1
|
Almeida‐Magana R, Au M, Al‐Hammouri T, Mathew M, Dinneen K, Mendes LST, Dinneen E, Vreuls W, Shaw G, Freeman A, Haider A. Accuracy of the LaserSAFE technique for detecting positive surgical margins during robot-assisted radical prostatectomy: blind assessment and inter-rater agreement analysis. Histopathology 2025; 86:433-440. [PMID: 39403832 PMCID: PMC11707496 DOI: 10.1111/his.15336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/11/2024] [Accepted: 09/22/2024] [Indexed: 01/11/2025]
Abstract
INTRODUCTION AND OBJECTIVES Fluorescence confocal microscopy (FCM) is a new imaging modality capable of generating digital microscopic resolution scans of fresh surgical specimens, and holds potential as an alternative to frozen section (FS) analysis for intra-operative assessment of surgical margins. Previously, we described the LaserSAFE technique as an application of FCM for margin assessment in robot-assisted radical prostatectomy (RARP) using the Histolog® scanner. This study describes the accuracy and inter-rater agreement of FCM imaging compared to corresponding paraffin-embedded analysis (PA) among four blinded pathologists for the presence of positive surgical margins (PSM). MATERIALS AND METHODS RARP specimens from patients enrolled in the control arm of the NeuroSAFE PROOF study (NCT03317990) were analysed from April 2022 to February 2023. Prostate specimens were imaged using the Histolog® scanner before formalin fixation and PA. Four trained assessors, blinded to PA, reviewed and analysed FCM images of the posterolateral prostatic surface. RESULTS A total of 31 prostate specimens were included in the study. PA per lateral side of the prostate identified 11 instances of positive margins. Among the four histopathologists included in our study, FCM achieved a sensitivity of 73-91 and specificity of 94-100% for the presence of PSM. Fleiss' Kappa for inter-rater agreement on PSM was 0.78 (95% confidence interval = 0.64-0.92), indicating substantial agreement. CONCLUSION This blinded analysis of FCM versus PA among histopathologists with different experience levels demonstrated high accuracy and substantial inter-rater agreement for diagnosing PSM. This supports the role of the FCM as an alternative to FS.
Collapse
Affiliation(s)
- Ricardo Almeida‐Magana
- Department of Targeted InterventionUniversity CollegeLondonUK
- Department of UrologyUniversity College London Hospitals NHS Foundation TrustLondonUK
| | - Matthew Au
- Department of Targeted InterventionUniversity CollegeLondonUK
| | - Tarek Al‐Hammouri
- Department of UrologyUniversity College London Hospitals NHS Foundation TrustLondonUK
- Centre for Medical ImagingUniversity CollegeLondonUK
| | - Manju Mathew
- Centre for Medical ImagingUniversity CollegeLondonUK
- Department of HistopathologyUniversity College London Hospitals NHS Foundation TrustLondonUK
| | - Kate Dinneen
- Department of HistopathologyUniversity College London Hospitals NHS Foundation TrustLondonUK
| | - Larissa S T Mendes
- Department of Targeted InterventionUniversity CollegeLondonUK
- Department of HistopathologyUniversity College London Hospitals NHS Foundation TrustLondonUK
| | - Eoin Dinneen
- Department of UrologyUniversity College London Hospitals NHS Foundation TrustLondonUK
| | - Willem Vreuls
- Department of HistopathologyCanisius Wilhelmina HospitalNijmegenthe Netherlands
- LabPONHengelothe Netherlands
| | - Greg Shaw
- Department of Targeted InterventionUniversity CollegeLondonUK
- Department of UrologyUniversity College London Hospitals NHS Foundation TrustLondonUK
| | - Alex Freeman
- Department of HistopathologyUniversity College London Hospitals NHS Foundation TrustLondonUK
| | - Aiman Haider
- Department of HistopathologyUniversity College London Hospitals NHS Foundation TrustLondonUK
| |
Collapse
|
2
|
Dinneen E, Almeida-Magana R, Al-Hammouri T, Fernandes I, Mayor N, Mendes L, Winkler M, Silvanto A, Haider A, Freeman A, Shaw G. Intraoperative margin assessment during radical prostatectomy: is microscopy frozen in time or ready for digital defrost? Histopathology 2024; 85:716-726. [PMID: 39104212 DOI: 10.1111/his.15290] [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: 08/07/2024]
Abstract
Intraoperative frozen section (IFS) is used with the intention to improve functional and oncological outcomes for patients undergoing radical prostatectomy (RP). High resource requirements of IFS techniques such as NeuroSAFE may preclude widespread adoption, even if there are benefits to patients. Recent advances in fresh-tissue microscopic digital imaging technologies may offer an attractive alternative, and there is a growing body of evidence regarding these technologies. In this narrative review, we discuss some of the familiar limitations of IFS and compare these to the attractive counterpoints of modern digital imaging technologies such as the speed and ease of image generation, the locality of equipment within (or near) the operating room, the ability to maintain tissue integrity, and digital transfer of images. Confocal laser microscopy (CLM) is the modality most frequently reported in the literature for margin assessment during RP. We discuss several imitations and obstacles to widespread dissemination of digital imaging technologies. Among these, we consider how the 'en-face' margin perspective will challenge urologists and pathologists to understand afresh the meaning of positive margin significance. As a part of this, discussions on how to describe, categorize, react to, and evaluate these technologies are needed to improve patient outcomes. Limitations of this review include its narrative structure and that the evidence base in this field is relatively immature but developing at pace.
Collapse
Affiliation(s)
- Eoin Dinneen
- Division of Surgery & Interventional Science, University College London, London, UK
- Department of Urology, Westmoreland Street Hospital, University College Hospital London, London, UK
| | - Ricardo Almeida-Magana
- Division of Surgery & Interventional Science, University College London, London, UK
- Department of Urology, Westmoreland Street Hospital, University College Hospital London, London, UK
| | - Tarek Al-Hammouri
- Division of Surgery & Interventional Science, University College London, London, UK
- Centre for Medical Imaging, University College London, London, UK
| | - Iona Fernandes
- Department of Urology, Westmoreland Street Hospital, University College Hospital London, London, UK
| | - Nikhil Mayor
- Department of Urology, Imperial College NHS Healthcare Trust, Charing Cross Hospital, London, UK
- Imperial Prostate, Division of Surgery, Imperial College London, London, UK
| | - Larissa Mendes
- Department of Histopathology, University College Hospital London, London, UK
| | - Mathias Winkler
- Department of Urology, Imperial College NHS Healthcare Trust, Charing Cross Hospital, London, UK
- Imperial Prostate, Division of Surgery, Imperial College London, London, UK
| | - Anna Silvanto
- Department of Histopathology, University College Hospital London, London, UK
| | - Aiman Haider
- Department of Histopathology, University College Hospital London, London, UK
| | - Alex Freeman
- Department of Histopathology, University College Hospital London, London, UK
| | - Greg Shaw
- Division of Surgery & Interventional Science, University College London, London, UK
- Department of Urology, Westmoreland Street Hospital, University College Hospital London, London, UK
| |
Collapse
|
3
|
Jiang S, Li Y, Guo Y, Gong B, Wei C, Liu W, Chen C, Pan F, Song J, He Q, Yang L, Zhou G. MRI-measured periprostatic to subcutaneous adipose tissue thickness ratio as an independent risk factor in prostate cancer patients undergoing radical prostatectomy. Sci Rep 2024; 14:20896. [PMID: 39245685 PMCID: PMC11381511 DOI: 10.1038/s41598-024-71862-w] [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: 05/09/2024] [Accepted: 09/02/2024] [Indexed: 09/10/2024] Open
Abstract
The purpose of this study is to evaluate whether the periprostatic adipose tissue thickness (PPATT) is an independent prognostic factor for prostate cancer patients after laparoscopic radical prostatectomy (LRP). This retrospective cohort study included consecutive prostate cancer patients who underwent LRP treatment at Wuhan Union Hospital from June 2, 2016, to September 7, 2023. PPATT was defined as the thickness of periprostatic fat and was obtained by measuring the shortest vertical distance from the pubic symphysis to the prostate on the midsagittal T2-weighted MR images. Subcutaneous adipose tissue thickness (SATT) was obtained by measuring the shortest vertical distance from the pubic symphysis to the skin at the same slice with PPATT. The primary outcome of the study was biochemical recurrence (BCR), and the secondary outcome was overall survival (OS). Multivariable Cox regression analysis was used to identify independent prognostic factors for prostate cancer survival and prognosis. Based on the optimal cutoff value, 162 patients were divided into a low PPATT/SATT group (n = 82) and a high PPATT/SATT group (n = 80). During the entire follow-up period (median 23.5 months), 26 patients in the high PPATT/SATT group experienced BCR (32.5%), compared to 18 in the low PPATT/SATT group (22.0%). Kaplan-Meier curve analysis indicated that the interval to BCR was significantly shorter in the high PPATT/SATT group (P = 0.037). Multivariable Cox regression analysis revealed that an increase in the PPATT/SATT ratio was associated with BCR (hazard ratio: 1.90, 95% CI, 1.03-3.51; P = 0.040). The PPATT/SATT ratio is a significant independent risk factor for BCR after LRP for prostate cancer patients.
Collapse
Affiliation(s)
- Shanshan Jiang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yi Li
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yusheng Guo
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Bingxin Gong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Chengcheng Wei
- Department of Urology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Weiwei Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Chao Chen
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Feng Pan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Jiyu Song
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qingliu He
- Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Department of Urology, The Second Affiliated Hospital of Fujian Medical University, No.34 North Zhongshan Road, Quanzhou, 362000, China.
| | - Lian Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| | - Guofeng Zhou
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1277 Jiefang Avenue, Wuhan, 430022, China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| |
Collapse
|
4
|
Leung D, Castellani D, Nicoletti R, Dilme RV, Sierra JM, Serni S, Franzese C, Chiacchio G, Galosi AB, Mazzucchelli R, Palagonia E, Dell'Oglio P, Galfano A, Bocciardi AM, Zhao X, Ng CF, Lee HY, Sakamoto S, Vasdev N, Rivas JG, Campi R, Teoh JYC. The Oncological and Functional Prognostic Value of Unconventional Histology of Prostate Cancer in Localized Disease Treated with Robotic Radical Prostatectomy: An International Multicenter 5-Year Cohort Study. Eur Urol Oncol 2024; 7:581-588. [PMID: 38185614 DOI: 10.1016/j.euo.2023.12.006] [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: 10/02/2023] [Revised: 12/03/2023] [Accepted: 12/11/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND AND OBJECTIVE The impact of prostate cancer of unconventional histology (UH) on oncological and functional outcomes after robot-assisted radical prostatectomy (RARP) and adjuvant radiotherapy (aRT) receipt is unclear. We compared the impact of cribriform pattern (CP), ductal adenocarcinoma (DAC), and intraductal carcinoma (IDC) in comparison to pure adenocarcinoma (AC) on short- to mid-term oncological and functional results and receipt of aRT after RARP. METHODS We retrospectively collected data for a large international cohort of men with localized prostate cancer treated with RARP between 2016 and 2020. The primary outcomes were biochemical recurrence (BCR)-free survival, erectile and continence function. aRT receipt was a secondary outcome. Kaplan-Meier survival and Cox regression analyses were performed. KEY FINDINGS AND LIMITATIONS A total of 3935 patients were included. At median follow-up of 2.8 yr, the rates for BCR incidence (AC 10.7% vs IDC 17%; p < 0.001) and aRT receipt (AC 4.5% vs DAC 6.3% [p = 0.003] vs IDC 11.2% [p < 0.001]) were higher with UH. The 5-yr BCR-free survival rate was significantly poorer for UH groups, with hazard ratios of 1.67 (95% confidence interval [CI] 1.16-2.40; p = 0.005) for DAC, 5.22 (95% CI 3.41-8.01; p < 0.001) for IDC, and 3.45 (95% CI 2.29-5.20; p < 0.001) for CP in comparison to AC. Logistic regression analysis revealed that the presence of UH doubled the risk of new-onset erectile dysfunction at 1 yr, in comparison to AC (grade group 1-3), with hazard ratios of 2.13 (p < 0.001) for DAC, 2.14 (p < 0.001) for IDC, and 2.01 (p = 0.011) for CP. Moreover, CP, but not IDC or DAC, was associated with a significantly higher risk of incontinence (odds ratio 1.97; p < 0.001). The study is limited by the lack of central histopathological review and relatively short follow-up. CONCLUSIONS AND CLINICAL IMPLICATIONS In a large cohort, UH presence was associated with worse short- to mid-term oncological outcomes after RARP. IDC independently predicted a higher rate of aRT receipt. At 1-yr follow-up after RP, patients with UH had three times higher risk of erectile dysfunction post RARP; CP was associated with a twofold higher incontinence rate. PATIENT SUMMARY Among patients with prostate cancer who undergo robot-assisted surgery to remove the prostate, those with less common types of prostate cancer have worse results for cancer control, erection, and urinary continence and a higher probability of receiving additional radiotherapy after surgery.
Collapse
Affiliation(s)
- David Leung
- Division of Urology, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Daniele Castellani
- Division of Urology, Azienda Ospedaliero-Universitaria, Ospedali Riuniti di Ancona, Università Politecnica delle Marche, Ancona, Italy
| | - Rossella Nicoletti
- Division of Urology, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China; Unit of Urological Robotic Surgery and Renal Transplantation, Careggi Hospital, University of Florence, Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | | | | | - Sergio Serni
- Unit of Urological Robotic Surgery and Renal Transplantation, Careggi Hospital, University of Florence, Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Carmine Franzese
- Division of Urology, Azienda Ospedaliero-Universitaria, Ospedali Riuniti di Ancona, Università Politecnica delle Marche, Ancona, Italy
| | - Giuseppe Chiacchio
- Division of Urology, Azienda Ospedaliero-Universitaria, Ospedali Riuniti di Ancona, Università Politecnica delle Marche, Ancona, Italy
| | - Andrea Benedetto Galosi
- Division of Urology, Azienda Ospedaliero-Universitaria, Ospedali Riuniti di Ancona, Università Politecnica delle Marche, Ancona, Italy
| | - Roberta Mazzucchelli
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, Azienda Ospedaliero-Universitaria delle Marche, Ancona, Italy
| | - Erika Palagonia
- Urology Department, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Paolo Dell'Oglio
- Urology Department, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Antonio Galfano
- Urology Department, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | | | - Xue Zhao
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Chi Fai Ng
- Division of Urology, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | | | - Shinichi Sakamoto
- Department of Urology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Nikhil Vasdev
- Department of Urology, Lister Hospital, East and North Herts NHS Trust, Stevenage, UK
| | - Juan Gomez Rivas
- Department of Urology, Hospital Clínico San Carlos, Madrid, Spain
| | - Riccardo Campi
- Unit of Urological Robotic Surgery and Renal Transplantation, Careggi Hospital, University of Florence, Florence, Italy; Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Jeremy Yuen-Chun Teoh
- Division of Urology, Department of Surgery, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
5
|
Frego N, Contieri R, Fasulo V, Maffei D, Avolio PP, Arena P, Beatrici E, Sordelli F, De Carne F, Lazzeri M, Saita A, Hurle R, Buffi NM, Casale P, Lughezzani G. Development of a microultrasound-based nomogram to predict extra-prostatic extension in patients with prostate cancer undergoing robot-assisted radical prostatectomy. Urol Oncol 2024; 42:159.e9-159.e16. [PMID: 38423852 DOI: 10.1016/j.urolonc.2024.01.033] [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: 11/07/2023] [Revised: 01/12/2024] [Accepted: 01/26/2024] [Indexed: 03/02/2024]
Abstract
OBJECTIVES To develop a microultrasound-based nomogram including clinicopathological parameters and microultrasound findings to predict the presence of extra-prostatic extension and guide the grade of nerve-sparing. MATERIAL AND METHODS All patients underwent microultrasound the day before robot-assisted radical prostatectomy. Variables significantly associated with extra-prostatic extension at univariable analysis were used to build the multivariable logistic model, and the regression coefficients were used to develop the nomogram. The model was subjected to 1000 bootstrap resamples for internal validation. The performance of the microultrasound-based model was evaluated using the area under the curve (AUC) of the receiver operating characteristic (ROC) curve, calibration plot, and decision curve analysis (DCA). RESULTS Overall, 122/295 (41.4%) patients had a diagnosis of extra-prostatic extension on definitive pathology. Microultrasound correctly identify extra-prostatic extension in 84/122 (68.9%) cases showing a sensitivity and a specificity of 68.9% and 84.4%, with an AUC of 76.6%. After 1000 bootstrap resamples, the predictive accuracy of the microultrasound-based model was 85.9%. The calibration plot showed a satisfactory concordance between predicted probabilities and observed frequencies of extra-prostatic extension. The DCA showed a higher clinical net-benefit compared to the model including only clinical parameters. Considering a 4% cut-off, nerve-sparing was recommended in 173 (58.6%) patients and extra-prostatic extension was detected in 32 (18.5%) of them. CONCLUSION We developed a microultrasound-based nomogram for the prediction of extra-prostatic extension that could aid in the decision whether to preserve or not neurovascular bundles. External validation and a direct comparison with mpMRI-based nomogram is crucial to corroborate our results.
Collapse
Affiliation(s)
- Nicola Frego
- Department of Urology, IRCCS - Humanitas Research Hospital, Milan, Italy; Department of Biomedical Science, Humanitas University, Milan, Italy
| | - Roberto Contieri
- Department of Urology, IRCCS - Humanitas Research Hospital, Milan, Italy; Department of Biomedical Science, Humanitas University, Milan, Italy
| | - Vittorio Fasulo
- Department of Urology, IRCCS - Humanitas Research Hospital, Milan, Italy; Department of Biomedical Science, Humanitas University, Milan, Italy
| | - Davide Maffei
- Department of Urology, IRCCS - Humanitas Research Hospital, Milan, Italy; Department of Biomedical Science, Humanitas University, Milan, Italy
| | - Pier Paolo Avolio
- Department of Urology, IRCCS - Humanitas Research Hospital, Milan, Italy; Department of Biomedical Science, Humanitas University, Milan, Italy
| | - Paola Arena
- Department of Urology, IRCCS - Humanitas Research Hospital, Milan, Italy; Department of Biomedical Science, Humanitas University, Milan, Italy
| | - Edoardo Beatrici
- Department of Urology, IRCCS - Humanitas Research Hospital, Milan, Italy; Department of Biomedical Science, Humanitas University, Milan, Italy
| | - Federica Sordelli
- Department of Urology, IRCCS - Humanitas Research Hospital, Milan, Italy; Department of Biomedical Science, Humanitas University, Milan, Italy
| | - Fabio De Carne
- Department of Urology, IRCCS - Humanitas Research Hospital, Milan, Italy; Department of Biomedical Science, Humanitas University, Milan, Italy
| | - Massimo Lazzeri
- Department of Urology, IRCCS - Humanitas Research Hospital, Milan, Italy
| | - Alberto Saita
- Department of Urology, IRCCS - Humanitas Research Hospital, Milan, Italy
| | - Rodolfo Hurle
- Department of Urology, IRCCS - Humanitas Research Hospital, Milan, Italy
| | - Nicolò Maria Buffi
- Department of Urology, IRCCS - Humanitas Research Hospital, Milan, Italy; Department of Biomedical Science, Humanitas University, Milan, Italy.
| | - Paolo Casale
- Department of Urology, IRCCS - Humanitas Research Hospital, Milan, Italy
| | - Giovanni Lughezzani
- Department of Urology, IRCCS - Humanitas Research Hospital, Milan, Italy; Department of Biomedical Science, Humanitas University, Milan, Italy
| |
Collapse
|
6
|
Li L, Shiradkar R, Gottlieb N, Buzzy C, Hiremath A, Viswanathan VS, MacLennan GT, Omil Lima D, Gupta K, Shen DL, Tirumani SH, Magi-Galluzzi C, Purysko A, Madabhushi A. Multi-scale statistical deformation based co-registration of prostate MRI and post-surgical whole mount histopathology. Med Phys 2024; 51:2549-2562. [PMID: 37742344 PMCID: PMC10960735 DOI: 10.1002/mp.16753] [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: 04/07/2023] [Revised: 09/07/2023] [Accepted: 09/12/2023] [Indexed: 09/26/2023] Open
Abstract
BACKGROUND Accurate delineations of regions of interest (ROIs) on multi-parametric magnetic resonance imaging (mpMRI) are crucial for development of automated, machine learning-based prostate cancer (PCa) detection and segmentation models. However, manual ROI delineations are labor-intensive and susceptible to inter-reader variability. Histopathology images from radical prostatectomy (RP) represent the "gold standard" in terms of the delineation of disease extents, for example, PCa, prostatitis, and benign prostatic hyperplasia (BPH). Co-registering digitized histopathology images onto pre-operative mpMRI enables automated mapping of the ground truth disease extents onto mpMRI, thus enabling the development of machine learning tools for PCa detection and risk stratification. Still, MRI-histopathology co-registration is challenging due to various artifacts and large deformation between in vivo MRI and ex vivo whole-mount histopathology images (WMHs). Furthermore, the artifacts on WMHs, such as tissue loss, may introduce unrealistic deformation during co-registration. PURPOSE This study presents a new registration pipeline, MSERgSDM, a multi-scale feature-based registration (MSERg) with a statistical deformation (SDM) constraint, which aims to improve accuracy of MRI-histopathology co-registration. METHODS In this study, we collected 85 pairs of MRI and WMHs from 48 patients across three cohorts. Cohort 1 (D1), comprised of a unique set of 3D printed mold data from six patients, facilitated the generation of ground truth deformations between ex vivo WMHs and in vivo MRI. The other two clinically acquired cohorts (D2 and D3) included 42 patients. Affine and nonrigid registrations were employed to minimize the deformation between ex vivo WMH and ex vivo T2-weighted MRI (T2WI) in D1. Subsequently, ground truth deformation between in vivo T2WI and ex vivo WMH was approximated as the deformation between in vivo T2WI and ex vivo T2WI. In D2 and D3, the prostate anatomical annotations, for example, tumor and urethra, were made by a pathologist and a radiologist in collaboration. These annotations included ROI boundary contours and landmark points. Before applying the registration, manual corrections were made for flipping and rotation of WMHs. MSERgSDM comprises two main components: (1) multi-scale representation construction, and (2) SDM construction. For the SDM construction, we collected N = 200 reasonable deformation fields generated using MSERg, verified through visual inspection. Three additional methods, including intensity-based registration, ProsRegNet, and MSERg, were also employed for comparison against MSERgSDM. RESULTS Our results suggest that MSERgSDM performed comparably to the ground truth (p > 0.05). Additionally, MSERgSDM (ROI Dice ratio = 0.61, landmark distance = 3.26 mm) exhibited significant improvement over MSERg (ROI Dice ratio = 0.59, landmark distance = 3.69 mm) and ProsRegNet (ROI Dice ratio = 0.56, landmark distance = 4.00 mm) in local alignment. CONCLUSIONS This study presents a novel registration method, MSERgSDM, for mapping ex vivo WMH onto in vivo prostate MRI. Our preliminary results demonstrate that MSERgSDM can serve as a valuable tool to map ground truth disease annotations from histopathology images onto MRI, thereby assisting in the development of machine learning models for PCa detection on MRI.
Collapse
Affiliation(s)
- Lin Li
- Deptartment of Biomedical Engineering, Case Western Reserve University, Cleveland, OH
| | - Rakesh Shiradkar
- Wallace H Coulter Department of Biomedical Engineering at Emory University and Georgia Institute of Technology
| | - Noah Gottlieb
- Deptartment of Biomedical Engineering, Case Western Reserve University, Cleveland, OH
| | - Christina Buzzy
- Deptartment of Biomedical Engineering, Case Western Reserve University, Cleveland, OH
| | - Amogh Hiremath
- Deptartment of Biomedical Engineering, Case Western Reserve University, Cleveland, OH
| | - Vidya Sankar Viswanathan
- Wallace H Coulter Department of Biomedical Engineering at Emory University and Georgia Institute of Technology
| | - Gregory T. MacLennan
- Department of Pathology and Urology, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Danly Omil Lima
- Department of Pathology and Urology, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Karishma Gupta
- Department of Pathology and Urology, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Daniel Lee Shen
- Department of Pathology and Urology, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | | | | | - Andrei Purysko
- Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
- Imaging Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Anant Madabhushi
- Wallace H Coulter Department of Biomedical Engineering at Emory University and Georgia Institute of Technology
- Atlanta Veterans Administration Medical Center
| |
Collapse
|
7
|
Hatayama T, Goto K, Fujiyama K, Goriki A, Kaneko M, Mita K. Risk classification system using the detailed positive surgical margin status for predicting biochemical recurrence after robot-assisted radical prostatectomy. Asia Pac J Clin Oncol 2024. [PMID: 38512888 DOI: 10.1111/ajco.14053] [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: 03/05/2023] [Revised: 09/01/2023] [Accepted: 02/25/2024] [Indexed: 03/23/2024]
Abstract
AIM This study aimed to evaluate the risk classification system using the detailed positive surgical margin (PSM) status to predict biochemical recurrence (BCR) after robot-assisted radical prostatectomy (RARP). METHODS We retrospectively analyzed 427 patients who underwent RARP between January 2016 and March 2020. We investigated risk factors for BCR using univariate and multivariate Cox proportional hazard regression models. The biochemical recurrence-free survival (BRFS) rate was assessed using the Kaplan-Meier method. RESULTS The median follow-up period was 43.4 months and 99 patients developed BCR. In the multivariate analysis, maximum PSM length > 5.0 mm and the International Society of Urological Pathology grade group (ISUP GG) at the PSM ≥3 were predictive factors for BCR in patients with a PSM. In the multivariate analysis, these factors were also independent predictive factors in the overall study population, including patients without a PSM. We classified the patients into four groups using these factors and found that the 1-year BRFS rates in the negative surgical margin (NSM) group, low-risk group (PSM and neither factor), intermediate-risk group (either factor), and high-risk group (both factors) were 94.9%, 94.5%, 83.1%, and 52.9%, respectively. The low-risk group showed similar BRFS to the NSM group (p = 0.985), while the high-risk group had significantly worse BRFS than the other groups (p < 0.001). CONCLUSION Maximum PSM length > 5.0 mm and ISUP GG at the PSM ≥3 were independent predictive factors for BCR after RARP. Risk classification for BCR using these factors is considered to be useful and might help urologists decide on additional treatment after RARP.
Collapse
Affiliation(s)
- Tomoya Hatayama
- Department of Urology, Hiroshima City North Medical Center Asa Citizens Hospital, Hiroshima, Japan
| | - Keisuke Goto
- Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kenta Fujiyama
- Department of Urology, Hiroshima City North Medical Center Asa Citizens Hospital, Hiroshima, Japan
| | - Akihiro Goriki
- Department of Urology, Hiroshima City North Medical Center Asa Citizens Hospital, Hiroshima, Japan
| | - Mayumi Kaneko
- Department of Diagnostic Pathology, Hiroshima City North Medical Center Asa Citizens Hospital, Hiroshima, Japan
| | - Koji Mita
- Department of Urology, Hiroshima City North Medical Center Asa Citizens Hospital, Hiroshima, Japan
| |
Collapse
|
8
|
Bidot S, Yin J, Zhou P, Zhang L, Deeb KK, Smith G, Hill CE, Xiu J, Bilen MA, Case KB, Tinsley M, Carthon B, Harik LR. Genetic Profiling of African American Patients With Prostatic Adenocarcinoma Metastatic to the Lymph Nodes: A Pilot Study. Arch Pathol Lab Med 2024; 148:310-317. [PMID: 37327205 DOI: 10.5858/arpa.2022-0274-oa] [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] [Accepted: 03/23/2023] [Indexed: 06/18/2023]
Abstract
CONTEXT.— Genetic profiling data of prostatic adenocarcinoma are derived from predominantly White patients. In African Americans, prostatic adenocarcinoma has a poorer prognosis, raising the possibility of distinct genetic alterations. OBJECTIVE.— To investigate the genomic alterations of prostatic adenocarcinoma metastatic to regional lymph nodes in African American patients, with an emphasis on SPOP mutation. DESIGN.— We retrospectively reviewed African American patients with pN1 prostatic adenocarcinoma managed with radical prostatectomy and lymph node dissection. Comprehensive molecular profiling was performed, and androgen receptor signaling scores were calculated. RESULTS.— Nineteen patients were included. The most frequent genetic alteration was SPOP mutations (5 of 17; 29.4% [95% CI: 10.3-56.0]). While most alterations were associated with a high androgen receptor signaling score, mutant SPOP was exclusively associated with a low median and interquartile range (IQR) androgen receptor signaling score (0.788 [IQR 0.765-0.791] versus 0.835 [IQR 0.828-0.842], P = .003). In mutant SPOP, mRNA expression of SPOP inhibitor G3BP1 and SPOP substrates showed a significantly decreased expression of AR (33.40 [IQR 28.45-36.30] versus 59.53 [IQR 53.10-72.83], P = .01), TRIM24 (3.95 [IQR 3.28-5.03] versus 9.80 [IQR 7.39-11.70], P = .008), and NCOA3 (15.19 [IQR 10.59-15.93] versus 21.88 [IQR 18.41-28.33], P = .046). CONCLUSIONS.— African American patients with metastatic prostate adenocarcinoma might have a higher prevalence of mutant SPOP (30%), compared to ∼10% in unselected cohorts with lower expressions of SPOP substrates. In our study, in patients with mutant SPOP, the mutation was associated with decreased SPOP substrate expression and androgen receptor signaling, raising concern for suboptimal efficacy of androgen deprivation therapy in this subset of patients.
Collapse
Affiliation(s)
- Samuel Bidot
- From the Departments of Pathology and Laboratory Medicine (Bidot, Zhang, Deeb, Smith, Hill, Tinsley, Harik)
| | - Jun Yin
- Department of Clinical and Translational Research, Caris Life Sciences, Phoenix, Arizona (Yin, Xiu)
| | - Pengbo Zhou
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York, New York (Zhou)
| | - Linsheng Zhang
- From the Departments of Pathology and Laboratory Medicine (Bidot, Zhang, Deeb, Smith, Hill, Tinsley, Harik)
| | - Kristin K Deeb
- From the Departments of Pathology and Laboratory Medicine (Bidot, Zhang, Deeb, Smith, Hill, Tinsley, Harik)
| | - Geoffrey Smith
- From the Departments of Pathology and Laboratory Medicine (Bidot, Zhang, Deeb, Smith, Hill, Tinsley, Harik)
| | - Charles E Hill
- From the Departments of Pathology and Laboratory Medicine (Bidot, Zhang, Deeb, Smith, Hill, Tinsley, Harik)
| | - Joanne Xiu
- Department of Clinical and Translational Research, Caris Life Sciences, Phoenix, Arizona (Yin, Xiu)
| | - Mehmet A Bilen
- Hematology and Oncology (Bilen, Carthon)
- Winship Cancer Institute of Emory University, Atlanta, GA (Bilen, Harik)
| | | | - Mazie Tinsley
- From the Departments of Pathology and Laboratory Medicine (Bidot, Zhang, Deeb, Smith, Hill, Tinsley, Harik)
| | | | - Lara R Harik
- From the Departments of Pathology and Laboratory Medicine (Bidot, Zhang, Deeb, Smith, Hill, Tinsley, Harik)
- Winship Cancer Institute of Emory University, Atlanta, GA (Bilen, Harik)
| |
Collapse
|
9
|
Varma M, Dormer J. Macroscopy of specimens from the genitourinary system. J Clin Pathol 2024; 77:177-183. [PMID: 38373783 DOI: 10.1136/jcp-2023-208831] [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: 08/03/2023] [Accepted: 09/12/2023] [Indexed: 02/21/2024]
Abstract
Macroscopic specimen examination is often critical for accurate histopathology reporting but has generally received insufficient attention and may be delegated to inexperienced staff with limited guidance and supervision. This review discusses issues around macroscopic examination of some common urological specimens; highlighting findings that are critical for patient management and others that are clinically irrelevant. Macroscopic findings are of limited value in completely submitted radical prostatectomy specimens but may be critical in orchidectomy specimens where identification of focal non-seminomatous components can significantly impact patient management. The maximum tumour dimension is often an important prognostic indicator, but specimen dimensions are generally of little clinical utility. Specimens should be carefully examined and judiciously sampled to identify clinically important focal abnormalities such as sarcomatoid change in a renal cell carcinoma and a minor non-seminomatous component in a predominant testicular seminoma. Meticulous macroscopic examination is key as less than 0.2% of the specimen (or macroscopically abnormal area) would be histologically examined even if the entire specimen/abnormal area is submitted for microscopic examination. Retroperitoneal pelvic lymph node dissection specimens for testicular cancer must be handled very differently from other lymph nodal block dissections. Current sampling protocols for transurethral resection of prostate specimens that are based on pre-MRI era data need to be reconsidered because they were specifically designed to detect occult prostate cancer, which would amount to histological cancer screening. Prostatic sampling of cystoprostatectomy specimens should be directed at accurately staging the known bladder cancer rather than detection of incidental prostate cancer.
Collapse
Affiliation(s)
- Murali Varma
- Cellular Pathology, University Hospital of Wales, Cardiff, UK
| | - John Dormer
- Cellular Pathology, University Hospitals of Leicester NHS Trust, Leicester, UK
| |
Collapse
|
10
|
Schouten D, van der Laak J, van Ginneken B, Litjens G. Full resolution reconstruction of whole-mount sections from digitized individual tissue fragments. Sci Rep 2024; 14:1497. [PMID: 38233535 PMCID: PMC10794243 DOI: 10.1038/s41598-024-52007-5] [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: 11/14/2023] [Accepted: 01/12/2024] [Indexed: 01/19/2024] Open
Abstract
Whole-mount sectioning is a technique in histopathology where a full slice of tissue, such as a transversal cross-section of a prostate specimen, is prepared on a large microscope slide without further sectioning into smaller fragments. Although this technique can offer improved correlation with pre-operative imaging and is paramount for multimodal research, it is not commonly employed due to its technical difficulty, associated cost and cumbersome integration in (digital) pathology workflows. In this work, we present a computational tool named PythoStitcher which reconstructs artificial whole-mount sections from digitized tissue fragments, thereby bringing the benefits of whole-mount sections to pathology labs currently unable to employ this technique. Our proposed algorithm consists of a multi-step approach where it (i) automatically determines how fragments need to be reassembled, (ii) iteratively optimizes the stitch using a genetic algorithm and (iii) efficiently reconstructs the final artificial whole-mount section on full resolution (0.25 µm/pixel). PythoStitcher was validated on a total of 198 cases spanning five datasets with a varying number of tissue fragments originating from different organs from multiple centers. PythoStitcher successfully reconstructed the whole-mount section in 86-100% of cases for a given dataset with a residual registration mismatch of 0.65-2.76 mm on automatically selected landmarks. It is expected that our algorithm can aid pathology labs unable to employ whole-mount sectioning through faster clinical case evaluation and improved radiology-pathology correlation workflows.
Collapse
Affiliation(s)
- Daan Schouten
- Department of Pathology, Radboud University Medical Centre, Nijmegen, The Netherlands.
| | - Jeroen van der Laak
- Department of Pathology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Bram van Ginneken
- Department of Radiology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Geert Litjens
- Department of Pathology, Radboud University Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
11
|
Pedraza AM, Gupta R, Musheyev D, Pino T, Shah A, Brody R, Wagaskar V, Kaufmann B, Gorin MA, Menon M, Tewari A. Microultrasound in the detection of the index lesion in prostate cancer. Prostate 2024; 84:79-86. [PMID: 37828815 DOI: 10.1002/pros.24628] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 08/20/2023] [Accepted: 09/19/2023] [Indexed: 10/14/2023]
Abstract
INTRODUCTION AND OBJECTIVE The natural progression of prostate cancer is primarily driven by an index lesion (IL). Studies have shown that different metastases within the same patient arise from a single precursor cell. Therefore, our aim is to assess the effectiveness of transrectal microultrasound (MUS) in comparison to multiparametric magnetic resonance imaging (mpMRI) for detecting the IL in prostate cancer. We used quarter-mount pathological results as the reference standard for this evaluation. MATERIALS AND METHODS Three hundred and sixty-three patients who underwent Robot-Assisted Radical Prostatectomy (RARP) from June 2021 to August 2022 were included. All received mpMRI and MUS before RARP. MUS was performed by experienced operators blinded to mpMRI and biopsy results. The IL in the radical prostatectomy specimen was defined as the lesion with extraprostatic extension, the highest Grade Group (GG), or the largest tumor volume if the GG was the same. The correlation between imaging and final pathology findings was performed. A descriptive statistical analysis is presented. RESULTS The patients' prostates were analyzed in 12 regions (anterior/posterior, right/left, apex/mid/base). A total of 4308 regions were identified. Of these, 935 were involved by the ILs. Compared with final pathology, MUS demonstrated a sensitivity, specificity, PPV, and NPV of 68.7%, 96.3%, 80.8%, and 93.1%, respectively, while mpMRI showed a sensitivity, specificity, PPV, and NPV of 68.6%, 97.2%, 86.1%, and 92.5%, respectively, for the detection of the IL. Most of the lesions missed by MUS were located in the anterior zone (62%). CONCLUSION MUS exhibits a diagnostic performance similar to mpMRI when it comes to detecting the IL in prostate cancer. MUS is a cost-effective option, offers real-time evaluation, and has no delay in the acquisition process.
Collapse
Affiliation(s)
- Adriana M Pedraza
- Department of Urologic Surgery, The Mount Sinai Hospital, Icahn School of Medicine, New York, New York, USA
| | - Raghav Gupta
- Department of Urologic Surgery, The Mount Sinai Hospital, Icahn School of Medicine, New York, New York, USA
| | - David Musheyev
- Department of Urologic Surgery, The Mount Sinai Hospital, Icahn School of Medicine, New York, New York, USA
| | - Tanisha Pino
- Department of Urologic Surgery, The Mount Sinai Hospital, Icahn School of Medicine, New York, New York, USA
| | - Akash Shah
- Department of Uro-oncologic Surgery, Kokilaben Dhirubhai Ambani Hospital, Four Bungalows, Mumbai, India
| | - Rachel Brody
- Department of Pathology, The Mount Sinai Hospital, Icahn School of Medicine, New York, New York, USA
| | - Vinayak Wagaskar
- Department of Urologic Surgery, The Mount Sinai Hospital, Icahn School of Medicine, New York, New York, USA
| | - Basil Kaufmann
- Department of Urologic Surgery, The Mount Sinai Hospital, Icahn School of Medicine, New York, New York, USA
| | - Michael A Gorin
- Department of Urologic Surgery, The Mount Sinai Hospital, Icahn School of Medicine, New York, New York, USA
| | - Mani Menon
- Department of Urologic Surgery, The Mount Sinai Hospital, Icahn School of Medicine, New York, New York, USA
| | - Ashutosh Tewari
- Department of Urologic Surgery, The Mount Sinai Hospital, Icahn School of Medicine, New York, New York, USA
| |
Collapse
|
12
|
Fan B, Zhang L, Wang Y, Dai Z, Pan H, Xie J, Wang H, Xin Z, Wang Y, Duan X, Luo J, Wang L, Liu Z. Value of three-dimensional visualization of preoperative prostatic magnetic resonance imaging based on measurements of anatomical structures in predicting positive surgical margin after radical prostatectomy. Front Endocrinol (Lausanne) 2023; 14:1228892. [PMID: 37859989 PMCID: PMC10582708 DOI: 10.3389/fendo.2023.1228892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 09/13/2023] [Indexed: 10/21/2023] Open
Abstract
Background Positive surgical margin (PSM) or apical positive surgical margin (APSM) is an established predictive factor of biochemical recurrence or disease progression in prostate cancer (PCa) patients after radical prostatectomy. Since there are limited usable magnetic resonance imaging (MRI)-based models, we sought to explore the role of three-dimensional (3D) visualization for preoperative MRI in the prediction of PSM or APSM. Methods From December 2016 to April 2022, 149 consecutive PCa patients who underwent radical prostatectomy were retrospectively selected from the Second Affiliated Hospital of Dalian Medical University. According to the presence of PSM or APSM, patients were divided into a PSM group (n=41) and a without PSM group (n=108) and into an APSM group (n=33) and a without APSM group (n=116). Twenty-one parameters, including prostate apical shape, PCa distance to the membranous urethra, and pubic angle, were measured on 3D visualization of MRI. The development of the nomogram models was built by the findings of multivariate logistic regression analysis for significant factors. Results To predict the probability of PSM, a longer PCa distance to the membranous urethra (OR=0.136, p=0.019) and the distance from the anterior peritoneum to the anterior border of the coccyx (work space AP, OR=0.240, p=0.030) were independent protective factors, while a type 3 prostate apical shape (OR=8.262, p=0.025) and larger pubic angle 2 (OR=5.303, p=0.029) were identified as independent risk factors. The nomogram model presented an area under the curve (AUC) of the receiver operating characteristic curve (ROC) of PSM of 0.777. In evaluating the incidence of APSM, we found that the distance to the membranous urethra (OR=0.135, p=0.014) was associated with a low risk of APSM, while larger pubic angle 1 (OR=4.666, p=0.043) was connected to a higher risk of APSM. The nomogram model showed that the AUC of APSM was 0.755. Conclusion As 3D visualization for preoperative MRI showed good performance in predicting PSM or APSM, the tool might be potentially valuable, which also needs to be validated by multicenter, large-scale, prospective studies.
Collapse
Affiliation(s)
- Bo Fan
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, the Liaoning Provincial Department of Science and Technology, Dalian, Liaoning, China
- Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Liaoning Provincial Development and Reform Commission, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Dalian Science and Technology Bureau, Dalian, Liaoning, China
| | - Luxin Zhang
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, the Liaoning Provincial Department of Science and Technology, Dalian, Liaoning, China
- Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Liaoning Provincial Development and Reform Commission, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Dalian Science and Technology Bureau, Dalian, Liaoning, China
| | - Yuchao Wang
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, the Liaoning Provincial Department of Science and Technology, Dalian, Liaoning, China
- Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Liaoning Provincial Development and Reform Commission, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Dalian Science and Technology Bureau, Dalian, Liaoning, China
| | - Zhihong Dai
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, the Liaoning Provincial Department of Science and Technology, Dalian, Liaoning, China
- Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Liaoning Provincial Development and Reform Commission, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Dalian Science and Technology Bureau, Dalian, Liaoning, China
| | - Heming Pan
- Department of Scientific Research, Dalian Neusoft University of Information, Dalian, Liaoning, China
| | - Jiaxin Xie
- Institute of Urology, Peking University, Beijing, China
| | - Hao Wang
- Department of Clinical Medicine, First Clinical School of Dalian Medical University, Dalian, Liaoning, China
| | - Zihan Xin
- Department of Clinical Medicine, First Clinical School of Dalian Medical University, Dalian, Liaoning, China
| | - Yutong Wang
- Department of Clinical Medicine, First Clinical School of Dalian Medical University, Dalian, Liaoning, China
| | - Xu Duan
- Department of Clinical Medicine, First Clinical School of Dalian Medical University, Dalian, Liaoning, China
| | - Jiawen Luo
- Department of Radiology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Liang Wang
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, the Liaoning Provincial Department of Science and Technology, Dalian, Liaoning, China
- Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Liaoning Provincial Development and Reform Commission, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Dalian Science and Technology Bureau, Dalian, Liaoning, China
| | - Zhiyu Liu
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Liaoning Provincial Key Laboratory of Urological Digital Precision Diagnosis and Treatment, the Liaoning Provincial Department of Science and Technology, Dalian, Liaoning, China
- Liaoning Engineering Research Center of Integrated Precision Diagnosis and Treatment Technology for Urological Cancer, Liaoning Provincial Development and Reform Commission, Dalian, Liaoning, China
- Dalian Key Laboratory of Prostate Cancer Research, Dalian Science and Technology Bureau, Dalian, Liaoning, China
| |
Collapse
|
13
|
Rao BV, Soni S, Kulkarni B, Bindhu MR, Ambekar A, Midha D, Kaushal S, Patil S, Jagdale R, Sundaram S, Kumar RM, Desai S, Menon S. Grossing and reporting of radical prostatectomy specimens: An evidence-based approach. Indian J Cancer 2023; 60:449-457. [PMID: 38155443 DOI: 10.4103/ijc.ijc_1550_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 09/05/2022] [Indexed: 12/30/2023]
Abstract
Radical prostatectomy (RP) constitutes the primary treatment option for patients with clinically localized, biopsy-proven prostate cancer that requires local treatment with curative intent. Accurate reporting of radical prostatectomy specimens is required to guide further risk stratification and management of patients. Hence, for the handling and reporting of RP specimens, a standardized protocol should be followed. Many general pathologists may not be well-versed with the guidelines for the handling of radical prostatectomy specimens. This article discusses a detailed approach to grossing techniques, including specimen description, fixation requirements, gross cut-up, and reporting of the grade and stage of RP specimens. This will enable the pathologist to aid in multidisciplinary management.
Collapse
Affiliation(s)
- B Vishal Rao
- Basavatarakam Indo American Cancer Hospital and Research Institute, Hyderabad, Telangana, India
| | - Shailesh Soni
- Muljibhai Patel Urological Hospital, Nadiad, Gujarat, India
| | - Bijal Kulkarni
- Kokilaben Dhirubhai Ambani Hospital and Research Centre, Mumbai, Maharashtra, India
| | - M R Bindhu
- Amrita Institute of Medical Sciences, Kochi, Kerala, India
| | | | - Divya Midha
- Tata Medical Centre Kolkata, West Bengal, India
| | | | - Sachin Patil
- Shri Siddhivinayak Ganapati Cancer Hospital, Miraj, Maharashtra, India
| | - Rakhi Jagdale
- Shri Siddhivinayak Ganapati Cancer Hospital, Miraj, Maharashtra, India
| | - Sandhya Sundaram
- Sri Ramachandra Institute of Higher Education and Research, Chennai, Tamil Nadu, India
| | | | - Sangeeta Desai
- Department of Pathology, Tata Medical Centre, Mumbai, Maharashtra, India
| | - Santosh Menon
- Department of Pathology, Tata Medical Centre, Mumbai, Maharashtra, India
| |
Collapse
|
14
|
Heetman JG, Hermsen R, Exterkate L, Küsters-Vandevelde HVN, Brouwer LJM, Somford DM, van den Bergh RCN, van Basten JPA. Immunohistochemical and histopathological validation of 18 F-PSMA-1007 PET/CT for intraprostatic cancerous lesions. Prostate 2023; 83:1332-1341. [PMID: 37455399 DOI: 10.1002/pros.24595] [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: 02/23/2023] [Revised: 06/02/2023] [Accepted: 06/21/2023] [Indexed: 07/18/2023]
Abstract
INTRODUCTION Prostate-specific membrane antigen (PSMA) is overexpressed in prostate cancer (PCa). In this study, we aim to immunohistochemically and histopathological validate the fluorine-18 (18 F)-PSMA-1007 positron emission tomography/computed tomography (PET/CT) for intraprostatic PCa lesions. METHODS Between February 2019 and October 2020, patients with biopsy-proven, treatment-naïve intermediate-to-high-risk PCa undergoing an 18 F-PSMA-1007 PET/CT before robot-assisted radical prostatectomy (RARP) were prospectively enrolled. For all PCa lesions found on whole-mount histopathology, location, size, International Society of Urological Pathology (ISUP) grade group (GG), and immune reactive score (IRS) were assessed after PSMA staining. ISUP GG ≥ 3 PCa was defined as clinically significant (cs) PCa. All lesions were matched on PSMA PET/CT and the maximum standardized uptake value (SUVmax) was measured. RESULTS A total of 125 lesions were analyzed in the 80 RARP specimens, of which 49 (40%) were csPCa and 76 (60%) non-csPCa. Linear multivariable regressions showed that an increase in SUVmax significantly correlated with a higher ISUP GG (p values between 0.021 and 0.001) and a higher IRS (p = 0.017). Logistic multivariable regression showed that csPCa significantly correlated with a higher SUVmax (odds ratio, OR: 1.17 [95% confidence interval, CI: 1.04-1.21, p = 0.005]), an increase in tumor length (OR: 1.05 [95% CI 1.01-1.10, p = 0.020]) and a higher IRS (OR; 1.24 [95% CI 1.07-1.47, p = 0.006]). A SUVmax threshold of 4 would have resulted in one (2%) missed lesion with csPCa. CONCLUSION This prospective study revealed that 18 F-PSMA-1007 PET/CT SUVmax is correlated with the ISUP GG and IRS, and thereby could be a tool to characterize intraprostatic PCa lesions.
Collapse
Affiliation(s)
- Joris G Heetman
- Department of Urology, Sint Antonius Hospital, Utrecht-Nieuwegein, The Netherlands
| | - Rick Hermsen
- Department of Nuclear Medicine, Canisius-Wilhelmina Hospital, Nijmegen, The Netherlands
| | - Leonie Exterkate
- Department of Urology, Canisius-Wilhelmina Hospital, Prosper Prostate Cancer Clinics, Nijmegen/Eindhoven, The Netherlands
| | | | - Lenneke J M Brouwer
- Department of Urology, Canisius-Wilhelmina Hospital, Prosper Prostate Cancer Clinics, Nijmegen/Eindhoven, The Netherlands
| | - Diederik M Somford
- Department of Urology, Canisius-Wilhelmina Hospital, Prosper Prostate Cancer Clinics, Nijmegen/Eindhoven, The Netherlands
| | | | - Jean-Paul A van Basten
- Department of Urology, Canisius-Wilhelmina Hospital, Prosper Prostate Cancer Clinics, Nijmegen/Eindhoven, The Netherlands
| |
Collapse
|
15
|
Montironi R, Cimadamore A, Mazzucchelli R, Lopez-Beltran A, Scarpelli M, Cheng L. Histopathology of Prostate Cancer and its Precursors. Appl Immunohistochem Mol Morphol 2023; 31:467-477. [PMID: 36222497 DOI: 10.1097/pai.0000000000001067] [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] [Received: 07/13/2021] [Accepted: 08/19/2022] [Indexed: 06/16/2023]
Abstract
Starting in the mid-1970s, we formed a group of pathologists with a major interest in uropathology. Originally, it included 2 (R.M. and M.S.). In the years the followed, the group was enlarged to include 4 more people, 2 in the mid- and late-1980s (A.L.B. and L.C.) and another in the mid-1990s (R.Ma.); a sixth (A.C.) joined the group ∼5 years ago. Two have reached the retirement age (R.M. and M.S.), while others are in the process of joining the group to replace them. A fruitful collaboration spanned for ∼45 years. This contribution is based on a series of personal recollections of the successive changes in the interpretation of prostate cancer and its precursors, starting in the mid-1970s. Here we have retraced our involvement steps, sharing issues related to them with a junior uropathologist (A.C.).
Collapse
Affiliation(s)
- Rodolfo Montironi
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - Alessia Cimadamore
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - Roberta Mazzucchelli
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - Antonio Lopez-Beltran
- Department of Morphological Sciences, Cordoba University Medical School, Cordoba, Spain
| | - Marina Scarpelli
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
16
|
Korpás KL, Beke L, Varga D, Bidiga L, Méhes G, Molnár S. Grade Group accuracy is improved by extensive prostate biopsy sampling, but unrelated to prostatectomy specimen sampling or use of immunohistochemistry. Pathol Oncol Res 2023; 29:1611157. [PMID: 37415848 PMCID: PMC10319996 DOI: 10.3389/pore.2023.1611157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 06/12/2023] [Indexed: 07/08/2023]
Abstract
Assessing the accurate Grade Group of a prostate needle biopsy specimen is essential for choosing the adequate therapeutic modality for prostate cancer patients. However, it is well-known that biopsy Grade Group tends to up- or downgrade significantly at radical prostatectomy. We aimed to investigate the correlation between accuracy and biopsy core number, performed immunohistochemical staining (IHC) or prostatectomy specimen sampling, with the latest also being correlated with higher detection rates of adverse pathological features, e.g., positive surgical margins, higher pathological stage or presence of perineural invasion (PnI status). The study cohort consisted of 315 consecutive patients diagnosed with prostate adenocarcinoma via transrectal ultrasound-guided needle biopsy who later underwent radical prostatectomy. We grouped and compared patients based on Grade Group accuracy, presence of IHC on biopsy, margin status, pathological stage, and PnI status. Inter-observer reproducibility was also calculated. Statistical analyzes included ANOVA, Tukey's multiple comparisons post hoc test, Chi-squared test, and Fleiss kappa statistics. Undergraded cases harboured a significantly lower number of biopsy cores (p < 0.05), than accurately graded cases. Using IHC did not affect grading accuracy significantly, nor did the number of slides from prostatectomy specimens. The mean number of slides was virtually identical when margin status, pathological stage and PnI status of prostatectomy specimens were compared. Inter-observer reproducibility at our institute was calculated as fair (overall kappa = 0.29). Grade Group accuracy is significantly improved by obtaining more cores at biopsy but is unrelated to performed IHC. The extent of sampling prostatectomy specimens, however, did not affect accuracy and failed to significantly improve detection of adverse pathological features.
Collapse
Affiliation(s)
| | - Lívia Beke
- Department of Pathology, Clinical Centre, University of Debrecen, Debrecen, Hungary
| | - Dániel Varga
- Department of Urology, Clinical Centre, University of Debrecen, Debrecen, Hungary
| | - László Bidiga
- Department of Pathology, Clinical Centre, University of Debrecen, Debrecen, Hungary
| | - Gábor Méhes
- Department of Pathology, Clinical Centre, University of Debrecen, Debrecen, Hungary
| | - Sarolta Molnár
- Department of Pathology, Clinical Centre, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
17
|
Karagoz A, Alis D, Seker ME, Zeybel G, Yergin M, Oksuz I, Karaarslan E. Anatomically guided self-adapting deep neural network for clinically significant prostate cancer detection on bi-parametric MRI: a multi-center study. Insights Imaging 2023; 14:110. [PMID: 37337101 DOI: 10.1186/s13244-023-01439-0] [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: 01/24/2023] [Accepted: 04/17/2023] [Indexed: 06/21/2023] Open
Abstract
OBJECTIVE To evaluate the effectiveness of a self-adapting deep network, trained on large-scale bi-parametric MRI data, in detecting clinically significant prostate cancer (csPCa) in external multi-center data from men of diverse demographics; to investigate the advantages of transfer learning. METHODS We used two samples: (i) Publicly available multi-center and multi-vendor Prostate Imaging: Cancer AI (PI-CAI) training data, consisting of 1500 bi-parametric MRI scans, along with its unseen validation and testing samples; (ii) In-house multi-center testing and transfer learning data, comprising 1036 and 200 bi-parametric MRI scans. We trained a self-adapting 3D nnU-Net model using probabilistic prostate masks on the PI-CAI data and evaluated its performance on the hidden validation and testing samples and the in-house data with and without transfer learning. We used the area under the receiver operating characteristic (AUROC) curve to evaluate patient-level performance in detecting csPCa. RESULTS The PI-CAI training data had 425 scans with csPCa, while the in-house testing and fine-tuning data had 288 and 50 scans with csPCa, respectively. The nnU-Net model achieved an AUROC of 0.888 and 0.889 on the hidden validation and testing data. The model performed with an AUROC of 0.886 on the in-house testing data, with a slight decrease in performance to 0.870 using transfer learning. CONCLUSIONS The state-of-the-art deep learning method using prostate masks trained on large-scale bi-parametric MRI data provides high performance in detecting csPCa in internal and external testing data with different characteristics, demonstrating the robustness and generalizability of deep learning within and across datasets. CLINICAL RELEVANCE STATEMENT A self-adapting deep network, utilizing prostate masks and trained on large-scale bi-parametric MRI data, is effective in accurately detecting clinically significant prostate cancer across diverse datasets, highlighting the potential of deep learning methods for improving prostate cancer detection in clinical practice.
Collapse
Affiliation(s)
- Ahmet Karagoz
- Department of Computer Engineering, Istanbul Technical University, Istanbul, Turkey
- Artificial Intelligence and Information Technologies, Hevi AI Health, Istanbul, Turkey
| | - Deniz Alis
- Artificial Intelligence and Information Technologies, Hevi AI Health, Istanbul, Turkey.
- Department of Radiology, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey.
| | - Mustafa Ege Seker
- School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Gokberk Zeybel
- School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Mert Yergin
- Artificial Intelligence and Information Technologies, Hevi AI Health, Istanbul, Turkey
| | - Ilkay Oksuz
- Department of Computer Engineering, Istanbul Technical University, Istanbul, Turkey
| | - Ercan Karaarslan
- Department of Radiology, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| |
Collapse
|
18
|
Galosi AB, Milanese G, Montesi L, Cimadamore A, Franzese C, Palagonia E, Chiacchio G, Castellani D. The pathway of isolated seminal vesicle invasion has a different impact on biochemical recurrence after radical prostatectomy and pelvic lymphadenectomy. Urol Oncol 2023:S1078-1439(23)00095-9. [PMID: 37142452 DOI: 10.1016/j.urolonc.2023.03.011] [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: 11/24/2022] [Revised: 01/20/2023] [Accepted: 03/20/2023] [Indexed: 05/06/2023]
Abstract
OBJECTIVE Prostate cancer with seminal vesicle invasion (SVI) has been considered an aggressive cancer. To evaluate the prognostic significance of different patterns of isolated SVI in patients undergoing radical prostatectomy (RP) and pelvic lymphadenectomy. METHODS AND MATERIALS We retrospectively analyzed all patients who underwent RP between 2007 and 2019. Inclusion criteria were localized prostate adenocarcinoma, SVI at RP, at least 24-months follow-up, and no adjuvant treatment. Patterns of SVI were following Ohori's classification: type 1: direct spread along the ejaculatory duct from inside; type 2: seminal vesicle invasion outside the prostate, through the capsule; type 3: the presence of cancer island(s) in the seminal vesicle with no continuity from the primary tumor (discontinuous metastases). Patients with type 3 SVI (isolated or in association) were included in the same group. Biochemical recurrence (BCR) was defined as any postoperative PSA ≥0.2 ng/ml. A logistic regression analysis was performed to assess predictors of BCR. Time to BCR was investigated using the Kaplan-Meier analysis with the log-rank test. RESULTS Sixty-one out of 1,356 patients were included. Median age was 67(7.2) years. Median PSA was 9.4(8.92) ng/ml. Mean follow-up was 85.28 ± 45.27 months. BCR occurred in 28(45.9%) patients. Logistic regression showed that a positive surgical margin (OR 19.964, 95%CI:1.172-29.322, P = 0.038) was predictor of BCR. Kaplan-Meier analysis demonstrated that patients with pattern 3 had a significantly shorter time to BCR compared to other groups (log-rank, P = 0.016). The estimated time to BCR was 48.7 months in type 3, 60.9 months in pattern 1 + 2, 74.8, and 100.8 months in isolated patterns 1 and 2, respectively. In patients with negative surgical margins, pattern 3 confirmed a shorter time to BCR compared to other types of invasions, with an estimated time to BCR of 30.8 months. CONCLUSIONS Patients with type 3 SVI demonstrated a shorter time to BCR compared to other patterns.
Collapse
Affiliation(s)
- Andrea B Galosi
- Urology Division, Azienda Ospedaliero-Universitaria delle Marche, School of Urology, Polytechnic University of the Marche Region, Ancona, Italy
| | - Giulio Milanese
- Urology Division, Azienda Ospedaliero-Universitaria delle Marche, School of Urology, Polytechnic University of the Marche Region, Ancona, Italy
| | - Lorenzo Montesi
- Urology Division, Azienda Ospedaliero-Universitaria delle Marche, School of Urology, Polytechnic University of the Marche Region, Ancona, Italy
| | - Alessia Cimadamore
- Section of Pathological Anatomy, Azienda Ospedaliero-Universitaria delle Marche, Marche Polytechnic University, Ancona, Italy
| | - Carmine Franzese
- Urology Division, Azienda Ospedaliero-Universitaria delle Marche, School of Urology, Polytechnic University of the Marche Region, Ancona, Italy
| | - Erika Palagonia
- Urology Division, Azienda Ospedaliero-Universitaria delle Marche, School of Urology, Polytechnic University of the Marche Region, Ancona, Italy
| | - Giuseppe Chiacchio
- Urology Division, Azienda Ospedaliero-Universitaria delle Marche, School of Urology, Polytechnic University of the Marche Region, Ancona, Italy
| | - Daniele Castellani
- Urology Division, Azienda Ospedaliero-Universitaria delle Marche, School of Urology, Polytechnic University of the Marche Region, Ancona, Italy.
| |
Collapse
|
19
|
Ren J, Melamed J, Taneja SS, Wysock JS, Huang WC, Lepor H, Deng FM. Prostate magnetic resonance imaging-targeted biopsy global grade correlates better than highest grade with prostatectomy grade. Prostate 2023; 83:323-330. [PMID: 36461793 DOI: 10.1002/pros.24464] [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: 05/16/2022] [Revised: 10/31/2022] [Accepted: 11/21/2022] [Indexed: 12/05/2022]
Abstract
BACKGROUND Magnetic resonance imaging (MRI)-targeted prostate biopsy has become an increasingly common method of diagnosing prostate cancer. A previous study from our institution demonstrated that the biopsy global Grade Group (gGG, aggregate GG of all positive cores) and highest Grade Group (hGG in any core) both show substantial concordance with the Grade Group at radical prostatectomy (RPGG) while the discordance predominantly consists of upgrading in gGG and downgrading in hGG. We performed a larger cohort study focused on biopsy cases in which gGG and hGG differ, to determine their relative concordance with RPGG. METHODS We conducted a retrospective review of radical prostatectomy specimens with prior MRI-targeted biopsies from our institution between 2016 and 2020. Separate gGG and hGG were assigned to each MRI-targeted lesion. Targeted lesions with different gGG versus hGG were segregated from those with identical gGG and hGG. The concordance of biopsy GG with RPGG was evaluated using κ coefficient analysis. RESULTS Of the 489 lesions with MRI-targeted biopsies, 82 (17%) differed in gGG versus hGG. The gGG of 46 (56%), 33 (40%), and 3 (4%) lesions were unchanged, upgraded, and downgraded at radical prostatectomy, respectively (κ= 0.302, weighted κ = 0.334). The hGG of 24 (29%), 9 (11%), and 49 (60%) lesions were unchanged, upgraded, and downgraded at radical prostatectomy, respectively (κ = 0.040, weighted κ = 0.198). When stratified by the biopsy GG, gGG showed the highest concordance in GG2 (61%) and GG3 (54%) lesions. The hGG resulted in substantial downgrading (60%) with less optimal concordance regardless of the biopsy GG. Neither the prebiopsy prostate specific antigen level nor the PI-RADS score was predictive of upgrading of gGG. CONCLUSIONS When gGG and hGG differ, gGG method more accurately predicts the RPGG than hGG, particularly in GG2 and GG3 lesions which comprised the majority of targeted lesions.
Collapse
Affiliation(s)
- Joyce Ren
- Department of Pathology, New York University Langone School of Medicine, New York, New York, USA
| | - Jonathan Melamed
- Department of Pathology, New York University Langone School of Medicine, New York, New York, USA
| | - Samir S Taneja
- Department of Urology, New York University Langone School of Medicine, New York, New York, USA
| | - James S Wysock
- Department of Urology, New York University Langone School of Medicine, New York, New York, USA
| | - William C Huang
- Department of Urology, New York University Langone School of Medicine, New York, New York, USA
| | - Herbert Lepor
- Department of Urology, New York University Langone School of Medicine, New York, New York, USA
| | - Fang-Ming Deng
- Department of Pathology, New York University Langone School of Medicine, New York, New York, USA
- Department of Urology, New York University Langone School of Medicine, New York, New York, USA
| |
Collapse
|
20
|
Huang C, He S, He Q, Gong Y, Song G, Zhou L. Determination of Whether Apex or Non-Apex Prostate Cancer Is the Best Candidate for the Use of Prostate-Specific Antigen Density to Predict Pathological Grade Group Upgrading and Upstaging after Radical Prostatectomy. J Clin Med 2023; 12:jcm12041659. [PMID: 36836195 PMCID: PMC9967179 DOI: 10.3390/jcm12041659] [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: 01/03/2023] [Revised: 02/12/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
Objective: Previous studies have demonstrated that prostate-specific antigen density (PSAD) may aid in predicting Gleason grade group (GG) upgrading and pathological upstaging in patients with prostate cancer (PCa). However, the differences and associations between patients with apex prostate cancer (APCa) and non-apex prostate cancer (NAPCa) have not been described. The aim of this study was to explore the different roles of PSAD in predicting GG upgrading and pathological upstaging between APCa and NAPCa. Patients and Methods: Five hundred and thirty-five patients who underwent prostate biopsy followed by radical prostatectomy (RP) were enrolled. All patients were diagnosed with PCa and classified as either APCa or NAPCa. Clinical and pathological variables were collected. Univariate, multivariate, and receiver operating characteristic (ROC) analyses were performed. Results: Of the entire cohort, 245 patients (45.8%) had GG upgrading. Multivariate analysis revealed that only PSAD (odds ratio [OR]: 4.149, p < 0.001) was an independent, significant predictor of upgrading. A total of 262 patients (49.0%) had pathological upstaging. Both PSAD (OR: 4.750, p < 0.001) and percentage of positive cores (OR: 5.108, p = 0.002) were independently significant predictors of upstaging. Of the 374 patients with NAPCa, 168 (44.9%) displayed GG upgrading. Multivariate analysis also showed PSAD (OR: 8.176, p < 0.001) was an independent predictor of upgrading. Upstaging occurred in 159 (42.5%) patients with NAPCa, and PSAD (OR: 4.973, p < 0.001) and percentage of positive cores (OR: 3.994, p = 0.034) were independently predictive of pathological upstaging. Conversely, of the 161 patients with APCa, 77 (47.8%) were identified with GG upgrading, and 103 (64.0%) patients with pathological upstaging. Multivariate analysis demonstrated that there were no significant predictors, including PSAD, for predicting GG upgrading (p = 0.462) and pathological upstaging (p = 0.100). Conclusions: PSAD may aid in the prediction of GG upgrading and pathological upstaging in patients with PCa. However, this may only be practical in patients with NAPCa but not with APCa. Additional biopsy cores taken from the prostatic apex region may help improve the accuracy of PSAD in predicting GG upgrading and pathological upstaging after RP.
Collapse
Affiliation(s)
- Cong Huang
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Institute of Urology, Peking University, National Urological Cancer Center of China, Beijing 100034, China
| | - Shiming He
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Institute of Urology, Peking University, National Urological Cancer Center of China, Beijing 100034, China
| | - Qun He
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Institute of Urology, Peking University, National Urological Cancer Center of China, Beijing 100034, China
| | - Yanqing Gong
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Institute of Urology, Peking University, National Urological Cancer Center of China, Beijing 100034, China
| | - Gang Song
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Institute of Urology, Peking University, National Urological Cancer Center of China, Beijing 100034, China
- Correspondence: (G.S.); (L.Z.)
| | - Liqun Zhou
- Department of Urology, Peking University First Hospital, Beijing 100034, China
- Institute of Urology, Peking University, National Urological Cancer Center of China, Beijing 100034, China
- Correspondence: (G.S.); (L.Z.)
| |
Collapse
|
21
|
Novikov FV, Tesljuk OS, Efremov GD. [Comparison of the efficiency of surgical excision methods for detecting extraprostatic extension and positive resection margin in prostate cancer]. Arkh Patol 2023; 85:48-51. [PMID: 37814850 DOI: 10.17116/patol20238505148] [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: 10/11/2023]
Abstract
On the samples of 26 prostatectomies, the method of excision of the prostate gland according to Kim was tested. This method increased the number of blocks by 30.2% and increased the detectability of extraprostatic extension by 41.7% and positive surgical margin by 40.0% compared to the method of alternate prostate sections. Also, the method according to Kim reduced the number of blocks of prostate tissue by 34.3% compared to the method of complete prostate excision.
Collapse
Affiliation(s)
- F V Novikov
- N. Lopatkin Scientific Research Institute of Urology and Interventional Radiology - branch of the National Medical Research Radiological Center, Moscow, Russia
| | - O S Tesljuk
- N. Lopatkin Scientific Research Institute of Urology and Interventional Radiology - branch of the National Medical Research Radiological Center, Moscow, Russia
| | - G D Efremov
- N. Lopatkin Scientific Research Institute of Urology and Interventional Radiology - branch of the National Medical Research Radiological Center, Moscow, Russia
| |
Collapse
|
22
|
Preoperative Multiparametric Prostate Magnetic Resonance Imaging Structured Report Informs Risk for Positive Apical Surgical Margins During Radical Prostatectomy. J Comput Assist Tomogr 2023; 47:38-44. [PMID: 35995580 DOI: 10.1097/rct.0000000000001377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND The prostatic apex is the most frequent location of positive surgical margin (PSM) after surgery. Data regarding the ability of multiparametric magnetic resonance imaging (mpMRI) to prospectively identify men at risk for apical PSMs (aPSMs) using a structured report are lacking. OBJECTIVES The aims of the study are to determine and to compare the rate of aPSM in men with versus without prospectively flagged at-risk prostate lesions during clinical mpMRI interpretation using whole-mount histopathology as the reference standard. METHODS This single-center, retrospective study of prospectively collected data included treatment-naive men with abnormal 3T mpMRI (PI-RADS v2 score ≥3) between January 2016 and December 2018 followed by surgery. During routine clinical interpretation, radiologists flagged prostate lesions abutting the apical most gland and/or encircling the distal most prostatic urethra using standardized language available as a "pick list" option in the structured report. Logistic regression was used to compare the rate of PSM in 2 groups (flagged vs nonflagged men). Propensity score covariate adjustment corrected for potential selection bias according to age, prostate-specific antigen (PSA), PSA density, grade group, and pT stage. The estimate was further adjusted by including surgeon as a covariate. RESULTS A total of 428 men were included. A statistically significant higher proportion of aPSMs was noted in flagged (56% [51/91]) compared with nonflagged apical lesions (31% [105/337]; adjusted odds ratio, 2.5; 95% confidence interval, 1.6-4.1; P < 0.01). The difference in aPSM between both groups also varied according to the surgeon performing the RP. Prostate-specific antigen, PSA density, lesion size, apical location, Prostate Imaging Reporting & Data System score, grade group, pT stage, and surgeon's experience were associated with higher PSM rate. Biochemical recurrence, defined as PSA greater than 0.2 ng/mL on 2 measurements after RP, was significantly associated with PSM status (propensity score adjusted odds ratio, 3.1; 95% confidence interval, 1.8-5.3; P < 0.0001); however, patients flagged by radiologists did not have a significant difference in biochemical recurrence rates as compared with nonflagged patients ( P = 0.11). CONCLUSIONS Standard language built into structured reports for mpMRI of the prostate helps identify preoperatively patients at risk for aPSM. CLINICAL IMPACT Multiparametric MRI is able to identify patients at increased risk for aPSM, and this information can be conveyed in a structured report to urologists, facilitating patient counseling and treatment decisions.
Collapse
|
23
|
Pedraza AM, Parekh S, Joshi H, Grauer R, Wagaskar V, Zuluaga L, Gupta R, Barthe F, Nasri J, Pandav K, Patel D, Gorin MA, Menon M, Tewari AK. Side-specific, Microultrasound-based Nomogram for the Prediction of Extracapsular Extension in Prostate Cancer. EUR UROL SUPPL 2022; 48:72-81. [PMID: 36743400 PMCID: PMC9895764 DOI: 10.1016/j.euros.2022.12.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2022] [Indexed: 12/29/2022] Open
Abstract
Background Prediction of extracapsular extension (ECE) is essential to achieve a balance between oncologic resection and neural tissue preservation. Microultrasound (MUS) is an attractive alternative to multiparametric magnetic resonance imaging (mpMRI) in the staging scenario. Objective To create a side-specific nomogram integrating clinicopathologic parameters and MUS findings to predict ipsilateral ECE and guide nerve sparing. Design setting and participants Prospective data were collected from consecutive patients who underwent robotic-assisted radical prostatectomy from June 2021 to May 2022 and had preoperative MUS and mpMRI. A total of 391 patients and 612 lobes were included in the analysis. Outcome measurements and statistical analysis ECE on surgical pathology was the primary outcome. Multivariate regression analyses were carried out to identify predictors for ECE. The resultant multivariable model's performance was visualized using the receiver-operating characteristic curve. A nomogram was developed based on the coefficients of the logit function for the MUS-based model. A decision curve analysis (DCA) was performed to assess clinical utility. Results and limitations The areas under the receiver-operating characteristic curve (AUCs) of the MUS-based model were 81.4% and 80.9% (95% confidence interval [CI] 75.6, 84.6) after internal validation. The AUC of the mpMRI-model was also 80.9% (95% CI 77.2, 85.7). The DCA demonstrated the net clinical benefit of the MUS-based nomogram and its superiority compared with MUS and MRI alone for detecting ECE. Limitations of our study included its sample size and moderate inter-reader agreement. Conclusions We developed a side-specific nomogram to predict ECE based on clinicopathologic variables and MUS findings. Its performance was comparable with that of a mpMRI-based model. External validation and prospective trials are required to corroborate our results. Patient summary The integration of clinical parameters and microultrasound can predict extracapsular extension with similar results to models based on magnetic resonance imaging findings. This can be useful for tailoring the preservation of nerves during surgery.
Collapse
Affiliation(s)
- Adriana M. Pedraza
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA.,Corresponding authors at: Department of Urology, Icahn School of Medicine at Mount Sinai, New York City, NY 10029, USA. Tel. +1 2122416500
| | - Sneha Parekh
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Himanshu Joshi
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA.,Institute for Healthcare Delivery Science, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ralph Grauer
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Vinayak Wagaskar
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Laura Zuluaga
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Raghav Gupta
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Flora Barthe
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Jordan Nasri
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Krunal Pandav
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Dhruti Patel
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Michael A. Gorin
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Mani Menon
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Ashutosh K. Tewari
- Department of Urology, Icahn School of Medicine at Mount Sinai, New York City, NY, USA.,Corresponding authors at: Department of Urology, Icahn School of Medicine at Mount Sinai, New York City, NY 10029, USA. Tel. +1 2122416500
| |
Collapse
|
24
|
Dias N, Colandrea G, Botelho F, Rodriguez-Sanchez L, Barbé Y, Macek P, Cathelineau X. Accuracy of PET-choline in nodal staging of localized very high-risk prostate cancer. Arch Ital Urol Androl 2022; 94:401-405. [PMID: 36576464 DOI: 10.4081/aiua.2022.4.401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 10/30/2022] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVES Localized very high-risk prostate cancer (VHR PCa) has long suffered from the inex-istence of good lymph node staging methods other than invasive surgery, as computed tomogra-phy has low sensitivity for nodal disease. With the rising use of positron emission tomography (PET), it is clinically meaningful to know its value for these patients. Our goal was to evaluate the real-life diagnostic accuracy of PET Choline in nodal staging, comparing it with the gold standard of extended pelvic lymph node dissection (ePLND). MATERIALS AND METHODS We reviewed data from a high-volume center, including patients with VHR PCa according to current NCCN guidelines who underwent community 18F-fluorocholine PET/CT; followed by ro-botic assisted laparoscopic prostatectomy (RALP) and ePLND between 2010 and 2021. RESULTS We included 44 patients and 88 lymph node regions. Among those, 14/44 (31.8%) patients and 20/88 (22.7%) regions had nodal disease present on definitive pathology. In comparison with ePLND, we found a sensitivity of 64.3% (95% CI, 39.2-89.4%), specificity of 83.3% (95% CI, 70.0- 96.7%), PPV of 64.3% (95% CI, 39.2-89.4%), and NPV of 83.3% (95% CI, 70.0-96.7%) for nodal disease on a patient-based analysis; and sensitivity of 35.0% (95% CI, 14.1-60.0%), specificity of 88.2% (95% CI, 80.6-95.9%), PPV of 46.7% (95% CI, 21.4-71.9%), and NPV of 82.2% (95% CI, 73.4-91.0%) on a region-based analysis. CONCLUSIONS In our view 18F-fluorocholine PET/CT doesn't meet the criteria to be a standard exam for pre-operative staging for patients with VHR PCa, mostly due to its low sensitivity. However, other radiotracers should continue to be investigated in this setting.
Collapse
Affiliation(s)
- Nuno Dias
- Urology Department, Institut Mutualiste Montsouris, Paris, France; Urology Department, São João Hospitalar and University Center, Porto.
| | - Gianmarco Colandrea
- Urology Department, Institut Mutualiste Montsouris, Paris, France; Unit of Urology, Division of Experimental Oncology, URI Urological Research Institute, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan.
| | - Francisco Botelho
- Urology Department, São João Hospitalar and University Center, Porto; Institute for Life and Health Sciences Investigation, School of Medicine, Minho University, Braga.
| | | | - Yann Barbé
- Urology Department, Institut Mutualiste Montsouris, Paris.
| | - Petr Macek
- Urology Department, Institut Mutualiste Montsouris, Paris.
| | | |
Collapse
|
25
|
Ucar T, Gunduz N, Demirci E, Culpan M, Gunel H, Kir G, Atis RG, Yildirim A. Comparison of 68Ga-PSMA PET/CT and mp-MRI in regard to local staging for prostate cancer with histopathological results: A retrospective study. Prostate 2022; 82:1462-1468. [PMID: 35915579 DOI: 10.1002/pros.24420] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 06/07/2022] [Accepted: 07/11/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND Imaging modalities are used to diagnose and clinical grading of clinically significant prostate cancer. In this study, 68Ga-PSMA PET/CT (PSMA) and multiparametric prostate MRI (mp-MRI) were compared in regard to locating intraprostatic tumor and locoregional staging. METHODS After ethics committee approval, a total of 49 patients with prostate cancer who had mp-MRI and PSMA before radical prostatectomy were included. Preoperative and postoperative PSA, transrectal ultrasound-guided prostate biopsy (TRUS-Bx) ISUP grade, radical prostatectomy ISUP grade, body mass index (BMI), TRUS prostate volume, mp-MRI tumor mapping, PSMAtumor mapping, pathologic tumor mapping, extraprostatic extension (EPE), seminal vesicle invasion (SVI), lymph node invasion (LNI), and bladder neck invasion (BNI)were retrospectively evaluated. Index tumor was located by uroradiologist, nuclear medicine specialist, and uropathologist on a 12-sector prostate pathology map and compared with each other in terms of accuracy and locoregional clinical staging. RESULTS Mean age of the patients was 66.18 ± 6.67 years and the mean of preoperative PSA results was 21.11 ± 32.56 ng/ml. Nearly half of the patients' (44.9%) pathology was reported as ISUP grade 4 and 5% and 18.4% of patients were surgical margin positive. According to the pathological findings, 362 out of 588 sectors were tumor-positive, 174 out of 362 sectors were tumor-positive in mp-MRI, and 175 out of 362 sectors were tumor-positive in PSMA. Both PSMA and mp-MRI were comparable (p = 0.823) and accurate to detect the location of the intraprostatic index tumor (AUC = 0.66 vs. 0.69 respectively, p = 0.82). The sensitivity and the specificity of the PSMA and mp-MRI for localizing intraprostatic index tumors were 42.5% versus 49.5% and 90.7% versus 88.6% respectively. mp-MRI was more accurate than PSMA in terms of EPE (AUC = 0.8 vs. AUC = 0.57 respectively, p = 0.027) and both methods were comparable in terms of SVI (AUC = 0.75 vs. AUC = 0.75, p = 0.886) and BNI (AUC = 0.51 vs. AUC = 0.59, p = 0.597). PSMA and mp-MRI were comparable in terms of LNI (AUC = 0.76 vs. AUC = 0.64, p = 0.39). CONCLUSION mp-MRI should be considered for its high accuracy in the diagnosis of EPE, especially before decision-making for nerve-sparing surgery in high-risk patients. Both imaging modalities were accurate for localizing intraprostatic index tumor. PSMA is accurate for detecting LNI.
Collapse
Affiliation(s)
- Taha Ucar
- Department of Urology, Nigde Omer Halis Demir University Research and Training Hospital, Nigde, Turkey
| | - Nesrin Gunduz
- Department of Radiology, Istanbul Medeniyet University Prof. Dr. Suleyman Yalcin Research and Training Hospital, Istanbul, Turkey
| | - Emre Demirci
- Department of Nuclear Medicine, Yeditepe University, Istanbul, Turkey
| | - Meftun Culpan
- Department of Urology, Istanbul Medeniyet University Prof. Dr. Suleyman Yalcin Research and Training Hospital, Istanbul, Turkey
| | - Humeyra Gunel
- Department of Pathology, Istanbul Medeniyet University Prof. Dr. Suleyman Yalcin Research and Training Hospital, Istanbul, Turkey
| | - Gozde Kir
- Department of Pathology, Istanbul Medeniyet University Prof. Dr. Suleyman Yalcin Research and Training Hospital, Istanbul, Turkey
| | - Ramazan Gokhan Atis
- Department of Urology, Istanbul Medeniyet University Prof. Dr. Suleyman Yalcin Research and Training Hospital, Istanbul, Turkey
| | - Asif Yildirim
- Department of Urology, Istanbul Medeniyet University Prof. Dr. Suleyman Yalcin Research and Training Hospital, Istanbul, Turkey
| |
Collapse
|
26
|
French AFU Cancer Committee Guidelines - Update 2022-2024: prostate cancer - Diagnosis and management of localised disease. Prog Urol 2022; 32:1275-1372. [DOI: 10.1016/j.purol.2022.07.148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 07/11/2022] [Indexed: 11/17/2022]
|
27
|
Picot F, Shams R, Dallaire F, Sheehy G, Trang T, Grajales D, Birlea M, Trudel D, Ménard C, Kadoury S, Leblond F. Image-guided Raman spectroscopy navigation system to improve transperineal prostate cancer detection. Part 1: Raman spectroscopy fiber-optics system and in situ tissue characterization. JOURNAL OF BIOMEDICAL OPTICS 2022; 27:JBO-220045GRR. [PMID: 36045491 PMCID: PMC9433338 DOI: 10.1117/1.jbo.27.9.095003] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 08/16/2022] [Indexed: 05/28/2023]
Abstract
SIGNIFICANCE The diagnosis of prostate cancer (PCa) and focal treatment by brachytherapy are limited by the lack of precise intraoperative information to target tumors during biopsy collection and radiation seed placement. Image-guidance techniques could improve the safety and diagnostic yield of biopsy collection as well as increase the efficacy of radiotherapy. AIM To estimate the accuracy of PCa detection using in situ Raman spectroscopy (RS) in a pilot in-human clinical study and assess biochemical differences between in vivo and ex vivo measurements. APPROACH A new miniature RS fiber-optics system equipped with an electromagnetic (EM) tracker was guided by trans-rectal ultrasound-guided imaging, fused with preoperative magnetic resonance imaging to acquire 49 spectra in situ (in vivo) from 18 PCa patients. In addition, 179 spectra were acquired ex vivo in fresh prostate samples from 14 patients who underwent radical prostatectomy. Two machine-learning models were trained to discriminate cancer from normal prostate tissue from both in situ and ex vivo datasets. RESULTS A support vector machine (SVM) model was trained on the in situ dataset and its performance was evaluated using leave-one-patient-out cross validation from 28 normal prostate measurements and 21 in-tumor measurements. The model performed at 86% sensitivity and 72% specificity. Similarly, an SVM model was trained with the ex vivo dataset from 152 normal prostate measurements and 27 tumor measurements showing reduced cancer detection performance mostly attributable to spatial registration inaccuracies between probe measurements and histology assessment. A qualitative comparison between in situ and ex vivo measurements demonstrated a one-to-one correspondence and similar ratios between the main Raman bands (e.g., amide I-II bands, phenylalanine). CONCLUSIONS PCa detection can be achieved using RS and machine learning models for image-guidance applications using in situ measurements during prostate biopsy procedures.
Collapse
Affiliation(s)
- Fabien Picot
- Polytechnique Montréal, Department of Engineering Physics, Montreal, Quebec, Canada
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
| | - Roozbeh Shams
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
- Polytechnique Montréal, Medical Laboratory, Montreal, Quebec, Canada
| | - Frédérick Dallaire
- Polytechnique Montréal, Department of Engineering Physics, Montreal, Quebec, Canada
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
| | - Guillaume Sheehy
- Polytechnique Montréal, Department of Engineering Physics, Montreal, Quebec, Canada
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
| | - Tran Trang
- Polytechnique Montréal, Department of Engineering Physics, Montreal, Quebec, Canada
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
| | - David Grajales
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
- Polytechnique Montréal, Medical Laboratory, Montreal, Quebec, Canada
| | - Mirela Birlea
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
| | - Dominique Trudel
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
| | - Cynthia Ménard
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
| | - Samuel Kadoury
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
- Polytechnique Montréal, Medical Laboratory, Montreal, Quebec, Canada
| | - Frédéric Leblond
- Polytechnique Montréal, Department of Engineering Physics, Montreal, Quebec, Canada
- Centre de recherche du Centre hospitalier de l’Université de Montréal, Montreal, Quebec, Canada
- Institut du cancer de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
28
|
Dinneen E, Grierson J, Almeida-Magana R, Clow R, Haider A, Allen C, Heffernan-Ho D, Freeman A, Briggs T, Nathan S, Mallett S, Brew-Graves C, Muirhead N, Williams NR, Pizzo E, Persad R, Aning J, Johnson L, Oxley J, Oakley N, Morgan S, Tahir F, Ahmad I, Dutto L, Salmond JM, Kelkar A, Kelly J, Shaw G. NeuroSAFE PROOF: study protocol for a single-blinded, IDEAL stage 3, multi-centre, randomised controlled trial of NeuroSAFE robotic-assisted radical prostatectomy versus standard robotic-assisted radical prostatectomy in men with localized prostate cancer. Trials 2022; 23:584. [PMID: 35869497 PMCID: PMC9306247 DOI: 10.1186/s13063-022-06421-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 05/24/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Robotic radical prostatectomy (RARP) is a first-line curative treatment option for localized prostate cancer. Postoperative erectile dysfunction and urinary incontinence are common associated adverse side effects that can negatively impact patients' quality of life. Preserving the lateral neurovascular bundles (NS) during RARP improves functional outcomes. However, selecting men for NS may be difficult when there is concern about incurring in positive surgical margin (PSM) which in turn risks adverse oncological outcomes. The NeuroSAFE technique (intra-operative frozen section examination of the neurovascular structure adjacent prostate margin) can provide real-time pathological consult to promote optimal NS whilst avoiding PSM. METHODS NeuroSAFE PROOF is a single-blinded, multi-centre, randomised controlled trial (RCT) in which men are randomly allocated 1:1 to either NeuroSAFE RARP or standard RARP. Men electing for RARP as primary treatment, who are continent and have good baseline erectile function (EF), defined by International Index of Erectile Function (IIEF-5) score > 21, are eligible. NS in the intervention arm is guided by the NeuroSAFE technique. NS in the standard arm is based on standard of care, i.e. a pre-operative image-based planning meeting, patient-specific clinical information, and digital rectal examination. The primary outcome is assessment of EF at 12 months. The primary endpoint is the proportion of men who achieve IIEF-5 score ≥ 21. A sample size of 404 was calculated to give a power of 90% to detect a difference of 14% between groups based on a feasibility study. Oncological outcomes are continuously monitored by an independent Data Monitoring Committee. Key secondary outcomes include urinary continence at 3 months assessed by the international consultation on incontinence questionnaire, rate of biochemical recurrence, EF recovery at 24 months, and difference in quality of life. DISCUSSION NeuroSAFE PROOF is the first RCT of intra-operative frozen section during radical prostatectomy in the world. It is properly powered to evaluate a difference in the recovery of EF for men undergoing RARP assessed by patient-reported outcome measures. It will provide evidence to guide the use of the NeuroSAFE technique around the world. TRIAL REGISTRATION NCT03317990 (23 October 2017). Regional Ethics Committee; reference 17/LO/1978.
Collapse
Affiliation(s)
- Eoin Dinneen
- Division of Surgery & Interventional Science, University College London, London, UK.
- University College Hospital London, Department of Urology, Westmoreland Street Hospital, 6-18 Westmoreland Street, W1G 8PH, London, UK.
| | - Jack Grierson
- Division of Surgery & Interventional Science, University College London, London, UK
- University College Hospital London, Department of Urology, Westmoreland Street Hospital, 6-18 Westmoreland Street, W1G 8PH, London, UK
| | | | - Rosie Clow
- Division of Surgery & Interventional Science, University College London, London, UK
- University College Hospital London, Department of Urology, Westmoreland Street Hospital, 6-18 Westmoreland Street, W1G 8PH, London, UK
| | - Aiman Haider
- University College Hospital London, Department of Histopathology, 235 Euston Road, Bristol, NW1 2BU, UK
| | - Clare Allen
- University College Hospital London, Department of Urology, Westmoreland Street Hospital, 6-18 Westmoreland Street, W1G 8PH, London, UK
| | - Daniel Heffernan-Ho
- University College Hospital London, Department of Urology, Westmoreland Street Hospital, 6-18 Westmoreland Street, W1G 8PH, London, UK
| | - Alex Freeman
- University College Hospital London, Department of Histopathology, 235 Euston Road, Bristol, NW1 2BU, UK
| | - Tim Briggs
- University College Hospital London, Department of Urology, Westmoreland Street Hospital, 6-18 Westmoreland Street, W1G 8PH, London, UK
| | - Senthil Nathan
- University College Hospital London, Department of Urology, Westmoreland Street Hospital, 6-18 Westmoreland Street, W1G 8PH, London, UK
| | - Susan Mallett
- Division of Medicine, University College London, Charles Bell House, 43-45 Foley Street, Sheffield, W1W 7JN, UK
| | - Chris Brew-Graves
- Division of Medicine, University College London, Charles Bell House, 43-45 Foley Street, Sheffield, W1W 7JN, UK
| | - Nicola Muirhead
- Division of Medicine, University College London, Charles Bell House, 43-45 Foley Street, Sheffield, W1W 7JN, UK
| | - Norman R Williams
- Division of Surgery & Interventional Science, University College London, London, UK
| | - Elena Pizzo
- Department of Applied Health Research, University College London, 1-19 Torrington Place, Glasgow, WC1E 7HB, UK
| | - Raj Persad
- North Bristol Hospitals Trust, Department of Urology, Southmead Hospital, Southmead Lane, Westbury-on-Trym, Bristol, BS10 5NB, UK
| | - Jon Aning
- North Bristol Hospitals Trust, Department of Urology, Southmead Hospital, Southmead Lane, Westbury-on-Trym, Bristol, BS10 5NB, UK
| | - Lyndsey Johnson
- North Bristol Hospitals Trust, Department of Urology, Southmead Hospital, Southmead Lane, Westbury-on-Trym, Bristol, BS10 5NB, UK
| | - Jon Oxley
- North Bristol Hospitals Trust, Department of Histopathology, Southmead Hospital, Southmead Lane, Westbury-on-Trym, BS10 5NB, Bristol, UK
| | - Neil Oakley
- Sheffield Teaching Hospitals NHS Trust, Department of Urology, Royal Hallamshire Hospital, Glossop Road, S10 2JF, UK
| | - Susan Morgan
- Sheffield Teaching Hospitals NHS Trust, Department of Histopathology, Royal Hallamshire Hospital, Glossop Road, S10 2JF, UK
| | - Fawzia Tahir
- Sheffield Teaching Hospitals NHS Trust, Department of Histopathology, Royal Hallamshire Hospital, Glossop Road, S10 2JF, UK
| | - Imran Ahmad
- Glasgow & Clyde NHS Trust, Department of Urology, Queen Elizabeth Hospital, 1345 Govan Road, Glasgow, UK
| | - Lorenzo Dutto
- Glasgow & Clyde NHS Trust, Department of Urology, Queen Elizabeth Hospital, 1345 Govan Road, Glasgow, UK
| | - Jonathan M Salmond
- Glasgow & Clude NHS Trust, Department of Histopathology, Queen Elizabeth Hospital, 1345 Govan Road, Glasgow, UK
| | - Anand Kelkar
- University College Hospital London, Department of Urology, Westmoreland Street Hospital, 6-18 Westmoreland Street, W1G 8PH, London, UK
- Barking Havering & Redbridge University Hospitals Trust, Rom Valley Way, Romford, RM7 0AG, UK
| | - John Kelly
- Division of Surgery & Interventional Science, University College London, London, UK
- University College Hospital London, Department of Urology, Westmoreland Street Hospital, 6-18 Westmoreland Street, W1G 8PH, London, UK
| | - Greg Shaw
- Division of Surgery & Interventional Science, University College London, London, UK
- University College Hospital London, Department of Urology, Westmoreland Street Hospital, 6-18 Westmoreland Street, W1G 8PH, London, UK
| |
Collapse
|
29
|
Vitek RA, Huang W, Geiger PG, Heninger E, Lang JM, Jarrard DF, Beebe DJ, Johnson BP. Fresh tissue procurement and preparation for multicompartment and multimodal analysis of the prostate tumor microenvironment. Prostate 2022; 82:836-849. [PMID: 35226381 PMCID: PMC9010374 DOI: 10.1002/pros.24326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 01/23/2022] [Accepted: 02/08/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Prostatic cancers include a diverse microenvironment of tumor cells, cancer-associated fibroblasts, and immune components. This tumor microenvironment (TME) is a known driving force of tumor survival after treatment, but the standard-of-care tissue freezing or fixation in pathology practice limit the use of available approaches/tools to study the TME's functionality in tumor resistance. Thus, there is a need for approaches that satisfy both clinical and laboratory endpoints for TME study. Here we present methods for clinical case identification, tissue processing, and analytical workflow that are compatible with standard histopathology while enabling molecular and functional interrogation of prostate TME components. METHODS We first performed a small retrospective review to identify cases where submission of alternate prostate tissue slices and a parallel live tissue processing protocol complement traditional histopathology and enable viable multicompartment analysis of the TME. Then, we tested its compatibility with commonly employed methods to study the microenvironment including quantification of components both in situ and after tissue dissociation. We also evaluated tissue digestion conditions and cell isolation techniques to aid various molecular and functional endpoints. RESULTS We identified Gleason Grade Group 3+ clinical cases where tumor volume was sufficient to allow slicing of unfixed tissue and distribution of alternating tissue slices to standard-of-care histopathology and viable multi-modal TME analyses. No single method was found that preserved cellular sub-types for all downstream readouts; instead, tissues were further divided so techniques could be catered to each endpoint. For instance, we show that incorporating the protease dispase into tissue dissociation improves viability for culture and functional analyses but hinders immune cell analysis by flow cytometry. We also found that flow activated cell sorting provides highly pure cell populations for quantitative reverse-transcription polymerase chain reaction and RNA-seq while isolation using antibody-labeled paramagnetic particles facilitated functional coculture experiments. CONCLUSIONS The identification of candidate cases and use of these techniques enable translational research and the development of molecular and functional assays to facilitate prostate TME study without compromising standard-of-care histopathological diagnosis. This allows bridging clinical histopathology and further interrogation of the prostate TME and promises to advance our understanding of tumor biology and unveil new predictive and prognostic markers of prostate cancer progression.
Collapse
Affiliation(s)
- Ross A. Vitek
- Department of Pathology and Laboratory MedicineUniversity of WisconsinMadisonWisconsinUSA
- Department of Biomedical EngineeringUniversity of WisconsinMadisonWisconsinUSA
| | - Wei Huang
- Department of Pathology and Laboratory MedicineUniversity of WisconsinMadisonWisconsinUSA
| | - Peter G. Geiger
- Department of Pathology and Laboratory MedicineUniversity of WisconsinMadisonWisconsinUSA
| | - Erika Heninger
- Carbone Cancer CenterUniversity of WisconsinMadisonWisconsinUSA
| | - Joshua M. Lang
- Carbone Cancer CenterUniversity of WisconsinMadisonWisconsinUSA
- Department of MedicineUniversity of WisconsinMadisonWisconsinUSA
| | | | - David J. Beebe
- Department of Pathology and Laboratory MedicineUniversity of WisconsinMadisonWisconsinUSA
- Department of Biomedical EngineeringUniversity of WisconsinMadisonWisconsinUSA
- Carbone Cancer CenterUniversity of WisconsinMadisonWisconsinUSA
| | - Brian P. Johnson
- Department of Pathology and Laboratory MedicineUniversity of WisconsinMadisonWisconsinUSA
- Department of Biomedical EngineeringUniversity of WisconsinMadisonWisconsinUSA
- Department of Pharmacology & ToxicologyMichigan State UniversityEast LansingMichiganUSA
| |
Collapse
|
30
|
Prognostic significance of percentage Gleason grade 5 prostatic adenocarcinoma in needle biopsies from patients treated by radical prostatectomy. Pathology 2022; 54:694-699. [DOI: 10.1016/j.pathol.2022.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 11/21/2022]
|
31
|
Dinneen E, Allen C, Strange T, Heffernan-Ho D, Banjeglav J, Lindsay J, Mulligan JP, Briggs T, Nathan S, Sridhar A, Grierson J, Haider A, Panayi C, Patel D, Freeman A, Aning J, Persad R, Ahmad I, Dutto L, Oakley N, Ambrosi A, Parry T, Kasivisvanathan V, Giganti F, Shaw G, Punwani S. Negative mpMRI Rules Out Extra-Prostatic Extension in Prostate Cancer before Robot-Assisted Radical Prostatectomy. Diagnostics (Basel) 2022; 12:1057. [PMID: 35626214 PMCID: PMC9139507 DOI: 10.3390/diagnostics12051057] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/18/2022] [Accepted: 04/20/2022] [Indexed: 11/23/2022] Open
Abstract
Background: The accuracy of multi-parametric MRI (mpMRI) in the pre-operative staging of prostate cancer (PCa) remains controversial. Objective: The purpose of this study was to evaluate the ability of mpMRI to accurately predict PCa extra-prostatic extension (EPE) on a side-specific basis using a risk-stratified 5-point Likert scale. This study also aimed to assess the influence of mpMRI scan quality on diagnostic accuracy. Patients and Methods: We included 124 men who underwent robot-assisted RP (RARP) as part of the NeuroSAFE PROOF study at our centre. Three radiologists retrospectively reviewed mpMRI blinded to RP pathology and assigned a Likert score (1-5) for EPE on each side of the prostate. Each scan was also ascribed a Prostate Imaging Quality (PI-QUAL) score for assessing the quality of the mpMRI scan, where 1 represents the poorest and 5 represents the best diagnostic quality. Outcome measurements and statistical analyses: Diagnostic performance is presented for the binary classification of EPE, including 95% confidence intervals and the area under the receiver operating characteristic curve (AUC). Results: A total of 231 lobes from 121 men (mean age 56.9 years) were evaluated. Of these, 39 men (32.2%), or 43 lobes (18.6%), had EPE. A Likert score ≥3 had a sensitivity (SE), specificity (SP), NPV, and PPV of 90.4%, 52.3%, 96%, and 29.9%, respectively, and the AUC was 0.82 (95% CI: 0.77-0.86). The AUC was 0.76 (95% CI: 0.64-0.88), 0.78 (0.72-0.84), and 0.92 (0.88-0.96) for biparametric scans, PI-QUAL 1-3, and PI-QUAL 4-5 scans, respectively. Conclusions: MRI can be used effectively by genitourinary radiologists to rule out EPE and help inform surgical planning for men undergoing RARP. EPE prediction was more reliable when the MRI scan was (a) multi-parametric and (b) of a higher image quality according to the PI-QUAL scoring system.
Collapse
Affiliation(s)
- Eoin Dinneen
- Division of Surgery & Interventional Science, University College London, Charles Bell House, 3rd Floor, 43-45 Foley Street, London W1W 7TS, UK; (J.G.); (V.K.); (F.G.); (G.S.)
- Department of Urology, University College Hospital London, Westmoreland Street Hospital, 16-18 Westmoreland Street, London W1G 8PH, UK; (J.B.); (J.L.); (J.-P.M.); (T.B.); (S.N.); (A.S.)
| | - Clare Allen
- Department of Radiology, University College London Hospitals, 235 Euston Road, London NW1 2BU, UK; (C.A.); (T.S.); (D.H.-H.); (S.P.)
| | - Tom Strange
- Department of Radiology, University College London Hospitals, 235 Euston Road, London NW1 2BU, UK; (C.A.); (T.S.); (D.H.-H.); (S.P.)
| | - Daniel Heffernan-Ho
- Department of Radiology, University College London Hospitals, 235 Euston Road, London NW1 2BU, UK; (C.A.); (T.S.); (D.H.-H.); (S.P.)
| | - Jelena Banjeglav
- Department of Urology, University College Hospital London, Westmoreland Street Hospital, 16-18 Westmoreland Street, London W1G 8PH, UK; (J.B.); (J.L.); (J.-P.M.); (T.B.); (S.N.); (A.S.)
| | - Jamie Lindsay
- Department of Urology, University College Hospital London, Westmoreland Street Hospital, 16-18 Westmoreland Street, London W1G 8PH, UK; (J.B.); (J.L.); (J.-P.M.); (T.B.); (S.N.); (A.S.)
| | - John-Patrick Mulligan
- Department of Urology, University College Hospital London, Westmoreland Street Hospital, 16-18 Westmoreland Street, London W1G 8PH, UK; (J.B.); (J.L.); (J.-P.M.); (T.B.); (S.N.); (A.S.)
| | - Tim Briggs
- Department of Urology, University College Hospital London, Westmoreland Street Hospital, 16-18 Westmoreland Street, London W1G 8PH, UK; (J.B.); (J.L.); (J.-P.M.); (T.B.); (S.N.); (A.S.)
| | - Senthil Nathan
- Department of Urology, University College Hospital London, Westmoreland Street Hospital, 16-18 Westmoreland Street, London W1G 8PH, UK; (J.B.); (J.L.); (J.-P.M.); (T.B.); (S.N.); (A.S.)
| | - Ashwin Sridhar
- Department of Urology, University College Hospital London, Westmoreland Street Hospital, 16-18 Westmoreland Street, London W1G 8PH, UK; (J.B.); (J.L.); (J.-P.M.); (T.B.); (S.N.); (A.S.)
| | - Jack Grierson
- Division of Surgery & Interventional Science, University College London, Charles Bell House, 3rd Floor, 43-45 Foley Street, London W1W 7TS, UK; (J.G.); (V.K.); (F.G.); (G.S.)
- Department of Histopathology, University College Hospital London, 235 Euston Road, London NW1 2BU, UK; (A.H.); (C.P.); (D.P.); (A.F.)
| | - Aiman Haider
- Department of Histopathology, University College Hospital London, 235 Euston Road, London NW1 2BU, UK; (A.H.); (C.P.); (D.P.); (A.F.)
| | - Christos Panayi
- Department of Histopathology, University College Hospital London, 235 Euston Road, London NW1 2BU, UK; (A.H.); (C.P.); (D.P.); (A.F.)
| | - Dominic Patel
- Department of Histopathology, University College Hospital London, 235 Euston Road, London NW1 2BU, UK; (A.H.); (C.P.); (D.P.); (A.F.)
| | - Alex Freeman
- Department of Histopathology, University College Hospital London, 235 Euston Road, London NW1 2BU, UK; (A.H.); (C.P.); (D.P.); (A.F.)
| | - Jonathan Aning
- North Bristol Hospitals Trust, Department of Urology, Southmead Hospital, Southmead Lane, Westbury-on-Trym, Bristol BS10 5NB, UK; (J.A.); (R.P.)
| | - Raj Persad
- North Bristol Hospitals Trust, Department of Urology, Southmead Hospital, Southmead Lane, Westbury-on-Trym, Bristol BS10 5NB, UK; (J.A.); (R.P.)
| | - Imran Ahmad
- Department of Urology, Queen Elizabeth University Hospital, NHS Greater Glasgow & Clyde, 1345 Govan Road, Glasgow G51 4TF, UK; (I.A.); (L.D.)
| | - Lorenzo Dutto
- Department of Urology, Queen Elizabeth University Hospital, NHS Greater Glasgow & Clyde, 1345 Govan Road, Glasgow G51 4TF, UK; (I.A.); (L.D.)
| | - Neil Oakley
- Department of Urology, Sheffield Teaching Hospitals NHS Trust, Royal Hallamshire Hospital, Glossop Road, Sheffield S10 2JF, UK;
| | - Alessandro Ambrosi
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, 20132 Milano, Italy;
| | - Tom Parry
- Centre for Medical Imaging, University College London, Charles Bell House, 2nd Floor, 43-45 Foley Street, London W1W 7TS, UK;
| | - Veeru Kasivisvanathan
- Division of Surgery & Interventional Science, University College London, Charles Bell House, 3rd Floor, 43-45 Foley Street, London W1W 7TS, UK; (J.G.); (V.K.); (F.G.); (G.S.)
- Department of Urology, University College Hospital London, Westmoreland Street Hospital, 16-18 Westmoreland Street, London W1G 8PH, UK; (J.B.); (J.L.); (J.-P.M.); (T.B.); (S.N.); (A.S.)
| | - Francesco Giganti
- Division of Surgery & Interventional Science, University College London, Charles Bell House, 3rd Floor, 43-45 Foley Street, London W1W 7TS, UK; (J.G.); (V.K.); (F.G.); (G.S.)
- Department of Radiology, University College London Hospitals, 235 Euston Road, London NW1 2BU, UK; (C.A.); (T.S.); (D.H.-H.); (S.P.)
| | - Greg Shaw
- Division of Surgery & Interventional Science, University College London, Charles Bell House, 3rd Floor, 43-45 Foley Street, London W1W 7TS, UK; (J.G.); (V.K.); (F.G.); (G.S.)
- Department of Urology, University College Hospital London, Westmoreland Street Hospital, 16-18 Westmoreland Street, London W1G 8PH, UK; (J.B.); (J.L.); (J.-P.M.); (T.B.); (S.N.); (A.S.)
| | - Shonit Punwani
- Department of Radiology, University College London Hospitals, 235 Euston Road, London NW1 2BU, UK; (C.A.); (T.S.); (D.H.-H.); (S.P.)
- Centre for Medical Imaging, University College London, Charles Bell House, 2nd Floor, 43-45 Foley Street, London W1W 7TS, UK;
| |
Collapse
|
32
|
Ueno Y, Tamada T, Sofue K, Urase Y, Hinata N, Fujisawa M, Murakami T. Do the variations in ROI placement technique have influence for prostate ADC measurements? Acta Radiol Open 2022; 11:20584601221086500. [PMID: 35368407 PMCID: PMC8973079 DOI: 10.1177/20584601221086500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/23/2022] [Indexed: 12/29/2022] Open
Abstract
Background Prostate apparent diffusion coefficient (ADC) values calculated from diffusion-weighted imaging have been used for evaluating prostate cancer (PCa) aggressiveness. However, the way of measuring ADC values has varied depending on the study. Purpose To investigate inter- and intra-reader variability and diagnostic performance of three kinds of shaped 2D regions of interests (ROIs) for tumor ADC measurements in PCa. Material and Methods Seventy-four patients with PCa undergoing 3-T MRI before surgery were included. Histologic findings from radical prostatectomy specimens were reviewed to define each patient’s dominant tumor. Three readers independently measured the tumor ADCs using three different ROI methods: freehand, large-circle, and small-circles ROIs. Readers repeated measurements after 3 weeks. Bland-Altman analysis was performed to evaluate the inter- and intra-reader variability. Receiver Operating Characteristic analysis was used for assessment of tumor aggressiveness for PCa. Results For intra-reader and inter-reader variability, the mean coefficient of repeatability for freehand ROIs, large-circle ROIs, and small-circles ROIs were as follows: 13.7%, 12.4%, and 11.5%; 9.4%, 9.7%, and 9.5%. For differentiating Gleason score (GS) = 3 + 3 from GS ≥ 3 + 4 tumors, the area under the curves were 0.90 for freehand ROIs, 0.89 for large-circle ROIs, and 0.94 small-circles ROIs ( p = 0.31). Conclusion The variations in ROI method did not have a major influence on intra-reader or inter-reader reproducibility or diagnostic performance for prostate ADC measurements.
Collapse
Affiliation(s)
- Yoshiko Ueno
- Department of Radiology, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Tsutomu Tamada
- Department of Radiology, Kobe University Graduate School of Medicine, Hyogo, Japan
- Department of Radiology, Kawasaki Medical School, Okayama, Japan
- Tsutomu Tamada, Department of Radiology, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, 701-0192, Japan.
| | - Keitaro Sofue
- Department of Radiology, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Yasuyo Urase
- Department of Radiology, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Nobuyuki Hinata
- Department of Urology, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Masato Fujisawa
- Department of Urology, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Takamichi Murakami
- Department of Radiology, Kobe University Graduate School of Medicine, Hyogo, Japan
| |
Collapse
|
33
|
Quantifying Tumor Heterogeneity from Multiparametric Magnetic Resonance Imaging of Prostate Using Texture Analysis. Cancers (Basel) 2022; 14:cancers14071631. [PMID: 35406403 PMCID: PMC8997150 DOI: 10.3390/cancers14071631] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 03/16/2022] [Indexed: 11/24/2022] Open
Abstract
Simple Summary Prostate cancer (PCa) occurs in males at a rate of 21.8%, predominantly at the customary primary site. High cure rates are possible through early detection and therapy when the tumor is still restricted to the prostate. These tumors do not grow rapidly, allowing for periods of up to 20 years between diagnosis and death. Multiparametric MRI (mp-MRI) is used as a non-invasive approach to diagnose PCa in subjects. This imaging method uses MR imaging with at least one functional MRI sequence to detect and characterize PCa. The use of multiparametric magnetic resonance imaging has refined the diagnosis of prostate cancer in radiology. Malignancy-modified critical features in tissue composition, such as heterogeneity, are associated with adverse tumor biology. Heterogeneity can be quantified through texture analysis, an effective technique for reviewing tumor images acquired in routine clinical practice. This study focused on identifying and quantifying tumor heterogeneity from prostate mp-MRI utilizing texture analysis. Abstract (1) Background: Multiparametric MRI (mp-MRI) is used to manage patients with PCa. Tumor identification via irregular sampling or biopsy is problematic and does not allow the comprehensive detection of the phenotypic and genetic alterations in a tumor. A non-invasive technique to clinically assess tumor heterogeneity is also in demand. We aimed to identify tumor heterogeneity from multiparametric magnetic resonance images using texture analysis (TA). (2) Methods: Eighteen patients with prostate cancer underwent mp-MRI scans before prostatectomy. A single radiologist matched the histopathology report to single axial slices that best depicted tumor and non-tumor regions to generate regions of interest (ROIs). First-order statistics based on the histogram analysis, including skewness, kurtosis, and entropy, were used to quantify tumor heterogeneity. We compared non-tumor regions with significant tumors, employing the two-tailed Mann–Whitney U test. Analysis of the area under the receiver operating characteristic curve (ROC-AUC) was used to determine diagnostic accuracy. (3) Results: ADC skewness for a 6 × 6 px filter was significantly lower with an ROC-AUC of 0.82 (p = 0.001). The skewness of the ADC for a 9 × 9 px filter had the second-highest result, with an ROC-AUC of 0.66; however, this was not statistically significant (p = 0.08). Furthermore, there were no substantial distinctions between pixel filter size groups from the histogram analysis, including entropy and kurtosis. (4) Conclusions: For all filter sizes, there was poor performance in terms of entropy and kurtosis histogram analyses for cancer diagnosis. Significant prostate cancer may be distinguished using a textural feature derived from ADC skewness with a 6 × 6 px filter size.
Collapse
|
34
|
Gatti M, Faletti R, Gentile F, Soncin E, Calleris G, Fornari A, Oderda M, Serafini A, Strazzarino GA, Vissio E, Bergamasco L, Cirillo S, Papotti MG, Gontero P, Fonio P. mEPE-score: a comprehensive grading system for predicting pathologic extraprostatic extension of prostate cancer at multiparametric magnetic resonance imaging. Eur Radiol 2022; 32:4942-4953. [PMID: 35290508 PMCID: PMC9213375 DOI: 10.1007/s00330-022-08595-9] [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: 10/11/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 11/24/2022]
Abstract
Objective To investigate the diagnostic accuracy of the PI-RADS v2.1 multiparametric magnetic resonance imaging (mpMRI) features in predicting extraprostatic extension (mEPE) of prostate cancer (PCa), as well as to develop and validate a comprehensive mpMRI-derived score (mEPE-score). Methods We retrospectively reviewed all consecutive patients admitted to two institutions for radical prostatectomy for PCa with available records of mpMRI performed between January 2015 and December 2020. Data from one institution was used for investigating diagnostic performance of each mEPE feature using radical prostatectomy specimens as benchmark. The results were implemented in a mEPE-score as follows: no mEPE features: 1; capsular abutment: 2; irregular or spiculated margin: 3; bulging prostatic contour, or asymmetry of the neurovascular bundles, or tumor-capsule interface > 1.0 cm: 4; ≥ 2 of the previous three parameters or measurable extraprostatic disease: 5. The performance of mEPE features was evaluated using the five diagnostic parameters and ROC curve analysis. Results Two-hundred patients were enrolled at site 1 and 76 at site 2. mEPE features had poor sensitivities ranging from 0.08 (0.00–0.15) to 0.71 (0.59–0.83), whereas specificity ranged from 0.68 (0.58–0.79) to 1.00. mEPE-score showed excellent discriminating ability (AUC > 0.8) and sensitivity = 0.82 and specificity = 0.77 with a threshold of 3. mEPE-score had AUC comparable to ESUR-score (p = 0.59 internal validation; p = 0.82 external validation), higher than or comparable to mEPE-grade (p = 0.04 internal validation; p = 0.58 external validation), and higher than early-and-late-EPE (p < 0.0001 internal and external validation). There were no significant differences between readers having different expertise with EPE-score (p = 0.32) or mEPE-grade (p = 0.45), but there were significant differences for ESUR-score (p = 0.02) and early-versus-late-EPE (p = 0.03). Conclusions The individual mEPE features have low sensitivity and high specificity. The use of mEPE-score allows for consistent and reliable assessment for pathologic EPE. Key Points • Individual PI-RADS v2.1 mpMRI features had poor sensitivities ranging from 0.08 (0.00–0.15) to 0.71 (0.59–0.83), whereas Sp ranged from 0.68 (0.58–0.79) to 1.00. • mEPE-score is an all-inclusive score for the assessment of pEPE with excellent discriminating ability (i.e., AUC > 0.8) and Se = 0.82, Sp = 0.77, PPV = 0.74, and NPV = 0.84 with a threshold of 3. • The diagnostic performance of the expert reader and beginner reader with pEPE-score was comparable (p = 0.32). Supplementary Information The online version contains supplementary material available at 10.1007/s00330-022-08595-9.
Collapse
Affiliation(s)
- Marco Gatti
- Department of Surgical Sciences, Radiology Unit, University of Turin, Via Genova 3, 10126, Turin, Italy.
| | - Riccardo Faletti
- Department of Surgical Sciences, Radiology Unit, University of Turin, Via Genova 3, 10126, Turin, Italy
| | - Francesco Gentile
- Department of Surgical Sciences, Radiology Unit, University of Turin, Via Genova 3, 10126, Turin, Italy
| | - Enrico Soncin
- Department of Surgical Sciences, Radiology Unit, University of Turin, Via Genova 3, 10126, Turin, Italy
| | - Giorgio Calleris
- Urology Unit, Department of Surgical Sciences, University of Turin, Turin, Italy
| | - Alberto Fornari
- Radiology Unit, Mauriziano Umberto I Hospital, 10128, Turin, Italy
| | - Marco Oderda
- Urology Unit, Department of Surgical Sciences, University of Turin, Turin, Italy
| | - Alessandro Serafini
- Department of Surgical Sciences, Radiology Unit, University of Turin, Via Genova 3, 10126, Turin, Italy
| | | | - Elena Vissio
- Pathology Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Laura Bergamasco
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Stefano Cirillo
- Radiology Unit, Mauriziano Umberto I Hospital, 10128, Turin, Italy
| | - Mauro Giulio Papotti
- Pathology Unit, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Paolo Gontero
- Urology Unit, Department of Surgical Sciences, University of Turin, Turin, Italy
| | - Paolo Fonio
- Department of Surgical Sciences, Radiology Unit, University of Turin, Via Genova 3, 10126, Turin, Italy
| |
Collapse
|
35
|
Urase Y, Ueno Y, Tamada T, Sofue K, Takahashi S, Hinata N, Harada K, Fujisawa M, Murakami T. Comparison of prostate imaging reporting and data system v2.1 and 2 in transition and peripheral zones: evaluation of interreader agreement and diagnostic performance in detecting clinically significant prostate cancer. Br J Radiol 2022; 95:20201434. [PMID: 33882243 PMCID: PMC8978254 DOI: 10.1259/bjr.20201434] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
OBJECTIVE To evaluate the interreader agreement and diagnostic performance of the Prostate Imaging Reporting and Data System (PI-RADS) v. 2.1, in comparison with v. 2. METHODS Institutional review board approval was obtained for this retrospective study. 77 consecutive patients who underwent a prostate multiparametric magnetic resonance imaging at 3.0 T before radical prostatectomy were included. Four radiologists (two experienced uroradiologists and two inexperienced radiologists) independently scored eight regions [six peripheral zones (PZ) and two transition zones (TZ)] using v. 2.1 and v. 2. Interreader agreement was assessed using κ statistics. To evaluate diagnostic performance for clinically significant prostate cancer (csPC), area under the curve (AUC) was estimated. RESULTS 228 regions were pathologically diagnosed as positive for csPC. With a cut-off ≥3, the agreement among all readers was better with v. 2.1 than v. 2 in TZ, PZ, or both zones combined (κ-value: TZ, 0.509 vs 0.414; PZ, 0.686 vs 0.568; both zones combined, 0.644 vs 0.531). With a cut-off ≥4, the agreement among all readers was also better with v. 2.1 than v. 2 in the PZ or both zones combined (κ-value: PZ, 0.761 vs 0.701; both zones combined, 0.756 vs 0.709). For all readers, AUC with v. 2.1 was higher than with v. 2 (TZ, 0.826-0.907 vs 0.788-0.856; PZ, 0.857-0.919 vs 0.853-0.902). CONCLUSION Our study suggests that the PI-RADS v. 2.1 could improve the interreader agreement and might contribute to improved diagnostic performance compared with v. 2. ADVANCES IN KNOWLEDGE PI-RADS v. 2.1 has a potential to improve interreader variability and diagnostic performance among radiologists with different levels of expertise.
Collapse
Affiliation(s)
- Yasuyo Urase
- Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yoshiko Ueno
- Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tsutomu Tamada
- Departmentof Radiology, Kawasaki Medical School, Kurashiki, Japan
| | - Keitaro Sofue
- Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan
| | | | - Nobuyuki Hinata
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kenichi Harada
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masato Fujisawa
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takamichi Murakami
- Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
36
|
Saïer E, Pinar U, De La Taille A, Irani J, Salomon L. Impact des marges chirurgicales positives après prostatectomie radicale chez les patients atteints d’un cancer de la prostate localisé pT2. Prog Urol 2022; 32:354-362. [DOI: 10.1016/j.purol.2022.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 01/25/2022] [Accepted: 01/28/2022] [Indexed: 10/19/2022]
|
37
|
Wang Y, You S, Su S, Yeon A, Lo EM, Kim S, Mohler JL, Freeman MR, Kim HL. Cholesterol-Lowering Intervention Decreases mTOR Complex 2 Signaling and Enhances Antitumor Immunity. Clin Cancer Res 2022; 28:414-424. [PMID: 34728526 PMCID: PMC8776603 DOI: 10.1158/1078-0432.ccr-21-1535] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 08/08/2021] [Accepted: 10/28/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE There is a need for strategies to prevent prostate cancer. Cholesterol-lowering interventions are employed widely and safely to reduce risk of cardiovascular disease and has been proposed for chemoprevention. Using preclinical models and a window-of-opportunity clinical trial, we describe an adaptive antitumor immunity resulting from cholesterol lowering. EXPERIMENTAL DESIGN Statins do not reliably lower serum cholesterol in mice. Therefore, oral ezetimibe was administered to mice to lower serum cholesterol to clinically relevant levels and evaluated the final adaptive immune response. T-lymphocytes-specific mTORC2 knockout mice were used to evaluate mTOR signaling and antitumor immunity. Pretreatment and posttreatment prostate tumors and lymphocytes were examined from a window-of-opportunity clinical trial where men with prostate cancer were treated with 2 to 6 weeks of aggressive cholesterol-lowering intervention prior to radical prostatectomy. RESULTS Mice treated with oral ezetimibe exhibited enhanced antitumor immunity against syngeneic cancers in a CD8+ lymphocyte-dependent manner, produced immunity that was transferrable through lymphocytes, and had enhanced central CD8+ T-cell memory. In mice and in patients undergoing prostatectomy, lowering serum cholesterol inhibited mTORC2 signaling in lymphocytes and increased infiltration of CD8+ lymphocytes into prostate tumors. T-lymphocyte-specific mTORC2 knockout mice demonstrated enhanced CD8+ lymphocyte function and antitumor capacity. In patients, cholesterol-lowering intervention prior to prostatectomy decreased the proliferation of normal prostate and low-grade adenocarcinomas. CONCLUSIONS Lowering serum cholesterol decreased signaling through mTORC2 and enhanced antitumor CD8+ T-cell memory. We provide a rationale for large-scale clinical testing of cholesterol lowering strategies for prostate cancer chemoprevention.
Collapse
Affiliation(s)
- Yanping Wang
- Department of Surgery/Division of Urology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Sungyong You
- Department of Surgery/Division of Urology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Shengchen Su
- Department of Surgery/Division of Urology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Austin Yeon
- Department of Surgery/Division of Urology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Eric M Lo
- Department of Surgery/Division of Urology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Sungjin Kim
- Biostatistics and Bioinformatics Core, Cedars-Sinai Medical Center, Los Angeles, California
| | - James L Mohler
- Department of Urology, Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Michael R Freeman
- Department of Surgery/Division of Urology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Hyung L Kim
- Department of Surgery/Division of Urology, Cedars-Sinai Medical Center, Los Angeles, California.
| |
Collapse
|
38
|
Duran A, Dussert G, Rouviére O, Jaouen T, Jodoin PM, Lartizien C. ProstAttention-Net: a deep attention model for prostate cancer segmentation by aggressiveness in MRI scans. Med Image Anal 2022; 77:102347. [DOI: 10.1016/j.media.2021.102347] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 12/20/2021] [Accepted: 12/31/2021] [Indexed: 11/27/2022]
|
39
|
Heuvel JO, de Wit-van der Veen BJ, van der Poel HG, van Leeuwen PJ, Bekers EM, Grootendorst MR, Vyas KN, Slump CH, Stokkel MPM. Cerenkov Luminescence Imaging in Prostate Cancer: Not the Only Light That Shines. J Nucl Med 2022; 63:29-35. [PMID: 33931467 PMCID: PMC8717187 DOI: 10.2967/jnumed.120.260034] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 04/01/2021] [Indexed: 12/19/2022] Open
Abstract
Cerenkov luminescence imaging (CLI) is a novel imaging technology that might have the ability to assess surgical margins intraoperatively during prostatectomy using 68Ga-prostate-specific membrane antigen (68Ga-PSMA-11). This study evaluated the accuracy of CLI compared with histopathology and, as an exploratory objective, investigated the characteristics of the identified chemiluminescence signal. Methods: After intravenous injection of a mean 68Ga-PSMA-11 activity of 69 MBq intraoperatively, all excised specimens were imaged with CLI. Areas of increased signal were marked for histopathologic comparison and scored for the likelihood of being a positive surgical margin (PSM) using a 5-point Likert scale. In addition, the chemiluminescence signal was investigated in 3 radioactive and 3 nonradioactive specimens using CLI. Results: In 15 patients, the agreement between CLI and histopathology was 60%; this improved to 83% when including close surgical margins (≤1 mm). In 6 hot spots, CLI correctly identified PSMs on histopathology, located at the apex and mid prostate. In all 15 patients, an increased signal at the prostate base was observed, without the presence of the primary tumor in this area in 8 patients. This chemiluminescence signal was also observed in nonradioactive prostate specimens, with a half-life of 48 ± 11 min. The chemiluminescence hampered the visual interpretation of 4 PSMs at the base. Conclusion: CLI was able to correctly identify margin status, including close margins, in 83% of the cases. The presence of a diathermy-induced chemiluminescent signal hampered image interpretation, especially at the base of the prostate. In the current form, CLI is most applicable to detect PSMs and close margins in the apex and mid prostate.
Collapse
Affiliation(s)
- Judith Olde Heuvel
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Technical Medical Centre, University of Twente, Enschede, The Netherlands
| | | | - Henk G van der Poel
- Department of Urology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Pim J van Leeuwen
- Department of Urology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Elise M Bekers
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands; and
| | | | - Kunal N Vyas
- Lightpoint Medical Ltd., Chesham, United Kingdom
| | - Cornelis H Slump
- Technical Medical Centre, University of Twente, Enschede, The Netherlands
| | - Marcel P M Stokkel
- Department of Nuclear Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
40
|
Radical or Not-So-Radical Prostatectomy: Do Surgical Margins Matter? Cancers (Basel) 2021; 14:cancers14010013. [PMID: 35008178 PMCID: PMC8749855 DOI: 10.3390/cancers14010013] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/17/2021] [Accepted: 12/20/2021] [Indexed: 02/02/2023] Open
Abstract
Simple Summary Prostate cancer is the second most common noncutaneous malignancy in men. Prostatectomy is a commonly used treatment modality for selected patients. The prostate’s ill-defined borders and its vicinity with vital structures complicate the wide excision of the organ, resulting in positive margins of resection. Neoplastic infiltration of margins of resection in prostatectomy specimens affects patients’ prognosis. The surgical technique and surgeons’ expertise affect the incidence of margin positivity. The location and the extent of positive margins diversify the risk of recurrence, with basal infiltration and multifocal foci of positive margins behaving more aggressively. Pathologists are encouraged to thoroughly report the status of margins of resection, as they provide important information for patients’ prognosis and enable the clinician to decide upon the most appropriate subsequent therapeutic steps. Abstract Prostate cancer is the second most common malignancy in men, and prostatectomy is the treatment of choice for most patients with at least low risk of progression. The presence of positive margins in the radical prostatectomy specimen is considered an adverse pathologic feature, and may prompt additional therapeutic intervention in the patients. The absence of a distinct capsule around the prostate and intraoperative manipulations that aim to minimize postoperative adverse effects, complicate its wide removal. Proper handling of the specimen during the gross processing is essential for accurate determination of the status of margins or resection. Positive margins, defined as the presence of neoplastic glands in the highlighted-with-ink margin of resection, range from 6–38%. The surgical technique, surgeon’s expertise and tumor (i.e., grade and stage) and patients’ (i.e., BMI) characteristics affect the rate of margin positivity. Extensive or multifocal and nonanterior/nonapical positive margins are linked with higher recurrence rates, especially in organ-confined disease, underscoring the need for treating these patients more aggressively. In summary, detailed description of the status of the margins should be performed in every pathology report to determine patients’ prognosis and the most appropriate therapeutic plan.
Collapse
|
41
|
Iakymenko OA, Briski LM, Punnen S, Nemov I, Lugo I, Jorda M, Parekh DJ, Gonzalgo ML, Kryvenko ON. Variance of Tumor Grade at Radical Prostatectomy With Assessment of Each Tumor Nodule Versus Global Grading. Arch Pathol Lab Med 2021; 146:1032-1036. [PMID: 34752602 DOI: 10.5858/arpa.2021-0279-oa] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2021] [Indexed: 11/06/2022]
Abstract
CONTEXT.— Multifocal prostate cancer at radical prostatectomy (RP) may be graded with assessment of each individual tumor nodule (TN) or global grading of all TNs in aggregate. OBJECTIVE.— To assess case-level grade variability between these 2 grading approaches. DESIGN.— We reviewed 776 RPs with multifocal prostate cancer with 2 or more separate TNs of different Grade Groups (GGs). Two separate grades were assigned to each RP: one based on the TN with the highest grade and a global grade based on the Gleason pattern volumes for all TNs. We then compared the results of these 2 methods. RESULTS.— The case-level grade changed by 1 or more GGs between the 2 grading methods in 35% (132 of 374) of GG3 through GG5 cases. Twelve percent (37 of 309) of GG2 cases with Gleason pattern 4 more than 5% based on individual TN grading decreased their Gleason pattern 4 to less than 5% based on the global approach. Minor tertiary pattern 5 (Gleason pattern 5 <5%) was observed in 6.8% (11 of 161) of GG4 (Gleason score 3 + 5 = 8 and 5 + 3 = 8) and GG5 cases with global grading. The risk of grade discrepancy between the 2 methods was associated with the highest-grade TN volume (inverse relationship), patient age, and number of TNs (P < .001, P = .003, and P < .001, respectively). CONCLUSIONS.— The global grading approach resulted in a lower grade in 35% of GG3 through GG5 cases compared with grading based on the highest-grade TN. Two significant risk factors for this discrepancy with a global grading approach occur when the highest-grade TN has a relatively small tumor volume and with the higher number of TNs per RP. The observed grade variability between the 2 grading schemes most likely limits the interchangeability of post-RP multi-institutional databases if those institutions use different grading approaches.
Collapse
Affiliation(s)
- Oleksii A Iakymenko
- From the Department of Pathology and Laboratory Medicine (Iakymenko, Briski, Nemov, Lugo, Jorda, Kryvenko), University of Miami Miller School of Medicine, Miami, Florida
| | - Laurence M Briski
- From the Department of Pathology and Laboratory Medicine (Iakymenko, Briski, Nemov, Lugo, Jorda, Kryvenko), University of Miami Miller School of Medicine, Miami, Florida
| | - Sanoj Punnen
- Department of Urology (Punnen, Jorda, Parekh, Gonzalgo, Kryvenko), University of Miami Miller School of Medicine, Miami, Florida.,The Sylvester Comprehensive Cancer Center (Punnen, Jorda, Parekh, Gonzalgo, Kryvenko), University of Miami Miller School of Medicine, Miami, Florida
| | - Ivan Nemov
- From the Department of Pathology and Laboratory Medicine (Iakymenko, Briski, Nemov, Lugo, Jorda, Kryvenko), University of Miami Miller School of Medicine, Miami, Florida
| | - Isabella Lugo
- From the Department of Pathology and Laboratory Medicine (Iakymenko, Briski, Nemov, Lugo, Jorda, Kryvenko), University of Miami Miller School of Medicine, Miami, Florida
| | - Merce Jorda
- From the Department of Pathology and Laboratory Medicine (Iakymenko, Briski, Nemov, Lugo, Jorda, Kryvenko), University of Miami Miller School of Medicine, Miami, Florida.,Department of Urology (Punnen, Jorda, Parekh, Gonzalgo, Kryvenko), University of Miami Miller School of Medicine, Miami, Florida.,The Sylvester Comprehensive Cancer Center (Punnen, Jorda, Parekh, Gonzalgo, Kryvenko), University of Miami Miller School of Medicine, Miami, Florida
| | - Dipen J Parekh
- Department of Urology (Punnen, Jorda, Parekh, Gonzalgo, Kryvenko), University of Miami Miller School of Medicine, Miami, Florida.,The Sylvester Comprehensive Cancer Center (Punnen, Jorda, Parekh, Gonzalgo, Kryvenko), University of Miami Miller School of Medicine, Miami, Florida
| | - Mark L Gonzalgo
- Department of Urology (Punnen, Jorda, Parekh, Gonzalgo, Kryvenko), University of Miami Miller School of Medicine, Miami, Florida.,The Sylvester Comprehensive Cancer Center (Punnen, Jorda, Parekh, Gonzalgo, Kryvenko), University of Miami Miller School of Medicine, Miami, Florida
| | - Oleksandr N Kryvenko
- From the Department of Pathology and Laboratory Medicine (Iakymenko, Briski, Nemov, Lugo, Jorda, Kryvenko), University of Miami Miller School of Medicine, Miami, Florida.,Department of Urology (Punnen, Jorda, Parekh, Gonzalgo, Kryvenko), University of Miami Miller School of Medicine, Miami, Florida.,The Sylvester Comprehensive Cancer Center (Punnen, Jorda, Parekh, Gonzalgo, Kryvenko), University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
42
|
Gun E, Ocal I. Cribriform glands are associated with worse outcome than other pattern 4 subtypes: A study of prognostic and clinicopathological characteristics of prostate adenocarcinoma with an emphasis on Grade Groups. Int J Clin Pract 2021; 75:e14722. [PMID: 34390077 DOI: 10.1111/ijcp.14722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 08/09/2021] [Indexed: 11/26/2022] Open
Abstract
AIM Although prostate adenocarcinoma is the most common cancer in men, survival is quite high and with the help of histopathological examination using the updated classification, patient management strategies are developing. We aimed to evaluate the correlation between the histopathological features and biochemical recurrence (BCR) in patients who underwent radical prostatectomy (RP) using the new classification. METHODS A total of 285 prostate adenocarcinoma cases that underwent RP between January 2009 and December 2017 and followed up for at least 3 months were included in the study. The cases were re-evaluated according to WHO-ISUP 2016 classification and the findings were recorded. RESULTS The mean age was 63,4 years. Gleason scores of the cases were as follows: 3+3 144 cases (50.5%), 3+4 81 cases (28.4%), 4+3 28 cases (9.8%), 4+4 7 cases (2.5%) , 3+5 6 cases (2.1%), 5+3 2 cases (0.7%), 4+5 17 cases (6%). There were 198 (69,5%) pT2, 54 (18,9%) pT3a and 33 (11,6%) pT3b cases. The mean follow-up time was 44,1 months and BCR was detected in 97 cases (34%). The relationship between the Group Grades and BCR was statistically significant. BCR rate increased as the tumour volume and the percentage of pattern 4 increased (P < .001).There was a significant correlation between preoperative PSA value, extraprostatic extension, seminal vesicle invasion, surgical margin positivity, tumour volume, pattern 4 percentage, presence of cribriform glands and BCR and recurrence-free survival in both univariate and multivariate analysis and recurrence-free survival was also affected by these parameters. Among the morphological subtypes of Pattern 4, recurrence-free survival decreased as the incidence of cribriform glands increased (P < .001). CONCLUSION Histopathological evaluation is important in predicting BCR in prostate adenocarcinoma, the Group Grade system seems to be helpful in this regard. More studies are needed to prove the relatively worse prognostic effect of cribriform glands.
Collapse
Affiliation(s)
- Eylul Gun
- Department of Pathology, Izmir Katip Celebi University Ataturk Training and Research Hospital, Izmir, Turkey
| | - Irfan Ocal
- Department of Pathology, Izmir Katip Celebi University Ataturk Training and Research Hospital, Izmir, Turkey
| |
Collapse
|
43
|
Samaratunga H, Delahunt B, Yaxley JW, Johannsen S, Egevad L. Intraductal Carcinoma of the Prostate: Extreme Nuclear Size Is Not a Diagnostic Parameter. Am J Surg Pathol 2021; 45:1527-1533. [PMID: 34265803 DOI: 10.1097/pas.0000000000001776] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
High-grade prostatic adenocarcinoma involving duct/acinar structures is labeled intraductal carcinoma of the prostate (IDCP). As numerous studies have shown that IDCP is associated with high stage disease with a significant negative impact on cancer-specific survival, accurate diagnosis is crucial to ensure appropriate patient management. The definition of IDCP recommended by 2016 World Health Organization (WHO) classification suggests that cases of IDCP with micropapillary or loose cribriform architecture without comedonecrosis should have cells with ≥6× nuclear enlargement. It is unclear how this size criterion was derived and which of the parameters of nuclear size (nuclear diameter, nuclear surface area, or nuclear perimeter) it relates to. To evaluate the extent of nuclear enlargement in IDCP, we performed morphometric analyses relating to each of these parameters in 100 radical prostatectomy specimens. One hundred nuclei from foci of IDCP and 50 nuclei from foci of normal luminal epithelium were examined for each patient. Diagnosis of IDCP was based on cells with definite features of carcinoma present within duct/acinar structures. Comparing the means of each of the parameters between IDCP cells and benign luminal cells, there was a statistically significant enlargement in nuclear perimeter (P<0.0005), nuclear area (P<0.0005), and nuclear diameter (P<0.0005); however, the difference in mean nuclear size was limited to factors of 1.3×, 1.6×, and 1.3×, respectively. Three patients each had rare large nuclei (largest perimeter 45, 45, and 44 μm; maximum nuclear area 135, 136, and 136 μm2; and the largest diameter 18 µm in each). For these rare cells, the nuclear size difference, when compared with benign nuclei was; nuclear perimeter 2.0×, 2.1×, and 2.1×; nuclear area 3.6×, 3.8×, and 3.8×; and nuclear maximum diameter 3.0×, 2.5×, and 2.5×. The definition of nuclear enlargement of ≥6× was not reached in any of our cases, all of which clearly showed features of duct invasive carcinoma. In these cases, reliance on nuclear size criteria would have resulted in underdiagnosis of IDCP. This is of concern as failure to recognize IDCP, particularly in needle biopsies, could lead to delays in the timely treatment of aggressive high-grade prostate cancer, resulting in cancer progression and suboptimal patient oncological outcomes.
Collapse
Affiliation(s)
- Hemamali Samaratunga
- Aquesta Uropathology
- Department of Pathology, University of Queensland School of Medicine
| | - Brett Delahunt
- Aquesta Uropathology
- Wellington School of Medicine and Health Sciences, University of Otago, Wellington, New Zealand
| | - John W Yaxley
- Department of Pathology, University of Queensland School of Medicine
- Wesley Hospital, Brisbane, QLD, Australia
| | | | - Lars Egevad
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
44
|
Hayee A, Lugo I, Iakymenko OA, Kwon D, Briski LM, Zhao W, Nemov I, Punnen S, Ritch CR, Pollack A, Jorda M, Stoyanova R, Parekh DJ, Gonzalgo ML, Kryvenko ON. Anterior or Posterior Prostate Cancer Tumor Nodule Location Predicts Likelihood of Certain Adverse Outcomes at Radical Prostatectomy. Arch Pathol Lab Med 2021; 146:833-839. [PMID: 34669939 DOI: 10.5858/arpa.2021-0104-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2021] [Indexed: 11/06/2022]
Abstract
CONTEXT.— Effect of tumor nodule (TN) location in the prostate on adverse radical prostatectomy (RP) outcomes is not well studied in contemporary cohort. OBJECTIVE.— To investigate the significance of TN location with respect to extraprostatic extension (EPE), seminal vesicle invasion (SV+), and positive margin status (SM+) in 1388 RPs. DESIGN.— Each TN at RP was independently graded, staged, and volumetrically assessed. TNs with at least 80% of their volume occupying either the anterior or posterior part of the prostate were categorized accordingly and included in our study, while all other TNs were excluded. RESULTS.— A total of 3570 separate TNs (median = 3 per RP; range = 1-7 per RP) were scored. There were 1320 of 3570 (37%) anterior TNs and 2250 of 3570 (63%) posterior TNs. Posterior TNs were more likely to be higher grade, and exhibit EPE (18% versus 9.4%) and SV+ (4% versus 0.15%), all P < .001. Anterior TNs with EPE were more likely to exhibit SM+ than posterior TNs with EPE (62% versus 30.8%, P < .001). TN location, grade, and volume were significant factors associated with adverse RP outcomes in our univariable analysis. When we controlled for grade and tumor volume in a multivariable analysis using anterior TN location as a reference, posterior TN location was an independent predictor of EPE and SV+ and was less likely to be associated with SM+ (odds ratio = 3.1, 81.5, and 0.7, respectively). CONCLUSIONS.— These associations may be useful in preoperative surgical planning, particularly with respect to improving radiographic analysis of prostate cancer.
Collapse
Affiliation(s)
- Amin Hayee
- From the Department of Pathology and Laboratory Medicine (Hayee, Lugo, Iakymenko, Briski, Nemov, Jorda, Kryvenko), at the University of Miami Miller School of Medicine, Miami, Florida
| | - Isabella Lugo
- From the Department of Pathology and Laboratory Medicine (Hayee, Lugo, Iakymenko, Briski, Nemov, Jorda, Kryvenko), at the University of Miami Miller School of Medicine, Miami, Florida
| | - Oleksii A Iakymenko
- From the Department of Pathology and Laboratory Medicine (Hayee, Lugo, Iakymenko, Briski, Nemov, Jorda, Kryvenko), at the University of Miami Miller School of Medicine, Miami, Florida
| | - Deukwoo Kwon
- Department of Public Health Sciences (Kwon), at the University of Miami Miller School of Medicine, Miami, Florida.,Sylvester Comprehensive Cancer Center (Kwon, Zhao, Punnen, Ritch, Pollack, Jorda, Parekh, Gonzalgo, Kryvenko), at the University of Miami Miller School of Medicine, Miami, Florida
| | - Laurence M Briski
- From the Department of Pathology and Laboratory Medicine (Hayee, Lugo, Iakymenko, Briski, Nemov, Jorda, Kryvenko), at the University of Miami Miller School of Medicine, Miami, Florida
| | - Wei Zhao
- Sylvester Comprehensive Cancer Center (Kwon, Zhao, Punnen, Ritch, Pollack, Jorda, Parekh, Gonzalgo, Kryvenko), at the University of Miami Miller School of Medicine, Miami, Florida
| | - Ivan Nemov
- From the Department of Pathology and Laboratory Medicine (Hayee, Lugo, Iakymenko, Briski, Nemov, Jorda, Kryvenko), at the University of Miami Miller School of Medicine, Miami, Florida
| | - Sanoj Punnen
- Department of Urology (Punnen, Ritch, Jorda, Parekh, Gonzalgo, Kryvenko), at the University of Miami Miller School of Medicine, Miami, Florida.,Sylvester Comprehensive Cancer Center (Kwon, Zhao, Punnen, Ritch, Pollack, Jorda, Parekh, Gonzalgo, Kryvenko), at the University of Miami Miller School of Medicine, Miami, Florida
| | - Chad R Ritch
- Department of Urology (Punnen, Ritch, Jorda, Parekh, Gonzalgo, Kryvenko), at the University of Miami Miller School of Medicine, Miami, Florida.,Sylvester Comprehensive Cancer Center (Kwon, Zhao, Punnen, Ritch, Pollack, Jorda, Parekh, Gonzalgo, Kryvenko), at the University of Miami Miller School of Medicine, Miami, Florida
| | - Alan Pollack
- Radiation Oncology (Pollack, Stoyanova), at the University of Miami Miller School of Medicine, Miami, Florida.,Sylvester Comprehensive Cancer Center (Kwon, Zhao, Punnen, Ritch, Pollack, Jorda, Parekh, Gonzalgo, Kryvenko), at the University of Miami Miller School of Medicine, Miami, Florida
| | - Merce Jorda
- From the Department of Pathology and Laboratory Medicine (Hayee, Lugo, Iakymenko, Briski, Nemov, Jorda, Kryvenko), at the University of Miami Miller School of Medicine, Miami, Florida.,Department of Urology (Punnen, Ritch, Jorda, Parekh, Gonzalgo, Kryvenko), at the University of Miami Miller School of Medicine, Miami, Florida.,Sylvester Comprehensive Cancer Center (Kwon, Zhao, Punnen, Ritch, Pollack, Jorda, Parekh, Gonzalgo, Kryvenko), at the University of Miami Miller School of Medicine, Miami, Florida
| | - Radka Stoyanova
- Radiation Oncology (Pollack, Stoyanova), at the University of Miami Miller School of Medicine, Miami, Florida
| | - Dipen J Parekh
- Department of Urology (Punnen, Ritch, Jorda, Parekh, Gonzalgo, Kryvenko), at the University of Miami Miller School of Medicine, Miami, Florida.,Sylvester Comprehensive Cancer Center (Kwon, Zhao, Punnen, Ritch, Pollack, Jorda, Parekh, Gonzalgo, Kryvenko), at the University of Miami Miller School of Medicine, Miami, Florida
| | - Mark L Gonzalgo
- Department of Urology (Punnen, Ritch, Jorda, Parekh, Gonzalgo, Kryvenko), at the University of Miami Miller School of Medicine, Miami, Florida.,Sylvester Comprehensive Cancer Center (Kwon, Zhao, Punnen, Ritch, Pollack, Jorda, Parekh, Gonzalgo, Kryvenko), at the University of Miami Miller School of Medicine, Miami, Florida
| | - Oleksandr N Kryvenko
- From the Department of Pathology and Laboratory Medicine (Hayee, Lugo, Iakymenko, Briski, Nemov, Jorda, Kryvenko), at the University of Miami Miller School of Medicine, Miami, Florida.,Department of Urology (Punnen, Ritch, Jorda, Parekh, Gonzalgo, Kryvenko), at the University of Miami Miller School of Medicine, Miami, Florida.,Sylvester Comprehensive Cancer Center (Kwon, Zhao, Punnen, Ritch, Pollack, Jorda, Parekh, Gonzalgo, Kryvenko), at the University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
45
|
Dinneen E, Oxley J, Shaw G. Re: Bernardo Rocco, Luca Sarchi, Simone Assumma, et al. Digital Frozen Sections with Fluorescence Confocal Microscopy During Robot-assisted Radical Prostatectomy: Surgical Technique. Eur Urol. In press. https://doi.org/10.1016/j.eururo.2021.03.021. Eur Urol 2021; 80:e120-e121. [PMID: 34489141 DOI: 10.1016/j.eururo.2021.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 08/20/2021] [Indexed: 10/20/2022]
Affiliation(s)
- Eoin Dinneen
- Division of Surgery and Interventional Science, University College London, London, UK; Department of Urology, University College Hospital London, London, UK.
| | - Jon Oxley
- Department of Histopathology, Southmead Hospital, North Bristol Hospitals Trust, Bristol, UK
| | - Greg Shaw
- Division of Surgery and Interventional Science, University College London, London, UK; Department of Urology, University College Hospital London, London, UK
| | | |
Collapse
|
46
|
Iakymenko OA, Lugo I, Briski LM, Nemov I, Punnen S, Kwon D, Pollack A, Stoyanova R, Parekh DJ, Jorda M, Gonzalgo ML, Kryvenko ON. Percentage of Gleason pattern 4 and tumor volume predict adverse pathological stage and margin status at radical prostatectomy in grade Group 2 and grade Group 3 prostate cancers. Prostate 2021; 81:866-873. [PMID: 34184782 DOI: 10.1002/pros.24183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 06/01/2021] [Accepted: 06/07/2021] [Indexed: 11/06/2022]
Abstract
BACKGROUND Increasing percentages of Gleason pattern 4 (GP4%) in radical prostatectomy (RP) correlate with an increased likelihood of nonorgan-confined disease and earlier biochemical recurrence (BCR). However, there are no detailed RP studies assessing the impact of GP4% and corresponding tumor volume (TV) on extraprostatic extension (EPE), seminal vesicle (SV) invasion (SV+), and positive surgical margin (SM) status (SM+). METHODS In 1301 consecutive RPs, we analyzed each tumor nodule (TN) for TV, Grade Group (GG), presence of focal versus nonfocal EPE, SV+ , and SM+. Using GG1 (GP4% = 0) TNs as a reference, we recorded GP4% for all GG2 or GG3 TNs. We performed a multivariable analysis (MVA) using a mixed effects logistic regression that tested significant variables for risk of EPE, SV+, and SM+, as well as a multinomial logistic regression model that tested significant variables for risks of nonorgan-confined disease (pT2+, pT3a, and pT3b) versus organ-confined disease (pT2). RESULTS We identified 3231 discrete TNs ranging from 1 to 7 (median: 2.5) per RP. These included GG1 (n = 2115), GG2 (n = 818), GG3 (n = 274), and GG4 (n = 24) TNs. Increasing GP4% weakly paralleled increasing TV (tau = 0.07, p < .001). In MVA, increasing GP4% and TV predicted a greater likelihood of EPE (odds ratio [OR]: 1.03 and 4.41), SV+ (OR: 1.03 and 3.83), and SM+ (1.01, p = .01 and 2.83), all p < .001. Our multinomial logistic regression model demonstrated an association between GP4% and the risk of EPE (i.e., pT3a and pT3b disease), as well as an association between TV and risk of upstaging (all p < .001). CONCLUSIONS Both GP4% and TV are independent predictors of adverse pathological stage and margin status at RP. However, the risks for adverse outcomes associated with GP4% are marginal, while those for TV are strong. The prognostic significance of GP4% on BCR-free survival has not been studied controlling for TV and other adverse RP findings. Whether adverse pathological stage and margin status associated with larger TV could decrease BCR-free survival to a greater extent than increasing RP GP4% remains to be studied.
Collapse
Affiliation(s)
- Oleksii A Iakymenko
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Isabella Lugo
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Laurence M Briski
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Ivan Nemov
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Sanoj Punnen
- Department of Urology, University of Miami Miller School of Medicine, Miami, Florida, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Deukwoo Kwon
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Alan Pollack
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, Florida, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Radka Stoyanova
- Department of Radiation Oncology, University of Miami Miller School of Medicine, Miami, Florida, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Dipen J Parekh
- Department of Urology, University of Miami Miller School of Medicine, Miami, Florida, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Merce Jorda
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
- Department of Urology, University of Miami Miller School of Medicine, Miami, Florida, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Mark L Gonzalgo
- Department of Urology, University of Miami Miller School of Medicine, Miami, Florida, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Oleksandr N Kryvenko
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
- Department of Urology, University of Miami Miller School of Medicine, Miami, Florida, USA
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
47
|
Costa DN, Jia L, Subramanian N, Xi Y, Rofsky NM, Recchimuzzi DZ, de Leon AD, Arraj P, Pedrosa I. Prospective PI-RADS v2.1 Atypical Benign Prostatic Hyperplasia Nodules With Marked Restricted Diffusion: Detection of Clinically Significant Prostate Cancer on Multiparametric MRI. AJR Am J Roentgenol 2021; 217:395-403. [PMID: 32876473 DOI: 10.2214/ajr.20.24370] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND. On the basis of expert consensus, PI-RADS version 2.1 (v2.1) introduced the transition zone (TZ) atypical benign prostatic hyperplasia (BPH) nodule, defined as a TZ lesion with an incomplete or absent capsule (T2 score, 2). PI-RADS v2.1 also included a revised scoring pathway whereby such nodules, if exhibiting marked restricted diffusion (DWI score, 4-5), are upgraded from overall PI-RADS category 2 to category 3 (2 + 1 TZ lesions). OBJECTIVE. The purpose of this study was to compare the rates of detection of clinically significant prostate cancer (csPCa) in prospectively reported 2 + 1 TZ lesions, as defined by PI-RADS v2.1, and conventional 3 + 0 TZ lesions with targeted biopsy as the reference standard. METHODS. This retrospective study included men with no known PCa or with treatment-naïve grade group (GG) 1 PCa who underwent 3-T multiparametric MRI of the prostate with prospective reporting by means of PI-RADS v2.1. Patients with at least one PI-RADS category 3 TZ lesion who underwent targeted biopsy formed the final sample. Biopsy results were summarized descriptively for 2 + 1 and 3 + 0 lesions. Generalized estimating equations were used to compare csPCa detection rates between groups. Associations between csPCa in 2 + 1 lesions and patient age, PSA level, prostate volume, PSA density, biopsy history, lesion size, and lesion ADC were tested with Kruskal-Wallis and Fisher exact tests. RESULTS. Among 1238 eligible patients who underwent MRI reported with PI-RADS v2.1, 2 + 1 lesions were reported in 6% (n = 69) and 3 + 0 TZ lesions in 7% (n = 87) of patients. No PCa, GG1 PCa, or csPCa was found in 84% (n = 41), 10% (n = 5), and 6% (n = 3) of 49 patients with 2 + 1 lesions who underwent targeted biopsy. Nor were they found in 74% (n = 45), 15% (n = 9), and 11% (n = 7) of 61 patients with 3 + 0 lesions who underwent targeted biopsy. The csPCa detection rate was not significantly different between 2 + 1 and 3 + 0 lesions (p = .31). All cases of csPCa were GG2, except for one 3 + 0 lesion with a GG3 tumor. No clinical or imaging variable was associated with csPCa in 2 + 1 lesions. CONCLUSION. The rate of csPCa in atypical BPH nodules with marked restricted diffusion was low (6%) and not significantly different from that of conventional 3 + 0 TZ lesions (11%). CLINICAL IMPACT. The results provide prospective clinical data about the revised TZ scoring criterion and pathway in PI-RADS v2.1 for atypical BPH nodules with marked restricted diffusion.
Collapse
Affiliation(s)
- Daniel N Costa
- Department of Radiology, UT Southwestern Medical Center, Clements Imaging Bldg (NE2.210), 5323 Harry Hines Blvd, Dallas, TX 75390
| | - Liwei Jia
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX
| | - Naveen Subramanian
- Department of Radiology, UT Southwestern Medical Center, Clements Imaging Bldg (NE2.210), 5323 Harry Hines Blvd, Dallas, TX 75390
| | - Yin Xi
- Department of Radiology, UT Southwestern Medical Center, Clements Imaging Bldg (NE2.210), 5323 Harry Hines Blvd, Dallas, TX 75390
| | - Neil M Rofsky
- Department of Radiology, UT Southwestern Medical Center, Clements Imaging Bldg (NE2.210), 5323 Harry Hines Blvd, Dallas, TX 75390
| | - Debora Z Recchimuzzi
- Department of Radiology, UT Southwestern Medical Center, Clements Imaging Bldg (NE2.210), 5323 Harry Hines Blvd, Dallas, TX 75390
| | - Alberto Diaz de Leon
- Department of Radiology, UT Southwestern Medical Center, Clements Imaging Bldg (NE2.210), 5323 Harry Hines Blvd, Dallas, TX 75390
| | - Patrick Arraj
- Department of Radiology, UT Southwestern Medical Center, Clements Imaging Bldg (NE2.210), 5323 Harry Hines Blvd, Dallas, TX 75390
| | - Ivan Pedrosa
- Department of Radiology, UT Southwestern Medical Center, Clements Imaging Bldg (NE2.210), 5323 Harry Hines Blvd, Dallas, TX 75390
| |
Collapse
|
48
|
Okubo Y, Sato S, Osaka K, Yamamoto Y, Suzuki T, Ida A, Yoshioka E, Suzuki M, Washimi K, Yokose T, Kishida T, Miyagi Y. Clinicopathological Analysis of the ISUP Grade Group And Other Parameters in Prostate Cancer: Elucidation of Mutual Impact of the Various Parameters. Front Oncol 2021; 11:695251. [PMID: 34395260 PMCID: PMC8356042 DOI: 10.3389/fonc.2021.695251] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/13/2021] [Indexed: 01/29/2023] Open
Abstract
Background Prostate cancer has become increasingly common worldwide. Although Grade group (GG) is widely accepted as an indicator of prostate cancer grade, there are malignancies that cannot be defined by GG alone. Moreover, the relationship between GG and other parameters remains unclear. Herein, we aimed to explore the biological characteristics of prostate cancer. Methods This study included 299 radical prostatectomy cases. The Chi-square test and analysis of variance were used to analyze the association of GG with binary and continuous variables. We then conducted morphological analyses. Multivariate analyses were performed to extract the data on risk factors for biochemical recurrence (BCR) and lymph node metastasis. Results The lymphatic, venous, perineural, and seminal vesicle invasion rates were 37/299 (12.4%), 25/299 (8.4%), 280/299 (93.6%), and 23/299 (7.7%), respectively. The extraprostatic extension (EPE), positive surgical margin, tertiary Gleason pattern 5, intraductal carcinoma of the prostate gland, and lymph node metastasis rates were 89/299 (29.8%), 106/299 (35.5%), 33/260 (12.7%), 56/299 (18.7%), and 23/299 (7.7%), respectively. As GG increased, various parameters became easier to visualize; however, there were differences between the parameters. Postoperative BCR was observed in 31/242 (12.8%) cases without preoperative hormone therapy; GG2, GG3, GG4, and GG5 accounted for 4, 7, 7, and 13 cases, respectively. Multivariate analyses revealed that GG and tumor diameter were significant risk factors for early BCR, whereas lymphatic invasion, EPE, and seminal vesicle invasion were significant risk factors for lymph node metastasis. For BCR, the odds ratios (ORs) for GG and tumor diameter were 2.253 (95% confidence interval (CI]): 1.297–3.912; P=0.004) and 1.074 (95% CI: 1.011–1.142; P=0.022), respectively. For lymph node metastasis, ORs for the presence of lymphatic invasion, EPE, and seminal vesicle invasion were 7.425 (95% CI: 1.688–22.583; P=0.004), 4.391 (95% CI: 1.037–18.589; P=0.044), and 5.755 (95% CI: 1.308–25.316; P=0.021), respectively. Conclusions We summarized various parameters correlating with each GG. Through multivariate analyses, we established the independent risk factors for early BCR and lymph node metastasis. In addition to GG, other important indices of malignancy were determined and weighted to provide a basis for future investigations.
Collapse
Affiliation(s)
- Yoichiro Okubo
- Department of Pathology, Kanagawa Cancer Center, Kanagawa, Japan
| | - Shinya Sato
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Kanagawa, Japan
| | - Kimito Osaka
- Department of Urology, Kanagawa Cancer Center, Kanagawa, Japan
| | - Yayoi Yamamoto
- Department of Radiology, Kanagawa Cancer Center, Kanagawa, Japan
| | - Takahisa Suzuki
- Department of Urology, Kanagawa Cancer Center, Kanagawa, Japan
| | - Arika Ida
- Department of Pathology, Kanagawa Cancer Center, Kanagawa, Japan
| | - Emi Yoshioka
- Department of Pathology, Kanagawa Cancer Center, Kanagawa, Japan
| | - Masaki Suzuki
- Department of Pathology, Kanagawa Cancer Center, Kanagawa, Japan.,Department of Pathology, University of Tokyo Hospital, Tokyo, Japan
| | - Kota Washimi
- Department of Pathology, Kanagawa Cancer Center, Kanagawa, Japan
| | - Tomoyuki Yokose
- Department of Pathology, Kanagawa Cancer Center, Kanagawa, Japan
| | - Takeshi Kishida
- Department of Urology, Kanagawa Cancer Center, Kanagawa, Japan
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Kanagawa, Japan
| |
Collapse
|
49
|
Comparison of Sensitivity and Specificity of Biparametric versus Multiparametric Prostate MRI in the Detection of Prostate Cancer in 431 Men with Elevated Prostate-Specific Antigen Levels. Diagnostics (Basel) 2021; 11:diagnostics11071223. [PMID: 34359307 PMCID: PMC8306749 DOI: 10.3390/diagnostics11071223] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 06/30/2021] [Accepted: 07/05/2021] [Indexed: 12/31/2022] Open
Abstract
(1) Background: the study of dynamic contrast enhancement (DCE) has a limited role in the detection of prostate cancer (PCa), and there is a growing interest in performing unenhanced biparametric prostate-MRI (bpMRI) instead of the conventional multiparametric-MRI (mpMRI). In this study, we aimed to retrospectively compare the performance of the mpMRI, which includes DCE study, and the unenhanced bpMRI, composed of only T2-weighted imaging and diffusion-weighted imaging (DWI), in PCa detection in men with elevated prostate-specific-antigen (PSA) levels. (2) Methods: a 1.5 T MRI, with an endorectal-coil, was performed on 431 men (aged 61.5 ± 8.3 years) with a PSA ≥4.0 ng/mL. The bpMRI and mpMRI tests were independently assessed in separate sessions by two readers with 5 (R1) and 3 (R2) years of experience. The histopathology or ≥2 years follow-up served as a reference standard. The sensitivity and specificity were calculated with their 95% CI, and McNemar’s and Cohen’s κ statistics were used. (3) Results: in 195/431 (45%) of histopathologically proven PCa cases, 62/195 (32%) were high-grade PCa (GS ≥ 7b) and 133/195 (68%) were low-grade PCa (GS ≤ 7a). The PCa could be excluded by histopathology in 58/431 (14%) and by follow-up in 178/431 (41%) of patients. For bpMRI, the sensitivity was 164/195 (84%, 95% CI: 79–89%) for R1 and 156/195 (80%, 95% CI: 74–86%) for R2; while specificity was 182/236 (77%, 95% CI: 72–82%) for R1 and 175/236 (74%, 95% CI: 68–80%) for R2. For mpMRI, sensitivity was 168/195 (86%, 95% CI: 81–91%) for R1 and 160/195 (82%, 95% CI: 77–87%) for R2; while specificity was 184/236 (78%, 95% CI: 73–83%) for R1 and 177/236 (75%, 95% CI: 69–81%) for R2. Interobserver agreement was substantial for both bpMRI (κ = 0.802) and mpMRI (κ = 0.787). (4) Conclusions: the diagnostic performance of bpMRI and mpMRI were similar, and no high-grade PCa was missed with bpMRI.
Collapse
|
50
|
olde Heuvel J, de Wit-van der Veen BJ, Huizing DM, van der Poel HG, van Leeuwen PJ, Bhairosing PA, Stokkel MP, Slump CH. State-of-the-art Intraoperative Imaging Technologies for Prostate Margin Assessment: A Systematic Review. Eur Urol Focus 2021; 7:733-741. [DOI: 10.1016/j.euf.2020.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 01/06/2020] [Accepted: 02/05/2020] [Indexed: 12/29/2022]
|