1
|
Giunta EF, Caroli P, Scarpi E, Altavilla A, Rossetti V, Marini I, Celli M, Casadei C, Lolli C, Schepisi G, Bleve S, Brighi N, Cursano MC, Paganelli G, Matteucci F, De Giorgi U. Correlation of [ 68Ga]Ga-PSMA PET/CT response and PSA decline in first-line enzalutamide for metastatic castration-resistant prostate cancer patients. Eur J Nucl Med Mol Imaging 2024; 52:326-334. [PMID: 39207484 PMCID: PMC11599341 DOI: 10.1007/s00259-024-06887-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024]
Abstract
PURPOSE to assess the utility of response monitoring to enzalutamide by using [68Ga]Ga-PSMA PET in mCRPC patients treated with enzalutamide as first-line therapy. METHODS patients underwent [68Ga]Ga-PSMA PET less than 8 weeks before and 3 months after starting enzalutamide. On the basis of EAU/EANM criteria, patients were categorized as PSMA responders (PET-R) or PSMA non-responders (PET-NR), whilst, based on PSA, they were classified as biochemical responders (PSA-R) or non-responders (PSA-NR). Survival analysis was performed using the Cox regression hazard model and the Kaplan-Meier method. RESULTS 69 patients were considered fully evaluable. We observed 47.8% of concordance between [68Ga]Ga-PSMA PET and PSA monitoring at 3 months after starting enzalutamide. For discordant cases, the PSA reduction has a weak impact on PFS and a significant impact on OS in PET-NR patients, whilst this change has no impact either for PFS and OS in PET-R ones. CONCLUSIONS [68Ga]Ga-PSMA PET could be a useful imaging tool for monitoring response to enzalutamide in mCRPC patients, being more informative than PSA in this setting, and possibly better guiding clinicians in therapeutic decisions.
Collapse
Affiliation(s)
- Emilio Francesco Giunta
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy.
| | - Paola Caroli
- Department of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Emanuela Scarpi
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Amelia Altavilla
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Virginia Rossetti
- Department of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Irene Marini
- Department of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Monica Celli
- Department of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Chiara Casadei
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Cristian Lolli
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giuseppe Schepisi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Sara Bleve
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Nicole Brighi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Maria Concetta Cursano
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giovanni Paganelli
- Department of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Federica Matteucci
- Department of Nuclear Medicine, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Ugo De Giorgi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| |
Collapse
|
2
|
Van Damme J, Tombal B, Michoux N, Van Nieuwenhove S, Pasoglou V, Triqueneaux P, Padhani AR, Lecouvet FE. Value of Whole-body Magnetic Resonance Imaging Using the MET-RADS-P Criteria for Assessing the Response to Intensified Androgen Deprivation Therapy in Metastatic Hormone-naïve and Castration-resistant Prostate Cancer. Eur Urol Oncol 2024:S2588-9311(24)00238-4. [PMID: 39505670 DOI: 10.1016/j.euo.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/27/2024] [Accepted: 10/14/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND AND OBJECTIVES We assessed the agreement between prostate-specific antigen (PSA) and imaging responses using whole-body magnetic resonance imaging (wbMRI). Our aim was to explore the potential prognostic value of PSA and wbMRI responses in metastatic hormone-naïve prostate cancer (mHNPC) and castration-resistant PC (mCRPC). METHODS wbMRI was prospectively performed in 37 patients with mHNPC and 51 with mCRPC before and after 6-12 mo of androgen deprivation therapy and an androgen receptor pathway inhibitor (ARPI). Imaging responses were defined according to the Metastasis Reporting and Data System for PC (MET-RADS-P) criteria. A PSA response was defined as PSA ≤0.2 ng/ml in mHNPC and a ≥50% decrease from the pretreatment level in mCRPC. Agreement between PSA and wbMRI responses was assessed using Cohen's κ. The association between time to subsequent treatment and overall survival (OS) was analyzed using Cox regression analysis. KEY FINDINGS AND LIMITATIONS Agreement between PSA and wbMRI responses was fair in mHNPC (κ = 0.30) but none to slight in mCRPC (κ = 0.15). In mHNPC, patients with a PSA or wbMRI response were less likely to receive subsequent treatments; wbMRI progression was associated with a significantly higher risk of death (hazard ratio 8.59; p = 0.002). In mCRPC, two-thirds of patients with a PSA response showed progression on wbMRI; neither PSA nor wbMRI progression changed the likelihood of starting a subsequent treatment or the risk of death. CONCLUSIONS AND CLINICAL IMPLICATIONS In mHNPC, wbMRI progression was associated with a higher risk of needing subsequent treatment and shorter OS. PATIENT SUMMARY We evaluated the agreement between routine PSA (prostate-specific antigen) test results and whole-body MRI (magnetic resonance imaging) scans for assessing the response of metastatic prostate cancer to treatment. There was disagreement between the PSA and MRI results, mainly for patients with cancer that was resistant to hormone-based treatment. Combining PSA with whole-body MRI might provide a more accurate picture of the response of advanced prostate cancer to treatment.
Collapse
Affiliation(s)
- Julien Van Damme
- Department of Urology, Chirurgie Expérimentale et Transplantation, Institut du Cancer Roi Albert II/Institut de Recherche Expérimentale & Clinique, Cliniques Universitaires Saint Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Bertrand Tombal
- Department of Urology, Chirurgie Expérimentale et Transplantation, Institut du Cancer Roi Albert II/Institut de Recherche Expérimentale & Clinique, Cliniques Universitaires Saint Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Nicolas Michoux
- Department of Radiology and Medical Imaging, Institut du Cancer Roi Albert II/Institut de Recherche Expérimentale & Clinique, Cliniques Universitaires Saint Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Sandy Van Nieuwenhove
- Department of Radiology and Medical Imaging, Institut du Cancer Roi Albert II/Institut de Recherche Expérimentale & Clinique, Cliniques Universitaires Saint Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Vassiliki Pasoglou
- Department of Radiology and Medical Imaging, Institut du Cancer Roi Albert II/Institut de Recherche Expérimentale & Clinique, Cliniques Universitaires Saint Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Perrine Triqueneaux
- Department of Radiology and Medical Imaging, Institut du Cancer Roi Albert II/Institut de Recherche Expérimentale & Clinique, Cliniques Universitaires Saint Luc, Université Catholique de Louvain, Brussels, Belgium
| | - Anwar R Padhani
- Paul Strickland Scanner Centre, Mount Vernon Hospital, Northwood, UK
| | - Frederic E Lecouvet
- Department of Radiology and Medical Imaging, Institut du Cancer Roi Albert II/Institut de Recherche Expérimentale & Clinique, Cliniques Universitaires Saint Luc, Université Catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
3
|
Mollica V, Marchetti A, Fraccascia N, Nanni C, Tabacchi E, Malizia C, Argalia G, Rosellini M, Tassinari E, Paccapelo A, Fanti S, Massari F. A prospective study on the early evaluation of response to androgen receptor-targeted agents with 11C-Choline, 68Ga-PSMA, and 18F-FACBC PET in metastatic castration-resistant prostate cancer: a single-center experience. ESMO Open 2024; 9:103448. [PMID: 38718704 PMCID: PMC11090858 DOI: 10.1016/j.esmoop.2024.103448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 03/30/2024] [Accepted: 04/08/2024] [Indexed: 05/27/2024] Open
Abstract
BACKGROUND The early identification of responsive and resistant patients to androgen receptor-targeting agents (ARTA) in metastatic castration-resistant prostate cancer (mCRPC) is not completely possible with prostate-specific antigen (PSA) assessment and conventional imaging. Considering its ability to determine metabolic activity of lesions, positron emission tomography (PET) assessment might be a promising tool. PATIENTS AND METHODS We carried out a monocentric prospective study in patients with mCRPC treated with ARTA to evaluate the role of different PET radiotracers: 49 patients were randomized to receive 11C-Choline, Fluorine 18 fluciclovine (anti-1-amino-3-18F-fluorocyclobutane-1-carboxylic acid - FACBC) (18F-FACBC), or Gallium-68-prostate-specific-membrane-antigen (68Ga-PSMA) PET, one scan before therapy and one 2 months later. The primary aim was to investigate the performance of three novel PET radiotracers for the early evaluation of response to ARTA in metastatic CRPC patients; the outcome evaluated was biochemical response (PSA reduction ≥50%). The secondary aim was to investigate the prognostic role of several semiquantitative PET parameters and their variations with the different radiotracers in terms of biochemical progression-free survival (bPFS) and overall survival (OS). The study was promoted by the Italian Department of Health (code RF-2016-02364809). RESULTS Regarding the primary endpoint, at log-rank test a statistically significant correlation was found between metabolic tumor volume (MTV) (P = 0.018) and total lesion activity (TLA) (P = 0.025) percentage variation among the two scans with 68Ga-PSMA PET and biochemical response. As for the secondary endpoints, significant correlations with bPFS were found for 68Ga-PSMA total MTV and TLA at the first scan (P = 0.001 and P = 0.025, respectively), and MTV percentage variation (P = 0.031). For OS, statistically significant correlations were found for different 68Ga-PSMA and 18F-FACBC parameters and for major maximum standardized uptake value at the first 11C-Choline PET scan. CONCLUSIONS Our study highlighted that 11C-Choline, 68Ga-PSMA, and 18F-FACBC semiquantitative PET parameters and their variations present a prognostic value in terms of OS and bPFS, and MTV and TLA variations with 68Ga-PSMA PET a correlation with biochemical response, which could help to assess the response to ARTA.
Collapse
Affiliation(s)
- V Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna; Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna.
| | - A Marchetti
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna; Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna
| | - N Fraccascia
- UOC Medicina Nucleare-Centro PET/TC, Ente Ecclesiastico Ospedale Generale Regionale 'F. Miulli', Acquaviva delle Fonti, Bari
| | - C Nanni
- Division of Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna
| | - E Tabacchi
- Division of Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna
| | - C Malizia
- Division of Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna
| | - G Argalia
- Nuclear Medicine, Department of Radiological Sciences, University Hospital of Marche, Ancona
| | - M Rosellini
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna; Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna
| | - E Tassinari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna; Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna
| | - A Paccapelo
- Research and Innovation Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - S Fanti
- Division of Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna
| | - F Massari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna; Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna
| |
Collapse
|
4
|
Oldan JD, Almaguel F, Voter AF, Duran A, Gafita A, Pomper MG, Hope TA, Rowe SP. PSMA-Targeted Radiopharmaceuticals for Prostate Cancer Diagnosis and Therapy. Cancer J 2024; 30:176-184. [PMID: 38753752 DOI: 10.1097/ppo.0000000000000718] [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: 05/18/2024]
Abstract
ABSTRACT Prostate cancer (PCa) is the most common noncutaneous malignancy in men. Until recent years, accurate imaging of men with newly diagnosed PCa, or recurrent or low-volume metastatic disease, was limited. Further, therapeutic options for men with advanced, metastatic, castration-resistant disease were increasingly limited as a result of increasing numbers of systemic therapies being combined in the upfront metastatic setting. The advent of urea-based, small-molecule inhibitors of prostate-specific membrane antigen (PSMA) has partially addressed those shortcomings in diagnosis and therapy of PCa. On the diagnostic side, there are multiple pivotal phase III trials with several different agents having demonstrated utility in the initial staging setting, with generally modest sensitivity but very high specificity for determining otherwise-occult pelvic nodal involvement. That latter statistic drives the utility of the scan by allowing imaging interpreters to read with very high sensitivity while maintaining a robust specificity. Other pivotal phase III trials have demonstrated high detection efficiency in patients with biochemical failure, with high positive predictive value at the lesion level, opening up possible new avenues of therapy such as metastasis-directed therapy. Beyond the diagnostic aspects of PSMA-targeted radiotracers, the same urea-based chemical scaffolds can be altered to deliver therapeutic isotopes to PCa cells that express PSMA. To date, one such agent, when combined with best standard-of-care therapy, has demonstrated an ability to improve overall survival, progression-free survival, and freedom from skeletal events relative to best standard-of-care therapy alone in men with metastatic, castration-resistant PCa who are post chemotherapy. Within the current milieu, there are a number of important future directions including the use of artificial intelligence to better leverage diagnostic findings, further medicinal chemistry refinements to the urea-based structure that may allow improved tumor targeting and decreased toxicities, and the incorporation of new radionuclides that may better balance efficacy with toxicities than those nuclides that are available.
Collapse
Affiliation(s)
- Jorge D Oldan
- From the Department of Radiology, University of North Carolina, Chapel Hill, NC
| | - Frankis Almaguel
- Department of Radiology, Loma Linda University School of Medicine, Loma Linda, CA
| | - Andrew F Voter
- The Russell H. Morgan Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Alfonso Duran
- Department of Radiology, Loma Linda University School of Medicine, Loma Linda, CA
| | - Andrei Gafita
- The Russell H. Morgan Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Martin G Pomper
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Thomas A Hope
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA
| | - Steven P Rowe
- From the Department of Radiology, University of North Carolina, Chapel Hill, NC
| |
Collapse
|
5
|
Bauckneht M, Ciccarese C, Laudicella R, Mosillo C, D'Amico F, Anghelone A, Strusi A, Beccia V, Bracarda S, Fornarini G, Tortora G, Iacovelli R. Theranostics revolution in prostate cancer: Basics, clinical applications, open issues and future perspectives. Cancer Treat Rev 2024; 124:102698. [PMID: 38359590 DOI: 10.1016/j.ctrv.2024.102698] [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: 11/22/2023] [Revised: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 02/17/2024]
Abstract
In the last years, theranostics has expanded the therapeutic options available for prostate cancer patients. In this review, we explore this dynamic field and its potential to revolutionize precision medicine for prostate cancer. We delve into the foundational principles, clinical applications, and emerging opportunities, emphasizing the potential synergy between radioligand therapy and other systemic treatments. Additionally, we address the ongoing challenges, including optimizing patient selection, assessing treatment responses, and determining the role of theranostics within the broader landscape of prostate cancer treatment.
Collapse
Affiliation(s)
- Matteo Bauckneht
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Health Sciences (DISSAL), University of Genova, Genova, Italy.
| | - Chiara Ciccarese
- Medical Oncology, Fondazione Policlinico A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Riccardo Laudicella
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98124 Messina, Italy
| | - Claudia Mosillo
- Oncologia Medica e Traslazionale, Azienda Ospedaliera Santa Maria di Terni, Terni, Italy
| | - Francesca D'Amico
- Department of Health Sciences (DISSAL), University of Genova, Genova, Italy
| | - Annunziato Anghelone
- Medical Oncology, Fondazione Policlinico A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Alessandro Strusi
- Medical Oncology, Fondazione Policlinico A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Viria Beccia
- Medical Oncology, Fondazione Policlinico A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Sergio Bracarda
- Oncologia Medica e Traslazionale, Azienda Ospedaliera Santa Maria di Terni, Terni, Italy
| | - Giuseppe Fornarini
- Medical Oncology 1, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Giampaolo Tortora
- Medical Oncology, Fondazione Policlinico A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; Università Cattolica del Sacro Cuore, Rome, Italy
| | - Roberto Iacovelli
- Medical Oncology, Fondazione Policlinico A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
6
|
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
|
7
|
Lindgren Belal S, Frantz S, Minarik D, Enqvist O, Wikström E, Edenbrandt L, Trägårdh E. Applications of Artificial Intelligence in PSMA PET/CT for Prostate Cancer Imaging. Semin Nucl Med 2024; 54:141-149. [PMID: 37357026 DOI: 10.1053/j.semnuclmed.2023.06.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 06/12/2023] [Indexed: 06/27/2023]
Abstract
Prostate-specific membrane antigen (PSMA) positron emission tomography/computed tomography (PET/CT) has emerged as an important imaging technique for prostate cancer. The use of PSMA PET/CT is rapidly increasing, while the number of nuclear medicine physicians and radiologists to interpret these scans is limited. Additionally, there is variability in interpretation among readers. Artificial intelligence techniques, including traditional machine learning and deep learning algorithms, are being used to address these challenges and provide additional insights from the images. The aim of this scoping review was to summarize the available research on the development and applications of AI in PSMA PET/CT for prostate cancer imaging. A systematic literature search was performed in PubMed, Embase and Cinahl according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. A total of 26 publications were included in the synthesis. The included studies focus on different aspects of artificial intelligence in PSMA PET/CT, including detection of primary tumor, local recurrence and metastatic lesions, lesion classification, tumor quantification and prediction/prognostication. Several studies show similar performances of artificial intelligence algorithms compared to human interpretation. Few artificial intelligence tools are approved for use in clinical practice. Major limitations include the lack of external validation and prospective design. Demonstrating the clinical impact and utility of artificial intelligence tools is crucial for their adoption in healthcare settings. To take the next step towards a clinically valuable artificial intelligence tool that provides quantitative data, independent validation studies are needed across institutions and equipment to ensure robustness.
Collapse
Affiliation(s)
- Sarah Lindgren Belal
- Department of Translational Medicine and Wallenberg Centre for Molecular Medicine, Lund University, Malmö, Sweden; Department of Surgery, Skåne University Hospital, Malmö, Sweden
| | - Sophia Frantz
- Department of Translational Medicine and Wallenberg Centre for Molecular Medicine, Lund University, Malmö, Sweden; Department of Health Technology Assessment South, Skåne University Hospital, Lund, Sweden
| | - David Minarik
- Department of Translational Medicine and Wallenberg Centre for Molecular Medicine, Lund University, Malmö, Sweden; Department of Radiation Physics, Skåne University Hospital, Malmö, Sweden
| | - Olof Enqvist
- Department of Electrical Engineering, Chalmers University of Technology, Gothenburg, Sweden; Department of Clinical Physiology and Nuclear Medicine, Malmö Sweden
| | - Erik Wikström
- Department of Health Technology Assessment South, Skåne University Hospital, Lund, Sweden
| | - Lars Edenbrandt
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Elin Trägårdh
- Department of Translational Medicine and Wallenberg Centre for Molecular Medicine, Lund University, Malmö, Sweden; Department of Clinical Physiology and Nuclear Medicine, Skåne University Hospital, Malmö, Sweden.
| |
Collapse
|
8
|
Oprea-Lager DE, MacLennan S, Bjartell A, Briganti A, Burger IA, de Jong I, De Santis M, Eberlein U, Emmett L, Fizazi K, Gillessen S, Herrmann K, Heskamp S, Iagaru A, Jereczek-Fossa BA, Kunikowska J, Lam M, Nanni C, O'Sullivan JM, Panebianco V, Sala E, Sathekge M, Sosnowski R, Tilki D, Tombal B, Treglia G, Tunariu N, Walz J, Yakar D, Dierckx R, Sartor O, Fanti S. European Association of Nuclear Medicine Focus 5: Consensus on Molecular Imaging and Theranostics in Prostate Cancer. Eur Urol 2024; 85:49-60. [PMID: 37743194 DOI: 10.1016/j.eururo.2023.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/17/2023] [Accepted: 09/04/2023] [Indexed: 09/26/2023]
Abstract
BACKGROUND In prostate cancer (PCa), questions remain on indications for prostate-specific membrane antigen (PSMA) positron emission tomography (PET) imaging and PSMA radioligand therapy, integration of advanced imaging in nomogram-based decision-making, dosimetry, and development of new theranostic applications. OBJECTIVE We aimed to critically review developments in molecular hybrid imaging and systemic radioligand therapy, to reach a multidisciplinary consensus on the current state of the art in PCa. DESIGN, SETTING, AND PARTICIPANTS The results of a systematic literature search informed a two-round Delphi process with a panel of 28 PCa experts in medical or radiation oncology, urology, radiology, medical physics, and nuclear medicine. The results were discussed and ratified in a consensus meeting. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Forty-eight statements were scored on a Likert agreement scale and six as ranking options. Agreement statements were analysed using the RAND appropriateness method. Ranking statements were analysed using weighted summed scores. RESULTS AND LIMITATIONS After two Delphi rounds, there was consensus on 42/48 (87.5%) of the statements. The expert panel recommends PSMA PET to be used for staging the majority of patients with unfavourable intermediate and high risk, and for restaging of suspected recurrent PCa. There was consensus that oligometastatic disease should be defined as up to five metastases, even using advanced imaging modalities. The group agreed that [177Lu]Lu-PSMA should not be administered only after progression to cabazitaxel and that [223Ra]RaCl2 remains a valid therapeutic option in bone-only metastatic castration-resistant PCa. Uncertainty remains on various topics, including the need for concordant findings on both [18F]FDG and PSMA PET prior to [177Lu]Lu-PSMA therapy. CONCLUSIONS There was a high proportion of agreement among a panel of experts on the use of molecular imaging and theranostics in PCa. Although consensus statements cannot replace high-certainty evidence, these can aid in the interpretation and dissemination of best practice from centres of excellence to the wider clinical community. PATIENT SUMMARY There are situations when dealing with prostate cancer (PCa) where both the doctors who diagnose and track the disease development and response to treatment, and those who give treatments are unsure about what the best course of action is. Examples include what methods they should use to obtain images of the cancer and what to do when the cancer has returned or spread. We reviewed published research studies and provided a summary to a panel of experts in imaging and treating PCa. We also used the research summary to develop a questionnaire whereby we asked the experts to state whether or not they agreed with a list of statements. We used these results to provide guidance to other health care professionals on how best to image men with PCa and what treatments to give, when, and in what order, based on the information the images provide.
Collapse
Affiliation(s)
| | - Steven MacLennan
- Academic Urology Unit, Institute of Applied Health Sciences, University of Aberdeen, Aberdeen, UK
| | - Anders Bjartell
- Department of Translational Medicine, Medical Faculty, Lund University, Lund, Sweden; Department of Urology, Skåne University Hospital, Skåne, Sweden
| | - Alberto Briganti
- Department of Urology, Vita e Salute San Raffaele University, Milan, Italy
| | - Irene A Burger
- Nuclear Medicine Department, Kantonspital Baden, Baden, Switzerland
| | - Igle de Jong
- Department of Urology, University Medical Center Groningen, Groningen, The Netherlands
| | - Maria De Santis
- Department of Urology, Charité Universitätsmedizin Berlin, Berlin, Germany; Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Uta Eberlein
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Louise Emmett
- Theranostics and Nuclear Medicine Department, St Vincent's Hospital Sydney, Sydney, Australia
| | - Karim Fizazi
- Department of Cancer Medicine, Institut Gustave Roussy, University of Paris Saclay, Villejuif, France
| | - Silke Gillessen
- Oncology Institute of Southern Switzerland (IOSI), Bellinzona, Switzerland; Department of Medical Oncology, Università della Svizzera Italiana, Lugano, Switzerland
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | - Sandra Heskamp
- Department of Medical Imaging-Nuclear Medicine Radboudumc, Nijmegen, The Netherlands
| | - Andrei Iagaru
- Division of Nuclear Medicine and Molecular Imaging, Stanford University Medical Center, Stanford, CA, USA
| | - Barbara Alicja Jereczek-Fossa
- Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy; Department of Radiation Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Jolanta Kunikowska
- Nuclear Medicine Department, Medical University of Warsaw, Warsaw, Poland
| | - Marnix Lam
- Department of Radiology and Nuclear Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Cristina Nanni
- Nuclear Medicine Unit, IRCCS Azienda Ospitaliero-Universitaria di Bologna, Bologna, Italy
| | - Joe M O'Sullivan
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK; Northern Ireland Cancer Centre, Belfast, UK
| | - Valeria Panebianco
- Department of Radiological Sciences, Oncology and Pathology, Sapienza University of Rome, Rome, Italy
| | - Evis Sala
- Department of Radiology, Università Cattolica del Sacro Cuore and Advanced Radiology Center, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Mike Sathekge
- Nuclear Medicine Department, University of Pretoria, Pretoria, South Africa; Nuclear Medicine Department, Steve Biko Academic Hospital, Pretoria, South Africa
| | - Roman Sosnowski
- Department of Urooncology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - 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
| | - Bertrand Tombal
- Department of Surgery, Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Giorgio Treglia
- Clinic of Nuclear Medicine, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland; Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Nina Tunariu
- Clinical Radiology, Drug Development Unit and Prostate Cancer Targeted Therapy Clinical Trials, Royal Marsden Hospital, London, UK
| | - Jochen Walz
- Department of Urology, Institut Paoli-Calmettes Cancer Centre, Marseille, France
| | - Derya Yakar
- Department of Radiology, University Medical Center of Groningen, Groningen, The Netherlands; Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Rudi Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University Medical Centre Groningen, Groningen, The Netherlands
| | - Oliver Sartor
- Departments of Medicine and Urology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Stefano Fanti
- Nuclear Medicine Division, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Policlinico S.Orsola, Bologna, Italy
| |
Collapse
|
9
|
Shagera QA, Karfis I, Kristanto P, Spyridon S, Diamand R, Santapau A, Peltier A, Roumeguère T, Flamen P, Artigas C. PSMA PET/CT for Response Assessment and Overall Survival Prediction in Patients with Metastatic Castration-Resistant Prostate Cancer Treated with Androgen Receptor Pathway Inhibitors. J Nucl Med 2023; 64:1869-1875. [PMID: 37770114 DOI: 10.2967/jnumed.123.265874] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 09/07/2023] [Indexed: 10/02/2023] Open
Abstract
We aimed to evaluate the role of prostate-specific membrane antigen (PSMA) PET/CT for response assessment and outcome prediction in patients with metastatic castration-resistant prostate cancer (mCRPC) treated with androgen receptor pathway inhibitors (ARPIs), including abiraterone acetate or enzalutamide. Methods: We retrospectively analyzed 30 ARPI-treated mCRPC patients who underwent 68Ga-PSMA-11 PET/CT within 8 wk before (baseline) and 12 ± 4 wk after treatment initiation. Total PSMA tumor volume was calculated using the fixed threshold method (SUV ≥ 3). Patients were categorized as PSMA responders (PSMA-Rs) or PSMA nonresponders (PSMA-NRs) on the basis of both European Association of Urology/European Association of Nuclear Medicine (EAU/EANM) criteria and Response Evaluation Criteria in PSMA PET/CT (RECIP) 1.0. PSMA-R included patients with a complete response, a partial response, or stable disease, and PSMA-NR included those with progressive disease. On the basis of prostate-specific antigen (PSA), patients were classified as biochemical responders if PSA decreased by at least 50% and as nonresponders if it did not. The Φ-coefficient was used to evaluate the correlation of PSMA- and PSA-based responses. Survival analysis was performed using the Cox regression hazard model and the Kaplan-Meier method. Predictive accuracy was tested for both response criteria. Results: On the basis of PSMA PET/CT, 13 (43%) patients were PSMA-NR according to the EAU/EANM criteria and 11 (37%) patients were PSMA-NR according to RECIP 1.0. Significant correlations were observed between PSMA- and PSA-based responses for both criteria (Φ = 0.79 and 0.66, respectively). After a median follow-up of 25 mo (interquartile range, 21-43 mo), the median overall survival was significantly longer for PSMA-R than PSMA-NR (54 vs. 22 mo) for both the EAU/EANM criteria and RECIP 1.0, with hazard ratios of 6.9 (95% CI, 1.9-26; P = 0.004) and 5.6 (95% CI, 1.69-18.26, P = 0.005), respectively. No significant difference in predictive accuracy was found between the 2 criteria (C-index, 0.79 vs. 0.76, respectively, P = 0.54). Flare phenomena at the second PSMA PET study were not observed in our cohort. Conclusion: Our results demonstrate that PSMA PET/CT is a valuable imaging biomarker for response assessment and overall survival prediction when performed at 3 mo after ARPI treatment initiation in mCRPC patients. Both proposed PSMA response criteria (EAU/EANM and RECIP 1.0) seem to perform equally well. No PSMA flare was observed. Prospective validation of these findings is strongly needed.
Collapse
Affiliation(s)
- Qaid Ahmed Shagera
- Department of Nuclear Medicine, Institut Jules Bordet, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Ioannis Karfis
- Department of Nuclear Medicine, Institut Jules Bordet, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Paulus Kristanto
- Data Centre, Unité de Gestion de l'Information, Institut Jules Bordet, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Sideris Spyridon
- Department of Oncology, Institut Jules Bordet, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium; and
| | - Romain Diamand
- Department of Urology, Institut Jules Bordet, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Albert Santapau
- Department of Nuclear Medicine, Institut Jules Bordet, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Alexandre Peltier
- Department of Urology, Institut Jules Bordet, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Thierry Roumeguère
- Department of Urology, Institut Jules Bordet, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Patrick Flamen
- Department of Nuclear Medicine, Institut Jules Bordet, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
| | - Carlos Artigas
- Department of Nuclear Medicine, Institut Jules Bordet, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium;
| |
Collapse
|
10
|
Onal C, Guler OC, Torun N, Oymak E, Reyhan M. The significance of metabolic response to neoadjuvant androgen deprivation therapy detected with [ 68Ga]Ga-PSMA-11-PET/CT in high-risk prostate cancer patients treated with definitive radiotherapy. Eur J Nucl Med Mol Imaging 2023; 50:3755-3764. [PMID: 37402832 DOI: 10.1007/s00259-023-06321-1] [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: 05/06/2023] [Accepted: 06/23/2023] [Indexed: 07/06/2023]
Abstract
PURPOSE We examined the prognostic significance of early changes in primary tumor SUV measured with Gallium-68-labeled prostate-specific membrane antigen positron emission tomography ([68Ga]Ga-PSMA-11-PET/CT) and serum PSA values after neoadjuvant androgen deprivation treatment (nADT) in high-risk prostate cancer (PCa) patients treated with definitive radiotherapy (RT). METHODS The clinical data and SUV parameters of 71 PCa patients were reviewed retrospectively. The serum PSA and primary tumor SUV values were calculated before and after the start of ADT. Using univariable and multivariable analyses, the prognostic factors predicting biochemical disease free survival (bDFS) and prostate cancer specific survival (PCSS) were investigated. In addition, logistic regression analysis was used to identify predictors of biochemical failure (BF). RESULTS All but one patient responded with a 98.8% reduction in serum PSA (21.8 ng/mL vs. 0.3 ng/mL; p < 0.001), and 64 patients (91.1%) had a median 66.6% decrease in primary tumor SUV after ADT (13.2 vs. 4.8, p < 0.001). The primary tumor SUV response rate was significantly higher in patients with Gleason score (GS) of 7 than in patients with GS > 7 (59.5% vs. 40.5%; p = 0.04), and it was significantly lower in patients with inadequate treatment response than in those with complete (CR) or partial response (PR) (1.1% vs. 66.1%; p < 0.001). There was a strong and significant correlation (Spearman = 0.41, p < 0.001) and a high concordance (91.5%) between PSA response and SUV response after ADT. With a median follow-up time of 76.1 months, the 5-year bDFS and PCSS rates were 77.2% and 92.2%, respectively. Nineteen patients (26.7%) patients had recurrence at a median of 44.6 months after the completion of RT. In multivariate analysis, lymph node metastasis, GS greater than 7, and SD/PD after nADT were independent predictors of worse bDFS. However, no significant factor for PCSS was identified. In the multivariable logistic regression analysis, advanced age, GS of > 7 disease, lymph node metastasis, and SD or PD after nADT were independent predictors of BF. CONCLUSION These results imply that the metabolic response measured with [68Ga]Ga-PSMA-11-PET/CT after nADT could be used to predict progression in high-risk PCa patients treated with definitive RT.
Collapse
Affiliation(s)
- Cem Onal
- Department of Radiation Oncology, Baskent University Faculty of Medicine, Adana Dr. Turgut Noyan Research and Treatment Center, Adana, 01120, Turkey.
- Department of Radiation Oncology, Baskent University Faculty of Medicine, Ankara, Turkey.
| | - Ozan Cem Guler
- Department of Radiation Oncology, Baskent University Faculty of Medicine, Adana Dr. Turgut Noyan Research and Treatment Center, Adana, 01120, Turkey
| | - Nese Torun
- Department of Nuclear Medicine, Baskent University Faculty of Medicine, Adana Dr. Turgut Noyan Research and Treatment Center, Adana, Turkey
| | - Ezgi Oymak
- Radiation Oncology Unit, Iskenderun Gelisim Hospital, Hatay, Turkey
| | - Mehmet Reyhan
- Department of Nuclear Medicine, Baskent University Faculty of Medicine, Adana Dr. Turgut Noyan Research and Treatment Center, Adana, Turkey
| |
Collapse
|
11
|
Esen B, Seymen H, Tarim K, Koseoglu E, Bolukbasi Y, Falay O, Selçukbiricik F, Molinas Mandel N, Kordan Y, Demirkol MO, Tilki D, Esen T. Diagnostic Performance of 68Ga-PSMA-11 Positron Emission Tomography/Computed Tomography to Monitor Treatment Response in Patients with Metastatic Prostate Cancer: The Concordance Between Biochemical Response and Prostate-specific Membrane Antigen Results. Eur Urol Focus 2023; 9:832-837. [PMID: 37032281 DOI: 10.1016/j.euf.2023.03.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/16/2023] [Accepted: 03/29/2023] [Indexed: 04/11/2023]
Abstract
BACKGROUND Treatment response is traditionally monitored using prostate-specific antigen (PSA) and conventional imaging in patients with metastatic prostate cancer (mPCa). OBJECTIVE To assess the diagnostic performance of prostate-specific membrane antigen (PSMA) positron emission tomography (PET)/computed tomography (CT) when monitoring mPCa patients receiving systemic treatment and also to investigate the concordance between PSMA PET response according to the PSMA PET progression (PPP) criteria and biochemical response. DESIGN, SETTING, AND PARTICIPANTS A total of 96 patients with 68Ga-PSMA-11 PET/CT-detected mPCa at baseline PSMA PET/CT (bPSMA) who underwent at least one follow-up scan after receiving systemic treatment were included in the study. PSA levels at bPSMA and follow-up PSMA PET (fPSMA) scans were recorded. The PPP criteria were used to define PSMA progression. Biochemical progression was defined as ≥25% increase in PSA. PSMA PET and PSA responses were dichotomized into progressive disease (PD) versus non-PD, and the concordance between PSA and PSMA responses was evaluated. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The concordance between PSA and PSMA PET responses was presented using frequencies, percentages, and Cohen's kappa test. RESULTS AND LIMITATIONS A total of 345 serial PSMA PET/CT (96 bPSMA and 249 fPSMA) scans were evaluated. The positivity rates of PSMA PET scans for PSA levels of <0.01, 0.01-0.2, 0.2-4, and >4 ng/ml were 55.6%, 75.0%, 100%, and 98.8%, respectively. PSA and PSMA responses showed moderate-to-high concordance (Cohen's κ = 0.623, p < 0.001). PSA-PSMA discordance was detected in 39 scans (17%). The most common cause of discordance was the discordant results between different metastatic lesions (16/28, 57.1%) in patients with PPP without PSA progression and local progression in prostate (n = 7/11, 63.6%) in patients with PSA progression without PPP. CONCLUSIONS PSMA PET/CT showed very high detection rates of malignant lesions even at very low PSA values and showed significant concordance with PSA response when monitoring treatment response in patients receiving systemic treatment for mPCa. PATIENT SUMMARY This study describes that prostate-specific membrane antigen positron emission tomography (PSMA PET), a new sensitive imaging tool, can detect malignant lesions even at very low prostate-specific antigen values when monitoring metastatic prostate cancer. The PSMA PET response and biochemical response showed significant concordance, and the reason for discordant results seems to be the different responses of metastatic lesions and prostatic lesions to systemic treatment.
Collapse
Affiliation(s)
- Baris Esen
- Department of Urology, Koc University Hospital, Istanbul, Turkey.
| | - Hulya Seymen
- Department of Nuclear Medicine, Koc University Hospital, Istanbul, Turkey
| | - Kayhan Tarim
- Department of Urology, Koc University Hospital, Istanbul, Turkey
| | - Ersin Koseoglu
- Department of Urology, Koc University Hospital, Istanbul, Turkey
| | - Yasemin Bolukbasi
- Department of Radiation Oncology, Koc University Hospital, Istanbul, Turkey
| | - Okan Falay
- Department of Nuclear Medicine, Koc University Hospital, Istanbul, Turkey
| | | | - Nil Molinas Mandel
- Department of Medical Oncology, Koc University Hospital, Istanbul, Turkey
| | - Yakup Kordan
- Department of Urology, Koc University Hospital, Istanbul, Turkey
| | | | - Derya Tilki
- Department of Urology, Koc University Hospital, Istanbul, Turkey; Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Tarık Esen
- Department of Urology, Koc University Hospital, Istanbul, Turkey
| |
Collapse
|
12
|
Eveslage M, Rassek P, Riegel A, Maksoud Z, Bauer J, Görlich D, Noto B. Diffusion-Weighted MRI for Treatment Response Assessment in Osteoblastic Metastases-A Repeatability Study. Cancers (Basel) 2023; 15:3757. [PMID: 37568573 PMCID: PMC10417276 DOI: 10.3390/cancers15153757] [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: 06/19/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
The apparent diffusion coefficient (ADC) is a candidate marker of treatment response in osteoblastic metastases that are not evaluable by morphologic imaging. However, it is unclear whether the ADC meets the basic requirement for reliable treatment response evaluation, namely a low variance of repeated measurements in relation to the differences found between viable and nonviable metastases. The present study addresses this question by analyzing repeated in vivo ADCmedian measurements of 65 osteoblastic metastases in nine patients, as well as phantom measurements. PSMA-PET served as a surrogate for bone metastasis viability. Measures quantifying repeatability were calculated and differences in mean ADC values according to PSMA-PET status were examined. The relative repeatability coefficient %RC of ADCmedian measurements was 5.8% and 12.9% for phantom and in vivo measurements, respectively. ADCmedian values of bone metastases ranged from 595×10-6mm2/s to 2090×10-6mm2/s with an average of 63% higher values in nonviable metastases compared with viable metastases (p < 0.001). ADC shows a small repeatability coefficient in relation to the difference in ADC values between viable and nonviable metastases. Therefore, ADC measurements fulfill the technical prerequisite for reliable treatment response evaluation in osteoblastic metastases.
Collapse
Affiliation(s)
- Maria Eveslage
- Institute of Biostatistics and Clinical Research, University of Münster, 48149 Münster, Germany
| | - Philipp Rassek
- Department of Nuclear Medicine, University Hospital Münster, 48149 Münster, Germany
| | - Arne Riegel
- Clinic for Radiology, University Hospital Münster, 48149 Münster, Germany
| | - Ziad Maksoud
- Clinic for Radiology, University Hospital Münster, 48149 Münster, Germany
| | - Jochen Bauer
- Clinic for Radiology, University Hospital Münster, 48149 Münster, Germany
| | - Dennis Görlich
- Institute of Biostatistics and Clinical Research, University of Münster, 48149 Münster, Germany
| | - Benjamin Noto
- Institute of Biostatistics and Clinical Research, University of Münster, 48149 Münster, Germany
- Department of Nuclear Medicine, University Hospital Münster, 48149 Münster, Germany
- Clinic for Radiology, University Hospital Münster, 48149 Münster, Germany
| |
Collapse
|
13
|
Esen B, Herrmann K, Bavbek S, Kordan Y, Tilki D, Esen T. Prostate-specific Membrane Antigen Positron Emission Tomography as a Biomarker to Assess Treatment Response in Patients with Advanced Prostate Cancer. Eur Urol Focus 2023:S2405-4569(23)00040-8. [PMID: 36842919 DOI: 10.1016/j.euf.2023.02.001] [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: 12/21/2022] [Revised: 01/11/2023] [Accepted: 02/08/2023] [Indexed: 02/28/2023]
Abstract
CONTEXT Prostate-specific membrane antigen (PSMA)-targeted positron emission tomography (PET) has superior accuracy for detection of metastatic lesions in patients with prostate cancer (PC). Although PSMA PET has a prominent role in primary and secondary imaging of PC, data on its role in assessing treatment response in advanced PC are limited. OBJECTIVE To review current data in the literature regarding the impact of antiandrogen therapy on PSMA expression of metastatic sites and the role of serial (baseline and at least 1 follow-up scan) PSMA PET to assess treatment response in patients with metastatic PC. EVIDENCE ACQUISITION A comprehensive literature search in the PubMed database was performed using the terms "PSMA expression prostate", "PSMA regulation", "PSMA PET response assessment", and "serial PSMA PET". EVIDENCE SYNTHESIS Serial PSMA PET studies (baseline and at least 1 follow-up scan) provide valuable data regarding PSMA expression changes after systemic treatment in patients with metastatic PC. PSMA PET-detected flare and upregulation of PSMA expression following hormonal intervention seem to be early events resolving after 3 mo of treatment. PSMA PET imaging is essential in selecting patients for 177Lu-PSMA radioligand therapy (RLT). Growing evidence favors its use in assessing treatment responses after RLT. Preliminary evidence indicates the value of PSMA PET for assessment of the treatment response in patients receiving systemic treatment other than RLT for metastatic PC. CONCLUSIONS PSMA flare following antiandrogen therapy seems to be an early event and thus PET scans should be performed no earlier than 3 mo after the start of treatment. PSMA PET has a promising role in tailoring treatment according to the specific needs of individual patients and assessing responses following systemic treatment in patients with advanced PC. PATIENT SUMMARY This review describes how a sensitive imaging method can be used to assess the tumor response to treatment for metastatic prostate cancer.
Collapse
Affiliation(s)
- Barış Esen
- Department of Urology, School of Medicine, Koç University, Istanbul, Turkey.
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium-University Hospital Essen, Essen, Germany
| | - Sevil Bavbek
- Department of Medical Oncology, VKF American Hospital, Istanbul, Turkey
| | - Yakup Kordan
- Department of Urology, School of Medicine, Koç University, Istanbul, Turkey
| | - Derya Tilki
- Department of Urology, School of Medicine, Koç University, Istanbul, Turkey; Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Tarık Esen
- Department of Urology, School of Medicine, Koç University, Istanbul, Turkey
| |
Collapse
|
14
|
Evaluation of Tumor Burden Response to Single-cycle of Lu-177 PSMA Treatment with Whole Body Scintigraphic Planar Images in Prostate Cancer Patients. JOURNAL OF BASIC AND CLINICAL HEALTH SCIENCES 2022. [DOI: 10.30621/jbachs.1189713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The aim of this study; evaluation of treatment response and survival with post-therapy images in patients who received one cycle of Lu-177 PSMA I&T treatment.
Collapse
|
15
|
Rodríguez-Fraile M, Tamayo Alonso P, Rosales Castillo JJ, de Arcocha-Torres M, Caresia-Aróztegui A, Puig Cózar-Santiago M, Orcajo-Rincon J, Simó Perdigó M, Delgado Bolton RC, Artigas Guix C. Utilidad de los radioligandos PSMA en el diagnóstico y tratamiento del carcinoma de próstata. Rev Esp Med Nucl Imagen Mol 2022. [DOI: 10.1016/j.remn.2021.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
16
|
Rodríguez-Fraile M, Tamayo Alonso P, Rosales JJ, de Arcocha-Torres M, Caresia-Aróztegui AP, Cózar-Santiago MP, Orcajo-Rincon J, Simó Perdigó M, Delgado Bolton RC, Artigas Guix C. The role of PSMA radioligands in the diagnosis and treatment of prostate carcinoma. Rev Esp Med Nucl Imagen Mol 2022; 41:126-135. [PMID: 35216940 DOI: 10.1016/j.remnie.2022.02.001] [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: 10/11/2021] [Accepted: 12/01/2021] [Indexed: 11/25/2022]
Abstract
Prostate cancer (PC) is the most common tumor in men in the West and the fifth leading cause of cancer-related death. The use of PSMA radioligands has represented an important advance both in its diagnosis, through PET molecular imaging, and in its treatment in advanced stages of the disease. This article reviews the contribution of PET studies with PSMA radioligands in initial staging, in tumor detection in biochemical recurrence (elevation of PSA) after treatment with curative intent, and in the more advanced stages of the disease (castration resistant PC or CRPC). The contribution of PSMA radioligand therapy (PSMA-RLT) in CRPC patients who progress to standard therapy is also analyzed.
Collapse
Affiliation(s)
- M Rodríguez-Fraile
- Servicio de Medicina Nuclear, Clínica Universidad de Navarra, Pamplona, Spain.
| | - P Tamayo Alonso
- Servicio de Medicina Nuclear, Hospital Universitario de Salamanca, Salamanca, Spain
| | - J J Rosales
- Servicio de Medicina Nuclear, Clínica Universidad de Navarra, Pamplona, Spain
| | | | | | | | | | - Marc Simó Perdigó
- Servicio de Medicina Nuclear Hospital Vall d'Hebron, Barcelona, Spain
| | - R C Delgado Bolton
- Servicio de Medicina Nuclear, Hospital San Pedro - Centro de Investigación Biomédica de La Rioja (CIBIR), La Rioja, Spain
| | - C Artigas Guix
- Servicio de Medicina Nuclear, Institut Jules Bordet, Université Libre de Bruxelles, Bruselas, Belgium
| | | |
Collapse
|
17
|
Oprea-Lager DE, Cysouw MC, Boellaard R, Deroose CM, de Geus-Oei LF, Lopci E, Bidaut L, Herrmann K, Fournier LS, Bäuerle T, deSouza NM, Lecouvet FE. Bone Metastases Are Measurable: The Role of Whole-Body MRI and Positron Emission Tomography. Front Oncol 2021; 11:772530. [PMID: 34869009 PMCID: PMC8640187 DOI: 10.3389/fonc.2021.772530] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/04/2021] [Indexed: 12/14/2022] Open
Abstract
Metastatic tumor deposits in bone marrow elicit differential bone responses that vary with the type of malignancy. This results in either sclerotic, lytic, or mixed bone lesions, which can change in morphology due to treatment effects and/or secondary bone remodeling. Hence, morphological imaging is regarded unsuitable for response assessment of bone metastases and in the current Response Evaluation Criteria In Solid Tumors 1.1 (RECIST1.1) guideline bone metastases are deemed unmeasurable. Nevertheless, the advent of functional and molecular imaging modalities such as whole-body magnetic resonance imaging (WB-MRI) and positron emission tomography (PET) has improved the ability for follow-up of bone metastases, regardless of their morphology. Both these modalities not only have improved sensitivity for visual detection of bone lesions, but also allow for objective measurements of bone lesion characteristics. WB-MRI provides a global assessment of skeletal metastases and for a one-step "all-organ" approach of metastatic disease. Novel MRI techniques include diffusion-weighted imaging (DWI) targeting highly cellular lesions, dynamic contrast-enhanced MRI (DCE-MRI) for quantitative assessment of bone lesion vascularization, and multiparametric MRI (mpMRI) combining anatomical and functional sequences. Recommendations for a homogenization of MRI image acquisitions and generalizable response criteria have been developed. For PET, many metabolic and molecular radiotracers are available, some targeting tumor characteristics not confined to cancer type (e.g. 18F-FDG) while other targeted radiotracers target specific molecular characteristics, such as prostate specific membrane antigen (PSMA) ligands for prostate cancer. Supporting data on quantitative PET analysis regarding repeatability, reproducibility, and harmonization of PET/CT system performance is available. Bone metastases detected on PET and MRI can be quantitatively assessed using validated methodologies, both on a whole-body and individual lesion basis. Both have the advantage of covering not only bone lesions but visceral and nodal lesions as well. Hybrid imaging, combining PET with MRI, may provide complementary parameters on the morphologic, functional, metabolic and molecular level of bone metastases in one examination. For clinical implementation of measuring bone metastases in response assessment using WB-MRI and PET, current RECIST1.1 guidelines need to be adapted. This review summarizes available data and insights into imaging of bone metastases using MRI and PET.
Collapse
Affiliation(s)
- Daniela E. Oprea-Lager
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Matthijs C.F. Cysouw
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Christophe M. Deroose
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
- Nuclear Medicine & Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Lioe-Fee de Geus-Oei
- Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
- Biomedical Photonic Imaging Group, University of Twente, Enschede, Netherlands
| | - Egesta Lopci
- Nuclear Medicine Unit, IRCCS – Humanitas Research Hospital, Milan, Italy
| | - Luc Bidaut
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- College of Science, University of Lincoln, Lincoln, United Kingdom
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen, and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | - Laure S. Fournier
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Paris Cardiovascular Research Center (PARCC), Institut National de la Santé et de la Recherche Médicale (INSERM), Radiology Department, Assistance Publique-Hôpitaux de Paris (AP-HP), Hopital europeen Georges Pompidou, Université de Paris, Paris, France
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
| | - Tobias Bäuerle
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Nandita M. deSouza
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- Division of Radiotherapy and Imaging, The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Frederic E. Lecouvet
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology, Institut de Recherche Expérimentale et Clinique (IREC), Cliniques Universitaires Saint Luc, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
18
|
Diagnostic performance of deep learning models for detecting bone metastasis on whole-body bone scan in prostate cancer. Eur J Nucl Med Mol Imaging 2021; 49:585-595. [PMID: 34363089 DOI: 10.1007/s00259-021-05481-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 06/21/2021] [Indexed: 12/22/2022]
Abstract
PURPOSE We evaluated the performance of deep learning classifiers for bone scans of prostate cancer patients. METHODS A total of 9113 consecutive bone scans (5342 prostate cancer patients) were initially evaluated. Bone scans were labeled as positive/negative for bone metastasis using clinical reports and image review for ground truth diagnosis. Two different 2D convolutional neural network (CNN) architectures were proposed: (1) whole body-based (WB) and (2) tandem architectures integrating whole body and local patches, here named as "global-local unified emphasis" (GLUE). Both models were trained using abundant (72%:8%:20% for training:validation:test sets) and limited training data (10%:40%:50%). The allocation of test sets was rotated across all images: therefore, fivefold and twofold cross-validation test results were available for abundant and limited settings, respectively. RESULTS A total of 2991 positive and 6142 negative bone scans were used as input. For the abundant training setting, the receiver operating characteristics curves of both the GLUE and WB models indicated excellent diagnostic ability in terms of the area under the curve (GLUE: 0.936-0.955, WB: 0.933-0.957, P > 0.05 in four of the fivefold tests). The overall accuracies of the GLUE and WB models were 0.900 and 0.889, respectively. With the limited training setting, the GLUE models showed significantly higher AUCs than the WB models (0.894-0.908 vs. 0.870-0.877, P < 0.0001). CONCLUSION Our 2D-CNN models accurately classified bone scans of prostate cancer patients. While both showed excellent performance with the abundant dataset, the GLUE model showed higher performance than the WB model in the limited data setting.
Collapse
|
19
|
Seitzer KE, Seifert R, Kessel K, Roll W, Schlack K, Boegemann M, Rahbar K. Lutetium-177 Labelled PSMA Targeted Therapy in Advanced Prostate Cancer: Current Status and Future Perspectives. Cancers (Basel) 2021; 13:3715. [PMID: 34359614 PMCID: PMC8371469 DOI: 10.3390/cancers13153715] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/09/2021] [Accepted: 07/21/2021] [Indexed: 11/17/2022] Open
Abstract
Patients suffering from metastatic castration-resistant prostate cancer (mCRPC) have a poor prognosis. As a further treatment option 177Lutetium (Lu) prostate-specific membrane antigen (PSMA) radioligand therapy gained a significant interest of many investigators. Several publications showed great response and prolonged survival with limited adverse events. However, to this point, it still remains unclear which patients benefit the most from 177Lu-PSMA therapy, and how to improve the treatment regimen to achieve best outcome while minimizing potential adverse events. The efficacy for mCRPC patients is a given fact, and with the newly published results of the VISION trial its approval is only a matter of time. Recently, investigators started to focus on treating prostate cancer patients in earlier disease stages and in combination with other compounds. This review gives a brief overview of the current state and the future perspectives of 177Lu labelled PSMA radioligand therapy.
Collapse
Affiliation(s)
- Konstantin Egon Seitzer
- Department of Urology, University Hospital Muenster, 48149 Muenster, Germany; (K.E.S.); (K.S.); (M.B.)
- Department of Nuclear Medicine, University Hospital Muenster, 48149 Muenster, Germany; (R.S.); (K.K.); (W.R.)
| | - Robert Seifert
- Department of Nuclear Medicine, University Hospital Muenster, 48149 Muenster, Germany; (R.S.); (K.K.); (W.R.)
- Department of Nuclear Medicine, University Hospital Essen, 45147 Essen, Germany
| | - Katharina Kessel
- Department of Nuclear Medicine, University Hospital Muenster, 48149 Muenster, Germany; (R.S.); (K.K.); (W.R.)
| | - Wolfgang Roll
- Department of Nuclear Medicine, University Hospital Muenster, 48149 Muenster, Germany; (R.S.); (K.K.); (W.R.)
| | - Katrin Schlack
- Department of Urology, University Hospital Muenster, 48149 Muenster, Germany; (K.E.S.); (K.S.); (M.B.)
| | - Martin Boegemann
- Department of Urology, University Hospital Muenster, 48149 Muenster, Germany; (K.E.S.); (K.S.); (M.B.)
| | - Kambiz Rahbar
- Department of Nuclear Medicine, University Hospital Muenster, 48149 Muenster, Germany; (R.S.); (K.K.); (W.R.)
| |
Collapse
|
20
|
PSMA Expression Predicts Early Biochemical Response in Patients with Metastatic Castration-Resistant Prostate Cancer under 177Lu-PSMA-617 Radioligand Therapy. Cancers (Basel) 2021; 13:cancers13122938. [PMID: 34208246 PMCID: PMC8230748 DOI: 10.3390/cancers13122938] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/30/2021] [Accepted: 06/09/2021] [Indexed: 01/02/2023] Open
Abstract
Simple Summary Prostate-specific membrane antigen (PSMA) is a promising target for both imaging and radioligand therapies (RLT) for men with prostate cancer. However, not all patients respond to RLT and some even progress early in the treatment course. We aimed to identify parameters to forecast which patients will achieve therapy response prior to commencing RLT. Therefore, we tested whether the tumor volume, the level of PSMA expression or a combination of both in metastases derived from PSMA-targeted molecular imaging prior to RLT can inform the treating physician whether a patient will respond to RLT. Compared to tumor volume, the level of PSMA-expression can better identify patients responding to RLT early in the treatment course. Abstract 177Lu-Prostate-specific membrane antigen (PSMA)-radioligand therapy (RLT) is a promising treatment option in patients with metastatic castration-resistant prostate cancer (mCRPC). We aimed to determine the predictive value of pretherapeutic PSMA-ligand positron emission tomography (PET) and established clinical parameters for early biochemical response after two cycles of RLT. In total, 71 mCRPC patients who had undergone PET/computed tomography (CT) with 68Ga-PSMA-11 prior to two cycles of 177Lu-PSMA-617 RLT were included. Malignant lesions on pretherapeutic PET/CTs were manually segmented and average maximum PSMA expression (maximum standardized uptake values, SUVmax), whole-body PSMA-tumor volume (TV), and whole-body total lesion (TL)-PSMA were calculated. We then tested the predictive performance of these parameters for early biochemical response (defined as prostate-sepcific antigen (PSA) decrease of ≥50% according to PCWG2) after two cycles of RLT, relative to established clinical parameters. Early PSA response was observed in 34/71 patients. PSA change after two cycles of RLT correlated with pretherapeutic SUVmax (r = −0.49; p < 0.001), but not with PSMA-TV (r = 0.02; p = 0.89) or TL-PSMA (r = −0.15; p = 0.22). A cut-off of 19.8 for SUVmax and 75.5 years for age was defined by receiver operating characteristics and revealed a significant outcome difference for early biochemical response between patients with adversely low vs. high PSMA expression and low vs. high age (p < 0.001). Multivariate analysis identified SUVmax (HR, 7.94, p = 0.001) and age (HR, 8.05, p = 0.002) as independent predictors for PSA response early in the treatment course. Thus, high age and high PSMA expression in patients scheduled for RLT identify patients with early biochemical response. This study provides a rationale for further prospective studies exploring PET-guided treatment intensification in selected patients.
Collapse
|