Hospital Zürich, Zürich, Switzerland
| | - Christian Fankhauser
- Department of Urology, University Hospital Zürich, Zürich, Switzerland
- Department of Urology, Cantonal Hospital Lucerne, Lucerne, Switzerland
| | | | - Wenguang Shao
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic and Developmental Sciences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Marija Buljan
- Empa - Swiss Federal Laboratories for Materials Science and Technology, St. Gallen, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | | | | | - Peter G Hains
- ProCan, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Roger R Reddel
- ProCan, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Phillip J Robinson
- ProCan, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Ruedi Aebersold
- Department of Biology, Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
- Faculty of Science, University of Zürich, Zürich, Switzerland
| | - Tiannan Guo
- iMarker Lab, Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, China
| | - Peter J Wild
- Goethe University Frankfurt, Dr. Senckenberg Institute of Pathology, University Hospital Frankfurt, Frankfurt am Main, Germany
- Frankfurt Institute for Advanced Studies, Frankfurt am Main, Germany
| |
Collapse
|
24
|
Boschheidgen M, Schimmöller L, Kastl R, Drewes LR, Jannusch K, Radke KL, Kirchner J, Ullrich T, Niegisch G, Albers P, Antoch G, Radtke JP. MRI characteristics and oncological follow-up of patients with ISUP grade group 4 or 5 prostate cancer. Abdom Radiol (NY) 2024; 49:192-201. [PMID: 37906272 PMCID: PMC10789849 DOI: 10.1007/s00261-023-04073-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 11/02/2023]
Abstract
OBJECTIVES To analyze multiparametric MRI (mpMRI) characteristics of patients with International Society of Urological Pathology (ISUP) grade group (GG) 4 or 5 prostate cancer (PC) and to correlate MRI parameters with the occurrence of biochemical recurrence (BCR) after radical prostatectomy (RPE). METHODS In this single-center cohort study consecutive patients with mpMRI and ISUP GG 4 or 5 PC were retrospectively analyzed. Clinical, MR-guided biopsy, and diagnostic mpMRI parameter were assessed. A subcohort of patients with RPE and follow-up was analyzed separately. A univariate and multivariate analyses were performed to determine parameters that are associated to patients with BCR after RPE. RESULTS 145 patients (mean age 70y, median PSA 10.9 ng/ml) were analyzed. 99% had a PI-RADS classification of 4 or 5, 48% revealed MRI T3 stage, and median diameter of the MRI index lesion (IL) was 15 mm. IL showed a median ADC value of 668 ×10-6 mm2/s and exhibited contrast enhancement in 94% of the cases. For patients with follow-up after RPE (n = 82; mean follow-up time 68 ± 27 m), MRI parameters were significantly different for contact length of the IL to the pseudocapsule (LCC), MRI T3 stage, and IL localization (p < 0.05). Higher PSAD and MRI T3 stage were independent parameters for the risk of BCR when incorporating clinical, biopsy, and MRI parameters. CONCLUSION ISUP GG 4 or 5 PC has distinctive characteristics on mpMRI and were detected on MRI in all cases. In addition, higher PSAD and MRI T3 stage were significant predictors for BCR after RPE.
Collapse
Affiliation(s)
- M Boschheidgen
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University Dusseldorf, Moorenstr. 5, 40225, Dusseldorf, Germany
| | - L Schimmöller
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University Dusseldorf, Moorenstr. 5, 40225, Dusseldorf, Germany.
- Department of Diagnostic, Interventional Radiology and Nuclear Medicine, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany.
| | - R Kastl
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University Dusseldorf, Moorenstr. 5, 40225, Dusseldorf, Germany
| | - L R Drewes
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University Dusseldorf, Moorenstr. 5, 40225, Dusseldorf, Germany
| | - K Jannusch
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University Dusseldorf, Moorenstr. 5, 40225, Dusseldorf, Germany
| | - K L Radke
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University Dusseldorf, Moorenstr. 5, 40225, Dusseldorf, Germany
| | - J Kirchner
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University Dusseldorf, Moorenstr. 5, 40225, Dusseldorf, Germany
| | - T Ullrich
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University Dusseldorf, Moorenstr. 5, 40225, Dusseldorf, Germany
| | - G Niegisch
- Department of Urology, Medical Faculty, University Dusseldorf, Moorenstr. 5, 40225, Dusseldorf, Germany
| | - P Albers
- Department of Urology, Medical Faculty, University Dusseldorf, Moorenstr. 5, 40225, Dusseldorf, Germany
| | - G Antoch
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University Dusseldorf, Moorenstr. 5, 40225, Dusseldorf, Germany
| | - J P Radtke
- Department of Urology, Medical Faculty, University Dusseldorf, Moorenstr. 5, 40225, Dusseldorf, Germany
| |
Collapse
|
25
|
Sushentsev N, Hamm G, Richings J, McLean MA, Menih IH, Ayyappan V, Mills IG, Warren AY, Gnanapragasam VJ, Barry ST, Goodwin RJA, Gallagher FA, Barrett T. Imaging tumor lactate is feasible for identifying intermediate-risk prostate cancer patients with postsurgical biochemical recurrence. Proc Natl Acad Sci U S A 2023; 120:e2312261120. [PMID: 38011568 PMCID: PMC10710070 DOI: 10.1073/pnas.2312261120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/27/2023] [Indexed: 11/29/2023] Open
Abstract
While radical prostatectomy remains the mainstay of prostate cancer (PCa) treatment, 20 to 40% of patients develop postsurgical biochemical recurrence (BCR). A particularly challenging clinical cohort includes patients with intermediate-risk disease whose risk stratification would benefit from advanced approaches that complement standard-of-care diagnostic tools. Here, we show that imaging tumor lactate using hyperpolarized 13C MRI and spatial metabolomics identifies BCR-positive patients in two prospective intermediate-risk surgical cohorts. Supported by spatially resolved tissue analysis of established glycolytic biomarkers, this study provides the rationale for multicenter trials of tumor metabolic imaging as an auxiliary tool to support PCa treatment decision-making.
Collapse
Affiliation(s)
- Nikita Sushentsev
- Department of Radiology, Addenbrooke’s Hospital and University of Cambridge, Cambridge Biomedical Campus, CB2 0QQCambridge, United Kingdom
| | - Gregory Hamm
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, Research & Development, AstraZeneca, CambridgeCB2 0AA, United Kingdom
| | - Jack Richings
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, Research & Development, AstraZeneca, CambridgeCB2 0AA, United Kingdom
| | - Mary A. McLean
- Department of Radiology, Addenbrooke’s Hospital and University of Cambridge, Cambridge Biomedical Campus, CB2 0QQCambridge, United Kingdom
| | - Ines Horvat Menih
- Department of Radiology, Addenbrooke’s Hospital and University of Cambridge, Cambridge Biomedical Campus, CB2 0QQCambridge, United Kingdom
| | - Vinay Ayyappan
- Department of Radiology, Addenbrooke’s Hospital and University of Cambridge, Cambridge Biomedical Campus, CB2 0QQCambridge, United Kingdom
| | - Ian G. Mills
- Patrick G. Johnston Centre for Cancer Research, Genito-Urinary and Prostate Focus Group, Queen’s University Belfast, BelfastBT9 7AE, United Kingdom
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, OxfordOX3 7DQ, United Kingdom
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen5021, Norway
| | - Anne Y. Warren
- Department of Pathology, Cambridge University Hospitals National Health Service Foundation Trust, CB2 0QQCambridge, United Kingdom
| | - Vincent J. Gnanapragasam
- Department of Urology, Cambridge University Hospitals National Health Service Foundation Trust, CambridgeCB2 0QQ, United Kingdom
- Cambridge Urology Translational Research and Clinical Trials Office, Cambridge Biomedical Campus, Addenbrooke’s Hospital, CambridgeCB2 0QQ, United Kingdom
| | - Simon T. Barry
- Bioscience, Discovery, Oncology Research & Development, AstraZeneca, CambridgeCB20AA, United Kingdom
| | - Richard J. A. Goodwin
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, Research & Development, AstraZeneca, CambridgeCB2 0AA, United Kingdom
| | - Ferdia A. Gallagher
- Department of Radiology, Addenbrooke’s Hospital and University of Cambridge, Cambridge Biomedical Campus, CB2 0QQCambridge, United Kingdom
| | - Tristan Barrett
- Department of Radiology, Addenbrooke’s Hospital and University of Cambridge, Cambridge Biomedical Campus, CB2 0QQCambridge, United Kingdom
| |
Collapse
|
26
|
Ah-Thiane L, Sargos P, Chapet O, Jolicoeur M, Terlizzi M, Salembier C, Boustani J, Prevost C, Gaudioz S, Derashodian T, Palumbo S, De Hertogh O, Créhange G, Zilli T, Supiot S. Managing postoperative biochemical relapse in prostate cancer, from the perspective of the Francophone group of Urological radiotherapy (GFRU). Cancer Treat Rev 2023; 120:102626. [PMID: 37734178 DOI: 10.1016/j.ctrv.2023.102626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 09/23/2023]
Abstract
Up to 50% of patients treated with radical surgery for localized prostate cancer may experience biochemical recurrence that requires appropriate management. Definitions of biochemical relapse may vary, but, in all cases, consist of an increase in a PSA without clinical or radiological signs of disease. Molecular imaging through to positron emission tomography has taken a preponderant place in relapse diagnosis, progressively replacing bone scan and CT-scan. Prostate bed radiotherapy is currently a key treatment, the action of which should be potentiated by androgen deprivation therapy. Nowadays perspectives consist in determining the best combination therapies, particularly thanks to next-generation hormone therapies, but not exclusively. Several trials are ongoing and should address these issues. We present here a literature review aiming to discuss the current management of biochemical relapse in prostate cancer after radical surgery, in lights of recent findings, as well as future perspectives.
Collapse
Affiliation(s)
- Loic Ah-Thiane
- Department of Radiation Oncology, ICO René Gauducheau, St-Herblain, France
| | - Paul Sargos
- Department of Radiation Oncology, Bergonie Institute, Bordeaux, France
| | - Olivier Chapet
- Department of Radiation Oncology, CHU Lyon Sud, Pierre-Bénite, France
| | - Marjory Jolicoeur
- Department of Radiation Oncology, Charles Le Moyne Hospital, Montreal, Canada
| | - Mario Terlizzi
- Department of Radiation Oncology, Gustave Roussy Cancer Center, Villejuif, France
| | - Carl Salembier
- Department of Radiation Oncology, Europe Hospitals Brussels, Belgium
| | - Jihane Boustani
- Department of Radiation Oncology, CHU Besançon, Besançon, France
| | - Célia Prevost
- Department of Radiation Oncology, CHU Lyon Sud, Pierre-Bénite, France
| | - Sonya Gaudioz
- Department of Radiation Oncology, CHU Lyon Sud, Pierre-Bénite, France
| | - Talar Derashodian
- Department of Radiation Oncology, Sindi Ahluwalia Hawkins Centre, Kelowna, Canada
| | - Samuel Palumbo
- Department of Radiation Oncology, CHU UCL Namur-Sainte Elisabeth, Namur, Belgium
| | - Olivier De Hertogh
- Department of Radiation Oncology, CHR Verviers East Belgium, Verviers, Belgium
| | - Gilles Créhange
- Department of Radiation Oncology, Curie Institute, Saint-Cloud, France
| | - Thomas Zilli
- Department of Radiation Oncology, Geneva University Hospital, Geneva, Switzerland
| | - Stéphane Supiot
- Department of Radiation Oncology, ICO René Gauducheau, St-Herblain, France.
| |
Collapse
|
27
|
Luzzago S, Colombo A, Mistretta FA, Alessi S, Di Trapani E, Summers PE, Piccinelli ML, Raimondi S, Vignati S, Clemente A, Rosati E, di Meglio L, d'Ascoli E, Scarabelli A, Zugni F, Belmonte M, Maggioni R, Ferro M, Fusco N, de Cobelli O, Musi G, Petralia G. Multiparametric MRI-based 5-year Risk Prediction Model for Biochemical Recurrence of Prostate Cancer after Radical Prostatectomy. Radiology 2023; 309:e223349. [PMID: 37987657 DOI: 10.1148/radiol.223349] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Background Current predictive tools to estimate the risk of biochemical recurrence (BCR) after treatment of prostate cancer do not consider multiparametric MRI (mpMRI) information. Purpose To develop a risk prediction tool that considers mpMRI findings to assess the risk of 5-year BCR after radical prostatectomy. Materials and Methods In this retrospective single-center analysis in 1459 patients with prostate cancer who underwent mpMRI before radical prostatectomy (in 2012-2015), the outcome of interest was 5-year BCR (two consecutive prostate-specific antigen [PSA] levels > 0.2 ng/mL [0.2 µg/L]). Patients were randomly divided into training (70%) and test (30%) sets. Kaplan-Meier plots were applied to the training set to estimate survival probabilities. Multivariable Cox regression models were used to test the relationship between BCR and different sets of exploratory variables. The C-index of the final model was calculated for the training and test sets and was compared with European Association of Urology, University of California San Francisco Cancer of the Prostate Risk Assessment, Memorial Sloan-Kettering Cancer Center, and Partin risk tools using the partial likelihood ratio test. Five risk categories were created. Results The median duration of follow-up in the whole cohort was 59 months (IQR, 32-81 months); 376 of 1459 (25.8%) patients had BCR. A multivariable Cox regression model (referred to as PIPEN, and composed of PSA density, International Society of Urological Pathology grade group, Prostate Imaging Reporting and Data System category, European Society of Urogenital Radiology extraprostatic extension score, nodes) fitted to the training data yielded a C-index of 0.74, superior to that of other predictive tools (C-index 0.70 for all models; P ≤ .01) and a median higher C-index on 500 test set replications (C-index, 0.73). Five PIPEN risk categories were identified with 5-year BCR-free survival rates of 92%, 84%, 71%, 56%, and 26% in very low-, low-, intermediate-, high-, and very high-risk patients, respectively (all P < .001). Conclusion A five-item model for predicting the risk of 5-year BCR after radical prostatectomy for prostate cancer was developed and internally verified, and five risk categories were identified. © RSNA, 2023 Supplemental material is available for this article. See also the editorial by Aguirre and Ortegón in this issue.
Collapse
Affiliation(s)
- Stefano Luzzago
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Alberto Colombo
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Francesco A Mistretta
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Sarah Alessi
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Ettore Di Trapani
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Paul E Summers
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Mattia Luca Piccinelli
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Sara Raimondi
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Silvano Vignati
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Alfredo Clemente
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Elisa Rosati
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Letizia di Meglio
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Elisa d'Ascoli
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Alice Scarabelli
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Fabio Zugni
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Maddalena Belmonte
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Roberta Maggioni
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Matteo Ferro
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Nicola Fusco
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Ottavio de Cobelli
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Gennaro Musi
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Giuseppe Petralia
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| |
Collapse
|
28
|
de Bie KCC, Veerman H, Bodar YJL, Meijer D, van Leeuwen PJ, van der Poel HG, Donswijk ML, Vis AN, Oprea-Lager DE. Higher Preoperative Maximum Standardised Uptake Values (SUV max) Are Associated with Higher Biochemical Recurrence Rates after Robot-Assisted Radical Prostatectomy for [ 68Ga]Ga-PSMA-11 and [ 18F]DCFPyL Positron Emission Tomography/Computed Tomography. Diagnostics (Basel) 2023; 13:2343. [PMID: 37510087 PMCID: PMC10378114 DOI: 10.3390/diagnostics13142343] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/01/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
This study aimed to investigate the association between the 68Ga- or 18F-radiolabeled prostate-specific membrane antigen (PSMA) tracer expression, represented by the maximum standardised uptake value (SUVmax) of the dominant intraprostatic lesion, and biochemical recurrence (BCR) in primary prostate cancer (PCa) patients prior to robot-assisted radical prostatectomy (RARP). This was a retrospective, multi-centre cohort study of 446 patients who underwent [68Ga]Ga-PSMA-11 (n = 238) or [18F]DCFPyL (n = 206) Positron Emission Tomography/Computed Tomography (PET/CT) imaging prior to RARP. SUVmax was measured in the dominant intraprostatic PCa lesions. [18F]DCFPyL patients were scanned 60 ([18F]DCFPyL-60; n = 106) or 120 ([18F]DCFPyL-120; n = 120) minutes post-injection of a radiotracer and were analysed separately. To normalise the data, SUVmax was log transformed for further analyses. During a median follow-up of 24 months, 141 (30.4%) patients experienced BCR. Log2SUVmax was a significant predictor for BCR (p < 0.001). In the multivariable analysis accounting for these preoperative variables: initial prostate-specific antigen (PSA), radiologic tumour stage (mT), the biopsy International Society of Urological Pathology grade group (bISUP) and the prostate imaging and reporting data system (PI-RADS), Log2SUVmax was found to be an independent predictor for BCR in [68Ga]Ga-PSMA-11 (HR 1.32, p = 0.04) and [18F]DCFPyL-120 PET/CT scans (HR 1.55, p = 0.04), but not in [18F]DCFPyL-60 ones (HR 0.92, p = 0.72). The PSMA expression of the dominant intraprostatic lesion proved to be an independent predictor for BCR in patients with primary PCa who underwent [68Ga]Ga-PSMA-11 or [18F]DCFPyL-120 PET/CT scans, but not in those who underwent [18F]DCFPyL-60 PET/CT scans.
Collapse
Affiliation(s)
- Katelijne C. C. de Bie
- Department of Urology, VU University, Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (H.V.); (Y.J.L.B.); (D.M.); (A.N.V.)
- Department of Radiology & Nuclear Medicine, VU University, Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands;
- Prostate Cancer Network The Netherlands, 1066 CX Amsterdam, The Netherlands;
| | - Hans Veerman
- Department of Urology, VU University, Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (H.V.); (Y.J.L.B.); (D.M.); (A.N.V.)
- Prostate Cancer Network The Netherlands, 1066 CX Amsterdam, The Netherlands;
- Department of Urology, Antoni van Leeuwenhoek Hospital—The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Yves J. L. Bodar
- Department of Urology, VU University, Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (H.V.); (Y.J.L.B.); (D.M.); (A.N.V.)
- Department of Radiology & Nuclear Medicine, VU University, Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands;
- Prostate Cancer Network The Netherlands, 1066 CX Amsterdam, The Netherlands;
| | - Dennie Meijer
- Department of Urology, VU University, Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (H.V.); (Y.J.L.B.); (D.M.); (A.N.V.)
- Department of Radiology & Nuclear Medicine, VU University, Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands;
- Prostate Cancer Network The Netherlands, 1066 CX Amsterdam, The Netherlands;
| | - Pim J. van Leeuwen
- Prostate Cancer Network The Netherlands, 1066 CX Amsterdam, The Netherlands;
- Department of Urology, Antoni van Leeuwenhoek Hospital—The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Henk G. van der Poel
- Prostate Cancer Network The Netherlands, 1066 CX Amsterdam, The Netherlands;
- Department of Urology, Antoni van Leeuwenhoek Hospital—The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Maarten L. Donswijk
- Department of Radiology and Nuclear Medicine, Antoni van Leeuwenhoek Hospital—The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands;
| | - André N. Vis
- Department of Urology, VU University, Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (H.V.); (Y.J.L.B.); (D.M.); (A.N.V.)
- Prostate Cancer Network The Netherlands, 1066 CX Amsterdam, The Netherlands;
| | - Daniela E. Oprea-Lager
- Department of Radiology & Nuclear Medicine, VU University, Amsterdam University Medical Centers, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands;
| |
Collapse
|
29
|
Peyrottes A, Achard V, Dariane C. How To Manage T3b Prostate Cancer in the Contemporary Era: The Benefits of Surgery. EUR UROL SUPPL 2023; 53:55-57. [PMID: 37287634 PMCID: PMC10241842 DOI: 10.1016/j.euros.2023.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2023] [Indexed: 06/09/2023] Open
Affiliation(s)
- Arthur Peyrottes
- Department of Urology, Hôpital européen Georges-Pompidou, AP-HP, Paris, France
- Prostate Group, Comité de Cancérologie de l’Association Française d’Urologie, Junior Member, Paris, France
| | - Verane Achard
- Department of Radiation Oncology, HFR Fribourg, Villars-sur-Glâne, Switzerland
| | - Charles Dariane
- Department of Urology, Hôpital européen Georges-Pompidou, AP-HP, Paris, France
- Prostate Group, Comité de Cancérologie de l’Association Française d’Urologie, Paris, France
- U1151 Inserm-INEM, Paris University, Paris, France
| |
Collapse
|
30
|
Diamand R, Peltier A, Roche JB, Lievore E, Lacetera V, Chiacchio G, Beatrici V, Mastroianni R, Simone G, Windisch O, Benamran D, Fourcade A, Nguyen TA, Fournier G, Fiard G, Ploussard G, Roumeguère T, Albisinni S. Risk stratification for early biochemical recurrence of prostate cancer in the era of multiparametric magnetic resonance imagining-targeted biopsy. Prostate 2023; 83:572-579. [PMID: 36705314 DOI: 10.1002/pros.24490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 11/23/2022] [Accepted: 01/06/2023] [Indexed: 01/28/2023]
Abstract
BACKGROUND Multiparametric magnetic resonance imaging (MRI) and MRI-targeted biopsy are nowadays recommended in the prostate cancer (PCa) diagnostic pathway. Ploussard and Mazzone have integrated these tools into novel risk classification systems predicting the risk of early biochemical recurrence (eBCR) in PCa patients who underwent radical prostatectomy (RP). We aimed to assess available risk classification systems and to define the best-performing. METHODS Data on 1371 patients diagnosed by MRI-targeted biopsy and treated by RP between 2014 and 2022 at eight European tertiary referral centers were analyzed. Risk classifications systems included were the European Association of Urology (EAU) and National Comprehensive Cancer Network (NCCN) risk groups, the Cancer of the Prostate Risk Assessment (CAPRA) score, the International Staging Collaboration for Cancer of the Prostate (STAR-CAP) classification, the Ploussard and Mazzone models, and ISUP grade group. Kaplan-Meier analyses were used to compare eBCR among risk classification systems. Performance was assessed in terms of discrimination quantified using Harrell's c-index, calibration, and decision curve analysis (DCA). RESULTS Overall, 152 (11%) patients had eBCR at a median follow-up of 31 months (interquartile range: 19-45). The 3-year eBCR-free survival rate was 91% (95% confidence interval [CI]: 89-93). For each risk classification system, a significant difference among survival probabilities was observed (log-rank test p < 0.05) except for NCCN classification (p = 0.06). The highest discrimination was obtained with the STAR-CAP classification (c-index 66%) compared to CAPRA score (63% vs. 66%, p = 0.2), ISUP grade group (62% vs. 66, p = 0.07), Ploussard (61% vs. 66%, p = 0.003) and Mazzone models (59% vs. 66%, p = 0.02), and EAU (57% vs. 66%, p < 0.001) and NCCN (57% vs. 66%, p < 0.001) risk groups. Risk classification systems demonstrated good calibration characteristics. At DCA, the CAPRA score showed the highest net benefit at a probability threshold of 9%-15%. CONCLUSIONS The performance of risk classification systems using MRI and MRI-targeted information was less optimistic when tested in a contemporary set of patients. CAPRA score and STAR-CAP classification were the best-performing and should be preferred for treatment decision-making.
Collapse
Affiliation(s)
- Romain Diamand
- Department of Urology, Jules Bordet Institute-Erasme Hospital, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Alexandre Peltier
- Department of Urology, Jules Bordet Institute-Erasme Hospital, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | | | - Elena Lievore
- Department of Urology, Clinique Saint-Augustin, Bordeaux, France
- Department of Urology, IRCCS IEO Istituto Europeo di Oncologia, Milan, Italy
| | - Vito Lacetera
- Department of Urology, Azienda Ospedaliera Ospedali Riuniti Marche Nord, Pesaro, Italy
| | - Giuseppe Chiacchio
- Department of Urology, Azienda Ospedaliera Ospedali Riuniti Marche Nord, Pesaro, Italy
| | - Valerio Beatrici
- Department of Urology, Azienda Ospedaliera Ospedali Riuniti Marche Nord, Pesaro, Italy
| | - Riccardo Mastroianni
- Department of Urology, IRCCS "Regina Elena" National Cancer Institute, Rome, Italy
| | - Giuseppe Simone
- Department of Urology, IRCCS "Regina Elena" National Cancer Institute, Rome, Italy
| | - Olivier Windisch
- Department of Urology, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| | - Daniel Benamran
- Department of Urology, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| | - Alexandre Fourcade
- Department of Urology, Hôpital Cavale Blanche, CHRU Brest, Brest, France
| | - Truong A Nguyen
- Department of Urology, Hôpital Cavale Blanche, CHRU Brest, Brest, France
| | - Georges Fournier
- Department of Urology, Hôpital Cavale Blanche, CHRU Brest, Brest, France
| | - Gaelle Fiard
- Department of Urology, Grenoble Alpes University Hospital, Université Grenoble Alpes, CNRS, Grenoble INP, TIMC, Grenoble, France
| | | | - Thierry Roumeguère
- Department of Urology, Jules Bordet Institute-Erasme Hospital, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Simone Albisinni
- Department of Urology, Jules Bordet Institute-Erasme Hospital, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
31
|
Fiard G, Seigneurin A, Roumiguié M, Albisinni S, Anract J, Assenmacher G, Barry Delongchamps N, Dariane C, Feyaerts A, Fourcade A, Fournier G, Gontero P, Mastroianni R, Oderda M, Peltier A, Roumeguère T, Saussez T, Simone G, Van Damme J, Descotes JL, Ploussard G, Diamand R. Prognostic significance of PI-RADS 5 lesions in patients treated by radical prostatectomy. World J Urol 2023; 41:1285-1291. [PMID: 36971827 DOI: 10.1007/s00345-023-04371-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/04/2023] [Indexed: 03/29/2023] Open
Abstract
PURPOSE To analyse the pathological features and survival of patients with a PI-RADS 5 lesion on pre-biopsy MRI. METHODS We extracted from a European multicentre prospectively gathered database the data of patients with a PI-RADS 5 lesion on pre-biopsy MRI, diagnosed using both systematic and targeted biopsies and subsequently treated by radical prostatectomy. The Kaplan-Meier model was used to assess the biochemical-free survival of the whole cohort and univariable and multivariable Cox models were set up to study factors associated with survival. RESULTS Between 2013 and 2019, 539 consecutive patients with a PI-RADS 5 lesion on pre-biopsy MRI were treated by radical prostatectomy and included in the analysis. Follow-up data were available for 448 patients. Radical prostatectomy and lymph node dissection specimens showed non-organ confined disease in 297/539 (55%), (including 2 patients with a locally staged pT2 lesion and lymph node involvement (LNI)). With a median follow-up of 25 months (12-39), the median biochemical recurrence-free survival was 54% at 2 years (95% CI 45-61) and 28% at 5 years (95% CI 18-39). Among the factors studied, MRI T stage [T3a vs T2 HR 3.57 (95%CI 1.78-7.16); T3b vs T2 HR 6.17 (95% CI 2.99-12.72)] and PSA density (HR 4.47 95% CI 1.55-12.89) were significantly associated with a higher risk of biochemical recurrence in multivariable analysis. CONCLUSION Patients with a PI-RADS 5 lesion on pre-biopsy MRI have a high risk of early biochemical recurrence after radical prostatectomy. MRI T stage and PSA density can be used to improve patient selection and counselling.
Collapse
|
32
|
Baboudjian M, Breda A, Roumeguère T, Uleri A, Roche JB, Touzani A, Lacetera V, Beauval JB, Diamand R, Simone G, Windisch O, Benamran D, Fourcade A, Fiard G, Durand-Labrunie C, Roumiguié M, Sanguedolce F, Oderda M, Barret E, Fromont G, Dariane C, Charvet AL, Gondran-Tellier B, Bastide C, Lechevallier E, Palou J, Ruffion A, Van Der Bergh RCN, Peltier A, Ploussard G. Expanding inclusion criteria for active surveillance in intermediate-risk prostate cancer: a machine learning approach. World J Urol 2023; 41:1301-1308. [PMID: 36920491 DOI: 10.1007/s00345-023-04353-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/26/2023] [Indexed: 03/16/2023] Open
Abstract
PURPOSE To develop new selection criteria for active surveillance (AS) in intermediate-risk (IR) prostate cancer (PCa) patients. METHODS Retrospective study including patients from 14 referral centers who underwent pre-biopsy mpMRI, image-guided biopsies and radical prostatectomy. The cohort included biopsy-naive IR PCa patients who met the following inclusion criteria: Gleason Grade Group (GGG) 1-2, PSA < 20 ng/mL, and cT1-cT2 tumors. We relied on a recursive machine learning partitioning algorithm developed to predict adverse pathological features (i.e., ≥ pT3a and/or pN + and/or GGG ≥ 3). RESULTS A total of 594 patients with IR PCa were included, of whom 220 (37%) had adverse features. PI-RADS score (weight:0.726), PSA density (weight:0.158), and clinical T stage (weight:0.116) were selected as the most informative risk factors to classify patients according to their risk of adverse features, leading to the creation of five risk clusters. The adverse feature rates for cluster #1 (PI-RADS ≤ 3 and PSA density < 0.15), cluster #2 (PI-RADS 4 and PSA density < 0.15), cluster #3 (PI-RADS 1-4 and PSA density ≥ 0.15), cluster #4 (normal DRE and PI-RADS 5), and cluster #5 (abnormal DRE and PI-RADS 5) were 11.8, 27.9, 37.3, 42.7, and 65.1%, respectively. Compared with the current inclusion criteria, extending the AS criteria to clusters #1 + #2 or #1 + #2 + #3 would increase the number of eligible patients (+ 60 and + 253%, respectively) without increasing the risk of adverse pathological features. CONCLUSIONS The newly developed model has the potential to expand the number of patients eligible for AS without compromising oncologic outcomes. Prospective validation is warranted.
Collapse
Affiliation(s)
- Michael Baboudjian
- Department of Urology, La Croix du Sud Hôpital, Quint Fonsegrives, France. .,Department of Urology, North Hospital, Aix-Marseille University, APHM, Marseille, France. .,Department of Urology, La Conception Hospital, Aix-Marseille University, APHM, Marseille, France. .,Department of Urology, Fundació Puigvert, Autonoma University of Barcelona, Barcelona, Spain.
| | - Alberto Breda
- Department of Urology, Fundació Puigvert, Autonoma University of Barcelona, Barcelona, Spain
| | - Thierry Roumeguère
- Department of Urology, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Alessandro Uleri
- Department of Urology, Fundació Puigvert, Autonoma University of Barcelona, Barcelona, Spain
| | | | - Alae Touzani
- Department of Urology, La Croix du Sud Hôpital, Quint Fonsegrives, France
| | - Vito Lacetera
- Azienda Ospedaliera Ospedali Riuniti Marche Nord, Pesaro, Italy
| | | | - Romain Diamand
- Department of Urology, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | - Guiseppe Simone
- Department of Urology, IRCCS "Regina Elena" National Cancer Institute, Rome, Italy
| | - Olivier Windisch
- Division of Urology, Geneva University Hospitals, Geneva, Switzerland
| | - Daniel Benamran
- Division of Urology, Geneva University Hospitals, Geneva, Switzerland
| | - Alexandre Fourcade
- Department of Urology, Hôpital Cavale Blanche, CHRU Brest, Brest, France
| | - Gaelle Fiard
- Department of Urology, Grenoble Alpes University Hospital, Université Grenoble Alpes, CNRS, Grenoble INP, TIMC, Grenoble, France
| | | | - Mathieu Roumiguié
- Department of Urology, Toulouse University Hospital, Toulouse, France
| | - Francesco Sanguedolce
- Department of Urology, Fundació Puigvert, Autonoma University of Barcelona, Barcelona, Spain
| | - Marco Oderda
- Division of Urology, Department of Surgical Sciences - Urology, Città Della Salute E Della Scienza Di Torino - Molinette Hospital, University of Turin, Turin, Italy
| | - Eric Barret
- Department of Urology, Institut Mutualiste Montsouris, Paris, France
| | | | - Charles Dariane
- Department of Urology, U1151 Inserm-INEM, Hôpital Européen Georges-Pompidou, APHP, Paris University, Necker, Paris, France
| | - Anne-Laure Charvet
- Department of Urology, La Conception Hospital, Aix-Marseille University, APHM, Marseille, France
| | - Bastien Gondran-Tellier
- Department of Urology, La Conception Hospital, Aix-Marseille University, APHM, Marseille, France
| | - Cyrille Bastide
- Department of Urology, North Hospital, Aix-Marseille University, APHM, Marseille, France
| | - Eric Lechevallier
- Department of Urology, La Conception Hospital, Aix-Marseille University, APHM, Marseille, France
| | - Joan Palou
- Department of Urology, Fundació Puigvert, Autonoma University of Barcelona, Barcelona, Spain
| | - Alain Ruffion
- Service d'urologie Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Lyon, France.,Equipe 2, Centre d'Innovation en Cancérologie de Lyon (EA 3738 CICLY), Faculté de Médecine Lyon Sud, Université Lyon 1, Lyon, France
| | | | - Alexandre Peltier
- Department of Urology, Jules Bordet Institute, Université Libre de Bruxelles, Brussels, Belgium
| | | |
Collapse
|
33
|
Urinary Continence Recovery after Robotic Radical Prostatectomy without Anterior or Posterior Reconstruction: Experience from a Tertiary Referral Center. J Clin Med 2023; 12:jcm12041358. [PMID: 36835893 PMCID: PMC9962972 DOI: 10.3390/jcm12041358] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 01/25/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND The aim of our study is to evaluate the prevalence and predictive factors of short- (30 d) and mid-term continence in a contemporary cohort of patients treated with robotic-assisted laparoscopic prostatectomy (RALP) without any posterior or anterior reconstruction at our referral academic center. METHODS Data from patients undergoing RALP between January 2017 and March 2021 were prospectively collected. RALP was performed by three highly experienced surgeons following the principles of the Montsouris technique, with a bladder-neck-sparing intent and maximal preservation of the membranous urethra (if oncologically safe) without any anterior/posterior reconstruction. (Self-assessed urinary incontinence (UI) was defined as the need of one or more pads per die (excluding the need for a safety pad/die. Univariable and multivariable logistic regression analysis was used to assess the independent predictors of early incontinence among routinely collected patient- and tumor-related variables). RESULTS A total of 925 patients were included; of these, 353 underwent RALP (38.2%) without nerve-sparing intent. The median patient age and BMI were 68 years (IQR 63-72) and 26 (IQR 24.0-28.0), respectively. Overall, 159 patients (17.2%) reported early (30 d) incontinence. In multivariable analysis adjusting for patient- and tumor-related features, a non-nerve-sparing procedure (OR: 1.57 [95% CI: 1.03-2.59], p = 0.035) was independently associated with the risk of urinary incontinence in the short-term period, while the absence of cardiovascular diseases before surgery (OR: 0.46 [95% CI: 0.320.67], p ≤ 0.01) was a protective factor for this outcome. At a median follow-up of 17 months (IQR 10-24), 94.5% of patients reported to be continent. CONCLUSIONS In experienced hands, most patients fully recover urinary continence after RALP at mid-term follow-up. On the contrary, the proportion of patients who reported early incontinence in our series was modest but not negligible. The implementation of surgical techniques advocating anterior and/or posterior fascial reconstruction might improve the early continence rate in candidates for RALP.
Collapse
|
34
|
Re: Oncologic Outcomes of Total Length Gleason Pattern 4 on Biopsy in Men with Grade Group 2 Prostate Cancer. Eur Urol 2023; 83:187-188. [PMID: 36446672 DOI: 10.1016/j.eururo.2022.09.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 09/22/2022] [Indexed: 11/27/2022]
|
35
|
The impact of local staging of prostate cancer determined on MRI or DRE at time of radical prostatectomy on progression-free survival: A Will Rogers phenomenon. Urol Oncol 2023; 41:106.e9-106.e16. [PMID: 36564258 DOI: 10.1016/j.urolonc.2022.10.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 09/01/2022] [Accepted: 10/20/2022] [Indexed: 12/24/2022]
Abstract
INTRODUCTION We aimed to test whether the current practice of using mpMRI stage might lead to a Will Rogers phenomenon with a stage migration compared to DRE in men undergoing radical prostatectomy. MATERIAL AND METHODS A total of 572 consecutive patients who underwent radical prostatectomy at a single institution (2007-2017) were included. Clinical stage using digital rectal examination was determined on table by the operating surgeon; mpMRI and pathological stage were recorded after tumor board review. Progression-free survival (PFS) was defined as no rising PSA, no adjuvant/salvage treatment, and no metastases or mortality. PFS was compared between groups and a model incorporating mpMRI into the EAU risk groups was created. RESULTS Median age was 63 years (IQR 58.5-67) and median PSA was 8.9 ng/ml (IQR 6.5-13.2). Using DRE stage, 20% were NCCN low risk, 43% were intermediate, and 37% high. Median follow-up was 48 months (IQR 22-73). Estimated PFS at 1, 3, and 5 years was 75%, 59%, and 54%, respectively. When comparing PFS between DRE and mpMRI stages, patients deemed T1 (P < 0.01) or T3 (P = 0.03) by mpMRI showed better outcomes than patients staged T1 or T3 by DRE. On univariable analysis lower risk for failure was seen for MRI T1 disease (HR 0.10 95%, CI 0.01-0.73, P = 0.02) or MRI T3 (HR 0.70, CI 0.51-0.97, P = 0.03). On multivariable analysis, only MRI T1 remained a significant predictor (HR 0.08, 95% CI 0.01-0.59, P = 0.01). The subsequent, modified EAU risk model using both DRE and mpMRI performed significantly better than the DRE model. CONCLUSION PFS based on mpMRI is not the same as DRE staging. Current risk groups which use DRE should be used with caution in whom local stage is based on mpMRI. Our modified EAU-risk categories can provide greater accuracy.
Collapse
|
36
|
Baboudjian M, Gondran-Tellier B, Touzani A, Martini A, Diamand R, Roche JB, Lacetera V, Beauval JB, Roumeguère T, Simone G, Benamran D, Fourcade A, Fiard G, van den Bergh RC, Peltier A, Ploussard G. Magnetic Resonance Imaging–based T-staging to Predict Biochemical Recurrence after Radical Prostatectomy: A Step Towards the iTNM Classification. Eur Urol Oncol 2022:S2588-9311(22)00169-9. [PMID: 36280445 DOI: 10.1016/j.euo.2022.09.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/13/2022] [Accepted: 09/30/2022] [Indexed: 11/04/2022]
Abstract
BACKGROUND Local staging of prostate cancer (PCa) still relies on digital rectal examination (DRE), which therefore remains the standard for risk stratification in guideline recommendations, clinical trials, and patient counseling. This issue is increasingly controversial as multiparametric magnetic resonance imaging (mpMRI) has become the most influential diagnostic tool for local staging of PCa over the past two decades. OBJECTIVE To compare various models of T category based on DRE or mpMRI to predict early biochemical recurrence (BCR) after radical prostatectomy (RP). DESIGN, SETTING, AND PARTICIPANTS A retrospective multicenter cohort study was conducted between 2014 and 2021. A total of 1436 patients were recruited across eight referral centers in France, Italy, Switzerland, and Belgium. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS BCR was defined as two prostate-specific antigen values of ≥0.2 ng/ml during follow-up. Harrell's concordance index (C index) was used to compare the discrimination of four models of T staging based on DRE (model 1: cT1 vs cT2 vs cT3) or mpMRI (model 2: organ-confined disease vs extracapsular extension [iECE] vs seminal vesicle invasion [iSVI]; model 3: Prostate Imaging-Reporting and Data System [PI-RADS] ≤3 vs PI-RADS 4 vs PI-RADS 5; and model 4: iT2a [PI-RADS ≤3] vs iT2b [PI-RADS 4] vs iT2c [PI-RADS 5 excluding ECE or SVI] vs iT3a [ECE] vs iT3b [SVI]) to predict BCR. RESULTS AND LIMITATIONS Overall, 74 (5%), 845 (59%), 482 (34%), and 35 (2%) patients had low-, intermediate-, high-, and very high-risk PCa, respectively, according to the Mazzone risk classification. After median follow-up of 16 mo, 113 patients experienced BCR. Although the new five-group mpMRI-based T classification system (model 4) had the highest prognostic discrimination (C index 0.694) for predicting early BCR on multivariable analysis, there was overlap between the 95% confidence intervals of the models. On sensitivity analysis, the new mpMRI-based T staging still had a higher C index than DRE for predicting BCR when excluding cN1 patients and comparing it with a five-group DRE-based T classification (cT1c vs cT2a vs cT2b vs cT2c vs cT3), but the overlap between the 95% confidence intervals of the models remained. The main limitation is the short follow-up. CONCLUSIONS We described an alternative mpMRI-based T staging for prediction of early BCR after RP for PCa. Our results need to be validated externally before they can be applied in clinical practice. PATIENT SUMMARY At present, digital rectal examination of the prostate is used to stage prostate cancer. We developed an alternative model for staging that uses information from magnetic resonance imaging (MRI) scans to predict cancer outcomes for men undergoing surgical removal of the prostate.
Collapse
|
37
|
Zhou Q, Yang C, Mou Z, Wu S, Dai X, Chen X, Ou Y, Zhang L, Sha J, Jiang H. Identification and validation of a poor clinical outcome subtype of primary prostate cancer with Midkine abundance. Cancer Sci 2022; 113:3698-3709. [PMID: 36018546 PMCID: PMC9633304 DOI: 10.1111/cas.15546] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 07/29/2022] [Accepted: 08/09/2022] [Indexed: 11/30/2022] Open
Abstract
Recent studies identified Midkine (MDK) as playing a key role in immune regulation. In this study, we aimed to discover the clinical significance and translational relevance in prostate cancer (PCa). We retrospectively analyzed 759 PCa patients who underwent radical prostatectomy from Huashan Hospital, Fudan University (training cohort, n = 369) and Chinese Prostate Cancer Consortium (validation cohort, n = 390). A total of 325 PCa patients from The Cancer Genome Atlas (TCGA) database (external cohort) were analyzed for exploration. Immune landscape and antitumor immunity were assessed through immunohistochemistry and flow cytometry. Patient‐derived explant culture system was applied for evaluating the targeting potential of MDK. We found that intratumoral MDK expression correlated with PCa progression, which indicated an unfavorable biochemical recurrence (BCR)‐free survival for postoperative PCa patients. Addition of MDK expression to the postoperative risk assessment tool CAPRA‐S could improve its prognostic value. Tumors with MDK abundance characterized the tumor‐infiltrating CD8+ T cells with less cytotoxicity production and increased immune checkpoint expression, which were accompanied by enriched immunosuppressive contexture. Moreover, MDK inhibition could reactivate CD8+ T cell antitumor immunity. MDK mRNA expression negatively correlated with androgen receptor activity signature and positively associated with radiotherapy‐related signature. In conclusion, intratumoral MDK expression could serve as an independent prognosticator for BCR in postoperative PCa patients. MDK expression impaired the antitumor function of CD8+ T cells through orchestrating an immunoevasive microenvironment, which could be reversed by MDK inhibition. Moreover, tumors with MDK enrichment possessed potential sensitivity to postoperative radiotherapy while resistance to adjuvant hormonal therapy of PCa. MDK could be considered as a potential therapeutic target for PCa.
Collapse
Affiliation(s)
- Quan Zhou
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Chen Yang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China.,National Clinical Research Center for Aging and Medicine, Fudan University, Shanghai, China
| | - Zezhong Mou
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Siqi Wu
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiyu Dai
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xinan Chen
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yuxi Ou
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Limin Zhang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jianjun Sha
- Department of Urology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Haowen Jiang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China.,National Clinical Research Center for Aging and Medicine, Fudan University, Shanghai, China
| |
Collapse
|
38
|
Bommelaere T, Villers A, Puech P, Ploussard G, Labreuche J, Drumez E, Leroy X, Olivier J. Risk Estimation of Metastatic Recurrence After Prostatectomy: A Model Using Preoperative Magnetic Resonance Imaging and Targeted Biopsy. EUR UROL SUPPL 2022; 41:24-34. [PMID: 35813259 PMCID: PMC9257652 DOI: 10.1016/j.euros.2022.04.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2022] [Indexed: 12/03/2022] Open
Abstract
Background The risk of prostate cancer metastatic is correlated with its volume and grade. These parameters are now best estimated preoperatively with magnetic resonance imaging (MRI) and MRI-guided biopsy. Objective To estimate the risk of metastatic recurrence after radical prostatectomy (RP) in our model versus conventional clinical European Association of Urology (EAU) classification. The secondary objective is biochemical recurrence (BCR). Design, setting, and participants A retrospective study was conducted of a cohort of 713 patients having undergone MRI-guided biopsies and RP between 2009 and 2018. The preoperative variables included prostate-specific antigen, cT stage, tumor volume (TV) based on the lesion’s largest diameter at MRI, percentage of Gleason pattern 4/5 (%GP4/5) at MRI-guided biopsy, and volume of GP4/5 (VolGP4/5) calculated as TV × %GP4/5. Outcome measurements and statistical analysis The variables’ ability to predict recurrence was determined in univariable and multivariable Fine-and-Gray models, according to the Akaike information criterion (AIC) and Harrell’s C-index. Results and limitations Overall, 176 (25%), 430 (60%), and 107 (15%) patients had low, intermediate, and high-risk disease, respectively, according to the EAU classification. During a median follow-up period of 57 mo, metastatic recurrence was observed in 48 patients with a 5-yr probability of 5.6% (95% confidence interval [CI] 3.9–7.7). VolGP4/5 (categories: <0.5, 0.5–1.0, 1.01–3.2, and >3.2 ml) was the parameter with the lowest AIC and the highest C-index for metastatic recurrence of 0.82 (95% CI 0.76–0.88), and for BCR it was 0.73 (95% CI 0.68–0.78). In a multivariable model that included %GP4/5 and TV, C-index values were 0.86 (95% CI 0.79–0.91) for metastatic recurrence and 0.77 (0.72–0.82) for BCR. The same results for EAU classification were 0.74 (0.67–0.80) and 0.67 (0.63–0.72), respectively. Limitations are related to short follow-up and expertise of radiologists and urologists. Conclusions We developed a preoperative risk tool integrating the VolGP4/5 based on MRI and MRI-guided biopsies to predict metastatic recurrence after RP. Our model showed higher accuracy than conventional clinical risk models. These findings might enable physicians to provide more personalized patient care. Patient summary Aggressiveness of prostate cancer evaluated before treatment by incorporating magnetic resonance imaging (MRI) and MRI-guided biopsy results gives a better estimate of the risk of metastatic recurrence than previous parameters not based on MRI.
Collapse
Affiliation(s)
| | - Arnauld Villers
- Department of Urology, University of Lille, Lille, France
- UMR8161/CNRS-Institut de Biologie de Lille, Lille, France
| | - Philippe Puech
- Department of Radiology, University of Lille, 59000 Lille, France
| | | | - Julien Labreuche
- Department of Biostatistics, CHU Lille, Lille, France
- ULR 2694 - METRICS: Évaluation des technologies de santé et des pratiques médicales, CHU Lille, University of Lille, Lille, France
| | - Elodie Drumez
- Department of Biostatistics, CHU Lille, Lille, France
- ULR 2694 - METRICS: Évaluation des technologies de santé et des pratiques médicales, CHU Lille, University of Lille, Lille, France
| | - Xavier Leroy
- Department of Histopathology, University of Lille, Lille, France
| | - Jonathan Olivier
- Department of Urology, University of Lille, Lille, France
- UMR8161/CNRS-Institut de Biologie de Lille, Lille, France
- Corresponding author. Department of Urology, Hôpital Huriez, Lille University, F-59000 Lille, France. Tel. + 33 320 444 235.
| |
Collapse
|
39
|
Montorsi F, Stabile A, Mazzone E, Gandaglia G, Briganti A. Re: Deepika Reddy, Max Peters, Taimur T. Shah, et al. Cancer Control Outcomes Following Focal Therapy Using High-intensity Focused Ultrasound in 1379 Men with Nonmetastatic Prostate Cancer: A Multi-institute 15-year Experience. Eur Urol 2022;81:407-13. Eur Urol 2022; 82:e73. [PMID: 35661628 DOI: 10.1016/j.eururo.2022.05.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 05/04/2022] [Indexed: 11/29/2022]
Affiliation(s)
- Francesco Montorsi
- Department of Urology and Division of Experimental Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Armando Stabile
- Department of Urology and Division of Experimental Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elio Mazzone
- Department of Urology and Division of Experimental Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giorgio Gandaglia
- Department of Urology and Division of Experimental Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alberto Briganti
- Department of Urology and Division of Experimental Oncology, Vita-Salute San Raffaele University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
40
|
Wan S, He Y, Zhang B, Yang Z, Du FM, Zhang CP, Fu YQ, Mi J. Overexpression of CDCA8 Predicts Poor Prognosis and Promotes Tumor Cell Growth in Prostate Cancer. Front Oncol 2022; 12:784183. [PMID: 35449575 PMCID: PMC9016845 DOI: 10.3389/fonc.2022.784183] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 03/07/2022] [Indexed: 12/24/2022] Open
Abstract
Human cell division cycle-related protein 8 (CDCA8) is an essential component of the vertebrate chromosomal passenger complex (CPC). CDCA8 was confirmed to play a role in promoting malignant tumor progression. However, the exact function of CDCA8 in the development and progression of prostate cancer (PCa) remains unclear. In this study, the database GSE69223 was downloaded by the gene expression omnibus (GEO) database, as well as CDCA8 expression differences in multiple tumor tissues and normal tissues were detected by The Cancer Genome Atlas (TCGA), TIMER, Oncomine, and Ualcan databases. Kaplan-Meier and Cox regression methods were used to analyze the correlation between CDCA8 expression and prognosis in PCa. We confirmed the expression of CDCA8 in PCa tissues by HPA. We also analyzed the association of CDCA8 expression with PCa clinical characteristics in the TCGA database. To further understand the role of CDCA8 in PCa, we assessed the effects of CDCA8 on PCa cell growth, proliferation, and migration in vitro studies. As a result, CDCA8 was significantly overexpressed in PCa cells compared with normal prostate cells. High CDCA8 expression predicts poor prognosis in PCa patients, and CDCA8 expression was higher in high-grade PCa. In addition, silencing of CDCA8 significantly inhibited PCa cell proliferation and migration. In summary, CDCA8 promoted the proliferation and migration of PCa cells.
Collapse
Affiliation(s)
- Shun Wan
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
| | - Yang He
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
| | - Bin Zhang
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
| | - Zhi Yang
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
| | - Fang-Ming Du
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
| | - Chun-Peng Zhang
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
| | - Yu-Qiang Fu
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
| | - Jun Mi
- Department of Urology, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
41
|
High Keratin-7 Expression in Benign Peri-Tumoral Prostatic Glands Is Predictive of Bone Metastasis Onset and Prostate Cancer-Specific Mortality. Cancers (Basel) 2022; 14:cancers14071623. [PMID: 35406395 PMCID: PMC8997075 DOI: 10.3390/cancers14071623] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 03/21/2022] [Indexed: 12/10/2022] Open
Abstract
BACKGROUND New predictive biomarkers are needed to accurately predict metastasis-free survival (MFS) and cancer-specific survival (CSS) in localized prostate cancer (PC). Keratin-7 (KRT7) overexpression has been associated with poor prognosis in several cancers and is described as a novel prostate progenitor marker in the mouse prostate. METHODS KRT7 expression was evaluated in prostatic cell lines and in human tissue by immunohistochemistry (IHC, on advanced PC, n = 91) and immunofluorescence (IF, on localized PC, n = 285). The KRT7 mean fluorescence intensity (MFI) was quantified in different compartments by digital analysis and correlated to clinical endpoints in the localized PC cohort. RESULTS KRT7 is expressed in prostatic cell lines and found in the basal and supra-basal compartment from healthy prostatic glands and benign peri-tumoral glands from localized PC. The KRT7 staining is lost in luminal cells from localized tumors and found as an aberrant sporadic staining (2.2%) in advanced PC. In the localized PC cohort, high KRT7 MFI above the 80th percentile in the basal compartment was significantly and independently correlated with MFS and CSS, and with hypertrophic basal cell phenotype. CONCLUSION High KRT7 expression in benign glands is an independent biomarker of MFS and CSS, and its expression is lost in tumoral cells. These results require further validation on larger cohorts.
Collapse
|
42
|
Roberts MJ, Morton A, Papa N, Franklin A, Raveenthiran S, Yaxley WJ, Coughlin G, Gianduzzo T, Kua B, McEwan L, Wong D, Delahunt B, Egevad L, Samaratunga H, Brown N, Parkinson R, Emmett L, Yaxley JW. Primary tumour PSMA intensity is an independent prognostic biomarker for biochemical recurrence-free survival following radical prostatectomy. Eur J Nucl Med Mol Imaging 2022; 49:3289-3294. [PMID: 35298693 PMCID: PMC9250456 DOI: 10.1007/s00259-022-05756-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 03/03/2022] [Indexed: 12/03/2022]
Abstract
Purpose The prognostic value of PSMA intensity on PSMA PET/CT due to underlying biology and subsequent clinical implications is an emerging topic of interest. We sought to investigate whether primary tumour PSMA PET intensity contributes to pre- and post-operative prediction of oncological outcomes following radical prostatectomy. Methods We performed a retrospective cohort study of 848 men who underwent all of multiparametric MRI (mpMRI), transperineal prostate biopsy, and 68 Ga-PSMA PET/CT prior to radical prostatectomy. PSMA intensity, quantified as maximum standard uptake value (SUVmax), and other clinical variables were considered relative to post-operative biochemical recurrence-free survival (BRFS) using Cox regression and Kaplan–Meier analysis. Results After a median follow-up of 41 months, 219 events occurred; the estimated 3-year BRFS was 79% and the 5-year BRFS was 70%. Increasing PSMA intensity was associated with less favourable BRFS overall (Log rank p < 0.001), and within subgroups of Gleason score category (Log rank p < 0.03). PSMA intensity was significantly associated with shorter time to biochemical recurrence, after adjusting for pre-operative (HR per 5-unit SUVmax increase = 1.15) and post-operative (HR per 5-unit SUVmax increase = 1.10) parameters. Conclusion These results in a large series of patients confirm PSMA intensity to be a novel, independent prognostic factor for BRFS. Supplementary Information The online version contains supplementary material available at 10.1007/s00259-022-05756-2.
Collapse
Affiliation(s)
- Matthew J Roberts
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, 4006, Australia. .,Faculty of Medicine, University of Queensland Centre for Clinical Research, Brisbane, Australia. .,Department of Urology, Redcliffe Hospital, Brisbane, Australia.
| | - Andrew Morton
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Nathan Papa
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Anthony Franklin
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | | | - William J Yaxley
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Geoffrey Coughlin
- Faculty of Medicine, The University of Queensland, Brisbane, Australia.,The Wesley Hospital, Brisbane, Australia
| | - Troy Gianduzzo
- Faculty of Medicine, The University of Queensland, Brisbane, Australia.,The Wesley Hospital, Brisbane, Australia
| | - Boon Kua
- Faculty of Medicine, The University of Queensland, Brisbane, Australia.,The Wesley Hospital, Brisbane, Australia
| | - Louise McEwan
- I-MED Radiology, The Wesley Hospital, Brisbane, Queensland, Australia
| | - David Wong
- I-MED Radiology, The Wesley Hospital, Brisbane, Queensland, Australia
| | - Brett Delahunt
- Department of Pathology and Molecular Medicine, Wellington School of Medicine and Health Sciences, University of Otago, Wellington, New Zealand
| | - Lars Egevad
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Hemamali Samaratunga
- Faculty of Medicine, The University of Queensland, Brisbane, Australia.,Aquesta Uropathology, Toowong, Queensland, Australia
| | - Nicholas Brown
- Faculty of Medicine, The University of Queensland, Brisbane, Australia.,I-MED Radiology, The Wesley Hospital, Brisbane, Queensland, Australia
| | - Robert Parkinson
- I-MED Radiology, The Wesley Hospital, Brisbane, Queensland, Australia
| | - Louise Emmett
- Faculty of Medicine, University of New South Wales, Sydney, Australia.,Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, Australia
| | - John W Yaxley
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, 4006, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, Australia.,The Wesley Hospital, Brisbane, Australia
| |
Collapse
|
43
|
Radziszewski P, Ślusarczyk A. Re: Risk Stratification of Patients Candidate to Radical Prostatectomy Based on Clinical and Multiparametric Magnetic Resonance Imaging Parameters: Development and External Validation of Novel Risk Groups. Eur Urol 2022; 82:240-241. [DOI: 10.1016/j.eururo.2022.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 03/15/2022] [Indexed: 11/15/2022]
|
44
|
Morote J, Campistol M, Triquell M, Celma A, Regis L, de Torres I, Semidey ME, Mast R, Santamaria A, Planas J, Trilla E. Improving the Early Detection of Clinically Significant Prostate Cancer in Men in the Challenging Prostate Imaging-Reporting and Data System 3 Category. EUR UROL SUPPL 2022; 37:38-44. [PMID: 35243388 PMCID: PMC8883194 DOI: 10.1016/j.euros.2021.12.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2021] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Prostate Imaging-Reporting and Data System (PI-RADS) category 3 is a challenging scenario for detection of clinically significant prostate cancer (csPCa) and some tools can improve the selection of appropriate candidates for prostate biopsy. OBJECTIVE To assess the performance of the European Randomized Study of Screening for Prostate Cancer (ERSPC) magnetic resonance imaging (MRI) model, the new Proclarix test, and prostate-specific antigen density (PSAD) in selecting candidates for prostate biopsy among men in the PI-RADS 3 category. DESIGN SETTING AND PARTICIPANTS We conducted a head-to-head prospective analysis of 567 men suspected of having PCa for whom guided and systematic biopsies were scheduled between January 2018 and March 2020 in a single academic institution. A PI-RADS v.2 category 3 lesion was identified in 169 men (29.8%). OUTCOME MEASUREMENT AND STATISTICAL ANALYSIS csPCa, insignificant PCa (iPCa), and unnecessary biopsy rates were analysed. csPCa was defined as grade group ≥2. Receiver operating characteristic (ROC) curves, decision curve analysis curves, and clinical utility curves were plotted. RESULTS AND LIMITATIONS PCa was detected in 53/169 men (31.4%) with a PI-RADS 3 lesion, identified as csPCa in 25 (14.8%) and iPCa in 28 (16.6%). The area under the ROC curve for csPCa detection was 0.703 (95% confidence interval [CI] 0.621-0.768) for Proclarix, 0.657 (95% CI 0.547-0.766) for the ERSPC MRI model, and 0.612 (95% CI 0.497-0.727) for PSAD (p = 0.027). The threshold with the highest sensitivity was 10% for Proclarix, 1.5% for the ERSPC MRI model, and 0.07 ng/ml/cm3 for PSAD, which yielded sensitivity of 100%, 91%, and 84%, respectively. Some 21.3%, 26.2%, and 7.1% of biopsies would be avoided with Proclarix, PSAD, and the ERSPC MRI model, respectively. Proclarix showed a net benefit over PSAD and the ERSPC MRI model. Both Proclarix and PSAD reduced iPCa overdetection from 16.6% to 11.3%, while the ERSPC MRI model reduced iPCa overdetection to 15.4%. CONCLUSIONS Proclarix was more accurate in selecting appropriate candidates for prostate biopsy among men in the PI-RADS 3 category when compared to PSAD and the ERSPC MRI model. Proclarix detected 100% of csPCa cases and would reduce prostate biopsies by 21.3% and iPCa overdetection by 5.3%. PATIENT SUMMARY We compared three methods and found that the Proclarix test can optimise the detection of clinically significant prostate cancer in men with a score of 3 on the Prostate Imaging-Reporting and Data System for magnetic resonance imaging scans.
Collapse
Affiliation(s)
- Juan Morote
- Department of Urology, Vall d’Hebron Hospital, Barcelona, Spain
- Prostate Cancer Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
- Department of Radiology, Vall d’Hebron Hospital, Barcelona, Spain
| | - Miriam Campistol
- Prostate Cancer Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
| | - Marina Triquell
- Prostate Cancer Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
| | - Anna Celma
- Department of Urology, Vall d’Hebron Hospital, Barcelona, Spain
- Prostate Cancer Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
| | - Lucas Regis
- Department of Urology, Vall d’Hebron Hospital, Barcelona, Spain
- Prostate Cancer Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
| | - Inés de Torres
- Prostate Cancer Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
- Department of Pathology, Vall d’Hebron Hospital, Barcelona, Spain
- Universitat Autònoma of Barcelona, Barcelona, Spain
| | - Maria E. Semidey
- Prostate Cancer Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
- Department of Radiology, Vall d’Hebron Hospital, Barcelona, Spain
- Department of Pathology, Vall d’Hebron Hospital, Barcelona, Spain
| | - Richard Mast
- Department of Radiology, Vall d’Hebron Hospital, Barcelona, Spain
| | - Anna Santamaria
- Prostate Cancer Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
| | - Jacques Planas
- Department of Urology, Vall d’Hebron Hospital, Barcelona, Spain
- Prostate Cancer Research Group, Vall d’Hebron Research Institute, Barcelona, Spain
| | - Enrique Trilla
- Department of Urology, Vall d’Hebron Hospital, Barcelona, Spain
- Universitat Autònoma of Barcelona, Barcelona, Spain
| |
Collapse
|
45
|
Gandaglia G, Montorsi F. Metastases-directed Therapies in the Prostate-specific Membrane Antigen Era: Not All That Glitters Is Curable. Eur Urol Oncol 2022; 5:52-53. [DOI: 10.1016/j.euo.2022.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 01/21/2022] [Accepted: 01/21/2022] [Indexed: 11/28/2022]
|
46
|
Active surveillance for intermediate-risk prostate cancer. World J Urol 2022; 40:79-86. [PMID: 35044491 DOI: 10.1007/s00345-021-03893-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 11/15/2021] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION Active surveillance (AS) is an established approach in the management of low-risk, localized prostate cancer. While the use of AS to manage intermediate-risk (IR) disease has gradually increased over time, there remains uncertainty with regards to its safety, with only a minority of IR patients currently being managed with this approach. MATERIALS AND METHODS We conducted a narrative review based on an analysis of the literature focusing on articles describing AS for IR prostate cancer. We focus on the uncertainty surrounding AS in IR disease by discussing variations in the definitions and guideline recommendations associated with IR disease, and describing the limitations of the evidence from observational studies and randomized trials. CONCLUSION The safety of AS for IR disease remains unknown, given the lack of randomized trials and the limitations of the current observational studies. Further research is needed to identify select patients with IR prostate cancer that can be managed safely with AS.
Collapse
|
47
|
Ferro M, de Cobelli O, Musi G, del Giudice F, Carrieri G, Busetto GM, Falagario UG, Sciarra A, Maggi M, Crocetto F, Barone B, Caputo VF, Marchioni M, Lucarelli G, Imbimbo C, Mistretta FA, Luzzago S, Vartolomei MD, Cormio L, Autorino R, Tătaru OS. Radiomics in prostate cancer: an up-to-date review. Ther Adv Urol 2022; 14:17562872221109020. [PMID: 35814914 PMCID: PMC9260602 DOI: 10.1177/17562872221109020] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 05/30/2022] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PCa) is the most common worldwide diagnosed malignancy in male population. The diagnosis, the identification of aggressive disease, and the post-treatment follow-up needs a more comprehensive and holistic approach. Radiomics is the extraction and interpretation of images phenotypes in a quantitative manner. Radiomics may give an advantage through advancements in imaging modalities and through the potential power of artificial intelligence techniques by translating those features into clinical outcome prediction. This article gives an overview on the current evidence of methodology and reviews the available literature on radiomics in PCa patients, highlighting its potential for personalized treatment and future applications.
Collapse
Affiliation(s)
- Matteo Ferro
- Department of Urology, European Institute of Oncology, IRCCS, Milan, Italy, via Ripamonti 435 Milano, Italy
| | - Ottavio de Cobelli
- Department of Urology, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Gennaro Musi
- Department of Urology, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hematology-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Francesco del Giudice
- Department of Urology, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Giuseppe Carrieri
- Department of Urology and Organ Transplantation, University of Foggia, Foggia, Italy
| | - Gian Maria Busetto
- Department of Urology and Organ Transplantation, University of Foggia, Foggia, Italy
| | | | - Alessandro Sciarra
- Department of Urology, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Martina Maggi
- Department of Urology, Policlinico Umberto I, Sapienza University of Rome, Rome, Italy
| | - Felice Crocetto
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples ‘Federico II’, Naples, Italy
| | - Biagio Barone
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples ‘Federico II’, Naples, Italy
| | - Vincenzo Francesco Caputo
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples ‘Federico II’, Naples, Italy
| | - Michele Marchioni
- Department of Medical, Oral and Biotechnological Sciences, G. d’Annunzio, University of Chieti, Chieti, Italy; Urology Unit, ‘SS. Annunziata’ Hospital, Chieti, Italy
- Department of Urology, ASL Abruzzo 2, Chieti, Italy
| | - Giuseppe Lucarelli
- Department of Emergency and Organ Transplantation, Urology, Andrology and Kidney Transplantation Unit, University of Bari, Bari, Italy
| | - Ciro Imbimbo
- Department of Neurosciences, Reproductive Sciences and Odontostomatology, University of Naples ‘Federico II’, Naples, Italy
| | - Francesco Alessandro Mistretta
- Department of Urology, European Institute of Oncology, IRCCS, Milan, Italy
- Università degli Studi di Milano, Milan, Italy
| | - Stefano Luzzago
- Department of Urology, European Institute of Oncology, IRCCS, Milan, Italy
- Università degli Studi di Milano, Milan, Italy
| | - Mihai Dorin Vartolomei
- Department of Cell and Molecular Biology, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mures, Târgu Mures, Romania
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Luigi Cormio
- Urology and Renal Transplantation Unit, Department of Medical and Surgical Sciences, University of Foggia, Foggia, Italy
- Urology Unit, Bonomo Teaching Hospital, Foggia, Italy
| | | | - Octavian Sabin Tătaru
- Institution Organizing University Doctoral Studies, I.O.S.U.D., George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mures, Târgu Mures, Romania
| |
Collapse
|