1
|
Hausmann D, Rupp N, Kuzmanic B, Spielhofer N, Heimer J, Koelzer V, Nowak M, Gampp C, Hefermehl L, Kubik-Huch RA, Singer G, Burger IA. How Prostate Cancer Growth Patterns Impact Detection and Interreader Agreement on Multiparametric MRI. Acad Radiol 2024:S1076-6332(24)00828-6. [PMID: 39643467 DOI: 10.1016/j.acra.2024.10.040] [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: 08/15/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 12/09/2024]
Abstract
RATIONALE AND OBJECTIVES Multiparametric MRI (mpMRI) substantially improves the detection of significant prostate carcinoma (PCa) compared to systematic biopsy. Nevertheless, mpMRI can overlook aggressive forms of PCa. Recent studies showed, that infiltrative growth (INF) has less restricted diffusion. This study aims to explore the impact of growth patterns on the detection of lesions. MATERIALS AND METHODS This retrospective study analyzed 52 patients who underwent radical prostatectomy, with preoperative mpMRI. For each patient, one dominant lesion was identified on one whole-mount prostatectomy section. Two pathologists (P1, P2) independently classified the growth pattern whether as expansive (EXP) being defined with at least three 5mm² regions of interest consisting entirely of carcinoma without benign glands or else as infiltrative (INF). Two radiologists (R1, R2) independently classified selected lesions according to PI-RADSv2.1. based on pathological localization. Apparent diffusion coefficient (ADC) values were measured in correlation with matched histopathology slides. Interreader-agreement was evaluated using weighted Cohen's Kappa. The relationship between PI-RADS scores and pathological diagnoses was analyzed using logistic regression. RESULTS Pathologic lesion characterization regarding growth patterns achieved almost perfect agreement (κ = 0.88), so did PI-RADS classification of mpMRI (κ = 0.90). PI-RADS scores correlated significantly with EXP growth patterns. Average ADC values were lower for EXP lesions (0.83×10-3 mm2/s, CI: 0.72-0.94×10-3 mm2/s) compared to INF lesions (0.97×10-3 mm2/s, CI: 0.86-1.07×10-3 mm2/s; p = 0.08). On T2 images, 8 of 28 (29%) INF lesions and 1 of 24 (4%) EXP lesions were not visible. CONCLUSION PCa missed on mpMRI more frequently demonstrate INF growth patterns. Lesions with EXP growth patterns show lower ADC values and have higher PI-RADS scores.
Collapse
Affiliation(s)
- Daniel Hausmann
- Department of Radiology, Kantonsspital Baden, affiliated Hospital for Research and Teaching of the Faculty of Medicine of the University of Zurich, Baden, Switzerland; Department of Clinical Radiology and Nuclear Medicine, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| | - N Rupp
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Switzerland
| | - B Kuzmanic
- Department of Pathology, Kantonsspital Baden, Baden, Switzerland
| | - N Spielhofer
- Department of Nuclear Medicine, Kantonsspital Baden, affiliated Hospital for Research and Teaching of the Faculty of Medicine, University of Zurich, Baden, Switzerland; Department of Health Science and Technology ETH Zurich, Zurich, Switzerland
| | - J Heimer
- Department of Mathematics, Seminar for Statistics, ETH Zurich, Zurich, Switzerland
| | - V Koelzer
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Switzerland
| | - M Nowak
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Switzerland
| | - C Gampp
- Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - L Hefermehl
- Department of Urology, Kantonsspital Baden, Baden, Switzerland
| | - R A Kubik-Huch
- Department of Radiology, Kantonsspital Baden, affiliated Hospital for Research and Teaching of the Faculty of Medicine of the University of Zurich, Baden, Switzerland
| | - G Singer
- Department of Pathology, Kantonsspital Baden, Baden, Switzerland
| | - I A Burger
- Department of Nuclear Medicine, Kantonsspital Baden, affiliated Hospital for Research and Teaching of the Faculty of Medicine, University of Zurich, Baden, Switzerland; Department of Health Science and Technology ETH Zurich, Zurich, Switzerland; Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
2
|
Huang RR, Zuo C, Mona CE, Holzgreve A, Morrissey C, Nelson PS, Brady L, True L, Sisk A, Czernin J, Calais J, Ye H. FAP and PSMA Expression by Immunohistochemistry and PET Imaging in Castration-Resistant Prostate Cancer: A Translational Pilot Study. J Nucl Med 2024; 65:1952-1958. [PMID: 39477498 PMCID: PMC11619584 DOI: 10.2967/jnumed.124.268037] [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: 05/08/2024] [Accepted: 09/25/2024] [Indexed: 12/08/2024] Open
Abstract
Prostate-specific membrane antigen (PSMA) is a theranostic target for metastatic prostate cancer (PCa). However, castration-resistant PCa (CRPC) may lose PSMA expression after systemic therapy. Fibroblast activation protein (FAP), expressed by carcinoma-associated fibroblasts in various cancer types, including PCa, has the potential to be an alternative target. In this study, we evaluated FAP expression in CRPC to assess its potential, using PSMA as a comparison. Methods: FAP expression was assessed using immunohistochemistry in 116 CRPC tumors: 78 adenocarcinomas, 11 small cell carcinomas, and 27 anaplastic carcinomas. Correlation analysis between manual scoring and automated scoring was performed on 54 whole-slide sections of metastatic CRPC. Paired FAP and PSMA stains were assessed in tissue microarray cores of CRPC (n = 62), consisting of locally advanced CRPC (n = 9) and metastatic CRPC (n = 53). FAP and PSMA positivity was defined by an immunohistochemistry score of at least 10. To explore the correlation of PSMA and FAP inhibitor (FAPi) PET imaging and immunohistochemistry, a preliminary analysis of 4 patients included in a [68Ga]-FAPi-46 imaging trial (NCT04457232) was conducted. Results: Manual and automated scoring of FAP yielded results with strong correlations. Overall, FAP expression in CRPC was notably lower than PSMA expression (median immunoscores, 14 vs. 72; P < 0.001). Different histologic subtypes of CRPC demonstrated distinct levels of PSMA expression, whereas their FAP expression levels were comparable. Among the 19 PSMA-negative tumors, 11 (58%) exhibited FAP positivity. FAP expression levels in lymph node metastases were significantly lower than those in nonnodal metastases (P = 0.021). Liver metastases showed significant enrichment of tumors with strong FAP expression compared with nonliver lesions (P = 0.016). In the 4 clinical trial patients, the biopsied metastatic lesions showed lower uptake on FAPi PET than on PSMA PET (median SUVmax, 9.6 vs. 14.5), consistent with FAP expression that was lower than PSMA expression in the corresponding tumor biopsy samples (median immunoscores, 30 vs. 160). Conclusion: Because of the low FAP expression levels in CRPC, the utility of FAPi PET imaging may be limited. Although FAPi PET imaging may be further tested in PSMA-negative CRPC, such as small cell carcinoma, other molecular imaging modalities should be evaluated as alternative choices.
Collapse
Affiliation(s)
- Rong Rong Huang
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Chunlai Zuo
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California
- Department of Pathology, Rocky Mountain Regional VA Medical Center, Aurora, Colorado
| | - Christine E Mona
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Adrien Holzgreve
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, Washington
| | - Peter S Nelson
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Lauren Brady
- Divisions of Human Biology and Clinical Research, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Lawrence True
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington; and
| | - Anthony Sisk
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Johannes Czernin
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Jeremie Calais
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, California;
| | - Huihui Ye
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California;
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
3
|
Rupp NJ, Freiberger SN, Ferraro DA, Laudicella R, Heimer J, Muehlematter UJ, Poyet C, Moch H, Eberli D, Rüschoff JH, Burger IA. Immunohistochemical ERG positivity is associated with decreased PSMA expression and lower visibility in corresponding [ 68Ga]Ga-PSMA-11 PET scans of primary prostate cancer. Eur J Nucl Med Mol Imaging 2024; 52:305-313. [PMID: 39083067 PMCID: PMC11599624 DOI: 10.1007/s00259-024-06856-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/17/2024] [Indexed: 11/27/2024]
Abstract
PURPOSE TMPRSS2:ERG gene fusion negatively regulates PSMA expression in prostate adenocarcinoma (PCa) cell lines. Therefore, immunohistochemical (IHC) ERG expression, a surrogate for an underlying ERG rearrangement, and PSMA expression patterns in radical prostatectomy (RPE) specimens of primary PCa, including corresponding PSMA-PET scans were investigated. METHODS Two cohorts of RPE samples (total n=148): In cohort #1 (n=62 patients) with available RPE and preoperative [68Ga]Ga-PSMA-11 PET, WHO/ISUP grade groups, IHC-ERG (positive vs. negative) and IHC-PSMA expression (% PSMA-negative tumour area, PSMA%neg) were correlated with the corresponding SUVmax. In the second cohort #2 (n=86 patients) including RPE only, same histopathological parameters were evaluated. RESULTS Cohort #1: PCa with IHC-ERG expression (35.5%) showed significantly lower IHC-PSMA expression and lower SUVmax values on the corresponding PET scans. Eight of 9 PCa with negative PSMA-PET scans had IHC-ERG positivity, and confirmed TMPRSS2::ERG rearrangement. In IHC-PSMA positive PCa, IHC-ERG positivity was significantly associated with lower SUVmax values. In cohort #2, findings of higher IHC-PSMA%neg and IHC-ERG expression was confirmed with only 0-10% PSMA%neg tumour areas in IHC-ERG-negative PCa. CONCLUSION IHC-ERG expression is significantly associated with more heterogeneous and lower IHC-PSMA tissue expression in two independent RPE cohorts. There is a strong association of ERG positivity in RPE tissue with lower [68Ga]Ga-PSMA-11 uptake on corresponding PET scans. Results may serve as a base for future biomarker development to enable tumour-tailored, individualized imaging approaches.
Collapse
Affiliation(s)
- Niels J Rupp
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstrasse 12, 8091, Zürich, Switzerland.
- University of Zurich, Zurich, Switzerland.
| | - Sandra N Freiberger
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstrasse 12, 8091, Zürich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Daniela A Ferraro
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Radiology and Oncology, Faculdade de Medicina FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Riccardo Laudicella
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, Nuclear Medicine Unit, University of Messina, Messina, Italy
| | - Jakob Heimer
- Seminar for Statistics, Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Urs J Muehlematter
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Cédric Poyet
- Department of Urology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstrasse 12, 8091, Zürich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Daniel Eberli
- University of Zurich, Zurich, Switzerland
- Department of Urology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Jan H Rüschoff
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstrasse 12, 8091, Zürich, Switzerland
| | - Irene A Burger
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Department of Nuclear Medicine, Cantonal Hospital Baden, affiliated Hospital for Research and Teaching of the Faculty of Medicine of the University of Zurich, Baden, Switzerland
| |
Collapse
|
4
|
Ahmadi Bidakhvidi N, Gevaert T, De Schepper M, Baldewijns M, Havinga E, Deckers W, Laenen A, Devos G, Giesen A, Joniau S, Koole M, Everaerts W, Deroose CM, Goffin K. Comparison of PSMA immunohistochemistry scoring systems to parametric [ 18F]PSMA-1007 PET/MRI in primary prostate cancer. Eur J Nucl Med Mol Imaging 2024:10.1007/s00259-024-06903-7. [PMID: 39269656 DOI: 10.1007/s00259-024-06903-7] [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: 05/23/2024] [Accepted: 08/25/2024] [Indexed: 09/15/2024]
Abstract
PURPOSE Quantification of PSMA expression via PSMA PET is well-established, however quantification of PSMA via immunohistochemistry (IHC) is not standardized. Our aim was to determine the most optimal PSMA IHC scoring system to quantify PSMA expression with PSMA PET as reference standard. METHODS Primary intermediate- and high-risk prostate cancer patients received an [18F]PSMA-1007 PET/MRI followed by radical prostatectomy. SUVmax, SUVmean and Ki of the prostate tumor was determined. Prostate tumors were stained with anti-PSMA antibodies and scored by 2 readers via 10 IHC scoring systems: histochemical score (H-score), immunoreactivity scorepredominant intensity (IRSpredominant intensity), IRS classificationpredominant intensity, IRSmean intensity, IRS classificationmean intensity, Allred score, predominant expression pattern, Shannon diversity index (SDI), percentage negatively stained cells and total percentage positively stained cells. Spearman's rank correlation coefficients (ρ) were calculated between PET parameters and IHC scoring systems. Interreader agreement for the IHC scoring systems was measured by the intraclass correlation coefficient (ICC). RESULTS Fifty tumors in 46 patients were analysed. H-score had the best correlation with SUVmax (ρ 0.615 p < 0.0001) and SUVmean (ρ 0.570, p < 0.0001) and the second best correlation with Ki (ρ 0.411, p = 0.0030). SDI had the best correlation with Ki (ρ -0.440, p = 0.0014) and the second best correlation with SUVmax (ρ -0.516, p = 0.0001) and SUVmean (ρ -0.490, p = 0.0003). A moderate interreader agreement was observed for H-score (ICC 0.663, 95% CI 0.495-0.797) and SDI (ICC 0.546, 95% CI 0.354-0.725). CONCLUSION H-score had the best correlation with PSMA PET quantification and an acceptable interreader agreement. Therefore, we deem H-score the most optimal PSMA IHC scoring system.
Collapse
Affiliation(s)
- Niloefar Ahmadi Bidakhvidi
- Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Thomas Gevaert
- Urogenital, Abdominal and Plastic Surgery, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | | | | | | | - Wies Deckers
- Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Annouschka Laenen
- Interuniversity Institute for Biostatistics and Statistical Bioinformatics, Leuven, Belgium
| | - Gaëtan Devos
- Urogenital, Abdominal and Plastic Surgery, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Urology, University Hospitals Leuven, Leuven, Belgium
| | - Alexander Giesen
- Urogenital, Abdominal and Plastic Surgery, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Urology, University Hospitals Leuven, Leuven, Belgium
| | - Steven Joniau
- Urogenital, Abdominal and Plastic Surgery, Department of Development and Regeneration, KU Leuven, Leuven, Belgium
- Urology, University Hospitals Leuven, Leuven, Belgium
| | - Michel Koole
- Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Wouter Everaerts
- Urology, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Christophe M Deroose
- Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Karolien Goffin
- Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium.
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
5
|
Lancia A, Ingrosso G, Detti B, Festa E, Bonzano E, Linguanti F, Camilli F, Bertini N, La Mattina S, Orsatti C, Francolini G, Abenavoli EM, Livi L, Aristei C, de Jong D, Al Feghali KA, Siva S, Becherini C. Biology-guided radiotherapy in metastatic prostate cancer: time to push the envelope? Front Oncol 2024; 14:1455428. [PMID: 39314633 PMCID: PMC11417306 DOI: 10.3389/fonc.2024.1455428] [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: 06/26/2024] [Accepted: 08/19/2024] [Indexed: 09/25/2024] Open
Abstract
The therapeutic landscape of metastatic prostate cancer has undergone a profound revolution in recent years. In addition to the introduction of novel molecules in the clinics, the field has witnessed a tremendous development of functional imaging modalities adding new biological insights which can ultimately inform tailored treatment strategies, including local therapies. The evolution and rise of Stereotactic Body Radiotherapy (SBRT) have been particularly notable in patients with oligometastatic disease, where it has been demonstrated to be a safe and effective treatment strategy yielding favorable results in terms of disease control and improved oncological outcomes. The possibility of debulking all sites of disease, matched with the ambition of potentially extending this treatment paradigm to polymetastatic patients in the not-too-distant future, makes Biology-guided Radiotherapy (BgRT) an attractive paradigm which can be used in conjunction with systemic therapy in the management of patients with metastatic prostate cancer.
Collapse
Affiliation(s)
- Andrea Lancia
- Department of Radiation Oncology, San Matteo Hospital Foundation Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Pavia, Italy
| | | | - Beatrice Detti
- Radiotherapy Unit Prato, Usl Centro Toscana, Presidio Villa Fiorita, Prato, Italy
| | - Eleonora Festa
- Radiation Oncology Section, University of Perugia, Perugia, Italy
| | - Elisabetta Bonzano
- Department of Radiation Oncology, San Matteo Hospital Foundation Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Pavia, Italy
| | | | - Federico Camilli
- Radiation Oncology Section, University of Perugia, Perugia, Italy
| | - Niccolò Bertini
- Radiation Oncology Unit, Oncology Department, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - Salvatore La Mattina
- Department of Radiation Oncology, San Matteo Hospital Foundation Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Pavia, Italy
| | - Carolina Orsatti
- Radiation Oncology Unit, Oncology Department, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - Giulio Francolini
- Radiation Oncology Unit, Oncology Department, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | | | - Lorenzo Livi
- Radiation Oncology Unit, Oncology Department, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - Cynthia Aristei
- Radiation Oncology Section, University of Perugia, Perugia, Italy
| | - Dorine de Jong
- Medical Affairs, RefleXion Medical, Inc., Hayward, CA, United States
| | | | - Shankar Siva
- Department of Radiation Oncology, Sir Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Carlotta Becherini
- Radiation Oncology Unit, Oncology Department, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| |
Collapse
|
6
|
Sushentsev N, Hamm G, Flint L, Birtles D, Zakirov A, Richings J, Ling S, Tan JY, McLean MA, Ayyappan V, Horvat Menih I, Brodie C, Miller JL, Mills IG, Gnanapragasam VJ, Warren AY, Barry ST, Goodwin RJA, Barrett T, Gallagher FA. Metabolic imaging across scales reveals distinct prostate cancer phenotypes. Nat Commun 2024; 15:5980. [PMID: 39013948 PMCID: PMC11252279 DOI: 10.1038/s41467-024-50362-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 07/07/2024] [Indexed: 07/18/2024] Open
Abstract
Hyperpolarised magnetic resonance imaging (HP-13C-MRI) has shown promise as a clinical tool for detecting and characterising prostate cancer. Here we use a range of spatially resolved histological techniques to identify the biological mechanisms underpinning differential [1-13C]lactate labelling between benign and malignant prostate, as well as in tumours containing cribriform and non-cribriform Gleason pattern 4 disease. Here we show that elevated hyperpolarised [1-13C]lactate signal in prostate cancer compared to the benign prostate is primarily driven by increased tumour epithelial cell density and vascularity, rather than differences in epithelial lactate concentration between tumour and normal. We also demonstrate that some tumours of the cribriform subtype may lack [1-13C]lactate labelling, which is explained by lower epithelial lactate dehydrogenase expression, higher mitochondrial pyruvate carrier density, and increased lipid abundance compared to lactate-rich non-cribriform lesions. These findings highlight the potential of combining spatial metabolic imaging tools across scales to identify clinically significant metabolic phenotypes in prostate cancer.
Collapse
Affiliation(s)
- Nikita Sushentsev
- Department of Radiology, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| | - Gregory Hamm
- Integrated BioAnalysis, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Lucy Flint
- Integrated BioAnalysis, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Daniel Birtles
- Integrated BioAnalysis, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Aleksandr Zakirov
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Jack Richings
- Predictive AI & Data, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Stephanie Ling
- Integrated BioAnalysis, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Jennifer Y Tan
- Predictive AI & Data, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Mary A McLean
- Department of Radiology, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Vinay Ayyappan
- Department of Radiology, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Ines Horvat Menih
- Department of Radiology, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Cara Brodie
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Jodi L Miller
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Ian G Mills
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
- Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Vincent J Gnanapragasam
- Department of Urology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Division of Urology, Department of Surgery, University of Cambridge, Cambridge, UK
- Cambridge Urology Translational Research and Clinical Trials Office, Cambridge Biomedical Campus, Addenbrooke's Hospital, Cambridge, UK
| | - Anne Y Warren
- Department of Pathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Simon T Barry
- Bioscience, Early Oncology, AstraZeneca, Cambridge, UK
| | - Richard J A Goodwin
- Integrated BioAnalysis, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Tristan Barrett
- Department of Radiology, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Ferdia A Gallagher
- Department of Radiology, University of Cambridge and Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
7
|
Cattabriga A, Renzetti B, Galuppi F, Bartalena L, Gaudiano C, Brocchi S, Rossi A, Schiavina R, Bianchi L, Brunocilla E, Spinozzi L, Catanzaro C, Castellucci P, Farolfi A, Fanti S, Tunariu N, Mosconi C. Multiparametric Whole-Body MRI: A Game Changer in Metastatic Prostate Cancer. Cancers (Basel) 2024; 16:2531. [PMID: 39061171 PMCID: PMC11274871 DOI: 10.3390/cancers16142531] [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: 05/27/2024] [Revised: 06/24/2024] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
Prostate cancer ranks among the most prevalent tumours globally. While early detection reduces the likelihood of metastasis, managing advanced cases poses challenges in diagnosis and treatment. Current international guidelines support the concurrent use of 99Tc-Bone Scintigraphy and Contrast-Enhanced Chest and Abdomen CT for the staging of metastatic disease and response assessment. However, emerging evidence underscores the superiority of next-generation imaging techniques including PSMA-PET/CT and whole-body MRI (WB-MRI). This review explores the relevant scientific literature on the role of WB-MRI in metastatic prostate cancer. This multiparametric imaging technique, combining the high anatomical resolution of standard MRI sequences with functional sequences such as diffusion-weighted imaging (DWI) and bone marrow relative fat fraction (rFF%) has proved effective in comprehensive patient assessment, evaluating local disease, most of the nodal involvement, bone metastases and their complications, and detecting the increasing visceral metastases in prostate cancer. It does have the advantage of avoiding the injection of contrast medium/radionuclide administration, spares the patient the exposure to ionizing radiation, and lacks the confounder of FLARE described with nuclear medicine techniques. Up-to-date literature regarding the diagnostic capabilities of WB-MRI, though still limited compared to PSMA-PET/CT, strongly supports its widespread incorporation into standard clinical practice, alongside the latest nuclear medicine techniques.
Collapse
Affiliation(s)
- Arrigo Cattabriga
- Department of Radiology, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy; (B.R.); (F.G.); (L.B.); (C.G.); (S.B.); (C.M.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40136 Bologna, Italy; (R.S.); (L.B.); (E.B.); (L.S.); (C.C.); (S.F.)
| | - Benedetta Renzetti
- Department of Radiology, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy; (B.R.); (F.G.); (L.B.); (C.G.); (S.B.); (C.M.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40136 Bologna, Italy; (R.S.); (L.B.); (E.B.); (L.S.); (C.C.); (S.F.)
| | - Francesco Galuppi
- Department of Radiology, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy; (B.R.); (F.G.); (L.B.); (C.G.); (S.B.); (C.M.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40136 Bologna, Italy; (R.S.); (L.B.); (E.B.); (L.S.); (C.C.); (S.F.)
| | - Laura Bartalena
- Department of Radiology, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy; (B.R.); (F.G.); (L.B.); (C.G.); (S.B.); (C.M.)
| | - Caterina Gaudiano
- Department of Radiology, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy; (B.R.); (F.G.); (L.B.); (C.G.); (S.B.); (C.M.)
| | - Stefano Brocchi
- Department of Radiology, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy; (B.R.); (F.G.); (L.B.); (C.G.); (S.B.); (C.M.)
| | - Alice Rossi
- Radiology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Riccardo Schiavina
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40136 Bologna, Italy; (R.S.); (L.B.); (E.B.); (L.S.); (C.C.); (S.F.)
- Division of Urology, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy
| | - Lorenzo Bianchi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40136 Bologna, Italy; (R.S.); (L.B.); (E.B.); (L.S.); (C.C.); (S.F.)
- Division of Urology, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy
| | - Eugenio Brunocilla
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40136 Bologna, Italy; (R.S.); (L.B.); (E.B.); (L.S.); (C.C.); (S.F.)
- Division of Urology, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy
| | - Luca Spinozzi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40136 Bologna, Italy; (R.S.); (L.B.); (E.B.); (L.S.); (C.C.); (S.F.)
- Division of Urology, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy
| | - Calogero Catanzaro
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40136 Bologna, Italy; (R.S.); (L.B.); (E.B.); (L.S.); (C.C.); (S.F.)
- Division of Urology, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy
| | - Paolo Castellucci
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (P.C.); (A.F.)
| | - Andrea Farolfi
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (P.C.); (A.F.)
| | - Stefano Fanti
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40136 Bologna, Italy; (R.S.); (L.B.); (E.B.); (L.S.); (C.C.); (S.F.)
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (P.C.); (A.F.)
| | - Nina Tunariu
- Clinical Radiology, Royal Marsden Hospital & Institute of Cancer Research, London SW3 6JJ, UK;
| | - Cristina Mosconi
- Department of Radiology, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy; (B.R.); (F.G.); (L.B.); (C.G.); (S.B.); (C.M.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40136 Bologna, Italy; (R.S.); (L.B.); (E.B.); (L.S.); (C.C.); (S.F.)
| |
Collapse
|
8
|
Pepe P, Pepe L, Pennisi M. Negative biopsy histology in men with PI-RADS score 5: is it useful PSMA PET/CT evaluation? Arch Ital Urol Androl 2024; 96:12358. [PMID: 38934527 DOI: 10.4081/aiua.2024.12358] [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: 02/05/2024] [Accepted: 02/18/2024] [Indexed: 06/28/2024] Open
Abstract
INTRODUCTION To evaluate the accuracy of PSMA PET/CT in men with mpMRI PI-RADS score 5 negative biopsy histology. MATERIALS AND METHODS From January 2011 to January 2023, 180 men with PI-RADS score 5 underwent systematic plus mpMRI/TRUS biopsy; 25/180 (13.9%) patients had absence of cancer and six months from biopsy were submitted to: digital rectal examination, PSA and PSA density exams, mpMRI and 68GaPSMA PET/CT evaluation (standardized uptake value "SUVmax" was reported). RESULTS In 24/25 (96%) patients PSA and PSA density significantly decreased, moreover, the PI-RADS score was downgraded resulting < 3; in addition, median SUVmax was 7.5. Only 1/25 (4%) man had an increased PSA value (from 10.5 to 31 ng/ml) with a confirmed PI-RADS score 5, SUVmax of 32 and repeated prostate biopsy demonstrating a Gleason score 9/ISUP Grade Group 5 PCa. CONCLUSIONS The strict follow up of men with PI-RADS score 5 and negative histology reduce the risk of missing csPCa especially if PSMA PET/CT evaluation is in agreement with downgrading of mpMRI (PI-RADS score < 3).
Collapse
Affiliation(s)
| | - Ludovica Pepe
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", University of Messina.
| | | |
Collapse
|
9
|
Xiao L, Fang Z, Tang Y, Sun Y, Zhu Z, Li J, Zhou M, Yang N, Zheng K, Hu S. Evaluation of gastrin-releasing peptide receptor, prostate-specific membrane antigen, and neurotensin receptor 1 as potential biomarkers for accurate prostate cancer stratified diagnosis. EJNMMI Res 2024; 14:55. [PMID: 38880858 PMCID: PMC11180645 DOI: 10.1186/s13550-024-01116-3] [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: 02/13/2024] [Accepted: 06/02/2024] [Indexed: 06/18/2024] Open
Abstract
BACKGROUND Studies on single-target PET imaging of gastrin-releasing peptide receptor (GRPR), prostate-specific membrane antigen (PSMA), or neurotensin receptor 1(NTR1) have been reported. However, the performance of these three targets in the progression of PCa remains unclear. Our study aims to compare the expression of GRPR, PSMA, and NTR1 in patients with prostatic intraepithelial neoplasia (PIN), prostate cancer (PCa), and lymph node metastasis. We synthesized molecular probes targeting the markers to achieve a non-invasive precise detection of PCa patients with PET/CT imaging. METHODS In this study, the expression of GRPR, PSMA, and NTR1 was evaluated by immunohistochemistry in 34 PIN, 171 PCa, and 22 lymph node metastasis tissues of patients. The correlation between their expression and the clinicopathological parameters of PCa patients was assessed. Sixteen PCa patients with different Gleason scores (GS) underwent dual-tracer (68Ga-NOTA-RM26 and 68Ga-NOTA-PSMA617) PET/CT. RESULTS In the PIN stage, the expression of GRPR was significantly higher than that of PSMA and NTR1 (P < 0.001), while NTR1 expression was significantly higher than PSMA and GRPR expression in primary PCa (P = 0.001). High PSMA expression in PCa patients was associated with shorter progression-free survival (P = 0.037) and overall survival (P = 0.035). PCa patients with high GS had higher tumor uptake of 68Ga-NOTA-PSMA617 than those with low GS (P = 0.001), while PCa patients with low GS had higher tumor uptake of 68Ga-NOTA-RM26 than those with high GS (P = 0.001). CONCLUSIONS This study presents three novel biomarkers (PSMA, GRPR, and NTR1) as imaging agents for PET/CT, and may offer a promising approach for non-invasive precise detection and Gleason grade prediction of PCa patients.
Collapse
Affiliation(s)
- Ling Xiao
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha City, 410008, Hunan Province, P.R. China
| | - Zhihui Fang
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha City, 410008, Hunan Province, P.R. China
- Department of Nuclear Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yongxiang Tang
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha City, 410008, Hunan Province, P.R. China
| | - Yanyan Sun
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450000, China
| | - Zehua Zhu
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha City, 410008, Hunan Province, P.R. China
| | - Jian Li
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha City, 410008, Hunan Province, P.R. China
| | - Ming Zhou
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha City, 410008, Hunan Province, P.R. China
| | - Nengan Yang
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha City, 410008, Hunan Province, P.R. China
| | - Kai Zheng
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha City, 410008, Hunan Province, P.R. China
| | - Shuo Hu
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha City, 410008, Hunan Province, P.R. China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
10
|
Laudicella R, Comelli A, Schwyzer M, Stefano A, Konukoglu E, Messerli M, Baldari S, Eberli D, Burger IA. PSMA-positive prostatic volume prediction with deep learning based on T2-weighted MRI. LA RADIOLOGIA MEDICA 2024; 129:901-911. [PMID: 38700556 PMCID: PMC11168990 DOI: 10.1007/s11547-024-01820-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 04/16/2024] [Indexed: 05/28/2024]
Abstract
PURPOSE High PSMA expression might be correlated with structural characteristics such as growth patterns on histopathology, not recognized by the human eye on MRI images. Deep structural image analysis might be able to detect such differences and therefore predict if a lesion would be PSMA positive. Therefore, we aimed to train a neural network based on PSMA PET/MRI scans to predict increased prostatic PSMA uptake based on the axial T2-weighted sequence alone. MATERIAL AND METHODS All patients undergoing simultaneous PSMA PET/MRI for PCa staging or biopsy guidance between April 2016 and December 2020 at our institution were selected. To increase the specificity of our model, the prostatic beds on PSMA PET scans were dichotomized in positive and negative regions using an SUV threshold greater than 4 to generate a PSMA PET map. Then, a C-ENet was trained on the T2 images of the training cohort to generate a predictive prostatic PSMA PET map. RESULTS One hundred and fifty-four PSMA PET/MRI scans were available (133 [68Ga]Ga-PSMA-11 and 21 [18F]PSMA-1007). Significant cancer was present in 127 of them. The whole dataset was divided into a training cohort (n = 124) and a test cohort (n = 30). The C-ENet was able to predict the PSMA PET map with a dice similarity coefficient of 69.5 ± 15.6%. CONCLUSION Increased prostatic PSMA uptake on PET might be estimated based on T2 MRI alone. Further investigation with larger cohorts and external validation is needed to assess whether PSMA uptake can be predicted accurately enough to help in the interpretation of mpMRI.
Collapse
Affiliation(s)
- Riccardo Laudicella
- Department of Nuclear Medicine, University Hospital Zürich, University of Zurich, Zurich, Switzerland.
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, Messina, Italy.
- Ri.MED Foundation, Palermo, Italy.
| | | | - Moritz Schwyzer
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, Zurich, Switzerland
| | - Alessandro Stefano
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), Cefalù, Italy
| | | | - Michael Messerli
- Department of Nuclear Medicine, University Hospital Zürich, University of Zurich, Zurich, Switzerland
| | - Sergio Baldari
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, Messina, Italy
| | - Daniel Eberli
- Department of Urology, University Hospital of Zürich, Zurich, Switzerland
| | - Irene A Burger
- Department of Nuclear Medicine, University Hospital Zürich, University of Zurich, Zurich, Switzerland
- Department of Nuclear Medicine, Cantonal Hospital Baden, Baden, Switzerland
| |
Collapse
|
11
|
Droghetti M, Bianchi L, Presutti M, Vetrone L, Farolfi A, Mei R, Giunchi F, Degiovanni A, Mottaran A, Piazza P, Cangemi D, Castellucci P, D’Errico A, Schiavina R, Brunocilla E, Fanti S. Immunohistochemistry analysis of PSMA expression at prostatic biopsy in high-risk prostate cancer: potential implications for PSMA-PET patient selection. Front Oncol 2024; 14:1324631. [PMID: 38807770 PMCID: PMC11130476 DOI: 10.3389/fonc.2024.1324631] [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: 10/19/2023] [Accepted: 04/18/2024] [Indexed: 05/30/2024] Open
Abstract
Introduction Prostate-specific membrane antigen (PSMA) is a transmembrane protein expressed by normal prostatic tissue. Therefore, molecular imaging targeting PSMA (PSMA-PET) has gained particular interest and diffusion for PCa staging and restaging. Several factors may affect PSMA-PET results, and many tools have been proposed to improve patient selection. Furthermore, PSMA expression is not homogeneous among different tissues and within the prostate itself. The aims of this study were to evaluate immunohistochemistry (IHC) features of prostate biopsy samples and to assess their correlation with whole-mount specimens and PSMA-PET parameters. Methods We included consecutive high-risk PCa patients who underwent PSMA-PET for staging proposal at our institution from January 2022 to December 2022. The PET parameters selected were SUVmax, total volume (TV), and total lesion activity (TL). Each patient underwent multiparametric MRI (mpMRI) and fusion-targeted prostate biopsy prior to surgery. IHC analyses were performed on the index lesion cores. IHC visual score (VS) (1, 2, 3) and visual pattern (VP) (membranous, cytoplasmic, and combined) and the percentage of PSMA-negative tumor areas (PSMA%neg) within biopsy cores were evaluated. Results Forty-three patients who underwent robotic radical prostatectomy after PSMA-PET were available for analyses. Concordance between VS and VP at biopsy and final pathology showed a Cohen's kappa coefficient of 0.39 and 0.38, respectively. Patients with PSMA%neg <20% had a higher concordance in VS and VP (Cohen's kappa 0.49 and 0.4, respectively). No difference emerged in terms of median PSMA-TV (p = 0.3) and PSMA-TL (p = 0.9) according to VS at biopsy, while median SUVmax was higher in patients with VS 3 (p = 0.04). Higher SUVmax was associated with membranous and combined VP expression (p = 0.008). No difference emerged between patients with PSMA%neg <20% or PSMA%neg >20% on biopsy cores in terms of SUVmax, PSMA-TL, and PSMA-TV (p = 0.5, p = 0.5, and p = 0.9 respectively). Conclusions We found a correlation between IHC VS and VP on targeted biopsy cores and SUVmax at PSMA-PET. However, the correlation between the IHC parameters of biopsy cores and final pathology was not as high as expected. Nevertheless, the presence of PSMA%neg <20% seems to have a better concordance in terms of visual score.
Collapse
Affiliation(s)
- Matteo Droghetti
- Division of Urology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Lorenzo Bianchi
- Division of Urology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Università degli Studi di Bologna, Bologna, Italy
| | - Massimiliano Presutti
- Division of Urology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Luigia Vetrone
- Nuclear Medicine, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Andrea Farolfi
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Riccardo Mei
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Francesca Giunchi
- Pathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Alessio Degiovanni
- Pathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Angelo Mottaran
- Division of Urology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Pietro Piazza
- Division of Urology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Danilo Cangemi
- Division of Urology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Paolo Castellucci
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Antonietta D’Errico
- Pathology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Riccardo Schiavina
- Division of Urology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Università degli Studi di Bologna, Bologna, Italy
| | - Eugenio Brunocilla
- Division of Urology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Università degli Studi di Bologna, Bologna, Italy
| | - Stefano Fanti
- Università degli Studi di Bologna, Bologna, Italy
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
12
|
Berrens AC, Sorbi MA, Donswijk ML, de Barros HA, Azargoshasb S, van Oosterom MN, Rietbergen DDD, Bekers EM, van der Poel HG, van Leeuwen FWB, van Leeuwen PJ. Strong Correlation Between SUV max on PSMA PET/CT and Numeric Drop-In γ-Probe Signal for Intraoperative Identification of Prostate Cancer Lesions. J Nucl Med 2024; 65:548-554. [PMID: 38485277 DOI: 10.2967/jnumed.123.267075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/11/2024] [Indexed: 04/04/2024] Open
Abstract
Prostate-specific membrane antigen (PSMA) PET is used to select patients with recurrent prostate cancer for metastasis-directed therapy. A surgical approach can be achieved through radioguided surgery (RGS), using a Drop-In γ-probe that traces lesions that accumulate the radioactive signal. With the aim of guiding patient selection for salvage surgery, we studied the correlation between the SUVmax of lesions on preoperative PSMA PET/CT and their intraoperative counts/s measured using the Drop-In γ-probe. Methods: A secondary analysis based on the prospective, single-arm, and single-center feasibility study was conducted (NCT03857113). Patients (n = 29) with biochemical recurrence after previous curative-intent therapy and a maximum of 3 suggestive lesions within the pelvis on preoperative PSMA PET/CT were included. Patients treated with androgen deprivation therapy within 6 mo before surgery were excluded. All patients received an intravenous injection of 99mTc-PSMA-I&S 1 d before surgery. Radioguidance was achieved using a Drop-In γ-probe. Correlation was determined using the Spearman rank correlation coefficient (ρs). Subgroup analysis was based on the median SUVmax Results: In total, 33 lesions were visible on the PSMA PET/CT images, with a median overall SUVmax of 6.2 (interquartile range [IQR], 4.2-9.7). RGS facilitated removal of 31 lesions. The median Drop-In counts/s were 134 (IQR, 81-220) in vivo and 109 (IQR, 72-219) ex vivo. The intensity of the values correlated with SUVmax (ρs = 0.728 and 0.763, respectively; P < 0.001). Subgroup analysis based on median SUVmax in the group with an SUVmax of less than 6 showed no statistically significant correlation with the numeric signal in vivo (ρs = 0.382; P = 0.221) or the signal-to-background-ratio (ρs = 0.245; P = 0.442), whereas the group with an SUVmax of 6 or more showed respective statistically significant positive correlations (ρs = 0.774 [P < 0.001] and ρs = 0.647 [P = 0.007]). Conclusion: Our findings indicate that there is a direct relation between SUVmax on PSMA PET/CT and the readout recorded by the surgical Drop-In probe, thereby indicating that SUVmax can be used to select patients for PSMA RGS. For more definitive subgroup definitions for treatment recommendations, further studies are necessary to validate the present findings.
Collapse
Affiliation(s)
- Anne-Claire Berrens
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands;
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Malou A Sorbi
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Maarten L Donswijk
- Department of Nuclear Medicine, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Hilda A de Barros
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Samaneh Azargoshasb
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Matthias N van Oosterom
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Daphne D D Rietbergen
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Centre, Leiden, The Netherlands
- Department of Nuclear Medicine, Leiden University Medical Centre, Leiden, The Netherlands
| | - Elise M Bekers
- Department of Pathology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands; and
| | - Henk G van der Poel
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Department of Urology, Amsterdam University Medical Centre, Location VUmc, Amsterdam, The Netherlands
| | - Fijs W B van Leeuwen
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Pim J van Leeuwen
- Department of Urology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| |
Collapse
|
13
|
Jiang Z, Guo J, Hu L, Yang S, Meng B, Tang Q. Diagnostic performance of 18F‑DCFPyL PET vs. 68Ga‑PSMA PET/CT in patients with suspected prostate cancer: A systemic review and meta‑analysis. Oncol Lett 2024; 27:188. [PMID: 38486944 PMCID: PMC10938285 DOI: 10.3892/ol.2024.14321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 02/13/2024] [Indexed: 03/17/2024] Open
Abstract
In this systematic review and meta-analysis, the diagnostic performance of 68Ga-prostate-specific membrane antigen (PSMA) positron emission tomography (PET)/CT was compared with that of 18F-DCFPyL PET for patients with suspected prostate cancer (PCa). Up to September 2023, the PubMed, Embase and Web of Science databases were thoroughly searched for relevant papers. Studies examining the diagnostic performance of 18F-DCFPyL PET and 68Ga-PSMA PET/CT in patients with suspected PCa were included in the present review. The Quality Assessment of Diagnostic Performance Studies-2 tool was used to rate the diagnostic performance of each study. The diagnostic performance of 18F-DCFPyL PET and 68Ga-PSMA PET/CT for primary PCa was examined by 13 studies included, comprising 1,178 patients. The pooled sensitivity and specificity of 18F-DCFPyL PET were 0.92 (95% CI, 0.85-0.96) and 0.59 (95% CI, 0.08-0.96), respectively. For 68Ga-PSMA PET/CT, the pooled sensitivity and specificity were 0.96 (95% CI, 0.88-0.99) and 0.71 (95% CI, 0.57-0.82), respectively. 18F-DCFPyL PET and 68Ga-PSMA PET/CT both had an area under the receiver operating characteristic curve of 0.92 (95% CI, 0.89-0.94). In addition, the Fagan nomogram revealed that the post-test probabilities for 18F-DCFPyL PET and 68Ga-PSMA PET/CT could rise to 69 and 77% when the pre-test probability was set at 50%. In conclusion, a comparable diagnostic performance for patients with suspected PCa was determined for 18F-DCFPyL PET and 68Ga-PSMA PET/CT. However, it is crucial to keep in mind that the findings of the present meta-analysis come from investigations with modest sample sizes. Therefore, more extensive research is required to obtain more solid data.
Collapse
Affiliation(s)
- Zhibing Jiang
- Department of Clinical Medicine, Medical College, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Jinjing Guo
- Department of Clinical Medicine, Medical College, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Liang Hu
- Department of Clinical Medicine, Medical College, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Siyu Yang
- Department of Clinical Medicine, Medical College, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Bin Meng
- Department of Clinical Medicine, Medical College, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Qun Tang
- Department of Clinical Medicine, Medical College, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| |
Collapse
|
14
|
Jafari E, Zarei A, Dadgar H, Keshavarz A, Manafi-Farid R, Rostami H, Assadi M. A convolutional neural network-based system for fully automatic segmentation of whole-body [ 68Ga]Ga-PSMA PET images in prostate cancer. Eur J Nucl Med Mol Imaging 2024; 51:1476-1487. [PMID: 38095671 DOI: 10.1007/s00259-023-06555-z] [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: 09/23/2023] [Accepted: 11/30/2023] [Indexed: 03/22/2024]
Abstract
PURPOSE The aim of this study was development and evaluation of a fully automated tool for the detection and segmentation of mPCa lesions in whole-body [68Ga]Ga-PSMA-11 PET scans by using a nnU-Net framework. METHODS In this multicenter study, a cohort of 412 patients from three different center with all indication of PCa who underwent [68Ga]Ga-PSMA-11 PET/CT were enrolled. Two hundred cases of center 1 dataset were used for training the model. A fully 3D convolutional neural network (CNN) is proposed which is based on the self-configuring nnU-Net framework. A subset of center 1 dataset and cases of center 2 and center 3 were used for testing of model. The performance of the segmentation pipeline that was developed was evaluated by comparing the fully automatic segmentation mask with the manual segmentation of the corresponding internal and external test sets in three levels including patient-level scan classification, lesion-level detection, and voxel-level segmentation. In addition, for comparison of PET-derived quantitative biomarkers between automated and manual segmentation, whole-body PSMA tumor volume (PSMA-TV) and total lesions PSMA uptake (TL-PSMA) were calculated. RESULTS In terms of patient-level classification, the model achieved an accuracy of 83%, sensitivity of 92%, PPV of 77%, and NPV of 91% for the internal testing set. For lesion-level detection, the model achieved an accuracy of 87-94%, sensitivity of 88-95%, PPV of 98-100%, and F1-score of 93-97% for all testing sets. For voxel-level segmentation, the automated method achieved average values of 65-70% for DSC, 72-79% for PPV, 53-58% for IoU, and 62-73% for sensitivity in all testing sets. In the evaluation of volumetric parameters, there was a strong correlation between the manual and automated measurements of PSMA-TV and TL-PSMA for all centers. CONCLUSIONS The deep learning networks presented here offer promising solutions for automatically segmenting malignant lesions in prostate cancer patients using [68Ga]Ga-PSMA PET. These networks achieve a high level of accuracy in whole-body segmentation, as measured by the DSC and PPV at the voxel level. The resulting segmentations can be used for extraction of PET-derived quantitative biomarkers and utilized for treatment response assessment and radiomic studies.
Collapse
Affiliation(s)
- Esmail Jafari
- The Persian Gulf Nuclear Medicine Research Center, Department of Nuclear Medicine, Molecular Imaging, and Theranostics, Bushehr Medical University Hospital, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Amin Zarei
- IoT and Signal Processing Research Group, ICT Research Institute, Faculty of Intelligent Systems Engineering and Data Science, Persian Gulf University, Bushehr, Iran
| | - Habibollah Dadgar
- Cancer Research Center, RAZAVI Hospital, Imam Reza International University, Mashhad, Iran
| | - Ahmad Keshavarz
- IoT and Signal Processing Research Group, ICT Research Institute, Faculty of Intelligent Systems Engineering and Data Science, Persian Gulf University, Bushehr, Iran
| | - Reyhaneh Manafi-Farid
- Research Center for Nuclear Medicine, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Habib Rostami
- Computer Engineering Department, Faculty of Intelligent Systems Engineering and Data Science, Persian Gulf University, Bushehr, Iran
| | - Majid Assadi
- The Persian Gulf Nuclear Medicine Research Center, Department of Nuclear Medicine, Molecular Imaging, and Theranostics, Bushehr Medical University Hospital, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran.
| |
Collapse
|
15
|
Bagheri H, Mahdavi SR, Geramifar P, Neshasteh-Riz A, Sajadi Rad M, Dadgar H, Arabi H, Zaidi H. An Update on the Role of mpMRI and 68Ga-PSMA PET Imaging in Primary and Recurrent Prostate Cancer. Clin Genitourin Cancer 2024; 22:102076. [PMID: 38593599 DOI: 10.1016/j.clgc.2024.102076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 02/28/2024] [Accepted: 03/09/2024] [Indexed: 04/11/2024]
Abstract
The objective of this work was to review comparisons of the efficacy of 68Ga-PSMA-11 (prostate-specific membrane antigen) PET/CT and multiparametric magnetic resonance imaging (mpMRI) in the detection of prostate cancer among patients undergoing initial staging prior to radical prostatectomy or experiencing recurrent prostate cancer, based on histopathological data. A comprehensive search was conducted in PubMed and Web of Science, and relevant articles were analyzed with various parameters, including year of publication, study design, patient count, age, PSA (prostate-specific antigen) value, Gleason score, standardized uptake value (SUVmax), detection rate, treatment history, sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV), and PI-RADS (prostate imaging reporting and data system) scores. Only studies directly comparing PSMA-PET and mpMRI were considered, while those examining combined accuracy or focusing on either modality alone were excluded. In total, 24 studies comprising 1717 patients were analyzed, with the most common indication for screening being staging, followed by relapse. The findings indicated that 68Ga-PSMA-PET/CT effectively diagnosed prostate cancer in patients with suspected or confirmed disease, and both methods exhibited comparable efficacy in identifying lesion-specific information. However, notable heterogeneity was observed, highlighting the necessity for standardization of imaging and histopathology systems to mitigate inter-study variability. Future research should prioritize evaluating the combined diagnostic performance of both modalities to enhance sensitivity and reduce unnecessary biopsies. Overall, the utilization of PSMA-PET and mpMRI in combination holds substantial potential for significantly advancing the diagnosis and management of prostate cancer.
Collapse
Affiliation(s)
- Hamed Bagheri
- Radiation Biology Research Center, Iran University of Medical Science (IUMS), Tehran, Iran
| | - Seyed Rabi Mahdavi
- Radiation Biology Research Center and Department of Medical Physics, Iran University of Medical Sciences, Tehran, Iran.
| | - Parham Geramifar
- Department Nuclear Medicine, School of Medicine Shariati Hospital, Tehran, Iran
| | - Ali Neshasteh-Riz
- Radiation Biology Research Center, Iran University of Medical Science (IUMS), Tehran, Iran
| | - Masoumeh Sajadi Rad
- Radiation Biology Research Center, Iran University of Medical Science (IUMS), Tehran, Iran
| | - Habibollah Dadgar
- Imam Reza research Center, Nuclear Medicine and Molecular imaging department, RAZAVI Hospital, Mashhad, Iran
| | - Hossein Arabi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva, Switzerland
| | - Habib Zaidi
- Division of Nuclear Medicine and Molecular Imaging, Geneva University Hospital, Geneva, Switzerland; Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University 6Medical Center Groningen, Groningen, Netherlands; Department of Nuclear Medicine, University of Southern Denmark, Odense, Denmark; University Research and Innovation Center, Óbuda University, Budapest, Hungary.
| |
Collapse
|
16
|
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
|
17
|
Ren X, Nur Salihin Yusoff M, Hartini Mohd Taib N, Zhang L, Wang K. 68Ga-prostate specific membrane antigen-11 PET/CT versus multiparametric MRI in the detection of primary prostate cancer: A systematic review and head-to-head comparative meta-analysis. Eur J Radiol 2024; 170:111274. [PMID: 38147764 DOI: 10.1016/j.ejrad.2023.111274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 12/28/2023]
Abstract
PURPOSE The goal of this study was to evaluate the effectiveness of two diagnostic methods, 68Ga-PSMA-11 PET/CT and mpMRI, in detecting primary prostate cancer without limitations on the Gleason score. METHODS We conducted a comprehensive literature review, searching databases such as PubMed, Embase, and Web of Science until June 2023. Our objective was to identify studies that compared the efficacy of 68Ga-PSMA-11 PET/CT and mpMRI in detecting primary prostate cancer. To determine heterogeneity, the I2 statistic was used. Meta-regression analysis and leave-one-out sensitivity analysis were conducted to identify potential sources of heterogeneity. RESULTS Initially, 1286 publications were found, but after careful evaluation, only 16 studies involving 1227 patients were analyzed thoroughly. The results showed that the 68Ga-PSMA-11 PET/CT method had a pooled sensitivity and specificity of 0.87 (95 % CI: 0.80-0.92) and 0.80 (95 % CI: 0.69-0.89), respectively, for diagnosing prostatic cancer. Similarly, the values for mpMRI were determined as 0.84 (95 % CI: 0.75-0.92) and 0.74 (95 % CI: 0.61-0.86), respectively. There were no significant differences in diagnostic effectiveness observed when comparing two primary prostate cancer methodologies (pooled sensitivity P = 0.62, pooled specificity P = 0.50). Despite this, the funnel plots showed symmetry and the Egger test results (P values > 0.05) suggested there was no publication bias. CONCLUSIONS After an extensive meta-analysis, it was found that both 68Ga-PSMA-11 PET/CT and mpMRI demonstrate similar diagnostic effectiveness in detecting primary prostate cancer. Future larger prospective studies are warranted to investigate this issue further.
Collapse
Affiliation(s)
- Xiaolu Ren
- Department of Radiology, General Hospital of Ningxia Medical University, Yinchuan 750004, China; School of Health Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | | | - Nur Hartini Mohd Taib
- Department of Radiology, School of Medical Sciences, Universiti Sains Malaysia, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Li Zhang
- Department of Urology, People's Hospital of Wuzhong, Wuzhong 751100, China
| | - Kehua Wang
- Department of Vascular Surgery, General Hospital of Ningxia Medical University, Yinchuan 750004, China.
| |
Collapse
|
18
|
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: 5] [Impact Index Per Article: 5.0] [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
|
19
|
Pepe P, Pennisi M. Targeted Biopsy in Men High Risk for Prostate Cancer: 68Ga-PSMA PET/CT Versus mpMRI. Clin Genitourin Cancer 2023; 21:639-642. [PMID: 37394379 DOI: 10.1016/j.clgc.2023.06.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 06/06/2023] [Accepted: 06/12/2023] [Indexed: 07/04/2023]
Abstract
INTRODUCTION/BACKGROUND To evaluate the accuracy of 68Ga-PSMA PET/CT versus mpMRI targeted biopsy (TPBx) in the diagnosis of clinically significant prostate cancer (csPCa) in men high risk for PCa. MATERIALS AND METHODS From January 2021 to March 2023, 125 men with clinical parameters high risk for PCa were evaluated by mpMRI and 68Ga-PSMA PET/CT; median PSA was 32.5 ng/mL (range: 12-160 ng/mL) and 60/125 (48%) had abnormal digital rectal examination. The mpMRI lesions with PI-RADS scores ≥ 3 and/or 68Ga-PSMA areas characterized by a standardized uptake value (SUVmax) values ≥ 8 were submitted to TPBx (4 cores); in addition, all the patients underwent systematic transperineal prostate biopsy (18 cores) under sedation and antibiotic prophylaxis. RESULTS In 80 of 125 men (64%) a csPCa was found: 10 (12.5%) had a ISUP Grade Group 3 (GG), 45 (56.2%) a ISUP GG4 and 25 (31.2%) ISUP GG5. The median intraprostatic 68Ga-PSMA SUVmax was 42.3 (range:10.5-164) and 72 of 80 (90%) had a PI-RADS score ≥ 3. 68GaPSMA PET/CT showed the presence of metastases in 20 of 80 (25%) men: the median SUVmax in bone (15 cases) and nodes (40 cases) metastases was 55 and 47, respectively. Accuracy of 68Ga PSMA PET/CT (SUVmax cut-off ≥ 8) versus mpMRI PI-RADS score ≥ 3 in the diagnosis of csPCa was 92 versus 86.2%. CONCLUSION 68GaPSMA PET/CT demonstrated a good diagnostic accuracy as a single procedure for the diagnosis and staging of high-risk PCa.
Collapse
Affiliation(s)
- Pietro Pepe
- Urology Unit, Imaging Department, Cannizzaro Hospital, Catania, Italy.
| | - Michele Pennisi
- Urology Unit, Imaging Department, Cannizzaro Hospital, Catania, Italy
| |
Collapse
|
20
|
Koehler D, Berliner C, Shenas F, Karimzadeh A, Apostolova I, Klutmann S, Adam G, Sauer M. PSMA hybrid imaging in prostate cancer - current applications and perspectives. ROFO-FORTSCHR RONTG 2023; 195:1001-1008. [PMID: 37348528 DOI: 10.1055/a-2088-9543] [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: 06/24/2023]
Abstract
BACKGROUND Prostate cancer (PCa) is the most common malignancy in men and the second most common tumor-associated cause of death in the male population in Germany. Prostate-specific membrane antigen (PSMA)-targeted hybrid imaging using positron emission tomography (PET) in combination with CT or MRI represents a comparably new method that gained increasing importance in the diagnostic process of PCa in recent years. METHOD Current applications of PSMA hybrid imaging were summarized according to the German and European guidelines on PCa. New developments were elaborated based on a literature review of PubMed conducted in 10/22. RESULTS PSMA-PET/CT demonstrated higher detection rates for metastases in high-risk PCa and recurrent PCa after primary therapy than established imaging methods (CT, MRI, and bone scan). Despite promising results from prospective trials in both scenarios and substantial influence on clinical decision making, data regarding the influence of PSMA-PET on PCa-specific and overall survival are still lacking. Hence, PSMA PET/CT is recommended with a "weak" strength rating in most situations. However, its importance in new treatment options like metastasis-directed therapy or PSMA-radioligand therapy expands the scope of PSMA-PET in the clinical routine. CONCLUSION PSMA-targeting hybrid imaging represents the most sensitive diagnostic test in several stages of PCa and allows the development of new treatment strategies. Prospective studies are needed to evaluate the influence of PSMA-PET on patient survival. KEY POINTS · PSMA-PET/CT is superior to conventional imaging in the primary staging of high-risk prostate cancer.. · PSMA hybrid imaging can detect metastases in patients with biochemical recurrence at low PSA values.. · Clinical decision making is frequently influenced by results of PSMA-PET/CT.. CITATION FORMAT · Koehler D, Berliner C, Shenas F et al. PSMA hybrid imaging in prostate cancer - current applications and perspectives. Fortschr Röntgenstr 2023; 195: 1001 - 1008.
Collapse
Affiliation(s)
- Daniel Koehler
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Farzad Shenas
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Amir Karimzadeh
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ivayla Apostolova
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Susanne Klutmann
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gerhard Adam
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Sauer
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
21
|
Heetman JG, Hermsen R, Exterkate L, Küsters-Vandevelde HVN, Brouwer LJM, Somford DM, van den Bergh RCN, van Basten JPA. Immunohistochemical and histopathological validation of 18 F-PSMA-1007 PET/CT for intraprostatic cancerous lesions. Prostate 2023; 83:1332-1341. [PMID: 37455399 DOI: 10.1002/pros.24595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/02/2023] [Accepted: 06/21/2023] [Indexed: 07/18/2023]
Abstract
INTRODUCTION Prostate-specific membrane antigen (PSMA) is overexpressed in prostate cancer (PCa). In this study, we aim to immunohistochemically and histopathological validate the fluorine-18 (18 F)-PSMA-1007 positron emission tomography/computed tomography (PET/CT) for intraprostatic PCa lesions. METHODS Between February 2019 and October 2020, patients with biopsy-proven, treatment-naïve intermediate-to-high-risk PCa undergoing an 18 F-PSMA-1007 PET/CT before robot-assisted radical prostatectomy (RARP) were prospectively enrolled. For all PCa lesions found on whole-mount histopathology, location, size, International Society of Urological Pathology (ISUP) grade group (GG), and immune reactive score (IRS) were assessed after PSMA staining. ISUP GG ≥ 3 PCa was defined as clinically significant (cs) PCa. All lesions were matched on PSMA PET/CT and the maximum standardized uptake value (SUVmax) was measured. RESULTS A total of 125 lesions were analyzed in the 80 RARP specimens, of which 49 (40%) were csPCa and 76 (60%) non-csPCa. Linear multivariable regressions showed that an increase in SUVmax significantly correlated with a higher ISUP GG (p values between 0.021 and 0.001) and a higher IRS (p = 0.017). Logistic multivariable regression showed that csPCa significantly correlated with a higher SUVmax (odds ratio, OR: 1.17 [95% confidence interval, CI: 1.04-1.21, p = 0.005]), an increase in tumor length (OR: 1.05 [95% CI 1.01-1.10, p = 0.020]) and a higher IRS (OR; 1.24 [95% CI 1.07-1.47, p = 0.006]). A SUVmax threshold of 4 would have resulted in one (2%) missed lesion with csPCa. CONCLUSION This prospective study revealed that 18 F-PSMA-1007 PET/CT SUVmax is correlated with the ISUP GG and IRS, and thereby could be a tool to characterize intraprostatic PCa lesions.
Collapse
Affiliation(s)
- Joris G Heetman
- Department of Urology, Sint Antonius Hospital, Utrecht-Nieuwegein, The Netherlands
| | - Rick Hermsen
- Department of Nuclear Medicine, Canisius-Wilhelmina Hospital, Nijmegen, The Netherlands
| | - Leonie Exterkate
- Department of Urology, Canisius-Wilhelmina Hospital, Prosper Prostate Cancer Clinics, Nijmegen/Eindhoven, The Netherlands
| | | | - Lenneke J M Brouwer
- Department of Urology, Canisius-Wilhelmina Hospital, Prosper Prostate Cancer Clinics, Nijmegen/Eindhoven, The Netherlands
| | - Diederik M Somford
- Department of Urology, Canisius-Wilhelmina Hospital, Prosper Prostate Cancer Clinics, Nijmegen/Eindhoven, The Netherlands
| | | | - Jean-Paul A van Basten
- Department of Urology, Canisius-Wilhelmina Hospital, Prosper Prostate Cancer Clinics, Nijmegen/Eindhoven, The Netherlands
| |
Collapse
|
22
|
van Lith SAM, Pruis IJ, Tolboom N, Snijders TJ, Henssen D, Ter Laan M, Te Dorsthorst M, Leenders WPJ, Gotthardt M, Nagarajah J, Robe PA, De Witt Hamer P, Hendrikse H, Oprea-Lager DE, Yaqub M, Boellaard R, Wesseling P, Balvers RK, Verburg FA, Harteveld AA, Smits M, van den Bent M, van Zanten SEMV, van de Giessen E. PET Imaging and Protein Expression of Prostate-Specific Membrane Antigen in Glioblastoma: A Multicenter Inventory Study. J Nucl Med 2023; 64:1526-1531. [PMID: 37652540 DOI: 10.2967/jnumed.123.265738] [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: 03/16/2023] [Revised: 05/31/2023] [Indexed: 09/02/2023] Open
Abstract
Upregulation of prostate-specific membrane antigen (PSMA) in neovasculature has been described in glioblastoma multiforme (GBM), whereas vasculature in nonaffected brain shows hardly any expression of PSMA. It is unclear whether PSMA-targeting tracer uptake on PET is based on PSMA-specific binding to neovasculature or aspecific uptake in tumor. Here, we quantified uptake of various PSMA-targeting tracers in GBM and correlated this with PSMA expression in tumor biopsy samples from the same patients. Methods: Fourteen patients diagnosed with de novo (n = 8) or recurrent (n = 6) GBM underwent a preoperative PET scan after injection of 1.5 MBq/kg [68Ga]Ga-PSMA-11 (n = 7), 200 MBq of [18F]DCFpyl (n = 3), or 200 MBq of [18F]PSMA-1007 (n = 4). Uptake in tumor and tumor-to-background ratios, with contralateral nonaffected brain as background, were determined. In a subset of patients, PSMA expression levels from different regions in the tumor tissue samples (n = 40), determined using immunohistochemistry (n = 35) or RNA sequencing (n = 13), were correlated with tracer uptake on PET. Results: Moderate to high (SUVmax, 1.3-20.0) heterogeneous uptake was found in all tumors irrespective of the tracer type used. Uptake in nonaffected brain was low, resulting in high tumor-to-background ratios (6.1-359.0) calculated by dividing SUVmax of tumor by SUVmax of background. Immunohistochemistry showed variable PSMA expression on endothelial cells of tumor microvasculature, as well as on dispersed individual cells (of unknown origin), and granular staining of the neuropil. No correlation was found between in vivo uptake and PSMA expression levels (for immunohistochemistry, r = -0.173, P = 0.320; for RNA, r = -0.033, P = 0.915). Conclusion: Our results indicate the potential use of various PSMA-targeting tracers in GBM. However, we found no correlation between PSMA expression levels on immunohistochemistry and uptake intensity on PET. Whether this may be explained by methodologic reasons, such as the inability to measure functionally active PSMA with immunohistochemistry, tracer pharmacokinetics, or the contribution of a disturbed blood-brain barrier to tracer retention, should still be investigated.
Collapse
Affiliation(s)
- Sanne A M van Lith
- Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ilanah J Pruis
- Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Nelleke Tolboom
- Radiology and Nuclear Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Tom J Snijders
- Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Dylan Henssen
- Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mark Ter Laan
- Neurosurgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - William P J Leenders
- Biochemistry, Radboud University Medical Center, Nijmegen, The Netherlands
- Predica Diagnostics, Nijmegen, The Netherlands
| | - Martin Gotthardt
- Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - James Nagarajah
- Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Pierre A Robe
- Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Harry Hendrikse
- Radiology and Nuclear Medicine, Amsterdam UMC, VUmc, Amsterdam, The Netherlands
| | | | - Maqsood Yaqub
- Radiology and Nuclear Medicine, Amsterdam UMC, VUmc, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Radiology and Nuclear Medicine, Amsterdam UMC, VUmc, Amsterdam, The Netherlands
| | - Pieter Wesseling
- Pathology, Amsterdam UMC, VUmc, Amsterdam, The Netherlands
- Pathology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | | | | - Anita A Harteveld
- Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Marion Smits
- Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
- Medical Delta, Delft, The Netherlands; and
| | - Martin van den Bent
- Brain Tumor Center at Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, The Netherlands
| | | | | |
Collapse
|
23
|
Giovanella L, Garo ML, Cuzzocrea M, Paone G, Herrmann K. Prognostic role of early prostate specific antigen changes after [ 177 Lu]Lu-PSMA radioligand therapy of metastasized prostate cancer: A meta-analysis. Eur J Clin Invest 2023; 53:e14014. [PMID: 37194605 DOI: 10.1111/eci.14014] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/29/2023] [Accepted: 04/29/2023] [Indexed: 05/18/2023]
Abstract
BACKGROUND Approximately 10%-20% of prostate cancers progress to metastatic and castration-resistant forms (mCRPC). Radioligand (RLT) therapy with [177 Lu]Lu-prostate-specific membrane antigen (PSMA) is an emerging treatment for metastasized mCRPC and its efficacy is assessed not only but also by prostate specific antigen (PSA) measurement after 12 weeks or more after treatment. Our aim was to evaluate the role of early PSA measurement after RLT in predicting overall survival (OS) of mCRPC patients. METHODS A systematic search on PubMed, Web Of Science and Scopus was performed from January to December 2022. PRISMA guidelines for prognostic studies was adopted. Risk of bias was assessed using quality of prognostic studies (QUIPS). RESULTS Twelve studies at low-intermediate risk of bias, were included in the meta-analysis (1646 patients, mean age 70 years). About 50% of patients showed a PSA decline after 1-2 of [177 Lu]Lu-PSMA, and more than 30% reported a PSA decline ≥50%. The median OS range for patients with any PSA decline was 13-20 months, while for patients with stable or increased PSA, the median OS fell to 6-12 months. The OS rate for a PSA decline after the one-two [177 Lu]Lu-PSMA cycles was 0.39 (95% CI: 0.31-0.50), while OS for a PSA decline ≥50% was 0.69 (95% CI: 0.57-0.83). CONCLUSIONS A PSA decline is observed in almost 50% of mCRPC patients after 1-2 [177 Lu]Lu-PSMA cycles, with a significantly longer OS compared to stable or increased PSA levels, respectively. Accordingly, any PSA decline after 1-2 cycles of therapy should be regarded as a favourable prognostic factor for OS.
Collapse
Affiliation(s)
- Luca Giovanella
- Clinic for Nuclear Medicine and Molecular Imaging, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Clinic for Nuclear Medicine, University Hospital of Zürich, Zürich, Switzerland
| | - Maria Luisa Garo
- Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Cardiac Surgery, Department of Cardiovascular Surgery, Università Campus Bio-Medico, Rome, Italy
| | - Marco Cuzzocrea
- Clinic for Nuclear Medicine and Molecular Imaging, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Gaetano Paone
- Clinic for Nuclear Medicine and Molecular Imaging, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Ken Herrmann
- Clinic for Nuclear Medicine, Essen University Hospital, Essen, Germany
| |
Collapse
|
24
|
Siebinga H, Privé BM, Peters SMB, Nagarajah J, Dorlo TPC, Huitema ADR, de Wit‐van der Veen BJ, Hendrikx JJMA. Population pharmacokinetic dosimetry model using imaging data to assess variability in pharmacokinetics of 177 Lu-PSMA-617 in prostate cancer patients. CPT Pharmacometrics Syst Pharmacol 2023; 12:1060-1071. [PMID: 36760133 PMCID: PMC10431047 DOI: 10.1002/psp4.12914] [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: 08/30/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 02/11/2023] Open
Abstract
Studies to evaluate and optimize [177 Lu]Lu-PSMA treatment focus primarily on individual patient data. A population pharmacokinetic (PK) dosimetry model was developed to explore the potential of using imaging data as input for population PK models and to characterize variability in organ and tumor uptake of [177 Lu]Lu-PSMA-617 in patients with low volume metastatic prostate cancer. Simulations were performed to identify the effect of dose adjustments on absorbed doses in salivary glands and tumors. A six-compartment population PK model was developed, consisting of blood, salivary gland, kidneys, liver, tumor, and a lumped compartment representing other tissue (compartment 1-6, respectively), based on data from 10 patients who received [177 Lu]Lu-PSMA-617 (2 cycles, ~ 3 and ~ 6 GBq). Data consisted of radioactivity levels (decay corrected) in blood and tissues (9 blood samples and 5 single photon emission computed tomography/computed tomography scans). Observations in all compartments were adequately captured by individual model predictions. Uptake into salivary glands was saturable with an estimated maximum binding capacity (Bmax ) of 40.4 MBq (relative standard error 12.3%) with interindividual variability (IIV) of 59.3% (percent coefficient of variation [CV%]). IIV on other PK parameters was relatively minor. Tumor volume was included as a structural effect on the tumor uptake rate constant (k15 ), where a two-fold increase in tumor volume resulted in a 1.63-fold increase in k15 . In addition, interoccasion variability on k15 improved the model fit (43.5% [CV%]). Simulations showed a reduced absorbed dose per unit administered activity for salivary glands after increasing radioactivity dosing from 3 to 6 GBq (0.685 Gy/GBq vs. 0.421 Gy/GBq, respectively). All in all, population PK modeling could help to improve future radioligand therapy research.
Collapse
Affiliation(s)
- Hinke Siebinga
- Department of Pharmacy & PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Department of Nuclear MedicineThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Bastiaan M. Privé
- Department of Radiology and Nuclear MedicineRadboud University Medical CenterNijmegenThe Netherlands
| | - Steffie M. B. Peters
- Department of Radiology and Nuclear MedicineRadboud University Medical CenterNijmegenThe Netherlands
| | - James Nagarajah
- Department of Radiology and Nuclear MedicineRadboud University Medical CenterNijmegenThe Netherlands
| | - Thomas P. C. Dorlo
- Department of Pharmacy & PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Department of PharmacyUppsala UniversityUppsalaSweden
| | - Alwin D. R. Huitema
- Department of Pharmacy & PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Department of Clinical PharmacyUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
- Department of PharmacologyPrincess Máxima Center for Pediatric OncologyUtrechtThe Netherlands
| | | | - Jeroen J. M. A. Hendrikx
- Department of Pharmacy & PharmacologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- Department of Nuclear MedicineThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| |
Collapse
|
25
|
Cheng L, Yang T, Zhang J, Gao F, Yang L, Tao W. The Application of Radiolabeled Targeted Molecular Probes for the Diagnosis and Treatment of Prostate Cancer. Korean J Radiol 2023; 24:574-589. [PMID: 37271211 DOI: 10.3348/kjr.2022.1002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/26/2023] [Accepted: 03/30/2023] [Indexed: 06/06/2023] Open
Abstract
Radiopharmaceuticals targeting prostate-specific membrane antigens (PSMA) are essential for the diagnosis, evaluation, and treatment of prostate cancer (PCa), particularly metastatic castration-resistant PCa, for which conventional treatment is ineffective. These molecular probes include [68Ga]PSMA, [18F]PSMA, [Al18F]PSMA, [99mTc]PSMA, and [89Zr]PSMA, which are widely used for diagnosis, and [177Lu]PSMA and [225Ac]PSMA, which are used for treatment. There are also new types of radiopharmaceuticals. Due to the differentiation and heterogeneity of tumor cells, a subtype of PCa with an extremely poor prognosis, referred to as neuroendocrine prostate cancer (NEPC), has emerged, and its diagnosis and treatment present great challenges. To improve the detection rate of NEPC and prolong patient survival, many researchers have investigated the use of relevant radiopharmaceuticals as targeted molecular probes for the detection and treatment of NEPC lesions, including DOTA-TOC and DOTA-TATE for somatostatin receptors, 4A06 for CUB domain-containing protein 1, and FDG. This review focused on the specific molecular targets and various radionuclides that have been developed for PCa in recent years, including those mentioned above and several others, and aimed to provide valuable up-to-date information and research ideas for future studies.
Collapse
Affiliation(s)
- Luyi Cheng
- Department of Nuclear Medicine, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Tianshuo Yang
- Department of Nuclear Medicine, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Jun Zhang
- Department of Nuclear Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu, China
| | - Feng Gao
- Key Laboratory for Experimental Teratology of the Ministry of Education and Center for Experimental Nuclear Medicine, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Lingyun Yang
- JYAMS PET Research and Development Limited, Nanjing, Jiangsu, China
| | - Weijing Tao
- Department of Nuclear Medicine, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China.
| |
Collapse
|
26
|
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: 108] [Impact Index Per Article: 108.0] [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
|
27
|
Laudicella R, Spataro A, Crocè L, Giacoppo G, Romano D, Davì V, Lopes M, Librando M, Nicocia A, Rappazzo A, Celesti G, Torre FL, Pagano B, Garraffa G, Bauckneht M, Burger IA, Minutoli F, Baldari S. Preliminary Findings of the Role of FAPi in Prostate Cancer Theranostics. Diagnostics (Basel) 2023; 13:diagnostics13061175. [PMID: 36980482 PMCID: PMC10047910 DOI: 10.3390/diagnostics13061175] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/22/2023] Open
Abstract
Prostate cancer (PCa) is the most frequently diagnosed cancer worldwide and the second most common cause of cancer-related deaths among men. Progress in molecular imaging has magnified its clinical management; however, an unmet clinical need involves the identification of new imaging biomarkers that complement the gold standard of prostate-specific membrane antigen (PSMA) positron emission tomography (PET) in cases of clinically significant PCa that do not express PSMA. Fibroblast activation protein (FAP) is a type II transmembrane serine overexpressed in many solid cancers that can be imaged through quinoline-based PET tracers derived from an FAP inhibitor (FAPi). Preliminary results of FAPi application in PCa (in PSMA-negative lesions, and in comparison with fluorodeoxyglucose—FDG) are now available in the literature. FAP-targeting ligands for PCa are not limited to detection, but could also include therapeutic applications. In this preliminary review, we provide an overview of the clinical applications of FAPi ligands in PCa, summarising the main results and highlighting contemporary strengths and limitations.
Collapse
Affiliation(s)
- Riccardo Laudicella
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98124 Messina, Italy
- Nuclear Medicine Unit, S. Antonio Abate Hospital, 91016 Trapani, Italy
- Correspondence:
| | - Alessandro Spataro
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98124 Messina, Italy
| | - Ludovica Crocè
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98124 Messina, Italy
| | - Giulia Giacoppo
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98124 Messina, Italy
| | - Davide Romano
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98124 Messina, Italy
| | - Valerio Davì
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98124 Messina, Italy
| | - Maria Lopes
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98124 Messina, Italy
| | - Maria Librando
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98124 Messina, Italy
| | - Antonio Nicocia
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98124 Messina, Italy
| | - Andrea Rappazzo
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98124 Messina, Italy
| | - Greta Celesti
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98124 Messina, Italy
| | - Flavia La Torre
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98124 Messina, Italy
| | - Benedetta Pagano
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98124 Messina, Italy
| | - Giuseppe Garraffa
- Nuclear Medicine Unit, S. Antonio Abate Hospital, 91016 Trapani, Italy
| | - Matteo Bauckneht
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
- Department of Health Sciences (DISSAL), University of Genova, 16132 Genova, Italy
| | - Irene A Burger
- Department of Nuclear Medicine, Cantonal Hospital Baden, 5404 Baden, Switzerland
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
| | - Fabio Minutoli
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98124 Messina, Italy
| | - Sergio Baldari
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98124 Messina, Italy
| |
Collapse
|
28
|
Kleiburg F, Heijmen L, Gelderblom H, Kielbasa SM, Bovée JV, De Geus-Oei LF. Prostate-specific membrane antigen (PSMA) as a potential target for molecular imaging and treatment in bone and soft tissue sarcomas. Br J Radiol 2023; 96:20220886. [PMID: 36728839 PMCID: PMC10161918 DOI: 10.1259/bjr.20220886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Bone and soft tissue sarcomas are a group of rare malignant tumours with major histological and anatomical varieties. In a metastatic setting, sarcomas have a poor prognosis due to limited response rates to chemotherapy. Radioligand therapy targeting prostate-specific membrane antigen (PSMA) may offer a new perspective. PSMA is a type II transmembrane glycoprotein which is present in all prostatic tissue and overexpressed in prostate cancer. Despite the name, PSMA is not prostate-specific. PSMA expression is also found in a multitude of non-prostatic diseases including a subgroup of sarcomas, mostly in its neovascular endothelial cells. On PET/CT imaging, multiple sarcomas have also shown intense PSMA-tracer accumulation. PSMA expression and PSMA-tracer uptake seem to be highest in patients with aggressive and advanced sarcomas, who are also in highest need of new therapeutic options. Although these results provide a good rationale for the future use of PSMA-targeted radioligand therapy in a selection of sarcoma patients, more research is needed to gain insight into optimal patient selection methods, PSMA-targeting antibodies and tracers, administered doses of radioligand therapy, and their efficacy and tolerability. In this review, mRNA expression of the FOLH1 gene which encodes PSMA, PSMA immunohistochemistry, PSMA-targeted imaging and PSMA-targeted therapy in sarcomas will be discussed.
Collapse
Affiliation(s)
- Fleur Kleiburg
- Biomedical Photonic Imaging Group, University of Twente, Enschede, The Netherlands.,Department of Radiology, section of Nuclear Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Linda Heijmen
- Department of Radiology, section of Nuclear Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Szymon M Kielbasa
- Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, The Netherlands
| | - Judith Vmg Bovée
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Lioe-Fee De Geus-Oei
- Biomedical Photonic Imaging Group, University of Twente, Enschede, The Netherlands.,Department of Radiology, section of Nuclear Medicine, Leiden University Medical Center, Leiden, The Netherlands.,Department of Radiation Science and Technology, Technical University of Delft, Delft, The Netherlands
| |
Collapse
|
29
|
PSMA PET/CT imaging for biochemical recurrence of prostate cancer after radiotherapy: is it necessary to review the Phoenix criteria? Clin Transl Imaging 2023. [DOI: 10.1007/s40336-023-00543-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
30
|
Bauckneht M, Marini C, Cossu V, Campi C, Riondato M, Bruno S, Orengo AM, Vitale F, Carta S, Chiola S, Chiesa S, Miceli A, D’Amico F, Fornarini G, Terrone C, Piana M, Morbelli S, Signori A, Barboro P, Sambuceti G. Gene's expression underpinning the divergent predictive value of [18F]F-fluorodeoxyglucose and prostate-specific membrane antigen positron emission tomography in primary prostate cancer: a bioinformatic and experimental study. J Transl Med 2023; 21:3. [PMID: 36600265 PMCID: PMC9811737 DOI: 10.1186/s12967-022-03846-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 12/23/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Positron Emission Tomography (PET) imaging with Prostate-Specific Membrane Antigen (PSMA) and Fluorodeoxyglucose (FDG) represent promising biomarkers for risk-stratification of Prostate Cancer (PCa). We verified whether the expression of genes encoding for PSMA and enzymes regulating FDG cellular uptake are independent and additive prognosticators in PCa. METHODS mRNA expression of genes involved in glucose metabolism and PSMA regulation obtained from primary PCa specimens were retrieved from open-source databases and analyzed using an integrative bioinformatics approach. Machine Learning (ML) techniques were used to create predictive Progression-Free Survival (PFS) models. Cellular models of primary PCa with different aggressiveness were used to compare [18F]F-PSMA-1007 and [18F]F-FDG uptake kinetics in vitro. Confocal microscopy, immunofluorescence staining, and quantification analyses were performed to assess the intracellular and cellular membrane PSMA expression. RESULTS ML analyses identified a predictive functional network involving four glucose metabolism-related genes: ALDOB, CTH, PARP2, and SLC2A4. By contrast, FOLH1 expression (encoding for PSMA) did not provide any additive predictive value to the model. At a cellular level, the increase in proliferation rate and migratory potential by primary PCa cells was associated with enhanced FDG uptake and decreased PSMA retention (paralleled by the preferential intracellular localization). CONCLUSIONS The overexpression of a functional network involving four glucose metabolism-related genes identifies a higher risk of disease progression since the earliest phases of PCa, in agreement with the acknowledged prognostic value of FDG PET imaging. By contrast, the prognostic value of PSMA PET imaging is independent of the expression of its encoding gene FOLH1. Instead, it is influenced by the protein docking to the cell membrane, regulating its accessibility to tracer binding.
Collapse
Affiliation(s)
- Matteo Bauckneht
- grid.5606.50000 0001 2151 3065Department of Health Sciences, University of Genoa, 16132 Genoa, Italy ,grid.410345.70000 0004 1756 7871Nuclear Medicine Unit, IRCCS, Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Cecilia Marini
- grid.410345.70000 0004 1756 7871Nuclear Medicine Unit, IRCCS, Ospedale Policlinico San Martino, 16132 Genoa, Italy ,grid.428490.30000 0004 1789 9809CNR, Institute of Molecular Bioimaging and Physiology (IBFM), 20054 Milan, Italy
| | - Vanessa Cossu
- grid.5606.50000 0001 2151 3065Department of Health Sciences, University of Genoa, 16132 Genoa, Italy ,grid.410345.70000 0004 1756 7871Nuclear Medicine Unit, IRCCS, Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Cristina Campi
- grid.5606.50000 0001 2151 3065LISCOMP Lab, Department of Mathematics (DIMA), University of Genoa, 16132 Genoa, Italy
| | - Mattia Riondato
- grid.410345.70000 0004 1756 7871Nuclear Medicine Unit, IRCCS, Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Silvia Bruno
- grid.5606.50000 0001 2151 3065Department of Experimental Medicine, Human Anatomy, University of Genoa, 16132 Genoa, Italy
| | - Anna Maria Orengo
- grid.410345.70000 0004 1756 7871Nuclear Medicine Unit, IRCCS, Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Francesca Vitale
- grid.410345.70000 0004 1756 7871Nuclear Medicine Unit, IRCCS, Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Sonia Carta
- grid.410345.70000 0004 1756 7871Nuclear Medicine Unit, IRCCS, Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Silvia Chiola
- grid.410345.70000 0004 1756 7871Nuclear Medicine Unit, IRCCS, Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Sabrina Chiesa
- grid.410345.70000 0004 1756 7871Nuclear Medicine Unit, IRCCS, Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Alberto Miceli
- grid.5606.50000 0001 2151 3065Department of Health Sciences, University of Genoa, 16132 Genoa, Italy
| | - Francesca D’Amico
- grid.5606.50000 0001 2151 3065Department of Health Sciences, University of Genoa, 16132 Genoa, Italy
| | - Giuseppe Fornarini
- grid.410345.70000 0004 1756 7871Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Carlo Terrone
- grid.410345.70000 0004 1756 7871Department of Urology, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy ,grid.5606.50000 0001 2151 3065Department of Surgical and Diagnostic Integrated Sciences (DISC), University of Genova, 16132 Genoa, Italy
| | - Michele Piana
- grid.5606.50000 0001 2151 3065LISCOMP Lab, Department of Mathematics (DIMA), University of Genoa, 16132 Genoa, Italy ,grid.482259.00000 0004 1774 9464CNR-SPIN Genoa, 16132 Genoa, Italy
| | - Silvia Morbelli
- grid.5606.50000 0001 2151 3065Department of Health Sciences, University of Genoa, 16132 Genoa, Italy ,grid.410345.70000 0004 1756 7871Nuclear Medicine Unit, IRCCS, Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Alessio Signori
- grid.5606.50000 0001 2151 3065Department of Health Sciences, University of Genoa, 16132 Genoa, Italy
| | - Paola Barboro
- grid.410345.70000 0004 1756 7871Proteomic and Mass Spectrometry Unit, IRCCS, Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Gianmario Sambuceti
- grid.5606.50000 0001 2151 3065Department of Health Sciences, University of Genoa, 16132 Genoa, Italy ,grid.410345.70000 0004 1756 7871Nuclear Medicine Unit, IRCCS, Ospedale Policlinico San Martino, 16132 Genoa, Italy
| |
Collapse
|
31
|
Shahrokhi P, Emami-Ardekani A, Karamzade-Ziarati N. SSTR-based theranostics in neuroendocrine prostate cancer (NEPC). Clin Transl Imaging 2022. [DOI: 10.1007/s40336-022-00535-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
32
|
The Role of [ 68Ga]PSMA PET/CT for Clinical Suspicion of Prostate Cancer in Patients with or without Previous Negative Biopsy: A Systematic Review. Cancers (Basel) 2022; 14:cancers14205036. [PMID: 36291820 PMCID: PMC9600353 DOI: 10.3390/cancers14205036] [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: 09/14/2022] [Revised: 10/04/2022] [Accepted: 10/10/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary In this paper we systematically evaluate the evidence regarding the role of [68Ga]PSMA PET/CT for clinical suspicions of prostate cancer in patients with or without previous negative biopsy. A critical review of PubMed and Web of Science according to the PRISMA statement was conducted. Eighteen publications were selected for inclusion in the analysis. In 8 articles, there was a direct comparison with mpMRI. [68Ga]PSMA PET/CT resulted more accurate in identifying primary prostate cancer with PSA values between 4 and 20 ng/mL than mpMRI. Moreover, its use combined with MRI improved sensitivity for csPCa detection, thus potentially avoiding unnecessary biopsies. Overall, [68Ga]PSMA PET/CT resulted a promising technique in patients with clinical suspicion of PCa and precedent negative biopsy or contraindications to MRI. Abstract The purpose of the study is to systematically evaluate the evidence regarding the role of [68Ga]PSMA PET/CT for clinical suspicions of prostate cancer in patients with or without previous negative biopsy. We performed a critical review of PubMed and Web of Science according to the PRISMA statement. Eighteen publications were selected for inclusion in this analysis. QUADAS-2 evaluation was adopted for quality analyses. [68Ga]PSMA-11 was the radiotracer of choice in 15 studies, while [68Ga]PSMA-617 was used in another 3. In 8 articles, there was a direct comparison with mpMRI. The total number of patients included was 1379, ranging from 15 to 291, with a median age of 64 years (range: 42–90). The median baseline PSA value was 12.9 ng/mL, ranging from 0.85 to 4156 ng/mL. Some studies evaluated the PSMA uptake comparing the SUVmax of suspicious lesions with the SUVmax of the normal biodistribution to find out optimal cut-off points. In addition, some studies suggested a significant association between PSA levels, PSA density, and [68Ga]PSMA PET/CT finding. [68Ga]PSMA PET/CT seems to be more accurate in identifying primary prostate cancer with PSA values between 4 and 20 ng/mL than mpMRI. Moreover, in some trials, the combination of PSMA PET/CT and MRI improved the NPV in the detection of clinically significant prostate cancer (csPCa) than MRI alone. Our findings are limited by the small numbers of studies and patient heterogeneity. [68Ga]PSMA PET/CT is a promising technique in patients with clinical suspicion of PCa and precedent negative biopsy or contraindications to MRI. Furthermore, its use combined with MRI improves sensitivity for csPCa detection and can avoid unnecessary biopsies.
Collapse
|
33
|
Laudicella R, La Torre F, Davì V, Crocè L, Aricò D, Leonardi G, Russo S, Minutoli F, Burger IA, Baldari S. Prostate Cancer Biochemical Recurrence Resulted Negative on [ 68Ga]Ga-PSMA-11 but Positive on [ 18F]Fluoromethylcholine PET/CT. Tomography 2022; 8:2471-2474. [PMID: 36287804 PMCID: PMC9609559 DOI: 10.3390/tomography8050205] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/23/2022] [Accepted: 09/28/2022] [Indexed: 11/07/2022] Open
Abstract
For prostate cancer (PCa) biochemical recurrence (BCR), the primarily suggested imaging technique by the European Association of Urology (EAU) guidelines is prostate-specific membrane antigen (PSMA) positron emission tomography/computer tomography (PET/CT). Indeed, the increased detection rate of PSMA PET/CT for early BCR has led to a fast and wide acceptance of this novel technology. However, PCa is a very heterogeneous disease, not always easily assessable with the highly specific PSMA PET with around 10% of cases occuring without PSMA expression. In this paper, we present the case of a patient with PCa BCR that resulted negative on [68Ga]Ga-PSMA-11 PET/CT, but positive on [18F]Fluoromethylcholine (Choline) PET/CT.
Collapse
Affiliation(s)
- Riccardo Laudicella
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98125 Messina, Italy
- Department of Nuclear Medicine, University Hospital Zürich, University of Zürich, 8091 Zürich, Switzerland
- Department of Nuclear Medicine, Kantonsspital Baden, 5404 Baden, Switzerland
- Correspondence:
| | - Flavia La Torre
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98125 Messina, Italy
| | - Valerio Davì
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98125 Messina, Italy
| | - Ludovica Crocè
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98125 Messina, Italy
| | - Demetrio Aricò
- Department of Nuclear Medicine, Humanitas Oncological Centre of Catania, 95125 Catania, Italy
| | - Giuseppe Leonardi
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98125 Messina, Italy
| | - Simona Russo
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98125 Messina, Italy
| | - Fabio Minutoli
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98125 Messina, Italy
| | - Irene A. Burger
- Department of Nuclear Medicine, University Hospital Zürich, University of Zürich, 8091 Zürich, Switzerland
- Department of Nuclear Medicine, Kantonsspital Baden, 5404 Baden, Switzerland
| | - Sergio Baldari
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98125 Messina, Italy
| |
Collapse
|
34
|
Pepe P, Pepe L, Tamburo M, Marletta G, Pennisi M, Fraggetta F. Targeted prostate biopsy: 68Ga-PSMA PET/CT vs. mpMRI in the diagnosis of prostate cancer. Arch Ital Urol Androl 2022; 94:274-277. [DOI: 10.4081/aiua.2022.3.274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/06/2022] [Indexed: 11/23/2022] Open
Abstract
Introduction: To evaluate the diagnostic accuracy of 68Ga-prostate-specific membrane antigen (PSMA) positron emission tomography/computed tomog-raphy (PET/CT) vs. multiparametric magnetic resonance imag-ing (mpMRI) targeted biopsy (TPBx) in the diagnosis of clinical-ly significant prostate cancer (csPCa: Grade Group ≥ 2).
Materials and methods: From January 2021 to June 2022, 100 patients (median age: 66 years) with negative digital rectal examination underwent transperineal prostate biopsy for abnor-mal PSA values (median 7.5 ng/ml). Before prostate biopsy, all patients underwent mpMRI and 68Ga-PET/CT examinations and mpMRI (PI-RADS version 2 ≥ 3) or 68Ga-PET/CT index lesions suspicious for cancer (SUVmax > 5 g/ml) underwent cognitive targeted cores (mpMRI-TPBx and PSMA-TPBx: four cores) com-bined with extended systematic prostate biopsy (eSPBx: median 18 cores). The procedure was performed transperineally using a tru-cut 18-gauge needle under sedation and antibiotic prophy-laxis. Results: PCa was found in 58/100 (58.0%) men; in detail, 44/58 (75.9%) were csPCa; mpMRI and 68Ga-PSMA showed 66/100 (66%) and 62/100 (60%) lesions suspicious for PCa, respective-ly. 68Ga-PSMA-TPBx vs. mpMRI-TPBx vs. eSPBx diagnosed 42 (95.4%) vs. 36 (81.8%) vs. 30 (68.2%) csPCa, respectively; mpMRI-TPBx vs. 68Ga-PSMA-TPBx showed a diagnostic accuracy of 76.9% vs. 84.9% in diagnosing csPCa.
Conclusions: 68GaPSMA PET/CT TPBx demonstrated good accuracy in the diagnosis of csPCa, which was not inferior to mpMRI TPBx (84.9% vs. 76.9%) improving the detection rate for cancer of systematic biopsy.
Collapse
|
35
|
Prostate specific membrane antigen positron emission tomography in primary prostate cancer diagnosis: First-line imaging is afoot. Cancer Lett 2022; 548:215883. [PMID: 36027998 DOI: 10.1016/j.canlet.2022.215883] [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: 07/23/2022] [Accepted: 08/11/2022] [Indexed: 11/23/2022]
Abstract
Prostate specific membrane antigen positron emission tomography (PSMA PET) is an excellent molecular imaging technique for prostate cancer. Currently, PSMA PET for patients with primary prostate cancer is supplementary to conventional imaging techniques, according to guidelines. This supplementary function of PSMA PET is due to a lack of systematic review of its strengths, limitations, and potential development direction. Thus, we review PSMA ligands, detection, T, N, and M staging, treatment management, and false results of PSMA PET in clinical studies. We also discuss the strengths and challenges of PSMA PET. PSMA PET can greatly increase the detection rate of prostate cancer and accuracy of T/N/M staging, which facilitates more appropriate treatment for primary prostate cancer. Lastly, we propose that PSMA PET could become the first-line imaging modality for primary prostate cancer, and we describe its potential expanded application.
Collapse
|
36
|
Brown EL, DeWeerd RA, Zidel A, Pereira PMR. Preclinical antibody-PET imaging of PD-L1. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2022; 2:953202. [PMID: 39354977 PMCID: PMC11440863 DOI: 10.3389/fnume.2022.953202] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/11/2022] [Indexed: 10/03/2024]
Abstract
Programmed cell death protein-1/ligand-1 (PD-1/PD-L1) blockade, including antibody therapeutics, has transformed cancer treatment. However, a major challenge in the field relates to selecting patients who are likely to respond to immune checkpoint inhibitors. Indeed, biopsy-based diagnostic tests to determine immune checkpoint protein levels do not accurately capture the inherent spatial and temporal heterogeneity of PD-L1 tumor expression. As a result, not all PD-L1-positive tumors respond to immunotherapies, and some patients with PD-L1-negative tumors have shown clinical benefits. In 2018, a first-in-human study of the clinically-approved anti-PD-L1 antibody Atezolizumab labeled with the positron emitter zirconium-89 validated the ability of positron emission tomography (PET) to visualize PD-L1 expression in vivo and predict tumor response to immunotherapy. These studies have triggered the expansion of PD-L1-targeted immunoPET to assess PD-L1 protein levels and PD-L1 expression heterogeneity in real time and across the whole tumor. First, this mini-review introduces new PD-L1 PET imaging studies of the last 4 years, focusing on the expansion of preclinical tumor models and anti-PD-L1 antibodies/antibody fragments in development. Then, the review discusses how these preclinical models and targeting agents can be utilized to study spatial and temporal heterogeneity of PD-L1 expression.
Collapse
Affiliation(s)
- Emma L. Brown
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Rachel A. DeWeerd
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, United States
| | - Abbey Zidel
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Patricia M. R. Pereira
- Department of Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
37
|
The Role of PSMA PET/CT in the Primary Diagnosis and Follow-Up of Prostate Cancer-A Practical Clinical Review. Cancers (Basel) 2022; 14:cancers14153638. [PMID: 35892897 PMCID: PMC9367536 DOI: 10.3390/cancers14153638] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary The combination of positron emission tomography (PET)-diagnostics with ligands binding to the prostate-specific membrane antigen (PSMA) has been a diagnostic milestone in the situation of biochemical recurrence of prostate cancer and is gaining importance in primary diagnostics, providing a highly specific and sensitive diagnostic method in various clinical situations. However, the clinical application of this method requires a comprehensive knowledge of its advantages and disadvantages, potential pitfalls and influencing factors. This review aims to provide a practical clinical review of the currently available background data on PSMA PET/CT, as well as the clinical implications. Although a large amount of data already exist, a thorough analysis is complicated by study heterogeneity, showing the need for future systematic and prospective research. Abstract The importance of PSMA PET/CT in both primary diagnostics and prostate cancer recurrence has grown steadily since its introduction more than a decade ago. Over the past years, a vast amount of data have been published on the diagnostic accuracy and the impact of PSMA PET/CT on patient management. Nevertheless, a large heterogeneity between studies has made reaching a consensus difficult; this review aims to provide a comprehensive clinical review of the available scientific literature, covering the currently known data on physiological and pathological PSMA expression, influencing factors, the differences and pitfalls of various tracers, as well as the clinical implications in initial TNM-staging and in the situation of biochemical recurrence. This review has the objective of providing a practical clinical overview of the advantages and disadvantages of the examination in various clinical situations and the body of knowledge available, as well as open questions still requiring further research.
Collapse
|
38
|
Could 68Ga-PSMA PET/CT Evaluation Reduce the Number of Scheduled Prostate Biopsies in Men Enrolled in Active Surveillance Protocols? J Clin Med 2022; 11:jcm11123473. [PMID: 35743547 PMCID: PMC9225630 DOI: 10.3390/jcm11123473] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/13/2022] [Accepted: 06/13/2022] [Indexed: 02/06/2023] Open
Abstract
Background: To evaluate the accuracy of 68Ga-prostate specific membrane antigen (PSMA) PET/CT in the diagnosis of clinically significant prostate cancer (csPCa) (Grade Group > 2) in men enrolled in Active Surveillance (AS) protocol. Methods: From May 2013 to May 2021, 173 men with very low-risk PCa were enrolled in an AS protocol study. During the follow-up, 38/173 (22%) men were upgraded and 8/173 (4.6%) decided to leave the AS protocol. After four years from confirmatory biopsy (range: 48−52 months), 30/127 (23.6%) consecutive patients were submitted to mpMRI and 68Ga-PSMA PET/CT scan before scheduled repeated biopsy. All the mpMRI (PI-RADS > 3) and 68Ga-PET/TC standardised uptake value (SUVmax) > 5 g/mL index lesions underwent targeted cores (mpMRI-TPBx and PSMA-TPBx) combined with transperineal saturation prostate biopsy (SPBx: median 20 cores). Results: mpMRI and 68Ga-PSMA PET/CT showed 14/30 (46.6%) and 6/30 (20%) lesions suspicious for PCa. In 2/30 (6.6%) men, a csPCa was found; 68Ga-PSMA-TPBx vs. mpMRI-TPBx vs. SPBx diagnosed 1/2 (50%) vs. 1/2 (50%) vs. 2/2 (100%) csPCa, respectively. In detail, mpMRI and 68Ga-PSMA PET/TC demonstrated 13/30 (43.3%) vs. 5/30 (16.7%) false positive and 1 (50%) vs. 1 (50%) false negative results. Conclusion: 68Ga-PSMA PET/CT did not improve the detection for csPCa of SPBx but would have spared 24/30 (80%) scheduled biopsies showing a lower false positive rate in comparison with mpMRI (20% vs. 43.3%) and a negative predictive value of 85.7% vs. 57.1%, respectively.
Collapse
|
39
|
Rosenzweig B, Haramaty R, Davidson T, Lazarovich A, Shvero A, Haifler M, Gal J, Golan S, Shpitzer S, Hoffman A, Nativ O, Freifeld Y, Zreik R, Dotan ZA. Reply to Veerman et al. Comment on “Rosenzweig et al. Very Low Prostate PET/CT PSMA Uptake May Be Misleading in Staging Radical Prostatectomy Candidates. J. Pers. Med. 2022, 12, 410”. J Pers Med 2022; 12:jpm12060861. [PMID: 35743645 PMCID: PMC9225530 DOI: 10.3390/jpm12060861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 05/14/2022] [Indexed: 02/01/2023] Open
Affiliation(s)
- Barak Rosenzweig
- Department of Urology, Chaim Sheba Medical Center, Ramat Gan 5211401, Israel; (R.H.); (A.L.); (A.S.); (Z.A.D.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (T.D.); (M.H.); (S.G.)
- Correspondence: ; Tel.: +972-3-5302221
| | - Rennen Haramaty
- Department of Urology, Chaim Sheba Medical Center, Ramat Gan 5211401, Israel; (R.H.); (A.L.); (A.S.); (Z.A.D.)
| | - Tima Davidson
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (T.D.); (M.H.); (S.G.)
- Department of Nuclear Medicine, Chaim Sheba Medical Center, Ramat Gan 5211401, Israel
| | - Alon Lazarovich
- Department of Urology, Chaim Sheba Medical Center, Ramat Gan 5211401, Israel; (R.H.); (A.L.); (A.S.); (Z.A.D.)
| | - Asaf Shvero
- Department of Urology, Chaim Sheba Medical Center, Ramat Gan 5211401, Israel; (R.H.); (A.L.); (A.S.); (Z.A.D.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (T.D.); (M.H.); (S.G.)
| | - Miki Haifler
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (T.D.); (M.H.); (S.G.)
- Department of Urology, Shamir Medical Center, Tzrifin 6093000, Israel;
| | - Jonathan Gal
- Department of Urology, Shamir Medical Center, Tzrifin 6093000, Israel;
| | - Shay Golan
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (T.D.); (M.H.); (S.G.)
- Section of Urology, Rabin Medical Center, Petah Tikva 4941492, Israel;
| | - Sagi Shpitzer
- Section of Urology, Rabin Medical Center, Petah Tikva 4941492, Israel;
| | - Azik Hoffman
- Department of Urology, Rambam Health Center, Haifa 3109601, Israel; (A.H.); (O.N.)
- Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa 3200003, Israel; (Y.F.); (R.Z.)
| | - Omri Nativ
- Department of Urology, Rambam Health Center, Haifa 3109601, Israel; (A.H.); (O.N.)
| | - Yuval Freifeld
- Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa 3200003, Israel; (Y.F.); (R.Z.)
- Department of Urology, Carmel Medical Center, Haifa 3436212, Israel
| | - Rani Zreik
- Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa 3200003, Israel; (Y.F.); (R.Z.)
| | - Zohar A. Dotan
- Department of Urology, Chaim Sheba Medical Center, Ramat Gan 5211401, Israel; (R.H.); (A.L.); (A.S.); (Z.A.D.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (T.D.); (M.H.); (S.G.)
| |
Collapse
|
40
|
Diverse PSMA expression in primary prostate cancer: reason for negative [ 68Ga]Ga-PSMA PET/CT scans? Immunohistochemical validation in 40 surgical specimens. Eur J Nucl Med Mol Imaging 2022; 49:3938-3949. [PMID: 35556160 DOI: 10.1007/s00259-022-05831-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 05/01/2022] [Indexed: 11/04/2022]
Abstract
PURPOSE The purpose of this study was to immunohistochemically validate the primary tumor PSMA expression in prostate cancer (PCa) patients imaged with [68Ga]Ga-PSMA PET/CT prior to surgery, with special consideration of PET-negative cases. METHODS The study included 40 men with newly diagnosed treatment-naïve PCa imaged with [68Ga]Ga-PSMA I&T PET/CT as part of the diagnostic work-up prior to radical prostatectomy. All primary tumors were routinely stained with H&E. In addition, immunohistochemical staining of PSMA was performed and the immunoreactive score (IRS) was computed as semiquantitative measure. Subsequently, imaging findings were correlated to histopathologic results. RESULTS Eighty-three percent (33/40) of patients presented focal uptake of [68Ga]Ga-PSMA I&T in the primary tumor in at least one prostate lobe. Among PSMA-PET positive patients, one-third had lymph node metastases (LNM) detected by post-operative histopathology, while in PET negative patients, only 1 out of 7 presented with regional LN involvement; PSMA-avid distant lesions, predominantly in bones, were observed in 15% and 0% of patients, respectively. The median IRS classification of PSMA expression in tumor tissue was 2 (range, 1-3) both in PSMA-PET positive and negative prostate lobes, with significantly different interquartile range: 2-3 vs. 2-2, respectively (p = 0.03). The median volume of PSMA-PET positive tumors was 5.4 mL (0.2-32.9) as compared to 1.6 mL (0.3-18.3) of PET-negative tumors (p < 0.001). There was a significant but weak correlation between SUVmax and percentage of PSMA-positive tumor cells (r = 0.46, p < 0.001). A total of 35/44 (~80%) lobes were positive in PSMA-PET imaging, when a cut-off percentage of PSMA-positive cells was ≥ 90%, while 19/36 (~53%) lobes with < 90% PSMA-positive cells were PSMA-PET negative. CONCLUSION Positive [68Ga]Ga-PSMA I&T PET/CT scan of primary tumor of PCa results from a combination of factors, such as homogeneity and intensity of PSMA expression, tumor volume and grade, with a cutoff value of ≥ 90% PSMA-positive cells strongly determining PET-positivity. Focal accumulation of [68Ga]Ga-PSMA in the primary tumor may correlate positively with aggressiveness of prostate cancer, harboring higher risk of regional LN involvement and distant metastatic spread.
Collapse
|
41
|
Brada MD, Rushing EJ, Bächinger D, Zoller L, Burger IA, Hüllner MW, Moch H, Huber A, Eckhard AH, Rupp NJ. Immunohistochemical Expression Pattern of Theragnostic Targets SSTR2 and PSMA in Endolymphatic Sac Tumors: A Single Institution Case Series. Head Neck Pathol 2022; 16:1012-1018. [PMID: 35546652 PMCID: PMC9729512 DOI: 10.1007/s12105-022-01456-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 04/16/2022] [Indexed: 12/16/2022]
Abstract
BACKGROUND Endolymphatic sac tumors are rare neoplasia characterized by slow growth. However, their clinical impact should not be underestimated, considering their potential for local aggressive behavior and strong association with von Hippel-Lindau syndrome. Therefore, early detection with emerging theragnostic examinations such as 68Ga-DOTATATE-PET/CT might improve patient management and reduce morbidity. METHODS We report the clinicopathological features of seven endolymphatic sac tumors. In this cohort, we performed immunohistochemical analysis of somatostatin receptor 2A (SSTR2A) and prostate specific membrane antigen (PSMA) protein expression patterns; two targets providing rationale for novel imaging modalities such as PSMA- or SSTR-targeted PET. RESULTS The tumor cells of all cases were negative for prostate specific membrane antigen and somatostatin receptor 2A, however immunolabeling was consistently detected in intratumoral endothelial cells of endolymphatic sac tumors for PSMA (7/7 cases, 100%), and for SSTR2A (5/7 cases, 71%). CONCLUSIONS Our results show a high rate of PSMA and SSTR2A expression in the tumor vasculature of endolymphatic sac tumors. PSMA and SSTR2A can be targeted with appropriate radioligands for diagnostic and therapeutic purposes. This finding provides a rationale for prospective clinical studies to test this approach as a sensitive screening tool for patients with suspected endolymphatic sac tumors including an improved management of von Hippel-Lindau syndrome.
Collapse
Affiliation(s)
- Muriel D. Brada
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland ,Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Elisabeth J. Rushing
- Department of Neuropathology, University Hospital Zurich, Zurich, Switzerland ,Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - David Bächinger
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Zurich, Zurich, Switzerland ,Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Loris Zoller
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Zurich, Zurich, Switzerland ,Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Irene A. Burger
- Department of Nuclear Medicine, Baden Cantonal Hospital, Baden, Switzerland ,Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Martin W. Hüllner
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Holger Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland ,Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Alexander Huber
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Zurich, Zurich, Switzerland ,Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Andreas H. Eckhard
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Zurich, Zurich, Switzerland ,Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Niels J. Rupp
- Department of Pathology and Molecular Pathology, University Hospital Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland ,Faculty of Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
42
|
Laudicella R, Rüschoff JH, Ferraro DA, Brada MD, Hausmann D, Mebert I, Maurer A, Hermanns T, Eberli D, Rupp NJ, Burger IA. Infiltrative growth pattern of prostate cancer is associated with lower uptake on PSMA PET and reduced diffusion restriction on mpMRI. Eur J Nucl Med Mol Imaging 2022; 49:3917-3928. [PMID: 35435496 PMCID: PMC9399036 DOI: 10.1007/s00259-022-05787-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/29/2022] [Indexed: 01/21/2023]
Abstract
Purpose Recently, a significant association was shown between novel growth patterns on histopathology of prostate cancer (PCa) and prostate-specific membrane antigen (PSMA) uptake on [68Ga]PSMA-PET. It is the aim of this study to evaluate the association between these growth patterns and ADC (mm2/1000 s) values in comparison to [68Ga]PSMA uptake on PET/MRI. Methods We retrospectively evaluated patients who underwent [68Ga]PSMA PET/MRI for staging or biopsy guidance, followed by radical prostatectomy at our institution between 07/2016 and 01/2020. The dominant lesion per patient was selected based on histopathology and correlated to PET/MRI in a multidisciplinary meeting, and quantified using SUVmax for PSMA uptake and ADCmean for diffusion restriction. PCa growth pattern was classified as expansive (EXP) or infiltrative (INF) according to its properties of forming a tumoral mass or infiltrating diffusely between benign glands by two independent pathologists. Furthermore, the corresponding WHO2016 ISUP tumor grade was evaluated. The t test was used to compare means, Pearson’s test for categorical correlation, Cohen’s kappa test for interrater agreement, and ROC curve to determine the best cutoff. Results Sixty-two patients were included (mean PSA 11.7 ± 12.5). The interrater agreement between both pathologists was almost perfect with κ = 0.81. While 25 lesions had an EXP-growth with an ADCmean of 0.777 ± 0.109, 37 showed an INF-growth with a significantly higher ADCmean of 1.079 ± 0.262 (p < 0.001). We also observed a significant difference regarding PSMA SUVmax for the EXP-growth (19.2 ± 10.9) versus the INF-growth (9.4 ± 6.2, p < 0.001). Within the lesions encompassing the EXP- or the INF-growth, no significant correlation between the ISUP groups and ADCmean could be observed (p = 0.982 and p = 0.861, respectively). Conclusion PCa with INF-growth showed significantly lower SUVmax and higher ADCmean values compared to PCa with EXP-growth. Within the growth groups, ADCmean values were independent from ISUP grading. Supplementary information The online version contains supplementary material available at 10.1007/s00259-022-05787-9.
Collapse
Affiliation(s)
- Riccardo Laudicella
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Rämistrasse 10, 8091, Zurich, Switzerland
- Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, Nuclear Medicine Unit, University of Messina, Messina, Italy
| | - Jan H Rüschoff
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Daniela A Ferraro
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Rämistrasse 10, 8091, Zurich, Switzerland
- Department of Radiology and Oncology, Faculdade de Medicina, FMUSP, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Muriel D Brada
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Daniel Hausmann
- Department of Radiology, Kantonsspital Baden, Baden, Switzerland
- Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Iliana Mebert
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Rämistrasse 10, 8091, Zurich, Switzerland
- Department of Urology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Alexander Maurer
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Rämistrasse 10, 8091, Zurich, Switzerland
| | - Thomas Hermanns
- Department of Urology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Daniel Eberli
- Department of Urology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Niels J Rupp
- Department of Pathology and Molecular Pathology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Irene A Burger
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Rämistrasse 10, 8091, Zurich, Switzerland.
- Department of Nuclear Medicine, Kantonsspital Baden, Baden, Switzerland.
| |
Collapse
|
43
|
Laudicella R, Skawran S, Ferraro DA, Mühlematter UJ, Maurer A, Grünig H, Rüschoff HJ, Rupp N, Donati O, Eberli D, Burger IA. Quantitative imaging parameters to predict the local staging of prostate cancer in intermediate- to high-risk patients. Insights Imaging 2022; 13:75. [PMID: 35426518 PMCID: PMC9012878 DOI: 10.1186/s13244-022-01217-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/21/2022] [Indexed: 01/16/2023] Open
Abstract
Abstract
Objectives
PSMA PET/MRI showed the potential to increase the sensitivity for extraprostatic disease (EPD) assessment over mpMRI; however, the interreader variability for EPD is still high. Therefore, we aimed to assess whether quantitative PSMA and mpMRI imaging parameters could yield a more robust EPD prediction.
Methods
We retrospectively evaluated PCa patients who underwent staging mpMRI and [68Ga]PSMA-PET, followed by radical prostatectomy at our institution between 01.02.2016 and 31.07.2019. Fifty-eight cases with PET/MRI and 15 cases with PET/CT were identified. EPD was determined on histopathology and correlated with quantitative PSMA and mpMRI parameters assessed by two readers: ADC (mm2/1000 s), longest capsular contact (LCC, mm), tumor volume (cm3), PSMA-SUVmax and volume-based parameters using a fixed threshold at SUV > 4 to delineate PSMAtotal (g/ml) and PSMAvol (cm3). The t test was used to compare means, Pearson’s test for categorical correlation, and ROC curve to determine the best cutoff. Interclass correlation (ICC) was performed for interreader agreement (95% CI).
Results
Seventy-three patients were included (64.5 ± 6.0 years; PSA 14.4 ± 17.1 ng/ml), and 31 had EPD (42.5%). From mpMRI, only LCC reached significance (p = 0.005), while both volume-based PET parameters PSMAtotal and PSMAvol were significantly associated with EPD (p = 0.008 and p = 0.004, respectively). On ROC analysis, LCC, PSMAtotal, and PSMAvol reached an AUC of 0.712 (p = 0.002), 0.709 (p = 0.002), and 0.718 (p = 0.002), respectively. ICC was moderate–good for LCC 0.727 (0.565–0.828) and excellent for PSMAtotal and PSMAvol with 0.944 (0.990–0.996) and 0.985 (0.976–0.991), respectively.
Conclusions
Quantitative PSMA parameters have a similar potential as mpMRI LCC to predict EPD of PCa, with a significantly higher interreader agreement.
Collapse
|
44
|
Diagnostic Performance of Preoperative Choline-PET/CT in Patients Undergoing Salvage Lymph Node Dissection for Recurrent Prostate Cancer: A Multicenter Experience. Tomography 2022; 8:1090-1096. [PMID: 35448723 PMCID: PMC9031841 DOI: 10.3390/tomography8020089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/31/2022] [Accepted: 04/07/2022] [Indexed: 11/25/2022] Open
Abstract
We aimed to retrospectively analyze consecutive prostate cancer patients diagnosed with biochemical or clinical recurrence after local treatment with curative intent, with no evidence of distant metastases, who underwent positron emission tomography/computed tomography (PET/CT) with choline followed by salvage lymph node dissection (SLND) in three academic centers between 2013 and 2020. A total of 27 men were included in the analyses. Sensitivity, specificity, positive and negative predictive values, and accuracy of choline-PET/CT in predicting pathology-proven lymph node involvement were 75%, 43%, 79%, 38% and 67% on per-patient and 70%, 86%, 80%, 78%, and 79% on per-site analyses, respectively, with the differences in specificity and NPV between per-patient and per-site analyses being statistically significant (p = 0.03 and 0.04, respectively). The study provides further insight into the role of preoperative choline-PET/CT in patients undergoing SLND for recurrent PC.
Collapse
|
45
|
Feliciani G, Celli M, Ferroni F, Menghi E, Azzali I, Caroli P, Matteucci F, Barone D, Paganelli G, Sarnelli A. Radiomics Analysis on [68Ga]Ga-PSMA-11 PET and MRI-ADC for the Prediction of Prostate Cancer ISUP Grades: Preliminary Results of the BIOPSTAGE Trial. Cancers (Basel) 2022; 14:cancers14081888. [PMID: 35454793 PMCID: PMC9028386 DOI: 10.3390/cancers14081888] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/01/2022] [Accepted: 04/06/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Radiomics analysis is used on magnetic resonance imaging – apparent diffusion coefficient (MRI-ADC) maps and [68Ga]Ga-PSMA-11 PET uptake maps to assess unique tumor traits not visible to the naked eye and predict histology-proven ISUP grades in a cohort of 28 patients. Our study’s main goal is to report imaging features that can distinguish patients with low ISUP grades from those with higher grades (ISUP one+) by employing logistic regression statistical models based on MRI-ADC and 68Ga-PSMA data, as well as assess the features’ stability under small contouring variations. Our findings reveal that MRI-ADC and [68Ga]Ga-PSMA-11 PET imaging features-based models are equivalent and complementary for predicting low ISUP grade patients. These models can be employed in broader studies to confirm their ISUP grade prediction ability and eventually impact clinical workflow by reducing overdiagnosis of indolent, early-stage PCa. Abstract Prostate cancer (PCa) risk categorization based on clinical/PSA testing results in a substantial number of men being overdiagnosed with indolent, early-stage PCa. Clinically non-significant PCa is characterized as the presence of ISUP grade one, where PCa is found in no more than two prostate biopsy cores.MRI-ADC and [68Ga]Ga-PSMA-11 PET have been proposed as tools to predict ISUP grade one patients and consequently reduce overdiagnosis. In this study, Radiomics analysis is applied to MRI-ADC and [68Ga]Ga-PSMA-11 PET maps to quantify tumor characteristics and predict histology-proven ISUP grades. ICC was applied with a threshold of 0.6 to assess the features’ stability with variations in contouring. Logistic regression predictive models based on imaging features were trained on 31 lesions to differentiate ISUP grade one patients from ISUP two+ patients. The best model based on [68Ga]Ga-PSMA-11 PET returned a prediction efficiency of 95% in the training phase and 100% in the test phase whereas the best model based on MRI-ADC had an efficiency of 100% in both phases. Employing both imaging modalities, prediction efficiency was 100% in the training phase and 93% in the test phase. Although our patient cohort was small, it was possible to assess that both imaging modalities add information to the prediction models and show promising results for further investigations.
Collapse
Affiliation(s)
- Giacomo Feliciani
- Medical Physics Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (E.M.); (A.S.)
- Correspondence: ; Tel.: +39-327-4730398
| | - Monica Celli
- Nuclear Medicine and Radiometabolic Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.C.); (P.C.); (F.M.); (G.P.)
| | - Fabio Ferroni
- Radiology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (F.F.); (D.B.)
| | - Enrico Menghi
- Medical Physics Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (E.M.); (A.S.)
| | - Irene Azzali
- Biostatistics and Clinical Trials Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Paola Caroli
- Nuclear Medicine and Radiometabolic Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.C.); (P.C.); (F.M.); (G.P.)
| | - Federica Matteucci
- Nuclear Medicine and Radiometabolic Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.C.); (P.C.); (F.M.); (G.P.)
| | - Domenico Barone
- Radiology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (F.F.); (D.B.)
| | - Giovanni Paganelli
- Nuclear Medicine and Radiometabolic Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.C.); (P.C.); (F.M.); (G.P.)
| | - Anna Sarnelli
- Medical Physics Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (E.M.); (A.S.)
| |
Collapse
|
46
|
Rosenzweig B, Haramaty R, Davidson T, Lazarovich A, Shvero A, Haifler M, Gal J, Golan S, Shpitzer S, Hoffman A, Nativ O, Freifeld Y, Zreik R, Dotan ZA. Very Low Prostate PET/CT PSMA Uptake May Be Misleading in Staging Radical Prostatectomy Candidates. J Pers Med 2022; 12:jpm12030410. [PMID: 35330410 PMCID: PMC8951096 DOI: 10.3390/jpm12030410] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 02/26/2022] [Accepted: 03/02/2022] [Indexed: 02/05/2023] Open
Abstract
Purpose: to evaluate a unique subpopulation of radical prostatectomy (RP) candidates with “negative” prostate 68Ga-labeled prostate-specific membrane antigen (PSMA) positron emission tomography (PET) computed tomography (CT) imaging scans and to characterize the clinical implications of misleading findings. Materials and Methods: This case-control retrospective study compared the final histological outcomes of patients with “negative” pre-RP PSMA PET/CT prostate scans (with a prostate maximal standardized uptake value [SUVmax] below the physiologic uptake) to those with an “intense” prostatic tracer uptake (with a SUVmax above the physiologic uptake). The patients underwent an RP between March 2015 and July 2019 in five academic centers. Data on the demographics, comorbidities, prostate-specific antigen (PSA) and rectal exam findings, prior biopsies, imaging results, biopsies, and RP histology results were collected. Results: Ninety-seven of the 392 patients who underwent an RP had PSMA PET/CT imaging preoperatively. Fifty-two (54%) had a “negative” uptake (in the study group), and 45 (46%) had a “positive” uptake (in the control group). Only the lesion size and SUVmax values on the PSMA PET/CT differed between the groups preoperatively. On the histological analysis, only the ISUP score, seminal vesicles invasion, T stage, and positive margin rates differed between the groups (p < 0.05), while 50 (96%) study group patients harbored clinically significant disease (ISUP ≥ 2), with an extra-prostatic disease in 24 (46%), perineural invasion in 35 (67%), and positive lymph nodes in 4 (8%). Conclusions: Disease aggressiveness generally correlated with an intense PSMA uptake on the preoperative PSMA PET/CT, but a subpopulation of patients with clinically significant cancer and aggressive characteristics showed a deceptively weak PSMA uptake. These data raise a concern about the unqualified application of PSMA PET/CT for staging RP candidates.
Collapse
Affiliation(s)
- Barak Rosenzweig
- Department of Urology, Chaim Sheba Medical Center, Ramat Gan 5211401, Israel; (R.H.); (A.L.); (A.S.); (Z.A.D.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (T.D.); (M.H.); (S.G.)
- Correspondence: ; Tel.: +972-3-5302221
| | - Rennen Haramaty
- Department of Urology, Chaim Sheba Medical Center, Ramat Gan 5211401, Israel; (R.H.); (A.L.); (A.S.); (Z.A.D.)
| | - Tima Davidson
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (T.D.); (M.H.); (S.G.)
- Department of Nuclear Medicine, Chaim Sheba Medical Center, Ramat Gan 5211401, Israel
| | - Alon Lazarovich
- Department of Urology, Chaim Sheba Medical Center, Ramat Gan 5211401, Israel; (R.H.); (A.L.); (A.S.); (Z.A.D.)
| | - Asaf Shvero
- Department of Urology, Chaim Sheba Medical Center, Ramat Gan 5211401, Israel; (R.H.); (A.L.); (A.S.); (Z.A.D.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (T.D.); (M.H.); (S.G.)
| | - Miki Haifler
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (T.D.); (M.H.); (S.G.)
- Department of Urology, Shamir Medical Center, Tzrifin 6093000, Israel;
| | - Jonathan Gal
- Department of Urology, Shamir Medical Center, Tzrifin 6093000, Israel;
| | - Shay Golan
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (T.D.); (M.H.); (S.G.)
- Section of Urology, Rabin Medical Center, Petah Tikva 4941492, Israel;
| | - Sagi Shpitzer
- Section of Urology, Rabin Medical Center, Petah Tikva 4941492, Israel;
| | - Azik Hoffman
- Department of Urology, Rambam Health Center, Haifa 3109601, Israel; (A.H.); (O.N.)
- Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa 3200003, Israel; (Y.F.); (R.Z.)
| | - Omri Nativ
- Department of Urology, Rambam Health Center, Haifa 3109601, Israel; (A.H.); (O.N.)
| | - Yuval Freifeld
- Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa 3200003, Israel; (Y.F.); (R.Z.)
- Department of Urology, Carmel Medical Center, Haifa 3436212, Israel
| | - Rani Zreik
- Ruth and Bruce Rappaport Faculty of Medicine, Technion Israel Institute of Technology, Haifa 3200003, Israel; (Y.F.); (R.Z.)
| | - Zohar A. Dotan
- Department of Urology, Chaim Sheba Medical Center, Ramat Gan 5211401, Israel; (R.H.); (A.L.); (A.S.); (Z.A.D.)
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel; (T.D.); (M.H.); (S.G.)
| |
Collapse
|
47
|
Vlachostergios PJ, Niaz MJ, Thomas C, Christos PJ, Osborne JR, Margolis DJA, Khani F, Bander NH, Scherr DS, Tagawa ST. Pilot study of the diagnostic utility of 89 Zr-df-IAB2M and 68 Ga-PSMA-11 PET imaging and multiparametric MRI in localized prostate cancer. Prostate 2022; 82:483-492. [PMID: 34985786 DOI: 10.1002/pros.24294] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 12/14/2021] [Accepted: 12/17/2021] [Indexed: 01/06/2023]
Abstract
BACKGROUND Accurate diagnosis of localized prostate cancer (PCa) is limited by inadequacy of multiparametric (mp) MRI to fully identify and differentiate localized malignant tissue from benign pathologies. Prostate-specific membrane antigen (PSMA) represents an excellent target for molecular imaging. IAB2M, an 85-kD minibody derived from a de-immunized monoclonal antibody directed at the extracellular domain of human PSMA (huJ591), and PSMA-11, a small molecule ligand have been previously tested as probes for visualization of recurrent/metastatic PCa with PET/CT. This pilot, non-randomized trial studied their diagnostic utility in patients (pts) with localized PCa. METHODS Pts planned for radical prostatectomy (RP) were enrolled and underwent mpMRI and PET/CT imaging with 89 Zr-df-IAB2M and/or 68 Ga-PSMA-PET/CT. Image results were read by a radiologist blinded to clinical information and pathology results, mapped and compared to corresponding histopathology findings from all lesions, both clinically significant and nonsignificant. The detection rates of all three imaging modalities were measured and correlated. RESULTS 20 pts with median age of 64.5 (46-79) years and PSA level of 7.5 (1.6-36.56) ng/ml were enrolled. 19 pts underwent RP and were imaged pre-operatively with 89 Zr-Df-IAB2M PET/CT and mpMRI. Nine of those were imaged using 68 Ga-PSMA-11 as well. Out of 48 intraprostatic lesions verified on surgical pathology, IAB2M PET/CT was able to detect 36 (75%). A similar proportion of pathologically confirmed, clinically significant lesions (22/29, 76%) was detected. IAB2M PET/CT was also able to identify 14/19 (74%) extraprostatic lesions. The performance of mpMRI was inferior, with 24/48 detectable lesions (50%) and 18/29 clinically significant intraprostatic lesions (62%). Compared to the current standard (mpMRI), IAB2M PET/CT had a sensitivity of 88%, specificity 38%, positive predictive value 58%, and accuracy 63%. In 9 pts who underwent Ga-PSMA-11 as well, the latter yielded a detection rate of 70% (14/20), which was also seen in clinically significant lesions (10/14, 71%). Ga-PSMA-11 PET/CT also detected 4/6 (67%) extraprostatic lesions. CONCLUSIONS In this pilot study, the performance of 89 Zr-df-IAB2M was superior to mpMRI and similar to 68 Ga-PSMA-11 PET/CT. The higher detection rate of PSMA-PET supports its use as a diagnostic tool with consequent management change implications in men with localized PCa.
Collapse
Affiliation(s)
- Panagiotis J Vlachostergios
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Muhammad J Niaz
- Department of Urology, Weill Cornell Medicine, New York, New York, USA
| | - Charlene Thomas
- Division of Biostatistics, Department of Population Health Sciences, Weill Cornell Medicine, New York, New York, USA
| | - Paul J Christos
- Division of Biostatistics, Department of Population Health Sciences, Weill Cornell Medicine, New York, New York, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York, USA
| | - Joseph R Osborne
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York, USA
- Division of Molecular Imaging and Therapeutics, Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| | - Daniel J A Margolis
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York, USA
- Division of Body Imaging, Department of Radiology, Weill Cornell Medicine, New York, New York, USA
| | - Francesca Khani
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Neil H Bander
- Department of Urology, Weill Cornell Medicine, New York, New York, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York, USA
| | - Douglas S Scherr
- Department of Urology, Weill Cornell Medicine, New York, New York, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York, USA
| | - Scott T Tagawa
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, New York, USA
- Department of Urology, Weill Cornell Medicine, New York, New York, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
48
|
Zhou C, Tang Y, Deng Z, Yang J, Zhou M, Wang L, Hu S. Comparison of 68Ga-PSMA PET/CT and multiparametric MRI for the detection of low- and intermediate-risk prostate cancer. EJNMMI Res 2022; 12:10. [PMID: 35147810 PMCID: PMC8837766 DOI: 10.1186/s13550-022-00881-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 01/25/2022] [Indexed: 11/21/2022] Open
Abstract
Purpose To assess 68Ga-PSMA PET/CT for detection of low- and intermediate-risk prostate cancer (PCa), high-risk PCa in comparison with mpMRI, respectively, and to determine which of low- and intermediate-risk PCa are more likely to be detected by 68Ga-PSMA PET/CT. Methods We conducted a retrospective analysis of patients who had undergone a prostate biopsy and/or radical prostatectomy and who were scanned with 68Ga-PSMA PET/CT and mpMRI between June 2019 and March 2021. The mpMRI images were scored with the Prostate Imaging-Reporting and Data System Version 2.1 (PI-RADS)
and were classified as either negative (PI-RADS 1–3) or positive (PI-RADS 4–5). Suspicious 68Ga-PSMA PET/CT lesions were reviewed for each relevant patient and classified by double-trained board-certified nuclear medicine physicians. The results were evaluated with the histopathological outcome. All patients were classified according to the D’Amico classification, and the clinical data were combined for stratified analysis. Result A total of 101 patients who were pathologically diagnosed with PCa were analyzed. Of the 101 patients, 88 (80.6%) patients presented with a pathologic mpMRI, and 85 (79.1%) with a pathologic 68Ga-PSMA PET/CT. In the high-risk PCa cohort, 68Ga-PSMA PET/CT was positive in 64/66 (97.0%) patients and yielded a higher detection rate than that for the mpMRI patients (58/66, 87.9%; p < 0.05). However, mpMRI provided superior diagnostic confidence in identifying low- and intermediate-risk PCa (30/35, 85.7% vs. 21/35, 60.0%; p < 0.05). When the age threshold exceeded 62.5 years and the serum prostate specific antigen (PSA) threshold exceeded 9.4 ng/ml, a higher uptake of PSMA was more likely to occur in the lesions of low- and intermediate-risk PCa. Conclusion The diagnostic performance of 68Ga-PSMA PET/CT was superior to that of mpMRI in the high-risk PCa cohort, which was consistent with prior studies. Furthermore, in the initial diagnosis of low- and intermediate-risk PCa, we found that mpMRI showed a higher diagnostic accuracy than 68Ga-PSMA PET/CT did. Low- and intermediate-risk PCa patients with a PSA ≥ 9.4 ng/ml and age ≥ 62.5 years were more likely to have a positive 68Ga-PSMA PET/CT result. Supplementary Information The online version contains supplementary material available at 10.1186/s13550-022-00881-3.
Collapse
Affiliation(s)
- Chuanchi Zhou
- Department of Urology, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan, China.,Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yongxiang Tang
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Zhihe Deng
- Department of Vascular Surgery, The Second Affiliated Hospital of University of South China, Hengyang, China
| | - Jinhui Yang
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Ming Zhou
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Long Wang
- Department of Urology, The Third Xiangya Hospital, Central South University, 138 Tongzipo Road, Changsha, 410013, Hunan, China.
| | - Shuo Hu
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China. .,Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,National Clinical Research Center for Geriatric Disorders (XIANGYA), Changsha, China.
| |
Collapse
|
49
|
Qiu DX, Li J, Zhang JW, Chen MF, Gao XM, Tang YX, Zhang Y, Yi XP, Yin HL, Gan Y, Wang GL, Zu XB, Hu S, Cai Y. Dual-tracer PET/CT-targeted, mpMRI-targeted, systematic biopsy, and combined biopsy for the diagnosis of prostate cancer: a pilot study. Eur J Nucl Med Mol Imaging 2021; 49:2821-2832. [PMID: 34860277 DOI: 10.1007/s00259-021-05636-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 11/24/2021] [Indexed: 12/21/2022]
Abstract
PURPOSE Growing evidence proved the efficacy of multi-parametric MRI (mpMRI) and prostate-specific membrane antigen (PSMA) positron emission tomography/computed tomography (PET/CT)-guided targeted biopsy (TB) in prostate cancer (PCa) diagnosis, but there is no direct comparison between mpMRI-TB and PSMA PET/CT-TB. Gastrin-releasing peptide receptor (GRPR) is highly expressed in PCa, which can compensate for the unstable expression of PSMA in PCa. Therefore, we designed a study to compare the efficiency of mpMRI-TB, dual-tracer (GRPR and PSMA) PET/CT-TB, systematic biopsy, and combined biopsy for the diagnosis of prostate cancer. METHODS One hundred twelve suspicious PCa patients were enrolled from September 2020 to June 2021. Patients with anyone of positive dual-tracer PET/CT or mpMRI underwent TB, and all enrolled patients underwent systematic biopsy (SB) after TB. The primary outcome was the detection rates of PCa in different biopsy strategies. Secondary outcomes were the performance of three imaging methods, omission diagnostic rates, and upgrading and downgrading of biopsy samples relative to those of prostatectomy specimens in different biopsy strategies. McNemar's tests and Bonferroni correction in multiple comparisons were used to compare the primary and secondary outcomes. RESULTS In 112 men, clinically significant PCa (grade group[GG] ≥ 2) accounted for 34.82% (39/112), and nonclinically significant PCa (GG = 1) accounted for 4.46% (5/112). 68 Ga-PSMA PET/CT-TB achieved higher PCa detection rate (69.77%) and positive ratio of biopsy cores (0.44) compared with SB (39.29% and 0.12) and mpMRI-TB (36.14% and 0.23), respectively (P < 0.005). Dual-tracer PET/CT screen out patients for avoiding 52.67% (59/112) unnecessary biopsy, whereas dual-tracer PET/CT-TB plus SB achieved high detection rate (77.36%) without misdiagnosis of csPCa. CONCLUSION Dual-tracer PET/CT might screen patients for avoiding unnecessary biopsy. Dual-tracer PET/CT-TB plus SB might be a more effective and promising strategy for the definite diagnosis of clinically significant PCa than mpMRI-TB.
Collapse
Affiliation(s)
- Dong-Xu Qiu
- Department of Urology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Jian Li
- Department of PET Center, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Jin-Wei Zhang
- Department of Radiology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Min-Feng Chen
- Department of Urology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Xiao-Mei Gao
- Department of Pathology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Yong-Xiang Tang
- Department of PET Center, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Ye Zhang
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Xiao-Ping Yi
- Department of Radiology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Hong-Ling Yin
- Department of Pathology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Yu Gan
- Department of Urology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Gui-Lin Wang
- Department of Urology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Xiong-Bing Zu
- Department of Urology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China
| | - Shuo Hu
- Department of PET Center, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
| | - Yi Cai
- Department of Urology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
| |
Collapse
|
50
|
Calais J, Czernin J. PSMA Expression Assessed by PET Imaging Is a Required Biomarker for Selecting Patients for Any PSMA-Targeted Therapy. J Nucl Med 2021; 62:1489-1491. [PMID: 34725231 PMCID: PMC8612346 DOI: 10.2967/jnumed.121.263159] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- Jeremie Calais
- David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Johannes Czernin
- David Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|