1
|
Chen G, Li Y, Geng S, Lv L, Wang Y, Li X, Chen S, Shi B. Evaluating the Heterogeneity of Advanced Prostate Cancer by 18F-DCFPyL and 18F-FDG PET/CT in a Prospective Cohort. Prostate 2025; 85:749-757. [PMID: 40045414 DOI: 10.1002/pros.24881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/20/2025] [Accepted: 02/18/2025] [Indexed: 04/30/2025]
Abstract
PURPOSE 18F-DCFPyL (targeted PSMA) and 18F-FDG dual-tracer PET/CT combination with next-generation sequencing was applied in a prospective cohort of men with prostate cancer to identify the clinical and genetic characteristics with heterogeneous PET/CT imaging features. METHODS 104 men with documented prostate cancer underwent 18F-DCFPyL and 18F-FDG PET/CT, of which 83 underwent next-generation sequencing for detecting variation of AR, TP53, RB1, PTEN, etc. Lesions were classified into DCFPyL+FDG± lesions and DCFPyL-FDG+ lesions and analyzed for heterogeneous distribution. We divided the patients with positive lesions into DCFPyL+FDG± group and DCFPyL-FDG+ group, then compared the differences in clinical features and genetic mutations between the two groups with CRPC. RESULTS Overall, 92 men had positive lesions detected. By comparing lesion distribution with the DCFPyL+FDG ± , DCFPyL-FDG+ disease had higher proportions of visceral metastases (4.1% vs. 1.0%, p = 0.002). DCFPyL-FDG+ was more frequently found in CRPC cohorts, and in the CRPC cohort, patients with DCFPyL-FDG+ lesions often had worse PSA response. Exploratory analysis showed that TP53 and/or RB1 mutations might be a risk factor for DCFPyL-FDG+ disease (OR = 10.625, 95% CI 3.492-32.332, p < 0.001). CONCLUSION Patients with DCFPyL-FDG+ lesions were more likely to have visceral metastases detected, be found in castration-resistant cohorts, have TP53 and/or RB1 mutations detected, and have poor therapeutic response compared to patients with DCFPyL+FDG± lesions. Therefore, dual-tracer (18F-DCFPyL and 18F-FDG) PET/CT is recommended for patients with low PSMA expression incompatible with the true burden of the disease and those with TP53 and/or RB1 mutations to better evaluate the disease burden, tumor heterogeneity, and prognosis.
Collapse
Affiliation(s)
- GuangHao Chen
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - YueKai Li
- Department of Nuclear Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - ShangZhen Geng
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - LinChen Lv
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Yong Wang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Xin Li
- Department of Nuclear Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - ShouZhen Chen
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - BenKang Shi
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
2
|
Nieuwenhove SV, Lhommel R, Pasoglou V, Damme JV, Michoux N, Triqueneaux P, Tombal B, Lecouvet FE. Fast (< 30 min) "All-in-One" whole-body MRI for TNM staging in high-risk prostate cancer (PCa): Feasibility and comparison to 68Ga-Prostate Specific Membrane Antigen (PSMA)-PET/CT. Eur J Radiol 2025; 186:112033. [PMID: 40132471 DOI: 10.1016/j.ejrad.2025.112033] [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: 12/02/2024] [Revised: 02/27/2025] [Accepted: 03/03/2025] [Indexed: 03/27/2025]
Abstract
PURPOSE Next-generation imaging techniques, including PSMA-PET/CT and whole-body MRI (WB-MRI), are disrupting the management of prostate cancer (PCa). This study aimed to build a fast "All-In-One" WB-MRI protocol and to compare it to 68Ga-PSMA-PET/CT for local (T), nodal (N), and distant staging (M1a, M1b, M1c). METHODS Fifty-two PCa patients at high-risk for metastases underwent a fast "All-in-One" WB-MRI (combining biparametric prostate assessment based on rapid T2-weighted and diffusion-weighted imaging (DWI) following the PI-RADS v2.1 guidelines and the MET-RADS-P guidelines). This WB-MRI prococol was compared to routinely performed PSMA-PET/CT read according to PSMA-RADS 1.0. Inter-technique agreement on staging and proportion differences in positive findings were assessed using Gwet's AC1 and Exact test. RESULTS Fast "All-in-One" WB-MRI better detected local tumors (T) (87 % vs 77 %, p = 0.063), while PSMA-PET/CT detected more nodes (40 % vs 29 %, p = 0.031) with a median small axis of 5 mm. No proportion difference was observed in the detection of extra nodal metastases (M1b) (p > 0.999). Inter-technique agreement ranged from good (AC1M1a = 0.68, AC1N = 0.79) to very good (AC1T = 0.86, AC1EPE = 0.89, AC1M1b = 0.93, AC1M1c = 0.96). Fewer oligometastatic patients were detected with PSMA-PET/CT (11 %) compared to WB-MRI (19 %). CONCLUSION A fast (<30 min) "All-In-One" WB-MRI protocol can be implemented in clinical routine. It provides better anatomical characterization of the T2/T3 stage than 68Ga-PSMA-PET/CT, similar distant metastasis detection rate, but slightly inferior detection rate of nodal metastases.
Collapse
Affiliation(s)
- Sandy Van Nieuwenhove
- Institut du Cancer Roi Albert 2 (IRA2) and Institut de Recherche Expérimentale & Clinique (IREC) Department of Radiology (IMAG), Université Catholique de Louvain (UCLouvain) - Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Renaud Lhommel
- Institut du Cancer Roi Albert 2 (IRA2) and Institut de Recherche Expérimentale & Clinique (IREC), Department of Nuclear Medicine, Université Catholique de Louvain (UCLouvain) - Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Vassiliki Pasoglou
- Institut du Cancer Roi Albert 2 (IRA2) and Institut de Recherche Expérimentale & Clinique (IREC) Department of Radiology (IMAG), Université Catholique de Louvain (UCLouvain) - Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Julien Van Damme
- Institut du Cancer Roi Albert 2 (IRA2) and Institut de Recherche Expérimentale & Clinique (IREC) Department of Urology and Department of Experimental Surgery and Transplantation (CHEX), Université Catholique de Louvain (UCLouvain) - Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Nicolas Michoux
- Institut du Cancer Roi Albert 2 (IRA2) and Institut de Recherche Expérimentale & Clinique (IREC) Department of Radiology (IMAG), Université Catholique de Louvain (UCLouvain) - Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Perrine Triqueneaux
- Institut du Cancer Roi Albert 2 (IRA2) and Institut de Recherche Expérimentale & Clinique (IREC) Department of Radiology (IMAG), Université Catholique de Louvain (UCLouvain) - Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Bertrand Tombal
- Institut du Cancer Roi Albert 2 (IRA2) and Institut de Recherche Expérimentale & Clinique (IREC) Department of Urology and Department of Experimental Surgery and Transplantation (CHEX), Université Catholique de Louvain (UCLouvain) - Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Frédéric E Lecouvet
- Institut du Cancer Roi Albert 2 (IRA2) and Institut de Recherche Expérimentale & Clinique (IREC) Department of Radiology (IMAG), Université Catholique de Louvain (UCLouvain) - Cliniques Universitaires Saint Luc, Brussels, Belgium.
| |
Collapse
|
3
|
Muglia VF, Laschena L, Pecoraro M, de Lion Gouvea G, Colli LM, Panebianco V. Imaging assessment of prostate cancer recurrence: advances in detection of local and systemic relapse. Abdom Radiol (NY) 2025; 50:807-826. [PMID: 39254707 DOI: 10.1007/s00261-024-04412-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 09/11/2024]
Abstract
Prostate cancer (PCa) relapse, defined either by persistent PSA levels (after RP) or biochemical recurrence (BCR), is a common occurrence. The imaging evaluation of patients experiencing PCa relapse has undergone significant advancements in the past decade, notably with the introduction of new Positron Emission Tomography (PET) tracers such as Prostate-specific membrane antigen (PSMA), and the progress in functional Magnetic Resonance Imaging (MRI). This article will explore the role of traditional imaging, the evolution of MRI towards the development of the Prostate Magnetic Resonance Imaging for Local Recurrence Reporting (PI-RR) scoring system, and how next-generation imaging is enhancing diagnostic accuracy in the setting of PCa relapse, which is essential for adopting personalized strategies that may ultimately impact outcomes.
Collapse
Affiliation(s)
- Valdair Francisco Muglia
- Department of Medical Images, Oncology and Hematology, Ribeirao Preto Medical School, University of Sao Paulo, Hospital Clinicas Ribeirao Preto - Av. Bandeirantes 3900. Campus Monte Alegre -USP, Sao Paulo, 14049-900, Brazil.
| | - Ludovica Laschena
- Department of Radiological Sciences, Oncology and Pathology, Sapienza University, Rome, Italy
| | - Martina Pecoraro
- Department of Radiological Sciences, Oncology and Pathology, Sapienza University, Rome, Italy
| | - Gabriel de Lion Gouvea
- Department of Medical Images, Oncology and Hematology, Ribeirao Preto Medical School, University of Sao Paulo, Hospital Clinicas Ribeirao Preto - Av. Bandeirantes 3900. Campus Monte Alegre -USP, Sao Paulo, 14049-900, Brazil
| | - Leandro Machado Colli
- Department of Medical Images, Oncology and Hematology, Ribeirao Preto Medical School, University of Sao Paulo, Hospital Clinicas Ribeirao Preto - Av. Bandeirantes 3900. Campus Monte Alegre -USP, Sao Paulo, 14049-900, Brazil
| | - Valeria Panebianco
- Department of Radiological Sciences, Oncology and Pathology, Sapienza University, Rome, Italy
| |
Collapse
|
4
|
Hajati A, Herold A, Catalano OA, Harisinghani MG. Urologic Imaging of the Prostate: Cancer and Mimics. Urol Clin North Am 2025; 52:125-138. [PMID: 39537298 DOI: 10.1016/j.ucl.2024.07.012] [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: 11/16/2024]
Abstract
This article provides a comprehensive overview of prostate cancer imaging, including detection of clinically significant cancer and initial staging. The role of multiparametric MRI in detection and local staging is discussed, along with the use of conventional imaging and advanced techniques such as Prostate-Specific Membrane Antigen-Positron Emission Tomography (PSMA-PET) for staging of nodal and distant metastases. The article also highlights the importance of differentiating benign prostatic conditions from prostate cancer on imaging to improve diagnostic accuracy and reduce false-positive interpretations.
Collapse
Affiliation(s)
- Azadeh Hajati
- Department of Radiology, Division of Abdominal Imaging, Harvard Medical School, 55 Fruit Street, White Building, Room 270, Boston, MA 02114, USA
| | - Alexander Herold
- Department of Radiology, Division of Abdominal Imaging, Harvard Medical School, 55 Fruit Street, White Building, Room 270, Boston, MA 02114, USA; Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Onofrio Antonio Catalano
- Department of Radiology, Division of Abdominal Imaging, Harvard Medical School, 55 Fruit Street, White Building, Room 270, Boston, MA 02114, USA
| | - Mukesh G Harisinghani
- Department of Radiology, Division of Abdominal Imaging, Harvard Medical School, 55 Fruit Street, White Building, Room 270, Boston, MA 02114, USA.
| |
Collapse
|
5
|
Rodríguez-Pajuelo A, Guerra-Gómez M, Cuenca Cuenca JI, Freire-Macías JM, Jiménez-Hoyuela García JM, Álvarez-Pérez RM. Impact of delayed pelvic imaging on the staging of biochemical recurrence in prostate cancer patients using [ 18F]DCFPYL PET/CT: a retrospective evaluation. EJNMMI REPORTS 2025; 9:5. [PMID: 39890711 PMCID: PMC11785851 DOI: 10.1186/s41824-025-00238-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 01/03/2025] [Indexed: 02/03/2025]
Abstract
PURPOSE The aim of this study was to evaluate the added diagnostic value of additional second stage pelvic scanning as part of the [18F]DCFPYL PET/CT procedure in patients treated for prostate cancer (PCa) who have biochemical recurrence (BR). MATERIALS AND METHODS Consecutive patients with a diagnosis of PCa who underwent a dual-phase PSMA-PET scan between September 2022 and December 2023, were retrospectively included. We analyzed the number and maximum SUV (SUVmax) of lesions only in the pelvic region (prostate, locoregional lymph nodes and bone), based on PSMA-RADS version 2.0 and miTNM criteria. To assess the potential diagnostic benefit of additional delayed pelvic PET/CT imaging as part of the PSMA-PET procedure, the change in molecular TNM classification was evaluated after the procedure. RESULTS Additional delayed pelvic PET/CT imaging as part of the PSMA-PET procedure resulted in a change in molecular TNM classification in 22 out of 136 patients (16.2%). The highest percentage change was obtained in the miN classification (14/22 patients), followed by the miT classification (7/22) and lastly miM (1/22). Moreover, we found that patients in whom delayed pelvic imaging resulted in a change in molecular TNM classification were significantly older and had a higher PSA level than those in whom delayed imaging did not provide additional information. CONCLUSIONS Pelvic delayed imaging in patients with biochemical recurrence of prostate cancer undergoing PET/CT with [18F]DCFPYL shows a non-negligible influence on patient staging, modifying the miTNM classification in 16.2% of cases, with pelvic lymphatic involvement benefiting the most from the dual study.
Collapse
Affiliation(s)
| | - Miriam Guerra-Gómez
- Nuclear Medicine Department, University Hospital Virgen del Rocío, Seville, USA
| | | | | | | | | |
Collapse
|
6
|
Parillo M, Mallio CA. The Whole-Body MRI Reporting and Data System Guidelines for Prostate Cancer (MET-RADS-P), Multiple Myeloma (MY-RADS), and Cancer Screening (ONCO-RADS). Cancers (Basel) 2025; 17:275. [PMID: 39858056 PMCID: PMC11763526 DOI: 10.3390/cancers17020275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 01/10/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Whole-body magnetic resonance imaging (WB-MRI) is being employed with increasing frequency to evaluate a broader spectrum of patients with diverse types of cancer and for cancer screening purposes. While clinical guidelines support its use, a standardized radiological approach is still lacking. To improve consistency in the acquisition, interpretation, and reporting of WB-MRI examinations, three reporting and data systems (RADSs) have been recently suggested: METastasis Reporting and Data System for Prostate Cancer (MET-RADS-P), Myeloma Response Assessment and Diagnosis System (MY-RADS), and Oncologically Relevant Findings Reporting and Data System (ONCO-RADS). MET-RADS-P was developed to stage and monitor men with advanced prostate cancer using WB-MRI. It has emerged as a reliable imaging biomarker for predicting metastatic disease progression and assessing treatment response. MY-RADS was developed to stage and monitor patients with multiple myeloma using WB-MRI, emerging as a prognostic imaging biomarker. However, the evidence regarding inter-reader agreement for MY-RADS is currently limited. ONCO-RADS was developed to standardize the use of WB-MRI for cancer screening in individuals with cancer predisposition syndromes and in the general population. While initial findings are promising, the evidence supporting its use remains limited. To further validate and expand upon these promising preliminary findings, additional large-scale, prospective, multicenter studies are necessary.
Collapse
Affiliation(s)
- Marco Parillo
- Radiology, Multizonal Unit of Rovereto and Arco, APSS Provincia Autonoma Di Trento, 38123 Trento, Italy
| | - Carlo Augusto Mallio
- Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Roma, Italy
- Research Unit of Diagnostic Imaging and Interventional Radiology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 00128 Roma, Italy
| |
Collapse
|
7
|
Mew A, Chau E, Bera K, Ramaiya N, Tirumani SH. Recommendations from Imaging, Oncology, and Radiology Organizations to Guide Management in Prostate Cancer: Summary of Current Recommendations. Radiol Imaging Cancer 2025; 7:e240091. [PMID: 39792015 PMCID: PMC11791666 DOI: 10.1148/rycan.240091] [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] [Indexed: 01/12/2025]
Abstract
Prostate cancer is the second most common malignancy among male individuals in the United States and requires careful imaging approaches because of its varied presentations. This review examines prostate cancer imaging guidelines from leading organizations, including the American College of Radiology, American Urological Association, European Association of Urology, American Society of Clinical Oncology, and National Comprehensive Cancer Network, and serves as a reference highlighting commonalities and divergences in current imaging recommendations across prostate cancer states. We outline these organizations and their methods, focusing on their approaches to panel expertise, guideline development, evidence grading, and revision schedules. We then compare and contrast the role of various imaging modalities across states of prostate cancer management in the following categories: clinically suspected prostate cancer, clinically established prostate cancer: active surveillance or staging, monitoring metastatic disease, and posttreatment follow-up: recurrent or residual disease. Overall, there is consensus on the importance of multiparametric MRI in diagnosis and staging prior to active surveillance and the emerging role of prostate-specific membrane antigen (PSMA) PET/CT in metastatic and recurrent disease. However, there is disparity in imaging recommendations for detecting metastases in unfavorable intermediate-risk prostate cancer and views on current applications of PSMA PET/CT. Ultimately, variations in radiologic expertise exist among guideline panels, and there continue to be inconsistencies in imaging recommendations in prostate cancer. Keywords: Prostate, Genital/Reproductive, Oncology Supplemental material is available for this article. © RSNA, 2025.
Collapse
Affiliation(s)
- Andy Mew
- From Case Western Reserve University School of Medicine, Cleveland, Ohio (A.M.); University of Pittsburgh School of Medicine, Pittsburgh, Pa (E.C.); and Department of Radiology, University Hospitals, 11000 Euclid Ave, Bolwell B2600, Cleveland, OH 44115 (K.B., N.R., S.H.T.)
| | - Eva Chau
- From Case Western Reserve University School of Medicine, Cleveland, Ohio (A.M.); University of Pittsburgh School of Medicine, Pittsburgh, Pa (E.C.); and Department of Radiology, University Hospitals, 11000 Euclid Ave, Bolwell B2600, Cleveland, OH 44115 (K.B., N.R., S.H.T.)
| | - Kaustav Bera
- From Case Western Reserve University School of Medicine, Cleveland, Ohio (A.M.); University of Pittsburgh School of Medicine, Pittsburgh, Pa (E.C.); and Department of Radiology, University Hospitals, 11000 Euclid Ave, Bolwell B2600, Cleveland, OH 44115 (K.B., N.R., S.H.T.)
| | - Nikhil Ramaiya
- From Case Western Reserve University School of Medicine, Cleveland, Ohio (A.M.); University of Pittsburgh School of Medicine, Pittsburgh, Pa (E.C.); and Department of Radiology, University Hospitals, 11000 Euclid Ave, Bolwell B2600, Cleveland, OH 44115 (K.B., N.R., S.H.T.)
| | - Sree Harsha Tirumani
- From Case Western Reserve University School of Medicine, Cleveland, Ohio (A.M.); University of Pittsburgh School of Medicine, Pittsburgh, Pa (E.C.); and Department of Radiology, University Hospitals, 11000 Euclid Ave, Bolwell B2600, Cleveland, OH 44115 (K.B., N.R., S.H.T.)
| |
Collapse
|
8
|
von Stauffenberg F, Poyet C, Beintner-Skawran S, Maurer A, Schmid FA. Current Clinical Applications of PSMA-PET for Prostate Cancer Diagnosis, Staging, and Treatment. Cancers (Basel) 2024; 16:4263. [PMID: 39766162 PMCID: PMC11674651 DOI: 10.3390/cancers16244263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Over the past decade, prostate-specific membrane antigen positron emission tomography (PSMA-PET) has revolutionized prostate cancer (PCa) imaging, offering greater sensitivity and specificity compared to conventional imaging modalities such as CT, MRI, and bone scintigraphy. PSMA-PET is particularly valuable in staging newly diagnosed patients with intermediate- and high-risk disease, detecting biochemical recurrence, and evaluating metastatic cases. By utilizing radiotracers that accumulate specifically in PSMA-expressing cells, even small metastases can be detected, offering a detailed assessment of cancer extent and enabling more targeted diagnostic evaluations. Among the most utilized radiotracers, [68Ga]- and [18F]-labeled PSMA tracers enable precise imaging even with low disease burden. This diagnostic precision also supports advanced therapeutic approaches, including metastasis-directed therapy for oligometastatic cases and systemic treatment options, such as radioligand therapy, which presents new treatment perspectives for metastatic, castration-resistant PCa. This review examines the evolution of PSMA-PET in the diagnostics and therapy of PCa while comparing the current recommendations from leading clinical guidelines. The integration of PSMA-PET into clinical practice has redefined the management of PCa, improving diagnostic accuracy and enabling personalized treatment strategies, while lacking prospective long-term outcome data. As PSMA-PET continues to expand in clinical application, this review highlights its significant advancements while critically addressing limitations to ensure balanced and evidence-based implementation in prostate cancer care.
Collapse
Affiliation(s)
- Franz von Stauffenberg
- Department of Urology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland;
| | - Cédric Poyet
- Department of Urology, Stadtspital Triemli, 8063 Zurich, Switzerland;
| | - Stephan Beintner-Skawran
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland; (S.B.-S.); (A.M.)
| | - Alexander Maurer
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland; (S.B.-S.); (A.M.)
| | - Florian A. Schmid
- Department of Urology, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland;
| |
Collapse
|
9
|
Padhani AR, Tunariu N, Perez-Lopez R, Tombal B, Lecouvet FE. Evaluating prostate cancer bone metastases response with whole-body MRI: What we know and still need to know. Eur Radiol 2024; 34:7564-7566. [PMID: 38902565 DOI: 10.1007/s00330-024-10864-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 05/12/2024] [Accepted: 05/19/2024] [Indexed: 06/22/2024]
Affiliation(s)
- Anwar R Padhani
- Paul Strickland Scanner Centre, Mount Vernon Cancer Centre, Northwood, United Kingdom.
| | - Nina Tunariu
- Clinical Radiology, The Royal Marsden NHS Foundation Trust, London, UK
- Institute of Cancer Research, Downs Road, Sutton, United Kingdom
| | - Raquel Perez-Lopez
- Radiomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Betrand Tombal
- Department of Surgery, Institut du Cancer Roi Albert II (IRA2) - Institut de Recherche Expérimentale & Clinique (IREC) - UCLouvain - Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Frederic E Lecouvet
- Department of Medical Imaging, Institut du Cancer Roi Albert II (IRA2) - Institut de Recherche Expérimentale & Clinique (IREC) - UCLouvain - Cliniques Universitaires Saint Luc, Brussels, Belgium
| |
Collapse
|
10
|
Esfahani SA, Morris MJ, Sartor O, Frydenberg M, Fanti S, Calais J, Vapiwala N. Standardized template for clinical reporting of PSMA PET/CT scans. Eur J Nucl Med Mol Imaging 2024; 52:335-341. [PMID: 39143250 PMCID: PMC11599343 DOI: 10.1007/s00259-024-06857-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: 03/18/2024] [Accepted: 07/21/2024] [Indexed: 08/16/2024]
Abstract
PURPOSE Accurate diagnosis and staging of prostate cancer are crucial to improving patient care. Prostate-specific membrane antigen (PSMA)-targeted positron emission tomography with computed tomography (PET/CT) imaging has demonstrated superiority for initial staging and restaging in patients with prostate cancer. Referring physicians and PET/CT readers must agree on a consistent communication method and application of information derived from this imaging modality. While several guidelines have been published, a single PSMA PET/CT reporting template has yet to be widely adopted. Based on the consensus from community and academic physicians, we developed a standardized PSMA PET/CT reporting template for radiologists and nuclear medicine physicians to report and relay key imaging findings to referring physicians. The aim was to improve the quality, clarity, and utility of imaging results reporting to facilitate patient management decisions. METHODS Based on community and expert consensus, we developed a standardized PSMA PET/CT reporting template to deliver key imaging findings to referring clinicians. RESULTS Core category components proposed include a summary of any prior treatment history; presence, location, and degree of PSMA radiopharmaceutical uptake in primary and/or metastatic tumor(s), lesions with no uptake, and incidentally found lesions with positive uptake on PET/CT. CONCLUSIONS This article provides recommendations on best practices for standardized reporting of PSMA PET/CT imaging. The generated reporting template is a proposed supplement designed to educate and improve data communication between imaging experts and referring physicians.
Collapse
Affiliation(s)
- Shadi A Esfahani
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| | - Michael J Morris
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Oliver Sartor
- Department of Medical Oncology, Mayo Clinic, Minnesota, USA
| | - Mark Frydenberg
- Department of Surgery, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Australia
- Cabrini Institute, Cabrini Health, Malvern, Australia
| | - Stefano Fanti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Jeremie Calais
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA, USA
| | - Neha Vapiwala
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
11
|
Correia ETDO, Baydoun A, Li Q, Costa DN, Bittencourt LK. Emerging and anticipated innovations in prostate cancer MRI and their impact on patient care. Abdom Radiol (NY) 2024; 49:3696-3710. [PMID: 38877356 PMCID: PMC11390809 DOI: 10.1007/s00261-024-04423-4] [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: 03/30/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/16/2024]
Abstract
Prostate cancer (PCa) remains the leading malignancy affecting men, with over 3 million men living with the disease in the US, and an estimated 288,000 new cases and almost 35,000 deaths in 2023 in the United States alone. Over the last few decades, imaging has been a cornerstone in PCa care, with a crucial role in the detection, staging, and assessment of PCa recurrence or by guiding diagnostic or therapeutic interventions. To improve diagnostic accuracy and outcomes in PCa care, remarkable advancements have been made to different imaging modalities in recent years. This paper focuses on reviewing the main innovations in the field of PCa magnetic resonance imaging, including MRI protocols, MRI-guided procedural interventions, artificial intelligence algorithms and positron emission tomography, which may impact PCa care in the future.
Collapse
Affiliation(s)
| | - Atallah Baydoun
- Department of Radiation Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Qiubai Li
- Department of Radiology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Daniel N Costa
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Leonardo Kayat Bittencourt
- Department of Radiology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA.
- Department of Radiology, Case Western Reserve University, 11100 Euclid Ave, Cleveland, OH, 44106, USA.
| |
Collapse
|
12
|
Zhang X, Ma Z. Head-to-head comparison of PSMA PET/CT and mpMRI for detecting biochemical recurrence of prostate cancer: A systematic review and meta-analysis. Curr Urol 2024; 18:177-184. [PMID: 39219632 PMCID: PMC11337997 DOI: 10.1097/cu9.0000000000000242] [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: 07/21/2023] [Accepted: 12/06/2023] [Indexed: 09/04/2024] Open
Abstract
Objectives This study aimed to evaluate the performance of prostate-specific membrane antigen positron emission tomography/computed tomography (PSMA PET/CT) in comparison to multiparametric magnetic resonance imaging (mpMRI) for detecting biochemical recurrence of prostate cancer (PCa). Materials and methods We conducted a comprehensive search for articles published in PubMed, Web of Science, Embase, and the Cochrane Library, spanning the inception of the database until October 26, 2022, which included head-to-head comparisons of PSMA PET/CT and mpMRI for assessing the biochemical recurrence of PCa. Results A total of 5 studies including 228 patients were analyzed. The overall positivity rates of PSMA PET/CT and mpMRI for detecting biochemical recurrence of PCa after final treatment were 0.68 (95% confidence interval [CI], 0.52-0.89) and 0.56 (95% CI, 0.36-0.88), respectively. The positivity rates of PSMA PET/CT and mpMRI for detecting local recurrence, lymph node metastasis, and bone metastases were 0.37 (95% CI, 0.30-0.47) and 0.38 (95% CI, 0.22-0.67), 0.44 (95% CI, 0.35-0.56) and 0.25 (95% CI, 0.17-0.35), and 0.19 (95% CI, 0.11-0.31) and 0.12 (95% CI, 0.05-0.25), respectively. Compared with mpMRI, PSMA PET/CT exhibited a higher positivity rate for detecting biochemical recurrence and lymph node metastases, and no significant difference in the positivity rate of local recurrence was observed between these 2 imaging modalities. Conclusions Compared with mpMRI, PSMA PET/CT appears to have a higher positivity rate for detecting biochemical recurrence of PCa. Although both imaging methods showed similar positivity rates of detecting local recurrence, PSMA PET/CT outperformed PSMA PET/CT in detecting lymph node involvement and overall recurrence.
Collapse
Affiliation(s)
| | - Zhe Ma
- Department of Ultrasound, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| |
Collapse
|
13
|
Zugni F, Mariani L, Lambregts DMJ, Maggioni R, Summers PE, Granata V, Pecchi A, Di Costanzo G, De Muzio F, Cardobi N, Giovagnoni A, Petralia G. Whole-body MRI in oncology: acquisition protocols, current guidelines, and beyond. LA RADIOLOGIA MEDICA 2024; 129:1352-1368. [PMID: 38990426 DOI: 10.1007/s11547-024-01851-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 07/04/2024] [Indexed: 07/12/2024]
Abstract
Acknowledging the increasing use of whole-body magnetic resonance imaging (WB-MRI) in the oncological setting, we conducted a narrative review focusing on practical aspects of the examination and providing a synthesis of various acquisition protocols described in the literature. Firstly, we addressed the topic of patient preparation, emphasizing methods to enhance examination acceptance. This included strategies for reducing anxiety and patient distress, improving staff-patient interactions, and increasing overall patient comfort. Secondly, we analysed WB-MRI acquisition protocols recommended in existing imaging guidelines, such as MET-RADS-P, MY-RADS, and ONCO-RADS, and provided an overview of acquisition protocols reported in the literature regarding other expanding applications of WB-MRI in oncology, in patients with breast cancer, ovarian cancer, melanoma, colorectal and lung cancer, lymphoma, and cancers of unknown primary. Finally, we suggested possible acquisition parameters for whole-body images across MR systems from three different vendors.
Collapse
Affiliation(s)
- Fabio Zugni
- Division of Radiology, IEO European Institute of Oncology IRCCS, Milan, Italy.
| | - Leonardo Mariani
- Postgraduation School in Radiodiagnostics, University of Milan, Milan, Italy
| | - Doenja M J Lambregts
- Department of Radiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Roberta Maggioni
- Division of Radiology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Paul E Summers
- Division of Radiology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Vincenza Granata
- Division of Radiology, Istituto Nazionale Tumori Di Napoli, IRCCS "G. Pascale", Naples, Italy
| | - Annarita Pecchi
- Department of Radiology, University of Modena and Reggio Emilia, Modena, Italy
| | | | - Federica De Muzio
- Department of Radiology, Pineta Grande Hospital, Via Domitiana Km 30, Castel Volturno, Italy
| | - Nicolò Cardobi
- Radiology Unit, Department of Pathology and Diagnostics, University Hospital of Verona, Verona, Italy
| | - Andrea Giovagnoni
- Department of Radiology, University Hospital "Azienda Ospedaliera Universitaria Delle Marche", Ancona, Italy
- Department of Clinical, Special and Dental Sciences, Università Politecnica Delle Marche, Ancona, Italy
| | - Giuseppe Petralia
- Division of Radiology, IEO European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
14
|
Inaki A, Mizokami A, Wakabayashi H, Izumi K, Kadono Y, Toyama T, Takahara S, Murayama T, Kinuya S. Evaluation of 68Ga-PSMA-11 PET/CT: a Phase 1 clinical study in Japanese patients with primary, recurrent, or suspected recurrent prostate cancer. Ann Nucl Med 2024; 38:587-595. [PMID: 38750331 PMCID: PMC11281955 DOI: 10.1007/s12149-024-01931-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/09/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND Prostate-specific membrane antigen (PSMA)-targeted radiopharmaceuticals allow whole-body imaging to detect prostate cancer (PC). Positron emission tomography imaging using gallium-68 (68Ga)-PSMA-11 has been shown to have a favorable safety and tolerability profile and high diagnostic performance. The study evaluates the safety and pharmacokinetics of 68Ga-PSMA-11 in Japanese patients with primary, recurrent, or suspected recurrent prostate cancer. METHODS This single arm study enrolled Japanese patients with primary PC (n = 3), suspected recurrent PC following radical prostatectomy (n = 4), or suspected recurrent PC following radical radiotherapy (n = 3). All patients received a single intravenous dose of 68Ga-PSMA-11 2.0 MBq/kg (±10%) followed by PSMA PET imaging and safety and pharmacokinetic evaluations. Based on the blood concentrations of 68Ga-PSMA-11 and the radioactivity distribution rate in each organ/tissue, the absorbed doses in major organs/tissues and the whole-body effective dose were calculated by the Medical Internal Radiation Dose method. RESULTS Ten patients were enrolled. Mean age was 73.3 ± 4.8 years, and median prostate-specific antigen was 8.250 ng/mL. Five patients (50%) experienced a total of 6 adverse events, and no grade ≥ 2 adverse events or serious adverse events were reported. No clinically significant changes in vital signs, haematology parameters, or blood chemistry or ECG abnormalities were observed. The estimated whole body effective dose of 68Ga-PSMA-11 (mean ± standard deviation) was 2.524 × 10-2 ± 2.546 × 10-3 mSv/MBq. Time to maximum concentration (1.16 × 10-4 ± 1.3 × 10-5% ID/mL) in whole blood was 2.15 ± 0.33 min. CONCLUSIONS 68Ga-PSMA-11 has a favourable safety and tolerability profile in Japanese patients with primary, recurrent, or suspected recurrent prostate cancer, which is comparable to previous observations in other populations.
Collapse
Affiliation(s)
- Anri Inaki
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-Machi, Kanazawa-Shi, Ishikawa, 920-8641, Japan.
| | - Atsushi Mizokami
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Hospital, 13-1 Takara-Machi, Kanazawa-Shi, Ishikawa, 920-8641, Japan
| | - Hiroshi Wakabayashi
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-Machi, Kanazawa-Shi, Ishikawa, 920-8641, Japan
| | - Kouji Izumi
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Hospital, 13-1 Takara-Machi, Kanazawa-Shi, Ishikawa, 920-8641, Japan
| | - Yoshifumi Kadono
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Hospital, 13-1 Takara-Machi, Kanazawa-Shi, Ishikawa, 920-8641, Japan
| | - Tadashi Toyama
- Innovative Clinical Research Center, Kanazawa University Hospital, 13-1 Takara-Machi, Kanazawa-Shi, Ishikawa, 920-8641, Japan
| | - Shizuko Takahara
- Innovative Clinical Research Center, Kanazawa University Hospital, 13-1 Takara-Machi, Kanazawa-Shi, Ishikawa, 920-8641, Japan
| | - Toshinori Murayama
- Innovative Clinical Research Center, Kanazawa University Hospital, 13-1 Takara-Machi, Kanazawa-Shi, Ishikawa, 920-8641, Japan
- Department of Clinical Development, Kanazawa University Hospital, 13-1 Takara-Machi, Kanazawa-Shi, Ishikawa, 920-8641, Japan
| | - Seigo Kinuya
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-Machi, Kanazawa-Shi, Ishikawa, 920-8641, Japan
| |
Collapse
|
15
|
Alshamrani AFA. Diagnostic Accuracy of Molecular Imaging Techniques for Detecting Prostate Cancer: A Systematic Review. Diagnostics (Basel) 2024; 14:1315. [PMID: 39001206 PMCID: PMC11240585 DOI: 10.3390/diagnostics14131315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/17/2024] [Accepted: 06/19/2024] [Indexed: 07/16/2024] Open
Abstract
Molecular imaging modalities show valuable non-invasive techniques capable of precisely and selectively addressing molecular markers associated with prostate cancer (PCa). This systematic review provides an overview of imaging markers utilized in positron emission tomography (PET) methods, specifically focusing on the pathways and mediators involved in PCa. This systematic review aims to evaluate and analyse existing literature on the diagnostic accuracy of molecular imaging techniques for detecting PCa. The PubMed, EBSCO, ScienceDirect, and Web of Science databases were searched, identifying 32 studies that reported molecular imaging modalities for detecting PCa. Numerous imaging modalities and radiotracers were used to detect PCa, including 68Ga-prostate-specific membrane antigen (PSMA) PET/computed tomography (CT), 68Ga-PSMA-11 PET/magnetic resonance imaging (MRI), 18F-PSMA-1007 PET/CT, 18F-DCFPyL PET/MRI, 18F-choline PET/MRI, and 18F-fluoroethylcholine PET/MRI. Across 11 studies, radiolabelled 68Ga-PSMA PET/CT imaging had a pooled sensitivity of 80 (95% confidence interval [CI]: 35-93), specificity of 90 (95% CI: 71-98), and accuracy of 86 (95% CI: 64-96). The PSMA-ligand 68Ga-PET/CT showed good diagnostic performance and appears promising for detecting and staging PCa.
Collapse
Affiliation(s)
- Abdullah Fahad A Alshamrani
- Department of Diagnostic Radiology Technology, College of Applied Medical Sciences, Taibah University, Madinah 42353, Saudi Arabia
| |
Collapse
|
16
|
Tajaldeen A, Alrashidi M, Alsaadi MJ, Alghamdi SS, Alshammari H, Alsleem H, Jafer M, Aljondi R, Alqahtani S, Alotaibi A, Alzandi AM, Alahmari AM. Photoacoustic imaging in prostate cancer: A new paradigm for diagnosis and management. Photodiagnosis Photodyn Ther 2024; 47:104225. [PMID: 38821240 DOI: 10.1016/j.pdpdt.2024.104225] [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: 04/16/2024] [Revised: 05/24/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
The global health issue of prostate cancer (PCa) requires better diagnosis and treatment. Photoacoustic imaging (PAI) may change PCa management. This review examines PAI's principles, diagnostic role, and therapeutic guidance. PAI uses optical light excitation and ultrasonic detection for high-resolution functional and molecular imaging. PAI uses endogenous and exogenous contrast agents to distinguish cancerous and benign prostate tissues with greater sensitivity and specificity than PSA testing and TRUS-guided biopsy. In addition to diagnosing, PAI can guide and monitor PCa therapy. Its real-time imaging allows precise biopsies and brachytherapy seed placement. Photoacoustic temperature imaging allows non-invasive monitoring of thermal therapies like cryotherapy, improving treatment precision and success. Transurethral illumination probes, innovative contrast agents, integration with other imaging modalities, and machine learning analysis are being developed to overcome depth and data complexity restrictions. PAI could become an essential tool for PCa diagnosis and therapeutic guidance as the field advances.
Collapse
Affiliation(s)
- Abdulrahman Tajaldeen
- Department of Radiologic Technology, College of Applied Medical Sciences, University of Jeddah, Jeddah 21959, Saudi Arabia.
| | - Muteb Alrashidi
- Department of Radiological Sciences, College of Applied Medical Sciences, Imam Abdulrahman Bin Faisal University, Dammam 34212, Saudi Arabia
| | - Mohamed J Alsaadi
- Radiology and Medical Imaging Department, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Salem Saeed Alghamdi
- Department of Radiologic Technology, College of Applied Medical Sciences, University of Jeddah, Jeddah 21959, Saudi Arabia
| | - Hamed Alshammari
- Department of Radiological Sciences, College of Applied Medical Sciences, Imam Abdulrahman Bin Faisal University, Dammam 34212, Saudi Arabia
| | - Haney Alsleem
- Department of Radiological Sciences, College of Applied Medical Sciences, Imam Abdulrahman Bin Faisal University, Dammam 34212, Saudi Arabia
| | - Mustafa Jafer
- Department of Radiologic Technology, College of Applied Medical Sciences, University of Jeddah, Jeddah 21959, Saudi Arabia
| | - Rowa Aljondi
- Department of Radiologic Technology, College of Applied Medical Sciences, University of Jeddah, Jeddah 21959, Saudi Arabia
| | - Saeed Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, Najran University, Najran, Saudi Arabia
| | - Awatif Alotaibi
- Department of Radiological Sciences, College of Applied Medical Sciences, Imam Abdulrahman Bin Faisal University, Dammam 34212, Saudi Arabia
| | - Abdulrahman M Alzandi
- Department of Radiological Sciences, College of Applied Medical Sciences, Imam Abdulrahman Bin Faisal University, Dammam 34212, Saudi Arabia
| | | |
Collapse
|
17
|
Shore ND, Moul JW, Pienta KJ, Czernin J, King MT, Freedland SJ. Biochemical recurrence in patients with prostate cancer after primary definitive therapy: treatment based on risk stratification. Prostate Cancer Prostatic Dis 2024; 27:192-201. [PMID: 37679602 PMCID: PMC11096125 DOI: 10.1038/s41391-023-00712-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Nearly one-third of patients with prostate cancer (PCa) experience biochemical recurrence (BCR) after primary definitive treatment. BCR increases the risk of distant metastasis and mortality in patients with prognostically unfavorable features. These patients are best managed with a tailored treatment strategy incorporating risk stratification using clinicopathological factors, next-generation imaging, and genomic testing. OBJECTIVE This narrative review examines the utility of risk stratification for the management of patients with BCR in the context of clinical trial data, referencing the latest recommendations by European and US medical societies. METHODS PubMed was searched for relevant studies published through May 21 2023 on treatment of patients with BCR after radical prostatectomy (RP) or external beam radiotherapy (EBRT). RESULTS European and US guidelines support the risk-stratified management of BCR. Post-RP, salvage EBRT (with or without androgen deprivation therapy [ADT]) is an accepted treatment option for patients with BCR. Post-EBRT, local salvage therapies (RP, cryotherapy, high-intensity focused ultrasound, stereotactic body radiotherapy, and low-dose-rate and high-dose-rate brachytherapy) have demonstrated comparable relapse-free survival rates but differing adverse event profiles, short and long term. Local salvage therapies should be used for local-only relapses while ADT should be considered for regional or distant relapses. In practice, patients often receive ADT, with varying guidance for intermittent ADT vs. continuous ADT, due to consideration of quality-of-life effects. CONCLUSIONS Despite a lack of consensus for BCR treatment among guideline associations and medical societies, risk stratification of patients is essential for personalized treatment approaches, as it allows for an informed selection of therapeutic strategies and estimation of adverse events. In lower-risk disease, observation is recommended while in higher-risk disease, after failed repeat local therapy, ADT and/or clinical trial enrollment may be appropriate. Results from ongoing clinical studies of patients with BCR should provide consensus for management.
Collapse
Affiliation(s)
- Neal D Shore
- Carolina Urologic Research Center, Myrtle Beach, SC, USA
| | - Judd W Moul
- Duke Cancer Institute, Duke University, Durham, NC, USA
| | | | - Johannes Czernin
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Martin T King
- Brigham and Women's Hospital and Dana-Farber Cancer Institute, Boston, MA, USA
| | - Stephen J Freedland
- Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Veterans Affairs Medical Center, Durham, NC, USA.
| |
Collapse
|
18
|
Moul JW, Shore ND, Pienta KJ, Czernin J, King MT, Freedland SJ. Application of next-generation imaging in biochemically recurrent prostate cancer. Prostate Cancer Prostatic Dis 2024; 27:202-211. [PMID: 37679601 PMCID: PMC11096127 DOI: 10.1038/s41391-023-00711-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Biochemical recurrence (BCR) following primary interventional treatment occurs in approximately one-third of patients with prostate cancer (PCa). Next-generation imaging (NGI) can identify local and metastatic recurrence with greater sensitivity than conventional imaging, potentially allowing for more effective interventions. This narrative review examines the current clinical evidence on the utility of NGI for patients with BCR. METHODS A search of PubMed was conducted to identify relevant publications on NGI applied to BCR. Given other relevant recent reviews on the topic, this review focused on papers published between January 2018 to May 2023. RESULTS NGI technologies, including positron emission tomography (PET) radiotracers and multiparametric magnetic resonance imaging, have demonstrated increased sensitivity and selectivity for diagnosing BCR at prostate-specific antigen (PSA) concentrations <2.0 ng/ml. Detection rates range between 46% and 50%, with decreasing PSA levels for choline (1-3 ng/ml), fluciclovine (0.5-1 ng/ml), and prostate-specific membrane antigen (0.2-0.49 ng/ml) PET radiotracers. Expert working groups and European and US medical societies recommend NGI for patients with BCR. CONCLUSIONS Available data support the improved detection performance and selectivity of NGI modalities versus conventional imaging techniques; however, limited clinical evidence exists demonstrating the application of NGI to treatment decision-making and its impact on patient outcomes. The emergence of NGI and displacement of conventional imaging may require a reexamination of the current definitions of BCR, altering our understanding of early recurrence. Redefining the BCR disease state by formalizing the role of NGI in patient management decisions will facilitate greater alignment across research efforts and better reflect the published literature.
Collapse
Affiliation(s)
- Judd W Moul
- Duke Cancer Institute and Division of Urology, Duke University, Durham, NC, USA
| | - Neal D Shore
- Carolina Urologic Research Center, Myrtle Beach, SC, USA
| | | | - Johannes Czernin
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Martin T King
- Brigham and Women's Hospital and Dana-Farber Cancer Institute, Boston, MA, USA
| | - Stephen J Freedland
- Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Veterans Affairs Medical Center, Durham, NC, USA.
| |
Collapse
|
19
|
Saxena A, Andrews J, Bryce AH, Riaz IB. Optimal systemic therapy in men with low-volume prostate cancer. Curr Opin Urol 2024; 34:183-197. [PMID: 38445371 DOI: 10.1097/mou.0000000000001165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2024]
Abstract
PURPOSE OF REVIEW Low-volume prostate cancer is an established prognostic category of metastatic hormone-sensitive prostate cancer. However, the term is often loosely used to reflect the low burden of disease across different prostate cancer states. This review explores the definitions of low-volume prostate cancer, biology, and current evidence for treatment. We also explore future directions, including the impact of advanced imaging modalities, particularly prostate-specific membrane antigen (PSMA) positron emission tomography (PET) scans, on refining patient subgroups and treatment strategies for patients with low-volume prostate cancer. RECENT FINDINGS Recent investigations have attempted to redefine low-volume disease, incorporating factors beyond metastatic burden. Advanced imaging, especially PSMA PET, offers enhanced accuracy in detecting metastases, potentially challenging the conventional definition of low volume. The prognosis and treatment of low-volume prostate cancer may vary by the timing of metastatic presentation. Biomarker-directed consolidative therapy, metastases-directed therapy, and de-escalation of systemic therapies will be increasingly important, especially in patients with metachronous low-volume disease. SUMMARY In the absence of validated biomarkers, the management of low-volume prostate cancer as defined by CHAARTED criteria may be guided by the timing of metastatic presentation. For metachronous low-volume disease, we recommend novel hormonal therapy (NHT) doublets with or without consolidative metastasis-directed therapy (MDT), and for synchronous low-volume disease, NHT doublets with or without consolidative MDT and prostate-directed radiation. Docetaxel triplets may be a reasonable alternative in some patients with synchronous presentation. There is no clear role of docetaxel doublets in patients with low-volume disease. In the future, a small subset of low-volume diseases with oligometastases selected by genomics and advanced imaging like PSMA PET may achieve long-term remission with MDT with no systemic therapy.
Collapse
Affiliation(s)
| | | | - Alan Haruo Bryce
- Department of Oncology, City of Hope Cancer Center, Goodyear, Arizona, USA
| | | |
Collapse
|
20
|
Vulasala SS, Virarkar M, Karbasian N, Calimano-Ramirez LF, Daoud T, Amini B, Bhosale P, Javadi S. Whole-body MRI in oncology: A comprehensive review. Clin Imaging 2024; 108:110099. [PMID: 38401295 DOI: 10.1016/j.clinimag.2024.110099] [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: 10/09/2023] [Revised: 01/28/2024] [Accepted: 01/31/2024] [Indexed: 02/26/2024]
Abstract
Whole-Body Magnetic Resonance Imaging (WB-MRI) has cemented its position as a pivotal tool in oncological diagnostics. It offers unparalleled soft tissue contrast resolution and the advantage of sidestepping ionizing radiation. This review explores the diverse applications of WB-MRI in oncology. We discuss its transformative role in detecting and diagnosing a spectrum of cancers, emphasizing conditions like multiple myeloma and cancers with a proclivity for bone metastases. WB-MRI's capability to encompass the entire body in a singular scan has ushered in novel paradigms in cancer screening, especially for individuals harboring hereditary cancer syndromes or at heightened risk for metastatic disease. Additionally, its contribution to the clinical landscape, aiding in the holistic management of multifocal and systemic malignancies, is explored. The article accentuates the technical strides achieved in WB-MRI, its myriad clinical utilities, and the challenges in integration into standard oncological care. In essence, this review underscores the transformative potential of WB-MRI, emphasizing its promise as a cornerstone modality in shaping the future trajectory of cancer diagnostics and treatment.
Collapse
Affiliation(s)
- Sai Swarupa Vulasala
- Department of Radiology, University of Florida College of Medicine, Jacksonville, FL, United States.
| | - Mayur Virarkar
- Department of Radiology, University of Florida College of Medicine, Jacksonville, FL, United States
| | - Niloofar Karbasian
- Department of Radiology, McGovern Medical School at University of Texas Health Houston, Houston, TX, United States
| | - Luis F Calimano-Ramirez
- Department of Radiology, University of Florida College of Medicine, Jacksonville, FL, United States
| | - Taher Daoud
- Division of Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Behrang Amini
- Division of Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Priya Bhosale
- Division of Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sanaz Javadi
- Division of Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
21
|
Al Saffar H, Chen DC, Delgado C, Ingvar J, Hofman MS, Lawrentschuk N, Perera M, Murphy DG, Eapen R. The Current Landscape of Prostate-Specific Membrane Antigen (PSMA) Imaging Biomarkers for Aggressive Prostate Cancer. Cancers (Basel) 2024; 16:939. [PMID: 38473301 PMCID: PMC10931387 DOI: 10.3390/cancers16050939] [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: 01/10/2024] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
The review examines the vital role of prostate-specific membrane antigen (PSMA) positron emission tomography/computed tomography (PET/CT) in the diagnosis, staging, and treatment of prostate cancer (PCa). It focuses on the superior diagnostic abilities of PSMA PET/CT for identifying both nodal and distant PCa, and its potential as a prognostic indicator for biochemical recurrence and overall survival. Additionally, we focused on the variability of PSMA's expression and its impact on personalised treatment, particularly the use of [177Lu] Lu-PSMA-617 radioligand therapy. This review emphasises the essential role of PSMA PET/CT in enhancing treatment approaches, improving patient outcomes, and reducing unnecessary interventions, positioning it as a key element in personalised PCa management.
Collapse
Affiliation(s)
- Haidar Al Saffar
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia; (D.C.C.); (J.I.); (N.L.); (M.P.); (D.G.M.); (R.E.)
| | - David C. Chen
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia; (D.C.C.); (J.I.); (N.L.); (M.P.); (D.G.M.); (R.E.)
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia;
- Department of Surgery, Austin Health, Heidelberg, VIC 3084, Australia
| | - Carlos Delgado
- School of Medicine and Health Sciences, Tecnologico de Monterrey, Monterrey 64849, Mexico;
| | - Jacob Ingvar
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia; (D.C.C.); (J.I.); (N.L.); (M.P.); (D.G.M.); (R.E.)
| | - Michael S. Hofman
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia;
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Nathan Lawrentschuk
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia; (D.C.C.); (J.I.); (N.L.); (M.P.); (D.G.M.); (R.E.)
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3052, Australia
- Department of Surgery (Urology), Royal Melbourne Hospital, Melbourne, VIC 3052, Australia
- EJ Whitten Prostate Cancer Research Centre, Epworth Hospital, Richmond, VIC 3121, Australia
| | - Marlon Perera
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia; (D.C.C.); (J.I.); (N.L.); (M.P.); (D.G.M.); (R.E.)
- Department of Surgery, Austin Health, Heidelberg, VIC 3084, Australia
| | - Declan G. Murphy
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia; (D.C.C.); (J.I.); (N.L.); (M.P.); (D.G.M.); (R.E.)
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia;
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Renu Eapen
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia; (D.C.C.); (J.I.); (N.L.); (M.P.); (D.G.M.); (R.E.)
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia;
- Department of Surgery, Austin Health, Heidelberg, VIC 3084, Australia
| |
Collapse
|
22
|
Sood A, Kishan AU, Evans CP, Feng FY, Morgan TM, Murphy DG, Padhani AR, Pinto P, Van der Poel HG, Tilki D, Briganti A, Abdollah F. The Impact of Positron Emission Tomography Imaging and Tumor Molecular Profiling on Risk Stratification, Treatment Choice, and Oncological Outcomes of Patients with Primary or Relapsed Prostate Cancer: An International Collaborative Review of the Existing Literature. Eur Urol Oncol 2024; 7:27-43. [PMID: 37423774 DOI: 10.1016/j.euo.2023.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 05/06/2023] [Accepted: 06/07/2023] [Indexed: 07/11/2023]
Abstract
CONTEXT The clinical introduction of next-generation imaging methods and molecular biomarkers ("radiogenomics") has revolutionized the field of prostate cancer (PCa). While the clinical validity of these tests has thoroughly been vetted, their clinical utility remains a matter of investigation. OBJECTIVE To systematically review the evidence to date on the impact of positron emission tomography (PET) imaging and tissue-based prognostic biomarkers, including Decipher, Prolaris, and Oncotype Dx, on the risk stratification, treatment choice, and oncological outcomes of men with newly diagnosed PCa or those with biochemical failure (BCF). EVIDENCE ACQUISITION We performed a quantitative systematic review of the literature using the MEDLINE, EMBASE, and Web of Science databases (2010-2022) following the Preferred Reporting Items for Systematic Reviews and Meta-analyses statement guidelines. The validated Quality Assessment of Diagnostic Accuracy Studies 2 scoring system was used to assess the risk of bias. EVIDENCE SYNTHESIS A total of 148 studies (130 on PET and 18 on biomarkers) were included. In the primary PCa setting, prostate-specific membrane antigen (PSMA) PET imaging was not useful in improving T staging, moderately useful in improving N staging, but consistently useful in improving M staging in patients with National Comprehensive Cancer Network (NCCN) unfavorable intermediate- to very-high-risk PCa. Its use led to a management change in 20-30% of patients. However, the effect of these treatment changes on survival outcomes was not clear. Similarly, biomarkers in the pretherapy primary PCa setting increased and decreased the risk, respectively, in 7-30% and 32-36% of NCCN low-risk and 31-65% and 4-15% of NCCN favorable intermediate-risk patients being considered for active surveillance. A change in management was noted in up to 65% of patients, with the change being in line with the molecular risk-based reclassification, but again, the impact of these changes on survival outcomes remained unclear. Notably, in the postsurgical primary PCa setting, biomarker-guided adjuvant radiation therapy (RT) was associated with improved oncological control: Δ↓ 2-yr BCF by 22% (level 2b). In the BCF setting, the data were more mature. PSMA PET was consistently useful in improving disease localization-Δ↑ detection for T, N, and M staging was 13-32%, 19-58%, and 9-29%, respectively. Between 29% and 73% of patients had a change in management. Most importantly, these management changes were associated with improved survival outcomes in three trials: Δ↑ 4-yr disease-free survival by 24.3%, Δ↑ 6-mo metastasis-free survival (MFS) by 46.7%, and Δ↑ androgen deprivation therapy-free survival by 8 mo in patients who received PET-concordant RT (level 1b-2b). Biomarker testing in these patients also appeared to be helpful in risk stratifying and guiding the use of early salvage RT (sRT) and concomitant hormonal therapy. Patients with high-genomic-risk scores benefitted from treatment intensification: Δ↑ 8-yr MFS by 20% with the use of early sRT and Δ↑ 12-yr MFS by 11.2% with the use of hormonal therapy alongside early sRT, while low-genomic-risk score patients did equally well with initial conservative management (level 3). CONCLUSIONS Both PSMA PET imaging and tumor molecular profiling provide actionable information in the management of men with primary PCa and those with BCF. Emerging data suggest that radiogenomics-guided treatments translate into direct survival benefits for patients, however, additional prospective data are awaited. PATIENT SUMMARY In this review, we evaluated the utility of prostate-specific membrane antigen positron emission tomography and tumor molecular profiling in guiding the care of men with prostate cancer (PCa). We found that these tests augmented risk stratification, altered management, and improved cancer control in men with a new diagnosis of PCa or for those experiencing a relapse.
Collapse
Affiliation(s)
- Akshay Sood
- Department of Urology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Urology, The James Cancer Hospital and Solove Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| | - Amar U Kishan
- Department of Radiation Oncology and Urology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA
| | - Christopher P Evans
- Department of Urologic Surgery, University of California Davis, Sacramento, CA, USA
| | - Felix Y Feng
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Todd M Morgan
- Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - Declan G Murphy
- Department of Genitourinary Oncology, Peter MacCallum Cancer Centre, The University of Melbourne, Victoria, Australia
| | - Anwar R Padhani
- Paul Strickland Scanner Centre, Mount Vernon Cancer Centre, Northwood, Middlesex, UK
| | - Peter Pinto
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Henk G Van der Poel
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Derya Tilki
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Department of Urology, Koc University Hospital, Istanbul, Turkey
| | - Alberto Briganti
- Department of Urology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Firas Abdollah
- Vattikuti Urology Institute, Henry Ford Hospital, Detroit, MI, USA.
| |
Collapse
|
23
|
Farkas I, Sipka G, Bakos A, Maráz A, Bajory Z, Mikó Z, Czékus T, Urbán S, Varga L, Pávics L, Besenyi Z. Diagnostic value of [ 99mTc]Tc-PSMA-I&S-SPECT/CT for the primary staging and restaging of prostate cancer. Ther Adv Med Oncol 2024; 16:17588359231221342. [PMID: 38249326 PMCID: PMC10798073 DOI: 10.1177/17588359231221342] [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: 07/21/2023] [Accepted: 11/29/2023] [Indexed: 01/23/2024] Open
Abstract
Background A large number of studies have proved that prostate-specific membrane antigen-positron emission tomography/computer tomography (PSMA-PET/CT) provides excellent accuracy in primary staging and restaging of prostate cancer. Less data exist with PSMA-single photon emission computed tomography (SPECT)/CT investigations. Objective The aim of this study was to evaluate the performance of [99mTc]Tc-PSMA-I&S (for imaging and surgery) in prostate cancer. Design and methods We retrospectively analysed PSMA-SPECT/CT scans of 20 healthy volunteers and 100 male patients with prostate cancer. All of them had histologically confirmed prostate cancer. In all, 28 patients were examined for primary staging and 72 for biochemical recurrence or progressive disease. Whole body SPECT/CT imaging was carried out 6 h after the intravenous administration of 666 ± 102 MBq [99mTc]Tc-PSMA-I&S. Images were evaluated visually and semi-quantitatively. Results Patient-based sensitivity, specificity, positive predictive value, negative predictive value and accuracy for primary prostate cancer were 86%, 100%, 100%, 83% and 92%, respectively. For detecting metastases in primary staging, these values were 88%, 100%, 100%, 85% and 93%, respectively. The radiopharmaceutical uptake of primary prostate cancer was significantly higher than in normal prostate. The patient-based sensitivity, specificity, positive predictive value, negative predictive value and accuracy of the method in the visualization of local recurrence were 67%, 100%, 100%, 86% and 89%, and for detecting metastases in restaging were 91%, 92%, 98%, 75% and 91%, respectively. In restaging, detection rates were 37% under prostate-specific antigen level of 1 ng/mL, 74% between 1 and 5 ng/mL and 80% >5 ng/mL. Conclusion [99mTc]Tc-PSMA-I&S-SPECT/CT can be easily integrated into the routine diagnostic practice, and it provides usable data in primary staging and restaging of prostate cancer. Quantitative assessment of PSMA-SPECT/CT has the potential to be used to differentiate between physiological and pathological intraprostatic tracer uptake.
Collapse
Affiliation(s)
- István Farkas
- Department of Nuclear Medicine, University of Szeged, Szeged, Hungary
| | - Gábor Sipka
- Department of Nuclear Medicine, University of Szeged, Szeged, Hungary
| | - Annamária Bakos
- Department of Nuclear Medicine, University of Szeged, Szeged, Hungary
| | - Anikó Maráz
- Department of Oncotherapy, University of Szeged, Szeged, Hungary
| | - Zoltán Bajory
- Department of Urology, University of Szeged, Szeged, Hungary
| | - Zsófia Mikó
- Department of Nuclear Medicine, University of Szeged, Szeged, Hungary
| | - Tamás Czékus
- Department of Nuclear Medicine, University of Szeged, Szeged, Hungary
| | - Szabolcs Urbán
- Department of Nuclear Medicine, University of Szeged, Szeged, Hungary
| | - Linda Varga
- Department of Oncotherapy, University of Szeged, Szeged, Hungary
| | - László Pávics
- Department of Nuclear Medicine, University of Szeged, Szeged, Hungary
| | - Zsuzsanna Besenyi
- Department of Nuclear Medicine, University of Szeged, Korányi Fasor 6, H-6720 Szeged, Hungary
| |
Collapse
|
24
|
Ouvrard E, Kaseb A, Poterszman N, Porot C, Somme F, Imperiale A. Nuclear medicine imaging for bone metastases assessment: what else besides bone scintigraphy in the era of personalized medicine? Front Med (Lausanne) 2024; 10:1320574. [PMID: 38288299 PMCID: PMC10823373 DOI: 10.3389/fmed.2023.1320574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/28/2023] [Indexed: 01/31/2024] Open
Abstract
Accurate detection and reliable assessment of therapeutic responses in bone metastases are imperative for guiding treatment decisions, preserving quality of life, and ultimately enhancing overall survival. Nuclear imaging has historically played a pivotal role in this realm, offering a diverse range of radiotracers and imaging modalities. While the conventional bone scan using 99mTc marked bisphosphonates has remained widely utilized, its diagnostic performance is hindered by certain limitations. Positron emission tomography, particularly when coupled with computed tomography, provides improved spatial resolution and diagnostic performance with various pathology-specific radiotracers. This review aims to evaluate the performance of different nuclear imaging modalities in clinical practice for detecting and monitoring the therapeutic responses in bone metastases of diverse origins, addressing their limitations and implications for image interpretation.
Collapse
Affiliation(s)
- Eric Ouvrard
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe (ICANS), University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France
| | - Ashjan Kaseb
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe (ICANS), University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France
- Radiology, College of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Nathan Poterszman
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe (ICANS), University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France
| | - Clémence Porot
- Radiopharmacy, Institut de Cancérologie Strasbourg Europe (ICANS), Strasbourg, France
| | - Francois Somme
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe (ICANS), University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France
| | - Alessio Imperiale
- Nuclear Medicine and Molecular Imaging, Institut de Cancérologie Strasbourg Europe (ICANS), University Hospitals of Strasbourg, University of Strasbourg, Strasbourg, France
- IPHC, UMR 7178, CNRS/Unistra, Strasbourg, France
| |
Collapse
|
25
|
Sun J, Sheng J, Zhang LJ. Gastrointestinal tract. TRANSPATHOLOGY 2024:281-296. [DOI: 10.1016/b978-0-323-95223-1.00005-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
26
|
Mohseninia N, Zamani-Siahkali N, Harsini S, Divband G, Pirich C, Beheshti M. Bone Metastasis in Prostate Cancer: Bone Scan Versus PET Imaging. Semin Nucl Med 2024; 54:97-118. [PMID: 37596138 DOI: 10.1053/j.semnuclmed.2023.07.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 07/11/2023] [Indexed: 08/20/2023]
Abstract
Prostate cancer is the second most common cause of malignancy among men, with bone metastasis being a significant source of morbidity and mortality in advanced cases. Detecting and treating bone metastasis at an early stage is crucial to improve the quality of life and survival of prostate cancer patients. This objective strongly relies on imaging studies. While CT and MRI have their specific utilities, they also possess certain drawbacks. Bone scintigraphy, although cost-effective and widely available, presents high false-positive rates. The emergence of PET/CT and PET/MRI, with their ability to overcome the limitations of standard imaging methods, offers promising alternatives for the detection of bone metastasis. Various radiotracers targeting cell division activity or cancer-specific membrane proteins, as well as bone seeking agents, have been developed and tested. The use of positron-emitting isotopes such as fluorine-18 and gallium-68 for labeling allows for a reduced radiation dose and unaffected biological properties. Furthermore, the integration of artificial intelligence (AI) and radiomics techniques in medical imaging has shown significant advancements in reducing interobserver variability, improving accuracy, and saving time. This article provides an overview of the advantages and limitations of bone scan using SPECT and SPECT/CT and PET imaging methods with different radiopharmaceuticals and highlights recent developments in hybrid scanners, AI, and radiomics for the identification of prostate cancer bone metastasis using molecular imaging.
Collapse
Affiliation(s)
- Nasibeh Mohseninia
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University, Salzburg, Austria
| | - Nazanin Zamani-Siahkali
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University, Salzburg, Austria; Department of Nuclear Medicine, Research center for Nuclear Medicine and Molecular Imaging, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Sara Harsini
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | | | - Christian Pirich
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University, Salzburg, Austria
| | - Mohsen Beheshti
- Division of Molecular Imaging and Theranostics, Department of Nuclear Medicine, University Hospital, Paracelsus Medical University, Salzburg, Austria.
| |
Collapse
|
27
|
Efstathiou JA, Morgans AK, Bland CS, Shore ND. Novel hormone therapy and coordination of care in high-risk biochemically recurrent prostate cancer. Cancer Treat Rev 2024; 122:102630. [PMID: 38035646 DOI: 10.1016/j.ctrv.2023.102630] [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: 05/30/2023] [Accepted: 09/25/2023] [Indexed: 12/02/2023]
Abstract
Biochemical recurrence (BCR) occurs in 20-50% of patients with prostate cancer (PCa) undergoing primary definitive treatment. Patients with high-risk BCR have an increased risk of metastatic progression and subsequent PCa-specific mortality, and thus could benefit from treatment intensification. Given the increasing complexity of diagnostic and therapeutic modalities, multidisciplinary care (MDC) can play a crucial role in the individualized management of this patient population. This review explores the role for MDC when evaluating the clinical evidence for the evolving definition of high-risk BCR and the emerging therapeutic strategies, especially with novel hormone therapies (NHTs), for patients with either high-risk BCR or oligometastatic PCa. Clinical studies have used different characteristics to define high-risk BCR and there is no consensus regarding the definition of high-risk BCR nor for management strategies. Next-generation imaging and multigene panels offer potential enhanced patient identification and precision-based decision-making, respectively. Treatment intensification with NHTs, either alone or combined with radiotherapy or metastasis-directed therapy, has been promising in clinical trials in patients with high-risk BCR or oligometastases. As novel risk-stratification and treatment options as well as evidence-based literature evolve, it is important to involve a multidisciplinary team to identify patients with high-risk features at an earlier stage, and make informed decisions on the treatments that could optimize their care and long-term outcomes. Nevertheless, MDC data are scarce in the BCR or oligometastatic setting. Efforts to integrate MDC into the standard management of this patient population are needed, and will likely improve outcomes across this heterogeneous PCa patient population.
Collapse
Affiliation(s)
- Jason A Efstathiou
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA.
| | - Alicia K Morgans
- Dana-Farber Cancer Institute, 850 Brookline Ave, Dana 09-930, Boston, MA 02215, USA.
| | - Christopher S Bland
- US Oncology Medical Affairs, Pfizer Inc., 66 Hudson Boulevard, Hudson Yards, Manhattan, New York, NY 10001, USA.
| | - Neal D Shore
- Carolina Urologic Research Center, GenesisCare US, 823 82nd Pkwy, Myrtle Beach, SC, USA.
| |
Collapse
|
28
|
Pecoraro M, Dehghanpour A, Das JP, Woo S, Panebianco V. Evaluation of Prostate Cancer Recurrence with MR Imaging and Prostate Imaging for Recurrence Reporting Scoring System. Radiol Clin North Am 2024; 62:135-159. [PMID: 37973239 DOI: 10.1016/j.rcl.2023.06.013] [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: 11/19/2023]
Abstract
Detection of prostate cancer recurrence after whole-gland treatment with curative intent is critical to identify patients who may benefit from local salvage therapy. Among the different imaging modalities used in clinical practice, MR imaging is the most accurate in identifying local prostate cancer recurrence; indeed, it is an excellent technique for local recurrence detection superior to PET/CT, even at low PSA, but provides no information about extra-pelvic lymph nodes or bone metastasis. In 2021, a group of experts developed the Prostate Imaging for local Recurrence Reporting scoring system to standardize acquisition, interpretation, and reporting of prostate cancer recurrence.
Collapse
Affiliation(s)
- Martina Pecoraro
- Department of Radiological Sciences, Oncology and Pathology, Sapienza University, Policlinico Umberto I, Viale Regina Elena 324, Rome 00161, Italy
| | - Ailin Dehghanpour
- Department of Radiological Sciences, Oncology and Pathology, Sapienza University, Policlinico Umberto I, Viale Regina Elena 324, Rome 00161, Italy
| | - Jeeban Paul Das
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Sungmin Woo
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Valeria Panebianco
- Department of Radiological Sciences, Oncology and Pathology, Sapienza University, Policlinico Umberto I, Viale Regina Elena 324, Rome 00161, Italy.
| |
Collapse
|
29
|
Francolini G, Gaetano Allegra A, Detti B, Di Cataldo V, Caini S, Bruni A, Ingrosso G, D'Angelillo RM, Alitto AR, Augugliaro M, Triggiani L, Parisi S, Facchini G, Banini M, Simontacchi G, Desideri I, Meattini I, Valicenti RK, Livi L. Stereotactic Body Radiation Therapy and Abiraterone Acetate for Patients Affected by Oligometastatic Castrate-Resistant Prostate Cancer: A Randomized Phase II Trial (ARTO). J Clin Oncol 2023; 41:5561-5568. [PMID: 37733977 DOI: 10.1200/jco.23.00985] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/12/2023] [Accepted: 08/08/2023] [Indexed: 09/23/2023] Open
Abstract
PURPOSE ARTO (ClinicalTrials.gov identifier: NCT03449719) is a multicenter, phase II randomized clinical trial testing the benefit of adding stereotactic body radiation therapy (SBRT) to abiraterone acetate and prednisone (AAP) in patients with oligometastatic castrate-resistant prostate cancer (CRPC). MATERIALS AND METHODS All patients were affected by oligometastatic CRPC as defined as three or less nonvisceral metastatic lesions. Patients were randomly assigned 1:1 to receive either AAP alone (control arm) or AAP with concomitant SBRT to all the sites of disease (experimental arm). Primary end point was the rate of biochemical response (BR), defined as a prostate-specific antigen (PSA) decrease ≥50% from baseline measured at 6 months from treatment start. Complete BR (CBR), defined as PSA < 0.2 ng/mL at 6 months from treatment, and progression-free survival (PFS) were secondary end points. RESULTS One hundred and fifty-seven patients were enrolled between January 2019 and September 2022. BR was detected in 79.6% of patients (92% v 68.3% in the experimental v control arm, respectively), with an odds ratio (OR) of 5.34 (95% CI, 2.05 to 13.88; P = .001) in favor of the experimental arm. CBR was detected in 38.8% of patients (56% v 23.2% in the experimental v control arm, respectively), with an OR of 4.22 (95% CI, 2.12 to 8.38; P < .001). SBRT yielded a significant PFS improvement, with a hazard ratio for progression of 0.35 (95% CI, 0.21 to 0.57; P < .001) in the experimental versus control arm. CONCLUSION The trial reached its primary end point of biochemical control and PFS, suggesting a clinical advantage for SBRT in addition to first-line AAP treatment in patients with metastatic castration-resistant prostate cancer.
Collapse
Affiliation(s)
- Giulio Francolini
- Radiation Oncology Unit, Oncology Department, Azienda Ospedaliero Universitaria Careggi, Firenze, Italy
| | - Andrea Gaetano Allegra
- Department of Biomedical, Experimental and Clinical Sciences "M. Serio", University of Florence, Florence, Italy
| | - Beatrice Detti
- Radiation Oncology Unit, Oncology Department, Azienda Ospedaliero Universitaria Careggi, Firenze, Italy
| | - Vanessa Di Cataldo
- Radiation Oncology Unit, Oncology Department, Azienda Ospedaliero Universitaria Careggi, Firenze, Italy
| | - Saverio Caini
- Cancer Risk Factors and Lifestyle Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network (ISPO), Florence, Italy
| | - Alessio Bruni
- Radiation Oncology Unit, Department of Oncology and Hematology, University Hospital of Modena, Modena, Italy
| | - Gianluca Ingrosso
- Radiation Oncology Section, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Rolando Maria D'Angelillo
- Radiation Oncology, Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Anna Rita Alitto
- UOC di Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Matteo Augugliaro
- Unit of Radiotherapy, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Luca Triggiani
- Università degli Studi di Brescia, Department of Radiation Oncology, Brescia University, Brescia, Italy
| | - Silvana Parisi
- Radiation Oncology Unit, Department of Biomedical, Dental Science and Morphological and Functional Images, University of Messina, Messina, Italy
| | - Gaetano Facchini
- Medical Oncology Unit, SM delle Grazie Hospital, Pozzuoli, Italy
| | - Marco Banini
- Department of Biomedical, Experimental and Clinical Sciences "M. Serio", University of Florence, Florence, Italy
| | - Gabriele Simontacchi
- Radiation Oncology Unit, Oncology Department, Azienda Ospedaliero Universitaria Careggi, Firenze, Italy
| | - Isacco Desideri
- Department of Biomedical, Experimental and Clinical Sciences "M. Serio", University of Florence, Florence, Italy
| | - Icro Meattini
- Department of Biomedical, Experimental and Clinical Sciences "M. Serio", University of Florence, Florence, Italy
| | | | - Lorenzo Livi
- Department of Biomedical, Experimental and Clinical Sciences "M. Serio", University of Florence, Florence, Italy
| |
Collapse
|
30
|
Soliman M, Elfatairy K, Ellessy R, Velichko YS, Avery R, kelahan LC, Ross AE, Savas H. The utility of [ 18F]Fluciclovine PET/CT in Evaluating Nonmetastatic Castrate-Resistant Prostate Cancer Patients (nmCRPCp): diagnostic performance and impact on management. Br J Radiol 2023; 96:20230414. [PMID: 37750841 PMCID: PMC10646647 DOI: 10.1259/bjr.20230414] [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: 05/10/2023] [Revised: 07/30/2023] [Accepted: 09/05/2023] [Indexed: 09/27/2023] Open
Abstract
OBJECTIVE To evaluate the role of [18F]Fluciclovine PET/CT scan in restaging nmCRPCp and its impact on management. METHODS AND MATERIALS This retrospective study included all patients with nonmetastatic castrate-resistant prostate cancer, who underwent [18F]Fluciclovine PET/CT scans for restaging who had concern for disease progression. Two radiologists independently reviewed the PET/CT studies, assigned an overall impression, and reported the site and number of radiotracer activities in consensus and impact on management was recorded. Available tissue diagnosis and/or six-month clinical and imaging follow-up were used as reference standards. RESULTS Thirty-five patients were included in this study. At least one lesion was detected in 73% (26/35) of the scans. Management changed in 71% (25/35) of patients, (22 positives and three negative scans). 26.9% (7/26) of patients were found to have an oligometastatic disease. Based on the reference standards, the diagnostic performance of [18F]Fluciclovine PET/CT in detecting recurrence in nmCRCP has 86%, sensitivity, 83% specificity, 96.1% PPV, and 55.5% NPV. There was no relationship between the Gleason score and a positive PET/CT scan in our patient population. CONCLUSION Detecting the source of recurrence is challenging in nmCRCP patients when conventional imaging fails. Given the high PPV, sensitivity, and specificity, [18F]Fluciclovine PET/CT can be used instead of conventional imaging as a first-line choice due to its superiority over bone scan and added value of detecting soft tissue metastasis regardless of the initial Gleason score. ADVANCES IN KNOWLEDGE The study highlights the added value of [18F]Fluciclovine PET/CT in detecting soft tissue metastasis regardless of the initial Gleason score, which is not possible with conventional imaging such as bone scans.The study highlights the potential role of [18F]Fluciclovine PET/CT guiding management change for nonmetastatic castrate-resistant prostate cancer patients, particularly those with oligometastatic disease.
Collapse
Affiliation(s)
- Moataz Soliman
- Department of Radiology, Northwestern University, Chicago, IL, USA
| | - Kareem Elfatairy
- Department of Radiology, Bridgeport Hospital, Yale New Haven, CT, USA
| | - Reham Ellessy
- Department of Radiology, Cairo University, Cairo, Egypt
| | - Yury S Velichko
- Department of Radiology, Northwestern University, Chicago, IL, USA
| | - Ryan Avery
- Department of Radiology, Northwestern University, Chicago, IL, USA
| | - Linda C kelahan
- Department of Radiology, Northwestern University, Chicago, IL, USA
| | - Ashley E Ross
- Department of Urology, Northwestern University, Chicago, IL, USA
| | - Hatice Savas
- Department of Radiology, Northwestern University, Chicago, IL, USA
| |
Collapse
|
31
|
Ogu J, Jayasekera M, Villanueva-Meyer J, Bhargava P. Gradual normalization of superscan in prostate cancer: A case report and literature review. Radiol Case Rep 2023; 18:4323-4326. [PMID: 37789917 PMCID: PMC10542603 DOI: 10.1016/j.radcr.2023.09.015] [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: 05/11/2023] [Revised: 08/30/2023] [Accepted: 09/05/2023] [Indexed: 10/05/2023] Open
Abstract
This report presents the imaging findings in a patient with advanced prostate cancer and bone metastases. A superscan pattern on the initial whole-body bone scan suggested extensive disease. The patient responded well to definitive treatment, exhibiting clinical improvement based on decreased PSA levels and CT findings in 6-month follow-up. However, serial follow-up bone scans showed normalization in about 18 months. This paper aims to discuss the limitations of bone scintigraphy in evaluating treatment responses in patients with prostate cancer.
Collapse
Affiliation(s)
- Julliet Ogu
- Department of Radiology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Migara Jayasekera
- Department of Radiology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | | | - Peeyush Bhargava
- Department of Radiology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| |
Collapse
|
32
|
Francolini G, Garlatti P, Di Cataldo V, Triggiani L, Simoni N, Detti B, Lorenzetti V, Colombo F, Morelli V, Ganovelli M, Caprara L, Orsatti C, Burchini L, Frosini G, Bertini N, Loi M, Simontacchi G, Greto D, Desideri I, Meattini I, Livi L. Pattern of recurrence after stereotactic body radiotherapy for para-aortic oligo-recurrent prostate cancer, a multicentric analysis. LA RADIOLOGIA MEDICA 2023; 128:1423-1428. [PMID: 37597125 DOI: 10.1007/s11547-023-01701-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 08/09/2023] [Indexed: 08/21/2023]
Abstract
BACKGROUND M1a disease represents an intermediate status between loco-regional relapse and bone metastatic disease. Metastasis directed therapy (MDT), through stereotactic body RT (SBRT) may be offered to patients, aiming to exclusively treat sites of macroscopic relapse and avoiding wide prophylactic treatment volumes. This appears as a viable treatment, especially after the rise of PSMA tailored treatment approaches. MATERIALS AND METHODS Data about patients treated in two different institutions were retrieved from a prospectively collected dataset. All included patients were affected by oligo-recurrent M1a disease after definitive RT or radical prostatectomy, defined as ≤ 3 nodal lesions situated above aortic bifurcation and below renal arteries. Both castration resistant PCa (CRPC) and castration sensitive (CSPC) PCa patients were included. All imaging methods were allowed to detect recurrence (CT scan, Choline or PSMA PET/CT).All sites of recurrences were treated with SBRT. RESULTS Median PFS was 10 months (95% CI 8-17). Twelve patients died, with a median OS of 114 months (95% CI 85-114). Out of the 83 recurrences, 2 (2.4%), 11 (13.25%), 36 (43.37%) and 15 (18%) patients had respectively prostate bed only, pelvic nodal, para-aortic or distant relapse. Furthermore, 19 (22.9%) patients experienced a biochemical only relapse with negative imaging at re-staging. DISCUSSION MDT conferred a remarkable PFS outcome in a mixed cohort of CSPC and CRPC patients with m1a disease, with an optimal safety profile. Prospective trials are needed in order to compare MDT and ENRT for these patients, allowing to select the best treatment option.
Collapse
Affiliation(s)
- Giulio Francolini
- Radiation Oncology Unit, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy.
| | - Pietro Garlatti
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Vanessa Di Cataldo
- Radiation Oncology Unit, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Luca Triggiani
- Department of Radiation Oncology, University and Spedali Civili Hospital, Brescia, Italy
| | - Nicola Simoni
- Radiotherapy Unit, Azienda Ospedaliera Universitaria, 43126, Parma, Italy
| | - Beatrice Detti
- Radiation Oncology Unit, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Victoria Lorenzetti
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Federico Colombo
- Radiotherapy Unit, Azienda Ospedaliera Universitaria, 43126, Parma, Italy
| | - Vittorio Morelli
- Department of Radiation Oncology, University and Spedali Civili Hospital, Brescia, Italy
| | - Michele Ganovelli
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Luisa Caprara
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Carolina Orsatti
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Luca Burchini
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Giulio Frosini
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Niccolò Bertini
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Mauro Loi
- Radiation Oncology Unit, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Gabriele Simontacchi
- Radiation Oncology Unit, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Daniela Greto
- Radiation Oncology Unit, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Isacco Desideri
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Icro Meattini
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
| | - Lorenzo Livi
- Department of Biomedical, Experimental and Clinical Sciences "Mario Serio", University of Florence, Florence, Italy
| |
Collapse
|
33
|
Luzzago S, Colombo A, Mistretta FA, Alessi S, Di Trapani E, Summers PE, Piccinelli ML, Raimondi S, Vignati S, Clemente A, Rosati E, di Meglio L, d'Ascoli E, Scarabelli A, Zugni F, Belmonte M, Maggioni R, Ferro M, Fusco N, de Cobelli O, Musi G, Petralia G. Multiparametric MRI-based 5-year Risk Prediction Model for Biochemical Recurrence of Prostate Cancer after Radical Prostatectomy. Radiology 2023; 309:e223349. [PMID: 37987657 DOI: 10.1148/radiol.223349] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Background Current predictive tools to estimate the risk of biochemical recurrence (BCR) after treatment of prostate cancer do not consider multiparametric MRI (mpMRI) information. Purpose To develop a risk prediction tool that considers mpMRI findings to assess the risk of 5-year BCR after radical prostatectomy. Materials and Methods In this retrospective single-center analysis in 1459 patients with prostate cancer who underwent mpMRI before radical prostatectomy (in 2012-2015), the outcome of interest was 5-year BCR (two consecutive prostate-specific antigen [PSA] levels > 0.2 ng/mL [0.2 µg/L]). Patients were randomly divided into training (70%) and test (30%) sets. Kaplan-Meier plots were applied to the training set to estimate survival probabilities. Multivariable Cox regression models were used to test the relationship between BCR and different sets of exploratory variables. The C-index of the final model was calculated for the training and test sets and was compared with European Association of Urology, University of California San Francisco Cancer of the Prostate Risk Assessment, Memorial Sloan-Kettering Cancer Center, and Partin risk tools using the partial likelihood ratio test. Five risk categories were created. Results The median duration of follow-up in the whole cohort was 59 months (IQR, 32-81 months); 376 of 1459 (25.8%) patients had BCR. A multivariable Cox regression model (referred to as PIPEN, and composed of PSA density, International Society of Urological Pathology grade group, Prostate Imaging Reporting and Data System category, European Society of Urogenital Radiology extraprostatic extension score, nodes) fitted to the training data yielded a C-index of 0.74, superior to that of other predictive tools (C-index 0.70 for all models; P ≤ .01) and a median higher C-index on 500 test set replications (C-index, 0.73). Five PIPEN risk categories were identified with 5-year BCR-free survival rates of 92%, 84%, 71%, 56%, and 26% in very low-, low-, intermediate-, high-, and very high-risk patients, respectively (all P < .001). Conclusion A five-item model for predicting the risk of 5-year BCR after radical prostatectomy for prostate cancer was developed and internally verified, and five risk categories were identified. © RSNA, 2023 Supplemental material is available for this article. See also the editorial by Aguirre and Ortegón in this issue.
Collapse
Affiliation(s)
- Stefano Luzzago
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Alberto Colombo
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Francesco A Mistretta
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Sarah Alessi
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Ettore Di Trapani
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Paul E Summers
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Mattia Luca Piccinelli
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Sara Raimondi
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Silvano Vignati
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Alfredo Clemente
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Elisa Rosati
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Letizia di Meglio
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Elisa d'Ascoli
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Alice Scarabelli
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Fabio Zugni
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Maddalena Belmonte
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Roberta Maggioni
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Matteo Ferro
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Nicola Fusco
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Ottavio de Cobelli
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Gennaro Musi
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| | - Giuseppe Petralia
- From the Department of Urology (S.L., F.A.M., E.D.T., M.L.P., M.F., O.D.C., G.M.), Division of Radiology (A.C., S.A., P.E.S., F.Z., M.B.), Department of Experimental Oncology (S.R., S.V.), Division of Pathology (N.F.), and Precision Imaging and Research Unit, Department of Medical Imaging and Radiation Sciences (G.P.), European Institute of Oncology (IEO), IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy; Department of Oncology and Hematology-Oncology (S.L., F.A.M., N.F., O.D.C., G.M., G.P.) and Postgraduate School in Radiodiagnostics (E.R., L.D.M., E.D., A.S., R.M.), University of Milan, Milan, Italy; and Radiology and Radiotherapy Unit, Department of Precision Medicine, University of Campania L. Vanvitelli, Naples, Italy (A.C.)
| |
Collapse
|
34
|
Volpe F, Nappi C, Piscopo L, Zampella E, Mainolfi CG, Ponsiglione A, Imbriaco M, Cuocolo A, Klain M. Emerging Role of Nuclear Medicine in Prostate Cancer: Current State and Future Perspectives. Cancers (Basel) 2023; 15:4746. [PMID: 37835440 PMCID: PMC10571937 DOI: 10.3390/cancers15194746] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
Prostate cancer is the most frequent epithelial neoplasia after skin cancer in men starting from 50 years and prostate-specific antigen (PSA) dosage can be used as an early screening tool. Prostate cancer imaging includes several radiological modalities, ranging from ultrasonography, computed tomography (CT), and magnetic resonance to nuclear medicine hybrid techniques such as single-photon emission computed tomography (SPECT)/CT and positron emission tomography (PET)/CT. Innovation in radiopharmaceutical compounds has introduced specific tracers with diagnostic and therapeutic indications, opening the horizons to targeted and very effective clinical care for patients with prostate cancer. The aim of the present review is to illustrate the current knowledge and future perspectives of nuclear medicine, including stand-alone diagnostic techniques and theragnostic approaches, in the clinical management of patients with prostate cancer from initial staging to advanced disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Michele Klain
- Department of Advanced Biomedical Sciences, University of Naples Federico II, 80138 Naples, Italy; (F.V.); (C.N.); (L.P.); (E.Z.); (C.G.M.); (A.P.); (M.I.); (A.C.)
| |
Collapse
|
35
|
Hara T, Terakawa T, Okamura Y, Bando Y, Furukawa J, Harada K, Nakano Y, Fujisawa M. Real-world analysis of metastatic prostate cancer demonstrates increased frequency of PSA-imaging discordance with visceral metastases and upfront ARAT/docetaxel therapy. Prostate 2023; 83:1270-1278. [PMID: 37316357 DOI: 10.1002/pros.24588] [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: 02/27/2023] [Revised: 05/11/2023] [Accepted: 06/05/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND The objective of this study was to evaluate the background and treatment course of patients with metastatic prostate cancer (PC), with a particular focus on radiographic progression in the absence of prostate-specific antigen (PSA) progression. METHODS The study population consisted of 229 patients with metastatic hormone-sensitive PC (HSPC), who received prostate biopsy and androgen deprivation therapy at Kobe University Hospital between January 2008 and June 2022. Clinical characteristics were retrospectively evaluated using medical records. PSA progression-free status was defined as ≤1.05 times greater than that from 3 months before. Multivariate analyses were performed using the Cox proportional hazards regression model to identify parameters associated with time to progression on imaging without PSA elevation. RESULTS A total of 227 patients with metastatic HSPC without neuroendocrine PC were identified. The median follow-up period was 38.0 months, with a median overall survival of 94.9 months. Six patients exhibited disease progression on imaging without PSA elevation during HSPC treatment, three during first-line castration-resistant PC (CRPC) treatment, and two during late-line CRPC treatment. The rate of disease progression without PSA elevation at 3 years after treatment initiation was 7.4%. Multivariate analysis revealed that organ metastases and upfront treatment with docetaxel or androgen receptor axis-targeted therapy were independent prognostic factors for imaging progression without PSA elevation. CONCLUSIONS Disease progression on imaging without PSA elevation occurred not only during HSPC treatment and first-line CRPC treatment, but also during late-line CRPC treatment. Patients with visceral metastases or those treated with upfront androgen receptor axis-targeted or docetaxel may be more prone to such progression.
Collapse
Affiliation(s)
- Takuto Hara
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoaki Terakawa
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yasuyoshi Okamura
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yukari Bando
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Junya Furukawa
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kenichi Harada
- Department of Urology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yuzo Nakano
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masato Fujisawa
- Department of Urology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
36
|
Cereser L, Evangelista L, Giannarini G, Girometti R. Prostate MRI and PSMA-PET in the Primary Diagnosis of Prostate Cancer. Diagnostics (Basel) 2023; 13:2697. [PMID: 37627956 PMCID: PMC10453091 DOI: 10.3390/diagnostics13162697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/29/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Over the last years, prostate magnetic resonance imaging (MRI) has gained a key role in the primary diagnosis of clinically significant prostate cancer (csPCa). While a negative MRI can avoid unnecessary prostate biopsies and the overdiagnosis of indolent cancers, a positive examination triggers biopsy samples targeted to suspicious imaging findings, thus increasing the diagnosis of csPCa with a sensitivity and negative predictive value of around 90%. The limitations of MRI, including suboptimal positive predictive values, are fueling debate on how to stratify biopsy decisions and management based on patient risk and how to correctly estimate it with clinical and/or imaging findings. In this setting, "next-generation imaging" imaging based on radiolabeled Prostate-Specific Membrane Antigen (PSMA)-Positron Emission Tomography (PET) is expanding its indications both in the setting of primary staging (intermediate-to-high risk patients) and primary diagnosis (e.g., increasing the sensitivity of MRI or acting as a problem-solving tool for indeterminate MRI cases). This review summarizes the current main evidence on the role of prostate MRI and PSMA-PET as tools for the primary diagnosis of csPCa, and the different possible interaction pathways in this setting.
Collapse
Affiliation(s)
- Lorenzo Cereser
- Institute of Radiology, Department of Medicine, University of Udine, 20072 Milan, Italy;
- University Hospital S. Maria della Misericordia, Azienda Sanitaria-Universitaria Friuli Centrale (ASUFC), p.le S. Maria della Misericordia, 15, 33100 Udine, Italy
| | - Laura Evangelista
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072 Milan, Italy
- IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, 20089 Milan, Italy
| | - Gianluca Giannarini
- Urology Unit, University Hospital S. Maria della Misericordia, Azienda Sanitaria-Universitaria Friuli Centrale (ASUFC), p.le S. Maria della Misericordia, 15, 33100 Udine, Italy
| | - Rossano Girometti
- Institute of Radiology, Department of Medicine, University of Udine, 20072 Milan, Italy;
- University Hospital S. Maria della Misericordia, Azienda Sanitaria-Universitaria Friuli Centrale (ASUFC), p.le S. Maria della Misericordia, 15, 33100 Udine, Italy
| |
Collapse
|
37
|
Simon H, Henkel D, Chiron P, Helissey C. New perspectives on metabolic imaging in the management of prostate cancer in 2022: A focus on radiolabeled PSMA‑PET/CT (Review). Mol Clin Oncol 2023; 19:51. [PMID: 37323248 PMCID: PMC10265585 DOI: 10.3892/mco.2023.2647] [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: 11/07/2022] [Accepted: 01/04/2023] [Indexed: 06/17/2023] Open
Abstract
Nuclear medicine is an essential part of prostate cancer management concerning initial staging, patient follow-up and even therapy. Prostate-specific membrane antigen (PSMA) is a glutamate carboxypeptidase II transmembrane glycoprotein expressed by 80% of prostatic cells. The interest in this protein is due to its specificity for prostatic tissue. The use of 68GaPSMA PET/CT in the context of disease staging is thus well-established and recommended, especially for high-risk disease with metastases and lymph node involvement. However, the risk of false positives raises questions regarding its place in the management of patients with prostate cancer. The present study aimed to determine the use of PET-PSMA in the care of patients with prostate cancer but also to assess its limits of use.
Collapse
Affiliation(s)
- Hélène Simon
- Clinical Research Unit, Department of Oncology, Military Hospital Begin, 94160 Saint-Mandé, France
| | - Daniel Henkel
- Unité de Formation et de Recherche 5, University of Paris 8 Vincennes-St. Denis, 93200 Paris, France
| | - Paul Chiron
- Department of Urology, Military Hospital Begin, 94160 Saint-Mandé, France
| | - Carole Helissey
- Clinical Research Unit, Department of Oncology, Military Hospital Begin, 94160 Saint-Mandé, France
| |
Collapse
|
38
|
Boldrini L, Romano A, Chiloiro G, Corradini S, De Luca V, Verusio V, D'Aviero A, Castelluccia A, Alitto AR, Catucci F, Grimaldi G, Trapp C, Hörner-Rieber J, Marchesano D, Frascino V, Mattiucci GC, Valentini V, Gentile P, Gambacorta MA. Magnetic resonance guided SBRT reirradiation in locally recurrent prostate cancer: a multicentric retrospective analysis. Radiat Oncol 2023; 18:84. [PMID: 37218005 PMCID: PMC10201772 DOI: 10.1186/s13014-023-02271-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 04/26/2023] [Indexed: 05/24/2023] Open
Abstract
AIMS Reirradiation of prostate cancer (PC) local recurrences represents an emerging challenge for current radiotherapy. In this context, stereotactic body radiation therapy (SBRT) allows the delivery of high doses, with curative intent. Magnetic Resonance guided Radiation Therapy (MRgRT) has shown promising results in terms of safety, feasibility and efficacy of delivering SBRT thanks to the enhanced soft tissue contrast and the online adaptive workflow. This multicentric retrospective analysis evaluates the feasibility and efficacy of PC reirradiation, using a 0.35 T hybrid MR delivery unit. METHODS Patients affected by local recurrences of PC and treated in five institutions between 2019 and 2022 were retrospectively collected. All patients had undergone previous Radiation Therapy (RT) in definitive or adjuvant setting. Re-treatment MRgSBRT was delivered with a total dose ranging from 25 to 40 Gy in 5 fractions. Toxicity according to CTCAE v 5.0 and treatment response were assessed at the end of the treatment and at follow-up. RESULTS Eighteen patients were included in this analysis. All patients had previously undergone external beam radiation therapy (EBRT) up to a total dose of 59.36 to 80 Gy. Median cumulative biologically effective dose (BED) of SBRT re-treatment was 213,3 Gy (103,1-560), considering an α/β of 1.5. Complete response was achieved in 4 patients (22.2%). No grade ≥ 2 acute genitourinary (GU) toxicity events were recorded, while gastrointestinal (GI) acute toxicity events occurred in 4 patients (22.2%). CONCLUSION The low rates of acute toxicity of this experience encourages considering MRgSBRT a feasibile therapeutic approach for the treatment of clinically relapsed PC. Accurate gating of target volumes, the online adaptive planning workflow and the high definition of MRI treatment images allow delivering high doses to the PTV while efficiently sparing organs at risk (OARs).
Collapse
Affiliation(s)
- Luca Boldrini
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Istituto di Radiologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Angela Romano
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giuditta Chiloiro
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Stefanie Corradini
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Viola De Luca
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Valeria Verusio
- Istituto di Radiologia, Università Cattolica del Sacro Cuore, Rome, Italy.
| | - Andrea D'Aviero
- Radiation Oncology, Mater Olbia Hospital, Olbia, Sassari, Italy
| | - Alessandra Castelluccia
- Radiation Oncology, Ospedale San Pietro Fatebenefratelli di Roma, Rome, Italy
- Radiation Oncology, Department of Radiotherapy, Hospital "A. Perrino", ASL Brindisi, Brindisi, Italy
| | - Anna Rita Alitto
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | | | - Gianmarco Grimaldi
- Radiation Oncology, Ospedale San Pietro Fatebenefratelli di Roma, Rome, Italy
| | - Christian Trapp
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Juliane Hörner-Rieber
- Heidelberg Ion-Beam Therapy Center (HIT), Department of Radiation Oncology, Heidelberg University Hospital, Heidelberg, Germany
| | - Domenico Marchesano
- Radiation Oncology, Ospedale San Pietro Fatebenefratelli di Roma, Rome, Italy
| | - Vincenzo Frascino
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Gian Carlo Mattiucci
- Istituto di Radiologia, Università Cattolica del Sacro Cuore, Rome, Italy
- Radiation Oncology, Mater Olbia Hospital, Olbia, Sassari, Italy
| | - Vincenzo Valentini
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Istituto di Radiologia, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Piercarlo Gentile
- Radiation Oncology, Ospedale San Pietro Fatebenefratelli di Roma, Rome, Italy
| | - Maria Antonietta Gambacorta
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Istituto di Radiologia, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
39
|
Kim SH, Jeon YJ, Bak JK, Yoo BN, Park JW, Ha YC, Lee YK. Association of Androgen Deprivation Therapy with Osteoporotic Fracture in Patients with Prostate Cancer with Low Tumor Burden Using a Retrospective Population-Based Propensity-Score-Matched Cohort. Cancers (Basel) 2023; 15:2822. [PMID: 37345162 DOI: 10.3390/cancers15102822] [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: 02/23/2023] [Revised: 05/08/2023] [Accepted: 05/11/2023] [Indexed: 06/23/2023] Open
Abstract
This study evaluated the effect of androgen deprivation therapy (ADT) on osteoporotic fractures (OF) and its prognostic effect on overall survival in patients with localized or regional prostate cancer (PC) using the Korean National Insurance Dataset. A total of 8883 pairs of 1:1 propensity-score-matched patients with localized or regional PC were retrospectively enrolled between 2007 and 2016. All patients underwent at least 1 year of follow-up to evaluate therapeutic outcomes. Multivariate analysis was performed to determine the prognostic effect of ADT on OF. During a mean follow-up of 47.7 months, 977 (3.43%) patients developed OF, and the incidences of hip, spine, and wrist fractures were significantly different between ADT and non-ADT groups (p < 0.05). The ADT group had a significantly higher incidence of OF (hazard ratio 2.055, 95% confidence interval 1.747-2.417) than the non-ADT group (p < 0.05), and the incidence of spine/hip/wrist OF was significantly higher in the ADT group regardless of the PC stage (p < 0.05). Multivariate analysis failed to show any significant difference in overall survival between the two groups (p > 0.05). ADT resulted in a significantly higher incidence of OF among patients with localized and regional PC, but the overall survival did not differ between ADT and non-ADT groups.
Collapse
Affiliation(s)
- Sung Han Kim
- Department of Urology, Urologic Cancer Center, Research Institute and Hospital of National Cancer Center, Goyang 10408, Republic of Korea
| | - Ye Jhin Jeon
- Department Statistics, Yonsei University, Seoul 03722, Republic of Korea
| | - Jean Kyung Bak
- National Evidence-Based Healthcare Collaborating Agency (NECA), Seoul 04933, Republic of Korea
| | - Bit-Na Yoo
- National Evidence-Based Healthcare Collaborating Agency (NECA), Seoul 04933, Republic of Korea
| | - Jung-Wee Park
- Department of Orthopaedic Surgery, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seognam 13620, Republic of Korea
| | - Yong-Chan Ha
- Department of Orthopaedic Surgery, Seoul Bumin Hospital, Seoul 07590, Republic of Korea
| | - Young-Kyun Lee
- Department of Orthopaedic Surgery, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seognam 13620, Republic of Korea
| |
Collapse
|
40
|
Kapplinger JD, Lima LMF, Packard AT, Nathan MA, Young JR, Stish BJ, Hough DM. Safety and Efficacy of CT-Guided Percutaneous Biopsy of Suspicious Subcentimeter Pelvic and Retroperitoneal Lymph Nodes Detected by 11C-Choline PET in Patients With Prostate Cancer. AJR Am J Roentgenol 2023; 220:718-725. [PMID: 36475814 DOI: 10.2214/ajr.22.28321] [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: 12/12/2022]
Abstract
BACKGROUND. In patients with prostate cancer, PET using targeted radiotracers can identify increased activity in small morphologically normal lymph nodes, facilitating earlier detection of metastatic disease. OBJECTIVE. The purpose of this article was to assess the efficacy and safety of CT-guided biopsy of suspicious pelvic and retroperitoneal lymph nodes measuring smaller than 1 cm detected by 11C-choline PET in patients with prostate cancer, with comparison with nodes measuring 1 cm or larger. METHODS. This retrospective study included patients with prostate cancer who underwent CT-guided percutaneous biopsy of suspicious pelvic or retroperitoneal lymph nodes detected by 11C-choline PET/CT or PET/MRI (performed because of a rising or elevated PSA level or known recurrent or metastatic disease) between June 1, 2012, and March 20, 2020. Patient, lymph node, and procedural characteristics, as well as biopsy outcomes and complications, were recorded. Biopsies of lymph nodes measuring smaller than 1 cm and of lymph nodes measuring 1 cm and larger were compared. RESULTS. A total of 269 patients (mean age, 68.7 ± 6.8 [SD] years) were included. A total of 156 patients underwent biopsy of lymph nodes measuring smaller than 1 cm (range, 3-9 mm); 113 patients underwent biopsy of lymph nodes measuring 1 cm or larger (range, 10-35 mm). Lymph nodes smaller than 1 cm and lymph nodes 1 cm and larger showed no significant difference in diagnostic yield (89.7% vs 92.9%; p = .40). Diagnostic yield was not significantly different between nodes smaller than 1 cm and nodes 1 cm and larger for any individual anatomic location within the pelvis or retroperitoneum (all p > .05). Malignant yield was lower for nodes smaller than 1 cm than for nodes 1 cm and larger (44.9% vs 63.7%; p = .003). The single biopsied 3-mm node had a nondiagnostic specimen. Diagnostic yield and malignant yield were 100.0% and 40.0%, respectively, for 4-mm nodes, and 95.5% and 45.5%, respectively, for 5-mm nodes. Patients with nodes smaller than 1 cm and nodes 1 cm and larger showed no significant difference in minor (12.8% vs 7.1%; p = .16) or major (0.6% vs 2.7%; p = .31) complication rate. CONCLUSION. The findings support the safety and efficacy of CT-guided biopsy of suspicious subcentimeter pelvic and retroperitoneal lymph nodes detected on 11C-choline PET in patients with prostate cancer. CLINICAL IMPACT. Earlier diagnosis of metastatic lymphadenopathy will impact prognostic assessment and management decisions in patients with recurrent prostate cancer.
Collapse
Affiliation(s)
- Jamie D Kapplinger
- Department of Radiology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905
| | - Livia M Frota Lima
- Department of Radiology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905
| | - Annie T Packard
- Department of Radiology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905
| | - Mark A Nathan
- Department of Radiology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905
| | - Jason R Young
- Department of Radiology, Mayo Clinic, Jacksonville, FL
| | - Bradley J Stish
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN
| | - David M Hough
- Department of Radiology, Mayo Clinic, 200 1st St SW, Rochester, MN 55905
| |
Collapse
|
41
|
Fendler WP, Eiber M, Beheshti M, Bomanji J, Calais J, Ceci F, Cho SY, Fanti S, Giesel FL, Goffin K, Haberkorn U, Jacene H, Koo PJ, Kopka K, Krause BJ, Lindenberg L, Marcus C, Mottaghy FM, Oprea-Lager DE, Osborne JR, Piert M, Rowe SP, Schöder H, Wan S, Wester HJ, Hope TA, Herrmann K. PSMA PET/CT: joint EANM procedure guideline/SNMMI procedure standard for prostate cancer imaging 2.0. Eur J Nucl Med Mol Imaging 2023; 50:1466-1486. [PMID: 36604326 PMCID: PMC10027805 DOI: 10.1007/s00259-022-06089-w] [Citation(s) in RCA: 160] [Impact Index Per Article: 80.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 12/18/2022] [Indexed: 01/07/2023]
Abstract
Here we aim to provide updated guidance and standards for the indication, acquisition, and interpretation of PSMA PET/CT for prostate cancer imaging. Procedures and characteristics are reported for a variety of available PSMA small radioligands. Different scenarios for the clinical use of PSMA-ligand PET/CT are discussed. This document provides clinicians and technicians with the best available evidence, to support the implementation of PSMA PET/CT imaging in research and routine practice.
Collapse
Affiliation(s)
- Wolfgang P Fendler
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
- PET Committee of the German Society of Nuclear Medicine, Marburg, Germany
| | - Matthias Eiber
- Department of Nuclear Medicine, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
| | - Mohsen Beheshti
- Division of Molecular Imaging & Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Jamshed Bomanji
- Institute of Nuclear Medicine, UCLH NHS Foundation Trust, London, UK
| | - Jeremie Calais
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, USA
| | - Francesco Ceci
- Division of Nuclear Medicine and Theranostics, IEO European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Steve Y Cho
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | | | - Frederik L Giesel
- Department of Nuclear Medicine, University Hospital Düsseldorf, Medical Faculty, Heinrich-Heine-University and Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Karolien Goffin
- Department of Nuclear Medicine, Division of Nuclear Medicine and Molecular Imaging, University Hospital Leuven, KU Leuven, Louvain, Belgium
| | - Uwe Haberkorn
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Heather Jacene
- Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, USA
| | | | - Klaus Kopka
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- School of Science, Faculty of Chemistry and Food Chemistry, Technical University Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
| | - Bernd J Krause
- Department of Nuclear Medicine, University Medical Center, University of Rostock, Rostock, Germany
| | - Liza Lindenberg
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Charles Marcus
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Felix M Mottaghy
- Department of Nuclear Medicine, University Hospital RWTH Aachen University, Aachen, Germany
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
| | - Daniela E Oprea-Lager
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical Centers, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Joseph R Osborne
- Department of Radiology, Division of Molecular Imaging and Therapeutics, Weill Cornell Medicine, New York, NY, USA
| | - Morand Piert
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Michigan, Ann Arbor, MI, USA
| | - Steven P Rowe
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Heiko Schöder
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Simon Wan
- Institute of Nuclear Medicine, UCLH NHS Foundation Trust, London, UK
| | - Hans-Jürgen Wester
- Pharmaceutical Radiochemistry, Technische Universität München, Walther-Meißner-Str. 3, 85748, Garching, Germany
| | - Thomas A Hope
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany.
| |
Collapse
|
42
|
Capela A, Antunes P, Coelho CA, Garcia CL, Custódio S, Amorim R, Costa T, Vilela E, Teixeira M, Amarelo A, Silva J, Joaquim A, Viamonte S, Brito J, Alves AJ. Effects of walking football on adherence, safety, quality of life and physical fitness in patients with prostate cancer: Findings from the PROSTATA_MOVE randomized controlled trial. Front Oncol 2023; 13:1129028. [PMID: 37025594 PMCID: PMC10070742 DOI: 10.3389/fonc.2023.1129028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/10/2023] [Indexed: 04/08/2023] Open
Abstract
Aims To analyze the feasibility and impact of a walking football (WF) program on quality of life (QoL), cardiorespiratory fitness (CRF), muscle strength, and balance program in men with prostate cancer under androgen deprivation therapy (ADT). Methods Fifty patients with prostate cancer (stages IIb-IVb) under ADT were randomized to a 16-week WF program plus usual care (n=25) or usual care control group (n=25). The WF program consisted of three 90-minute sessions per week. Recruitment, withdrawal, adherence, enjoyment rate, and safety of the intervention were recorded throughout the study. Cardiorespiratory fitness was assessed before and after the interventions, while handgrip strength, lower limb muscle strength, static balance, and QoL were assessed before, during (week 8), and after (week 16) the interventions. Adverse events during sessions were also recorded. Results The WF group showed high levels of adherence (81.6 ± 15.9%) and enjoyment rate (4.5 ± 0.5 out of 5 points). In the intention-to-treat analysis, the WF group showed an improvement in chair sit-to-stand (p=0.035) compared to the control group. Within-group comparisons showed that handgrip strength in the dominant upper limb (p=0.024), maximal isometric muscle strength in the non-dominant lower limb (p=0.006), and balance in the dominant limb (p=0.009) improved over time in the WF group but not in the usual care group. The results obtained from the per-protocol analysis indicate that CRF improved significantly in the WF group as compared to the control group (p=0.035). Within-group analysis revealed that CRF (p=0.036), muscle strength in dominant (p=0.006) and non-dominant (p=0.001) lower limbs, and balance in the non-dominant lower limb (p=0.023) improved after 16 weeks of WF, but not in the control group. One major traumatic injury (muscle tear) was reported with a complete recovery before the end of the intervention. Conclusion This study suggests that WF is feasible, safe, and enjoyable in patients with prostate cancer under hormonal therapy. Furthermore, patients who adhere to the WF program can expect cardiorespiratory fitness, muscle strength, and balance improvements. Clinical trials registration clinicaltrials.gov, identifier NCT04062162.
Collapse
Affiliation(s)
- Andreia Capela
- ONCOMOVE® – Associação de Investigação de Cuidados de Suporte em Oncologia (AICSO), Vila Nova de Gaia, Portugal
- Centro Hospitalar Vila Nova de Gaia – Espinho, Entidade pública empresaríal (EPE), Vila Nova de Gaia, Portugal
| | - Pedro Antunes
- ONCOMOVE® – Associação de Investigação de Cuidados de Suporte em Oncologia (AICSO), Vila Nova de Gaia, Portugal
- Research Center in Sport Sciences, Health and Human Development (CIDESD), Sport Sciences Department, University of Beira Interior, Covilhã, Portugal
| | - César André Coelho
- ONCOMOVE® – Associação de Investigação de Cuidados de Suporte em Oncologia (AICSO), Vila Nova de Gaia, Portugal
| | - Catarina Laranjeiro Garcia
- ONCOMOVE® – Associação de Investigação de Cuidados de Suporte em Oncologia (AICSO), Vila Nova de Gaia, Portugal
- Research Center in Sport Sciences, Health and Human Development (CIDESD), Physical Education and Sport Sciences Department, University of Maia, Maia, Portugal
| | - Sandra Custódio
- ONCOMOVE® – Associação de Investigação de Cuidados de Suporte em Oncologia (AICSO), Vila Nova de Gaia, Portugal
- Centro Hospitalar Vila Nova de Gaia – Espinho, Entidade pública empresaríal (EPE), Vila Nova de Gaia, Portugal
| | - Rui Amorim
- Centro Hospitalar Vila Nova de Gaia – Espinho, Entidade pública empresaríal (EPE), Vila Nova de Gaia, Portugal
| | - Telma Costa
- ONCOMOVE® – Associação de Investigação de Cuidados de Suporte em Oncologia (AICSO), Vila Nova de Gaia, Portugal
- Centro Hospitalar Vila Nova de Gaia – Espinho, Entidade pública empresaríal (EPE), Vila Nova de Gaia, Portugal
| | - Eduardo Vilela
- Centro Hospitalar Vila Nova de Gaia – Espinho, Entidade pública empresaríal (EPE), Vila Nova de Gaia, Portugal
| | - Madalena Teixeira
- Centro Hospitalar Vila Nova de Gaia – Espinho, Entidade pública empresaríal (EPE), Vila Nova de Gaia, Portugal
| | - Anabela Amarelo
- ONCOMOVE® – Associação de Investigação de Cuidados de Suporte em Oncologia (AICSO), Vila Nova de Gaia, Portugal
- Centro Hospitalar Vila Nova de Gaia – Espinho, Entidade pública empresaríal (EPE), Vila Nova de Gaia, Portugal
| | - Joana Silva
- ONCOMOVE® – Associação de Investigação de Cuidados de Suporte em Oncologia (AICSO), Vila Nova de Gaia, Portugal
- Centro Hospitalar Vila Nova de Gaia – Espinho, Entidade pública empresaríal (EPE), Vila Nova de Gaia, Portugal
| | - Ana Joaquim
- ONCOMOVE® – Associação de Investigação de Cuidados de Suporte em Oncologia (AICSO), Vila Nova de Gaia, Portugal
- Centro Hospitalar Vila Nova de Gaia – Espinho, Entidade pública empresaríal (EPE), Vila Nova de Gaia, Portugal
| | - Sofia Viamonte
- ONCOMOVE® – Associação de Investigação de Cuidados de Suporte em Oncologia (AICSO), Vila Nova de Gaia, Portugal
- Centro Hospitalar Vila Nova de Gaia – Espinho, Entidade pública empresaríal (EPE), Vila Nova de Gaia, Portugal
| | - João Brito
- Portugal Football School, Federação Portuguesa de Futebol, Oeiras, Portugal
| | - Alberto J. Alves
- ONCOMOVE® – Associação de Investigação de Cuidados de Suporte em Oncologia (AICSO), Vila Nova de Gaia, Portugal
- Research Center in Sport Sciences, Health and Human Development (CIDESD), Physical Education and Sport Sciences Department, University of Maia, Maia, Portugal
| |
Collapse
|
43
|
Prostate and metastasis diffusion volume based on apparent diffusion coefficient as a prognostic factor in Hormone-naïve prostate Cancer. Clin Exp Metastasis 2023; 40:187-195. [PMID: 36914924 DOI: 10.1007/s10585-023-10200-2] [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: 10/16/2022] [Accepted: 02/20/2023] [Indexed: 03/16/2023]
Abstract
In this study, to assess the utility of whole-body DWI (WB-DWI) as an imaging biomarker for metastatic hormone-naïve prostate cancer (mHNPC), we evaluated tumor diffusion volume based on apparent diffusion coefficient (ADC) values. WB-DWI results obtained from 62 mHNPC patients were evaluated in this retrospective analysis. The association with castration resistant-free survival (CFS) was evaluated for both prostate and metastatic tumor diffusion volume (pDV and mDV, respectively) based on WB-DWI. The usefulness of pDV and mDV based on ADC values to predict CFS was also examined. During the follow-up period, 22 patients progressed to castration-resistant prostate cancer, and the median CFS was 42.6 months. The median mDV and pDV were 6.7 and 12.6 mL, respectively. mDV was a significant predictor of CFS (hazard ratio [HR]: 2.75; p = 0.022), while pDV was not significant. When DV was divided into groups by ADC values (× 10- 3 mm2/s) of 0.4-1.0 and 1.0-1.8 (× 10- 3 mm2/s), mDV with ADC values (× 10- 3 mm2/s) of 0.4-1.0 (mDV0.4-1.0) showed a more favorable association with CFS compared to total mDV. On multivariate analysis, mDV0.4-1.0 and Gleason grade group had a statistically significant association with CFS (HR: 4.0; p = 0.004, and HR: 3.4; p = 0.006, respectively), while pDV with ADC values (× 10- 3 mm2/s) of 0.4-1.0 did not have a significant association. mDV is useful for predicting CFS in mHNPC patients. mDV may be a better imaging biomarker when based on ADC values.
Collapse
|
44
|
Bauckneht M, Miceli A, Signori A, Albano D, Capitanio S, Piva R, Laudicella R, Franchini A, D'Amico F, Riondato M, Chiola S, Marini C, Fornarini G, Scarale A, Muni A, Bertagna F, Burger IA, Sambuceti G, Morbelli S. Combined forced diuresis and late acquisition on [ 68Ga]Ga-PSMA-11 PET/CT for biochemical recurrent prostate cancer: a clinical practice-oriented study. Eur Radiol 2023; 33:3343-3353. [PMID: 36892650 PMCID: PMC10121525 DOI: 10.1007/s00330-023-09516-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/22/2022] [Accepted: 02/03/2023] [Indexed: 03/10/2023]
Abstract
OBJECTIVES Increased detection of prostate cancer (PCa) recurrences using [68Ga]Ga-PSMA-11 PET/CT has been reported by adding forced diuresis or late-phase imaging to the standard protocol. However, the combination of these procedures in the clinical setting is still not standardized. METHODS One hundred prospectively recruited biochemical recurrent PCa patients were restaged with dual-phase [68Ga]Ga-PSMA-11 PET/CT from September 2020 to October 2021. All patients received a standard scan (60 min), followed by diuretics (140 min) and a late-phase abdominopelvic scan (180 min). PET readers with low (n = 2), intermediate (n = 2), or high (n = 2) experience rated (i) standard and (ii) standard + forced diuresis late-phase images in a stepwise fashion according to E-PSMA guidelines, scoring their level of confidence. Study endpoints were (i) accuracy against a composite reference standard, (ii) reader's confidence level, and (iii) interobserver agreement. RESULTS Forced diuresis late-phase imaging increased the reader's confidence category for local and nodal restaging (both p < 0.0001), and the interobserver agreement in identifying nodal recurrences (from moderate to substantial, p < 0.01). However, it significantly increased diagnostic accuracy exclusively for local uptakes rated by low-experienced readers (from 76.5 to 84%, p = 0.05) and for nodal uptakes rated as uncertain at standard imaging (from 68.1 to 78.5%, p < 0.05). In this framework, SUVmax kinetics resulted in an independent predictor of PCa recurrence compared to standard metrics, potentially guiding the dual-phase PET/CT interpretation. CONCLUSIONS The present results do not support the systematic combination of forced diuresis and late-phase imaging in the clinical setting, but allow the identification of patients-, lesions-, and reader-based scenarios that might benefit from it. KEY POINTS • Increased detection of prostate cancer recurrences has been reported by adding diuretics administration or an additional late abdominopelvic scan to the standard [68Ga]Ga-PSMA-11 PET/CT procedure. • We verified the added value of combined forced diuresis and delayed imaging, showing that this protocol only slightly increases the diagnostic accuracy of [68Ga]Ga-PSMA-11 PET/CT, thus not justifying its systematic use in clinics. • However, it can be helpful in specific clinical scenarios, e.g., when PET/CT is reported by low-experienced readers. Moreover, it increased the reader's confidence and the agreement among observers.
Collapse
Affiliation(s)
- Matteo Bauckneht
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Genoa, Italy. .,Department of Health Sciences (DISSAL), University of Genova, Genoa, Italy.
| | - Alberto Miceli
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Department of Health Sciences (DISSAL), University of Genova, Genoa, Italy
| | - Alessio Signori
- Department of Health Sciences (DISSAL), University of Genova, Genoa, Italy
| | - Domenico Albano
- Nuclear Medicine, University of Brescia and ASST Spedali Civili Brescia, Brescia, Italy
| | - Selene Capitanio
- Nuclear Medicine ASST, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Roberta Piva
- Nuclear Medicine Unit, Azienda Ospedaliera SS. Antonio E Biagio E Cesare Arrigo, Alessandria, Italy
| | - Riccardo Laudicella
- Nuclear Medicine, Cantonal Hospital Baden, Baden, Switzerland.,Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland.,Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, Messina, Italy
| | - Annalisa Franchini
- Nuclear Medicine ASST, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Francesca D'Amico
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Department of Health Sciences (DISSAL), University of Genova, Genoa, Italy
| | - Mattia Riondato
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Silvia Chiola
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Cecilia Marini
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,CNR Institute of Molecular Bioimaging and Physiology, Milan, Italy
| | - Giuseppe Fornarini
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Antonio Scarale
- Nuclear Medicine ASST, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Alfredo Muni
- Nuclear Medicine Unit, Azienda Ospedaliera SS. Antonio E Biagio E Cesare Arrigo, Alessandria, Italy
| | - Francesco Bertagna
- Nuclear Medicine, University of Brescia and ASST Spedali Civili Brescia, Brescia, Italy
| | - Irene A Burger
- Nuclear Medicine, Cantonal Hospital Baden, Baden, Switzerland.,Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Gianmario Sambuceti
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Department of Health Sciences (DISSAL), University of Genova, Genoa, Italy
| | - Silvia Morbelli
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,Department of Health Sciences (DISSAL), University of Genova, Genoa, Italy
| |
Collapse
|
45
|
Huo H, Shen S, He D, Liu B, Yang F. Head-to-head comparison of 68Ga-PSMA-11 PET/CT and 68Ga-PSMA-11 PET/MRI in the detection of biochemical recurrence of prostate cancer: summary of head-to-head comparison studies. Prostate Cancer Prostatic Dis 2023; 26:16-24. [PMID: 35931759 DOI: 10.1038/s41391-022-00581-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/20/2022] [Accepted: 07/25/2022] [Indexed: 12/13/2022]
Abstract
PURPOSE Our meta-analysis aimed to evaluate the diagnostic performance of 68Ga-PSMA-11 PET/CT vs. 68Ga-PSMA-11 PET/MRI for biochemical recurrence of prostate cancer. METHODS We searched for relevant articles in PubMed and Embase until February 2022. Studies evaluating head-to-head comparison of 68Ga-PSMA-11 PET/CT and 68Ga-PSMA-11 PET/MRI in men with prostate cancer biochemical recurrence were included. The quality of each study was assessed using the Quality Assessment of Diagnostic Performance Studies-2 (QUADAS-2) tool. RESULTS A total of 5 studies with 219 patients were included in the analysis. The pooled overall detection rates of 68Ga-PSMA-11 PET/CT and 68Ga-PSMA-11 PET/MRI in detecting recurrent PCa after definitive treatment were 0.89 (95% CI: 0.65-1.00), 0.92 (95% CI: 0.77-1.00), while the detection rates were 0.20 (95% CI: 0.05-0.41) and 0.29 (95% CI: 0.10-0.53) in local recurrence, 0.51 (95% CI: 0.33-0.69) and 0.52 (95% CI: 0.44-0.61) in lymph node metastasis, 0.18 (95% CI: 0.07-0.33) and 0.20 (95% CI: 0.09-0.35) in bone metastasis. There was no significant difference between the two imaging modalities in the overall detection rate (P = 0.82). In addition, detection rates were also not significantly different in local recurrence, lymph node metastasis, or bone metastasis (P = 0.54, 1.00, 0.82). CONCLUSIONS 68Ga-PSMA-11 PET/CT and 68Ga-PSMA-11 PET/MRI seem to have equivalent performance in detecting biochemical recurrence in prostate cancer. However, the results of the meta-analysis were drawn from studies with small samples. Further larger studies in this setting are warranted.
Collapse
Affiliation(s)
- Huasong Huo
- Department of Nuclear Medicine, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Shurui Shen
- Department of Nuclear Medicine, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Ding He
- Department of Nuclear Medicine, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Bin Liu
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, China.
| | - Fuwei Yang
- Department of neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, China.
| |
Collapse
|
46
|
Parghane RV, Mahajan A, Chakrabarty N, Basu S. Imaging Recommendations for Theranostic PET-CT in Oncology. Indian J Med Paediatr Oncol 2023. [DOI: 10.1055/s-0042-1760310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023] Open
Abstract
AbstractWe in this article have presented a review of the guideline recommendations on theranostic positron emission tomography-computed tomography (PET-CT) imaging which will be helpful to assist practitioners in providing appropriate patient care. Multiple guidelines by different societies and medical associations provide standards for diagnosis, imaging, and treatment of cancer patients. They have generated a number of recommendations related to 68Ga-DOTATATE and 68Ga-PSMA-11 PET-CT, which are the classical examples of theranostic PET-CT imaging in current practice.
Collapse
Affiliation(s)
- Rahul V. Parghane
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital Annexe, Parel, Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Abhishek Mahajan
- The Clatterbridge Cancer Centre NHS Foundation Trust, Pembroke Place, Liverpool, United Kingdom
| | - Nivedita Chakrabarty
- Department of Radiodiagnosis, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
| | - Sandip Basu
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Hospital Annexe, Parel, Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
| |
Collapse
|
47
|
Knill AK, Blackledge MD, Curcean A, Larkin J, Turajlic S, Riddell A, Koh DM, Messiou C, Winfield JM. Optimisation of b-values for the accurate estimation of the apparent diffusion coefficient (ADC) in whole-body diffusion-weighted MRI in patients with metastatic melanoma. Eur Radiol 2023; 33:863-871. [PMID: 36169688 PMCID: PMC9889461 DOI: 10.1007/s00330-022-09088-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 07/12/2022] [Accepted: 08/04/2022] [Indexed: 02/04/2023]
Abstract
OBJECTIVE To establish optimised diffusion weightings ('b-values') for acquisition of whole-body diffusion-weighted MRI (WB-DWI) for estimation of the apparent diffusion coefficient (ADC) in patients with metastatic melanoma (MM). Existing recommendations for WB-DWI have not been optimised for the tumour properties in MM; therefore, evaluation of acquisition parameters is essential before embarking on larger studies. METHODS Retrospective clinical data and phantom experiments were used. Clinical data comprised 125 lesions from 14 examinations in 11 patients with multifocal MM, imaged before and/or after treatment with immunotherapy at a single institution. ADC estimates from these data were applied to a model to estimate the optimum b-value. A large non-diffusing phantom was used to assess eddy current-induced geometric distortion. RESULTS Considering all tumour sites from pre- and post-treatment examinations together, metastases exhibited a large range of mean ADC values, [0.67-1.49] × 10-3 mm2/s, and the optimum high b-value (bhigh) for ADC estimation was 1100 (10th-90th percentile: 740-1790) s/mm2. At higher b-values, geometric distortion increased, and longer echo times were required, leading to reduced signal. CONCLUSIONS Theoretical optimisation gave an optimum bhigh of 1100 (10th-90th percentile: 740-1790) s/mm2 for ADC estimation in MM, with the large range of optimum b-values reflecting the wide range of ADC values in these tumours. Geometric distortion and minimum echo time increase at higher b-values and are not included in the theoretical optimisation; bhigh in the range 750-1100 s/mm2 should be adopted to maintain acceptable image quality but performance should be evaluated for a specific scanner. KEY POINTS • Theoretical optimisation gave an optimum high b-value of 1100 (10th-90th percentile: 740-1790) s/mm2 for ADC estimation in metastatic melanoma. • Considering geometric distortion and minimum echo time (TE), a b-value in the range 750-1100 s/mm2 is recommended. • Sites should evaluate the performance of specific scanners to assess the effect of geometric distortion and minimum TE.
Collapse
Affiliation(s)
- Annemarie K Knill
- The Institute of Cancer Research, London, UK
- The Royal Marsden NHS Foundation Trust, London, UK
| | | | - Andra Curcean
- The Institute of Cancer Research, London, UK
- The Royal Marsden NHS Foundation Trust, London, UK
| | - James Larkin
- The Royal Marsden NHS Foundation Trust, London, UK
| | - Samra Turajlic
- The Royal Marsden NHS Foundation Trust, London, UK
- The Francis Crick Institute, London, UK
| | | | - Dow Mu Koh
- The Institute of Cancer Research, London, UK
- The Royal Marsden NHS Foundation Trust, London, UK
| | - Christina Messiou
- The Institute of Cancer Research, London, UK.
- The Royal Marsden NHS Foundation Trust, London, UK.
| | - Jessica M Winfield
- The Institute of Cancer Research, London, UK
- The Royal Marsden NHS Foundation Trust, London, UK
| |
Collapse
|
48
|
Margolis D. Next-Generation Diffusion Imaging of Osseous Metastases. Radiol Imaging Cancer 2023; 5:e220167. [PMID: 36705560 PMCID: PMC9896219 DOI: 10.1148/rycan.220167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Daniel Margolis
- From the Department of Radiology, Weill Cornell Imaging, NewYork-Presbyterian, 525 E 68th St, Box 141, New York, NY 10021
| |
Collapse
|
49
|
Roberts MJ, Maurer T, Perera M, Eiber M, Hope TA, Ost P, Siva S, Hofman MS, Murphy DG, Emmett L, Fendler WP. Using PSMA imaging for prognostication in localized and advanced prostate cancer. Nat Rev Urol 2023; 20:23-47. [PMID: 36473945 DOI: 10.1038/s41585-022-00670-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2022] [Indexed: 12/12/2022]
Abstract
The use of prostate-specific membrane antigen (PSMA)-directed applications in modern prostate cancer management has evolved rapidly over the past few years, helping to establish new treatment pathways and provide further insights into prostate cancer biology. However, the prognostic implications of PSMA-PET have not been studied systematically, owing to rapid clinical implementation without long follow-up periods to determine intermediate-term and long-term oncological outcomes. Currently available data suggest that traditional prognostic factors and survival outcomes are associated with high PSMA expression (both according to immunohistochemistry and PET uptake) in men with localized and biochemically recurrent disease. Treatment with curative intent (primary and/or salvage) often fails when PSMA-positive metastases are present; however, the sensitivity of PSMA-PET in detecting all metastases is poor. Low PSMA-PET uptake in recurrent disease is a favourable prognostic factor; however, it can be associated with poor prognosis in conjunction with high 18F-fluorodeoxyglucose uptake in metastatic castration-resistant prostate cancer. Clinical trials embedding PSMA-PET for guiding management with reliable oncological outcomes are needed to support ongoing clinical use.
Collapse
Affiliation(s)
- Matthew J Roberts
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia.
- University of Queensland Centre for Clinical Research, Faculty of Medicine, Brisbane, Queensland, Australia.
- Department of Urology, Redcliffe Hospital, Brisbane, Queensland, Australia.
| | - Tobias Maurer
- Martini-Klinik Prostate Cancer Center, Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Marlon Perera
- Department of Surgery, Austin Health, Heidelberg, Victoria, Australia
| | - Matthias Eiber
- Department of Nuclear Medicine, Technical University of Munich, Munich, Germany
| | - Thomas A Hope
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Piet Ost
- Department of Radiation Oncology, Iridium Network, GZA Ziekenhuizen, Antwerp, Belgium
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Shankar Siva
- Peter MacCallum Cancer Centre, Radiation Oncology, Parkville, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, Melbourne University, Parkville, Victoria, Australia
| | - Michael S Hofman
- Sir Peter MacCallum Department of Oncology, Melbourne University, Parkville, Victoria, Australia
- Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Declan G Murphy
- Sir Peter MacCallum Department of Oncology, Melbourne University, Parkville, Victoria, Australia
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Louise Emmett
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, New South Wales, Australia
- Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University of Duisburg-Essen, Essen, Germany
- PET Committee of the German Society of Nuclear Medicine, Goettingen, Germany
| |
Collapse
|
50
|
Wennmann M, Klein A, Bauer F, Chmelik J, Grözinger M, Uhlenbrock C, Lochner J, Nonnenmacher T, Rotkopf LT, Sauer S, Hielscher T, Götz M, Floca RO, Neher P, Bonekamp D, Hillengass J, Kleesiek J, Weinhold N, Weber TF, Goldschmidt H, Delorme S, Maier-Hein K, Schlemmer HP. Combining Deep Learning and Radiomics for Automated, Objective, Comprehensive Bone Marrow Characterization From Whole-Body MRI: A Multicentric Feasibility Study. Invest Radiol 2022; 57:752-763. [PMID: 35640004 DOI: 10.1097/rli.0000000000000891] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES Disseminated bone marrow (BM) involvement is frequent in multiple myeloma (MM). Whole-body magnetic resonance imaging (wb-MRI) enables to evaluate the whole BM. Reading of such whole-body scans is time-consuming, and yet radiologists can transfer only a small fraction of the information of the imaging data set to the report. This limits the influence that imaging can have on clinical decision-making and in research toward precision oncology. The objective of this feasibility study was to implement a concept for automatic, comprehensive characterization of the BM from wb-MRI, by automatic BM segmentation and subsequent radiomics analysis of 30 different BM spaces (BMS). MATERIALS AND METHODS This retrospective multicentric pilot study used a total of 106 wb-MRI from 102 patients with (smoldering) MM from 8 centers. Fifty wb-MRI from center 1 were used for training of segmentation algorithms (nnU-Nets) and radiomics algorithms. Fifty-six wb-MRI from 8 centers, acquired with a variety of different MRI scanners and protocols, were used for independent testing. Manual segmentations of 2700 BMS from 90 wb-MRI were performed for training and testing of the segmentation algorithms. For each BMS, 296 radiomics features were calculated individually. Dice score was used to assess similarity between automatic segmentations and manual reference segmentations. RESULTS The "multilabel nnU-Net" segmentation algorithm, which performs segmentation of 30 BMS and labels them individually, reached mean dice scores of 0.88 ± 0.06/0.87 ± 0.06/0.83 ± 0.11 in independent test sets from center 1/center 2/center 3-8 (interrater variability between radiologists, 0.88 ± 0.01). The subset from the multicenter, multivendor test set (center 3-8) that was of high imaging quality was segmented with high precision (mean dice score, 0.87), comparable to the internal test data from center 1. The radiomic BM phenotype consisting of 8880 descriptive parameters per patient, which result from calculation of 296 radiomics features for each of the 30 BMS, was calculated for all patients. Exemplary cases demonstrated connections between typical BM patterns in MM and radiomic signatures of the respective BMS. In plausibility tests, predicted size and weight based on radiomics models of the radiomic BM phenotype significantly correlated with patients' actual size and weight ( P = 0.002 and P = 0.003, respectively). CONCLUSIONS This pilot study demonstrates the feasibility of automatic, objective, comprehensive BM characterization from wb-MRI in multicentric data sets. This concept allows the extraction of high-dimensional phenotypes to capture the complexity of disseminated BM disorders from imaging. Further studies need to assess the clinical potential of this method for automatic staging, therapy response assessment, or prediction of biopsy results.
Collapse
Affiliation(s)
| | - André Klein
- Medical Image Computing, German Cancer Research Center
| | | | | | | | | | | | - Tobias Nonnenmacher
- Department of Diagnostic and Interventional Radiology, University Hospital Heidelberg
| | | | - Sandra Sauer
- Department of Medicine V, Multiple Myeloma Section, University Hospital Heidelberg
| | - Thomas Hielscher
- Division of Biostatistics, German Cancer Research Center, Heidelberg
| | | | | | - Peter Neher
- Medical Image Computing, German Cancer Research Center
| | | | - Jens Hillengass
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY
| | | | - Niels Weinhold
- Department of Medicine V, Multiple Myeloma Section, University Hospital Heidelberg
| | - Tim Frederik Weber
- Department of Diagnostic and Interventional Radiology, University Hospital Heidelberg
| | | | | | | | | |
Collapse
|