1
|
Henríquez I, Malave B, Campos FL, Hidalgo EC, Muelas R, Ferrer C, Muñoz-Rodriguez J, Villamón AM, Pascual MC, Badia J, Fuertes J, Hinojosa-Salas P. PSMA PET/CT SUVmax as a prognostic biomarker in patients with metachronous metastatic hormone-sensitive prostate cancer (mHSPC). Clin Transl Oncol 2024:10.1007/s12094-024-03625-y. [PMID: 39073734 DOI: 10.1007/s12094-024-03625-y] [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: 04/08/2024] [Accepted: 07/19/2024] [Indexed: 07/30/2024]
Abstract
BACKGROUND Metastatic hormone-sensitive prostate cancer (mHSPC) is treatment-resistant and generally considered incurable. The development of prostate-specific membrane antigen positron emission-computed tomography (PSMA PET/CT) has generated immense expectations due to its diagnostic accuracy in prostate cancer (PCa). PSMA expression of the primary tumor, quantified by SUVmax, is a predictor of oncological outcomes. The role of PSMA-PET/CT SUVmax in metachronous mHSPC treated with ADT plus second-generation antiandrogens (ARSI) is unknown. The main aim of this study was to evaluate 68Ga-PSMA-11expression (SUVmax) as a potential prognostic biomarker in patients with metachronous mHSPC treated with ADT and first or second-generation antiandrogens. A second aim was to determine the association between PSMA SUVmax and PSA response to hormone therapy. MATERIAL AND METHODS Patients diagnosed with metachronous mHSPC between July 2017 and February 2023 who developed biochemical recurrence following radical surgery (with or without salvage radiotherapy and/or ADT) or external radiation therapy (with or without ADT) were included. All patients underwent 68 Ga-PSMA-11 PET/CT imaging and the SUVmax value was determined for all measurable locations. The SUVmax value was used for the semiquantitative analysis. The Wilcoxon method was used to compare responders (PSA reduction ≥ 50%) to non-responders (PSA reduction < 50%). The SUVmax value and hormone therapy were evaluated as independent variables relative to the PSA response rate or PSA reduction using the linear regression method. A mixed-effects model (ANOVA) was used for the comparisons. RESULTS A total of 82 patients were included. Median follow-up was 11.7 months. On the linear regression analysis, patients with a high SUVmax treated with ADT + ARSI showed a greater PSA response (p = 0.034) than those treated with ADT + first-generation antiandrogens. In the mixed-effects model, SUVmax was significant (p = 0.041). On the univariate analysis, PSA at recurrence (HR, 3.2; 95% CI: 1.07-13.6; p = 0.078) and the number of metastases (HR, 4.77; 95% CI 1.1-26.1: p = 0.002) were associated with the type of hormone therapy administered. CONCLUSIONS PSMA-PET/CT SUVmax is a prognostic biomarker that can be used to predict a PSA response to ADT + ARSI in patients with metachronous mHSPC. However, these findings need to be confirmed in larger prospective studies.
Collapse
Affiliation(s)
- Iván Henríquez
- Department of Radiation Oncology, Hospital Universitario Sant Joan, Pere i Virgili Health Research Institute (IISPV), Reus, Spain.
| | - Bárbara Malave
- Department of Radiation Oncology, Hospital Universitario Sant Joan, Pere i Virgili Health Research Institute (IISPV), Reus, Spain
| | | | | | - Rodrigo Muelas
- Department of Radiation Oncology. Hospital Provincial Castellón, Valencia, Spain
| | - Carlos Ferrer
- Department of Radiation Oncology. Hospital Provincial Castellón, Valencia, Spain
| | | | | | | | - Joan Badia
- Statistical Support Platform. Pere I Virgili Health Research Institute (IISPV), Instituto de Oncología de La Cataluña Sud (IOCS), Reus, Spain
| | - Jordi Fuertes
- Nuclear Medicine Department. Hospital, Universitario Sant Joan, Reus, Spain
| | | |
Collapse
|
2
|
Pijeira MSO, Nunes PSG, Chaviano SL, Diaz AMA, DaSilva JN, Ricci-Junior E, Alencar LMR, Chen X, Santos-Oliveira R. Medicinal (Radio) Chemistry: Building Radiopharmaceuticals for the Future. Curr Med Chem 2024; 31:5481-5534. [PMID: 37594105 DOI: 10.2174/0929867331666230818092634] [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: 03/10/2023] [Revised: 05/30/2023] [Accepted: 07/13/2023] [Indexed: 08/19/2023]
Abstract
Radiopharmaceuticals are increasingly playing a leading role in diagnosing, monitoring, and treating disease. In comparison with conventional pharmaceuticals, the development of radiopharmaceuticals does follow the principles of medicinal chemistry in the context of imaging-altered physiological processes. The design of a novel radiopharmaceutical has several steps similar to conventional drug discovery and some particularity. In the present work, we revisited the insights of medicinal chemistry in the current radiopharmaceutical development giving examples in oncology, neurology, and cardiology. In this regard, we overviewed the literature on radiopharmaceutical development to study overexpressed targets such as prostate-specific membrane antigen and fibroblast activation protein in cancer; β-amyloid plaques and tau protein in brain disorders; and angiotensin II type 1 receptor in cardiac disease. The work addresses concepts in the field of radiopharmacy with a special focus on the potential use of radiopharmaceuticals for nuclear imaging and theranostics.
Collapse
Affiliation(s)
- Martha Sahylí Ortega Pijeira
- Laboratory of Nanoradiopharmaceuticals and Synthesis of Novel Radiopharmaceuticals, Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Rio de Janeiro 21941906, Brazil
| | - Paulo Sérgio Gonçalves Nunes
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas SP13083-970, Brazil
| | - Samila Leon Chaviano
- Laboratoire de Biomatériaux pour l'Imagerie Médicale, Axe Médicine Régénératrice, Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, QC, Canada
| | - Aida M Abreu Diaz
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
- Institute de Génie Biomédical, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Jean N DaSilva
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
- Institute de Génie Biomédical, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Eduardo Ricci-Junior
- Laboratório de Desenvolvimento Galênico, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Luciana Magalhães Rebelo Alencar
- Laboratory of Biophysics and Nanosystems, Federal University of Maranhão, Av. dos Portugueses, 1966, Vila Bacanga, São Luís MA65080-805, Brazil
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore 117597, Singapore
| | - Ralph Santos-Oliveira
- Laboratory of Nanoradiopharmaceuticals and Synthesis of Novel Radiopharmaceuticals, Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Rio de Janeiro 21941906, Brazil
- Laboratory of Radiopharmacy and Nanoradiopharmaceuticals, Rio de Janeiro State University, Rio de Janeiro 23070200, Brazil
| |
Collapse
|
3
|
Hu X, Cao Y, Ji B, Zhao M, Wen Q, Chen B. Comparative study of 18F-DCFPyL PET/CT and 99mTc-MDP SPECT/CT bone imaging for the detection of bone metastases in prostate cancer. Front Med (Lausanne) 2023; 10:1201977. [PMID: 37588003 PMCID: PMC10425766 DOI: 10.3389/fmed.2023.1201977] [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: 04/07/2023] [Accepted: 06/27/2023] [Indexed: 08/18/2023] Open
Abstract
Purpose This study aimed to compare the diagnostic efficiency of 18F-DCFPyL PET/CT imaging and 99mTc-MDP SPECT/CT bone imaging for the detection of bone metastases in prostate cancer. Methods A retrospective analysis was conducted on 31 patients with confirmed prostate cancer between September 2020 and September 2022 at China-Japan Union Hospital of Jilin University. All patients underwent 18F-DCFPyL PET/CT and 99mTc-MDP SPECT/CT bone imaging. The gold standard was the pathology or Best Valuable Comparator (BVC) result based on clinical follow-up. Diagnostic performance indicators, including sensitivity, specificity, accuracy, positive predictive value (PPV), and negative predictive value (NPV), were analyzed at both the patient and lesion levels. The paired sample chi-square test was used to compare the two imaging methods. Receiver operating characteristic (ROC) curves were plotted, and the area under the curve (AUC) was calculated for each method. The AUC values were compared using the Z-test, and a p-value < 0.05 was considered statistically significant. Results Of the 31 prostate cancer patients, 18 were diagnosed with bone metastases, with a total of 84 bone metastatic lesions. At the patient level, 18F-DCFPyL PET/CT imaging showed superior diagnostic performance compared to 99mTc-MDP SPECT/CT bone imaging in all indicators: sensitivity (100% vs. 77.8%, p < 0.01), specificity (92.3% vs. 69.2%, p < 0.05), accuracy (96.8% vs. 74.2%, p < 0.01), PPV (94.7% vs. 77.8%, p < 0.01), and NPV (100% vs. 69.2%, p < 0.01). The AUC values for 18F-DCFPyL PET/CT imaging and 99mTc-MDP SPECT/CT bone imaging were 0.962 and 0.735 (Z = 2.168, p < 0.05). At the lesion level, 18F-DCFPyL PET/CT imaging showed superior diagnostic performance compared to 99mTc-MDP SPECT/CT bone imaging in all indicators: sensitivity (97.6% vs. 72.6%, p < 0.01), specificity (95.7% vs. 73.9%, p < 0.01), accuracy (97.2% vs. 72.9%, p < 0.01), PPV (98.8% vs. 91.0%, p < 0.01), and NPV (91.7% vs. 42.5%, p < 0.01). The AUC values for 18F-DCFPyL PET/CT imaging and 99mTc-MDP SPECT/CT bone imaging were 0.966 and 0.733 (Z = 3.541, p < 0.001). Conclusion Compared with 99mTc-MDP SPECT/CT bone imaging, 18F-DCFPyL PET/CT imaging demonstrated higher diagnostic efficiency for bone metastases in prostate cancer, and it can more accurately determine the presence of bone metastases. It is an important supplement to imaging examination for prostate cancer patients and has great potential and broad application prospects.
Collapse
Affiliation(s)
| | | | | | | | - Qiang Wen
- Department of Nuclear Medicine, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Bin Chen
- Department of Nuclear Medicine, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
4
|
Hill S, Kassam F, Verma S, Sidana A. Traditional and novel imaging modalities for advanced prostate cancer: A critical review. Urol Ann 2023; 15:249-255. [PMID: 37664103 PMCID: PMC10471808 DOI: 10.4103/ua.ua_170_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 04/26/2021] [Indexed: 09/05/2023] Open
Abstract
Accurate detection of metastatic prostate cancer in the setting of preoperative staging as well as posttreatment recurrence is crucial to provide patients with appropriate and timely treatment of their disease. This has traditionally been accomplished with a combination of computed tomography, magnetic resonance imaging, and bone scan. Recently, more novel imaging techniques have been developed to help improve the detection of advanced and metastatic prostate cancer. This review discusses the efficacy of the traditional imaging modalities as well as the novel imaging techniques in detecting metastatic prostate cancer. Articles discussed were gathered through a formal PubMed search.
Collapse
Affiliation(s)
- Spencer Hill
- Department of Urology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Farzaan Kassam
- Department of Urology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Sadhna Verma
- Department of Urology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Abhinav Sidana
- Department of Urology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| |
Collapse
|
5
|
Deng H, Wang Y, Zhou Y, Zhai D, Chen J, Hao S, Chen X. In vitro and in vivo Evaluation of Folic Acid Modified DOX-Loaded 32P-nHA Nanoparticles in Prostate Cancer Therapy. Int J Nanomedicine 2023; 18:2003-2015. [PMID: 37077940 PMCID: PMC10108875 DOI: 10.2147/ijn.s403887] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/24/2023] [Indexed: 04/21/2023] Open
Abstract
Background Prostate cancer (PCa) ranks second in the incidence of all malignancies in male worldwide. The presence of multi-organ metastases and tumor heterogeneity often leads to unsatisfactory outcomes of conventional radiotherapy treatments. This study aimed to develop a novel folate-targeted nanohydroxyapatite (nHA) coupling to deliver adriamycin (Doxorubicin, DOX), 32P, and 99mTc simultaneously for the diagnosis and treatment of prostate-specific membrane antigen (PSMA) positive prostate cancer. Methods The spherical nHA was prepared by the biomimetic method and characterized. Folic acid (FA) was coupled to nHA with polyethylene glycol (PEG), and the grafting ratio of PEG-nHA and FA-PEG-nHA was determined by the thermogravimetric analysis (TGA) method. In addition, 32P, 99mTc, and DOX were loaded on nHA by physisorption. And the labeling rate and stability of radionuclides were measured by a γ-counter. The loading and release of DOX at different pH were determined by the dialysis method. Targeting of FA-PEG-nHA loaded with 99mTc was verified by in vivo SPECT imaging. In vitro anti-tumor effect of 32P/DOX-FA-PEG-nHA was assessed with apoptosis assay. The safety of the nano-drugs was verified by histopathological analysis. Results The SEM images showed that the synthesized nHA was spherical with uniform particle size (average diameter of about 100nm). The grafting ratio is about 10% for PEG and about 20% for FA. The drug loading and the delayed release of DOX at different pH confirmed its long-term therapeutic ability. The labeling of 32P and 99mTc was stable and the labeling rate was great. SPECT showed that FA-PEG-nHA showed well in vivo tumor targeting and less damage to normal tissues. Conclusion FA-targeted nHA loaded with 32P, 99mTc, and DOX may be a new diagnostic and therapeutic strategy for targeting PSMA-positive prostate cancer tumors, which may achieve better therapeutic results while circumventing the severe toxic side effects of conventional chemotherapeutic agents.
Collapse
Affiliation(s)
- Hao Deng
- Department of Nuclear Medicine, Chongqing University Cancer Hospital, Chongqing, 400030, People’s Republic of China
| | - Yumei Wang
- Department of Nuclear Medicine, Chongqing University Cancer Hospital, Chongqing, 400030, People’s Republic of China
| | - Yue Zhou
- Department of Nuclear Medicine, Chongqing University Cancer Hospital, Chongqing, 400030, People’s Republic of China
| | - Dongliang Zhai
- Department of Nuclear Medicine, Chongqing University Cancer Hospital, Chongqing, 400030, People’s Republic of China
| | - Jie Chen
- Department of Nuclear Medicine, Chongqing University Cancer Hospital, Chongqing, 400030, People’s Republic of China
| | - Shilei Hao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400030, People’s Republic of China
- Shilei Hao, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, 174 Shazhengjie, Shapingba District, Chongqing, 400030, People’s Republic of China, Tel +86023-135 9463 5765, Email
| | - Xiaoliang Chen
- Department of Nuclear Medicine, Chongqing University Cancer Hospital, Chongqing, 400030, People’s Republic of China
- Correspondence: Xiaoliang Chen, Department of Nuclear Medicine, Chongqing University Cancer Hospital, No. 181 HanYu St, Shapingba District, Chongqing, 400030, People’s Republic of China, Tel/Fax +86 023-65079156, Email
| |
Collapse
|
6
|
Bukavina L, Luckenbaugh AN, Hofman MS, Hope T, Kamran SC, Murphy DG, Yamoah K, Ost P. Incorporating Prostate-specific Membrane Antigen Positron Emission Tomography in Management Decisions for Men with Newly Diagnosed or Biochemically Recurrent Prostate Cancer. Eur Urol 2022; 83:521-533. [PMID: 36404204 DOI: 10.1016/j.eururo.2022.10.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/15/2022] [Accepted: 10/28/2022] [Indexed: 11/19/2022]
Abstract
CONTEXT Prostate-specific membrane antigen (PSMA) is a promising molecular target for prostate cancer (PCa) that has allowed the development of a novel diagnostic approach to PCA in the primary and recurrent settings. OBJECTIVE To summarize available data and recommendations regarding the use of PSMA in newly diagnosed and recurrent PCa via a narrative review. EVIDENCE ACQUISITION A literature review was conducted using MEDLINE (via PubMed) and Scopus. The search strategy included meta-analyses, reviews, and original studies on staging and restaging with 68Ga-PSMA positron emission tomography (PET)/computed tomography (CT). EVIDENCE SYNTHESIS Studies comparing PSMA-targeted imaging and conventional imaging suggest superior performance of PSMA-targeted imaging in primary and recurrent PCa, albeit with several clinically relevant limitations. Pretreatment 68Ga-PSMA PET/CT allowed more accurate PCa staging in compared to routine practice for high-risk cases, and identified a number of otherwise unknown metastatic lesions. In biochemically recurrent PCa, PSMA PET can reveal sites of recurrence with greater sensitivity and specificity than conventional imaging, potentially detecting a major proportion of occult disease. This review will help providers in applying the most up-to-date and relevant literature to (1) determine which patients truly have oligometastatic disease and (2) ascertain who is most likely to experience a meaningful response to local consolidation in the biochemical recurrence setting. CONCLUSIONS Data on PSMA diagnostic studies in primary and recurrent PCa highlight the accuracy and clinical application of PSMA PET. While this review and the evidence to date might lead to a perception of superiority in metastasis directed therapy, fundamental lack of phase III clinical trials with clinically meaningful outcomes are yet to be determined. PATIENT SUMMARY PSMA (prostate-specific membrane antigen) scans have shown great promise for initial evaluation of prostate cancer (PCa) and in detection of PCa recurrence. The benefits are more apparent for initial staging of PCa. There are more limited clinical trial results for PCa recurrence on how best to use this new technique to guide cancer treatment.
Collapse
|
7
|
Rowe SP, Salavati A, Werner RA, Pienta KJ, Gorin MA, Pomper MG, Solnes LB. 18F-Labeled Radiotracers for Prostate-specific Membrane Antigen. PET Clin 2022; 17:585-593. [DOI: 10.1016/j.cpet.2022.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
8
|
Hu C, Dong L, Xue W, Pienta KJ. Prostate-Specific Membrane Antigen-Based PET Brings New Insights into the Management of Prostate Cancer. PET Clin 2022; 17:555-564. [PMID: 36153235 DOI: 10.1016/j.cpet.2022.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Prostate cancer (PCa) is the third most common cancer diagnosed in the world. Since its first identification in 1987 and its first molecular cloning in 1993, prostate-specific membrane antigen (PSMA) has been developed as a theragnostic imaging biomarker and therapeutic agent for PCa. For metastatic castration-resistant PCa, PSMA-based PET imaging can be applied to the monitoring of disease and response assessment with PSMA-based therapeutics. This novel imaging modality is bringing new insights into diagnosis, stratification, and clinical decision-making and treatment.
Collapse
Affiliation(s)
- Cong Hu
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai 200127, China
| | - Liang Dong
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai 200127, China
| | - Wei Xue
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai 200127, China
| | - Kenneth J Pienta
- Department of Urology, The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Baltimore, MD 21287, USA.
| |
Collapse
|
9
|
Chen J, Qi L, Tang Y, Tang G, Gan Y, Cai Y. Current role of prostate-specific membrane antigen-based imaging and radioligand therapy in castration-resistant prostate cancer. Front Cell Dev Biol 2022; 10:958180. [PMID: 36036001 PMCID: PMC9411749 DOI: 10.3389/fcell.2022.958180] [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: 05/31/2022] [Accepted: 07/11/2022] [Indexed: 11/29/2022] Open
Abstract
Castration-resistant prostate cancer (CRPC) is a therapy-resistant and lethal form of prostate cancer as well as a therapeutic challenge. Prostate-specific membrane antigen (PSMA) has been proved as a promising molecular target for optimizing the theranostics for CRPC patients. When combined with PSMA radiotracers, novel molecular imaging techniques such as positron emission tomography (PET) can provide more accurate and expedient identification of metastases when compared with conventional imaging techniques. Based on the PSMA-based PET scans, the accurate visualization of local and disseminative lesions may help in metastasis-directed therapy. Moreover, the combination of 68Ga-labeled PSMA-based PET imaging and radiotherapy using PSMA radioligand therapy (RLT) becomes a novel treatment option for CRPC patients. The existing studies have demonstrated this therapeutic strategy as an effective and well-tolerated therapy among CRPC patients. PSMA-based PET imaging can accurately detect CRPC lesions and describe their molecular features with quantitative parameters, which can be used to select the best choice of treatments, monitor the response, and predict the outcome of RLT. This review discussed the current and potential role of PSMA‐based imaging and RLT in the diagnosis, treatment, and prediction of prognosis of CRPC.
Collapse
Affiliation(s)
- Jiaxian Chen
- Department of Urology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Lin Qi
- Department of Urology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yongxiang Tang
- Department of PET Center, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Guyu Tang
- Department of Urology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Yu Gan
- Department of Urology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- *Correspondence: Yu Gan, ; Yi Cai,
| | - Yi Cai
- Department of Urology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, China
- *Correspondence: Yu Gan, ; Yi Cai,
| |
Collapse
|
10
|
Mena E, Rowe SP, Shih JH, Lindenberg L, Turkbey B, Fourquet A, Lin FI, Adler S, Eclarinal P, McKinney YL, Citrin DE, Dahut W, Wood BJ, Chang R, Levy E, Merino M, Gorin MA, Pomper MG, Pinto PA, Eary JF, Choyke PL, Pienta KJ. Predictors of 18F-DCFPyL PET/CT Positivity in Patients with Biochemical Recurrence of Prostate Cancer After Local Therapy. J Nucl Med 2022; 63:1184-1190. [PMID: 34916246 PMCID: PMC9364352 DOI: 10.2967/jnumed.121.262347] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 12/02/2021] [Indexed: 02/03/2023] Open
Abstract
Our objective was to investigate the factors predicting scan positivity and disease location in patients with biochemical recurrence (BCR) of prostate cancer (PCa) after primary local therapy using prostate-specific membrane antigen-targeted 18F-DCFPyL PET/CT. Methods: This was a 2-institution study including 245 BCR PCa patients after primary local therapy and negative results on conventional imaging. The patients underwent 18F-DCFPyL PET/CT. We tested for correlations of lesion detection rate and disease location with tumor characteristics, time from initial therapy, prostate-specific antigen (PSA) level, and PSA doubling time (PSAdt). Multivariate logistic regression analyses were used to determine predictors of a positive scan. Regression-based coefficients were used to develop nomograms predicting scan positivity and extrapelvic disease. Results: Overall, 79.2% (194/245) of patients had a positive 18F-DCFPyL PET/CT result, with detection rates of 48.2% (27/56), 74.3% (26/35), 84% (37/44), 96.7% (59/61), and 91.8% (45/49) for PSAs of <0.5, 0.5 to <1.0, 1.0 to <2.0, 2.0 to <5.0, and ≥5.0 ng/mL, respectively. Patients with lesions confined to the pelvis had lower PSAs than those with distant sites (1.6 ± 3.5 vs. 3.0 ± 6.3 ng/mL, P < 0.001). In patients treated with prostatectomy (n = 195), 24.1% (47/195) had a negative scan result, 46.1% (90/195) showed intrapelvic disease, and 29.7% (58/195) showed extrapelvic disease. In the postradiation subgroup (n = 50), 18F-DCFPyL PET/CT was always negative at a PSA lower than 1.0 ng/mL and extrapelvic disease was seen only when PSA was greater than 2.0 ng/mL. At multivariate analysis, PSA and PSAdt were independent predictive factors of scan positivity and the presence of extrapelvic disease in postsurgical patients, with area under the curve of 78% and 76%, respectively. PSA and PSAdt were independent predictors of the presence of extrapelvic disease in the postradiation cohort, with area under the curve of 85%. Time from treatment to scan was significantly longer for prostatectomy-bed-only recurrences than for those with bone or visceral disease (6.2 ± 6.4 vs. 2.4 ± 1.3 y, P < 0.001). Conclusion:18F-DCFPyL PET/CT offers high detection rates in BCR PCa patients. PSA and PSAdt are able to predict scan positivity and disease location. Furthermore, the presence of bone or visceral lesions is associated with shorter intervals from treatment than are prostate-bed-only recurrences. These tools might guide clinicians to select the most suitable candidates for 18F-DCFPyL PET/CT imaging.
Collapse
Affiliation(s)
- Esther Mena
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Steven P. Rowe
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Joanna H. Shih
- Division of Cancer Treatment and Diagnosis: Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Liza Lindenberg
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Baris Turkbey
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Aloyse Fourquet
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Frank I. Lin
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Stephen Adler
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Philip Eclarinal
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Yolanda L. McKinney
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Deborah E. Citrin
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - William Dahut
- Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Bradford J. Wood
- Center of Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Richard Chang
- Center of Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Elliot Levy
- Center of Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Maria Merino
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Michael A. Gorin
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Martin G. Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Peter A. Pinto
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Janet F. Eary
- Cancer Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Peter L. Choyke
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Kenneth J. Pienta
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
11
|
Miyahira AK, Soule HR. The History of Prostate-Specific Membrane Antigen as a Theranostic Target in Prostate Cancer: The Foundational Role of the Prostate Cancer Foundation. J Nucl Med 2021; 63:331-338. [PMID: 34675109 DOI: 10.2967/jnumed.121.262997] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/08/2021] [Indexed: 11/16/2022] Open
Abstract
Prostate-Specific Membrane Antigen (PSMA) is a credentialed imaging and therapy (theranostic) target for the detection and treatment of prostate cancer. PSMA-targeted positron emission tomography (PET) imaging and molecular radiotherapy (MRT) are promising evolving technologies that will improve the outcomes of prostate cancer patients. In anticipation of this new era in prostate cancer theranostics, this article will review the history of PSMA from discovery, through early and late stage clinical trials. Since 1993, the Prostate Cancer Foundation (PCF) has funded critical and foundational PSMA research that established this theranostic revolution. The history and role of PCF funding in this field will be discussed.
Collapse
|
12
|
Lapa C, Nestle U, Albert NL, Baues C, Beer A, Buck A, Budach V, Bütof R, Combs SE, Derlin T, Eiber M, Fendler WP, Furth C, Gani C, Gkika E, Grosu AL, Henkenberens C, Ilhan H, Löck S, Marnitz-Schulze S, Miederer M, Mix M, Nicolay NH, Niyazi M, Pöttgen C, Rödel CM, Schatka I, Schwarzenboeck SM, Todica AS, Weber W, Wegen S, Wiegel T, Zamboglou C, Zips D, Zöphel K, Zschaeck S, Thorwarth D, Troost EGC. Value of PET imaging for radiation therapy. Strahlenther Onkol 2021; 197:1-23. [PMID: 34259912 DOI: 10.1007/s00066-021-01812-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 06/09/2021] [Indexed: 12/13/2022]
Abstract
This comprehensive review written by experts in their field gives an overview on the current status of incorporating positron emission tomography (PET) into radiation treatment planning. Moreover, it highlights ongoing studies for treatment individualisation and per-treatment tumour response monitoring for various primary tumours. Novel tracers and image analysis methods are discussed. The authors believe this contribution to be of crucial value for experts in the field as well as for policy makers deciding on the reimbursement of this powerful imaging modality.
Collapse
Affiliation(s)
- Constantin Lapa
- Nuclear Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Ursula Nestle
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
- Department of Radiation Oncology, Kliniken Maria Hilf, Mönchengladbach, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Christian Baues
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Ambros Beer
- Department of Nuclear Medicine, Ulm University Hospital, Ulm, Germany
| | - Andreas Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Volker Budach
- Department of Radiation Oncology, Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Rebecca Bütof
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Stephanie E Combs
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
- Department of Radiation Sciences (DRS), Institute of Radiation Medicine (IRM), Neuherberg, Germany
| | - Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Matthias Eiber
- Department of Nuclear Medicine, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | - Christian Furth
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Cihan Gani
- German Cancer Consortium (DKTK), Partner Site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Eleni Gkika
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Anca-L Grosu
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Christoph Henkenberens
- Department of Radiotherapy and Special Oncology, Medical School Hannover, Hannover, Germany
| | - Harun Ilhan
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Steffen Löck
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Simone Marnitz-Schulze
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Matthias Miederer
- Department of Nuclear Medicine, University Hospital Mainz, Mainz, Germany
| | - Michael Mix
- Department of Nuclear Medicine, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Nils H Nicolay
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Maximilian Niyazi
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Christoph Pöttgen
- Department of Radiation Oncology, West German Cancer Centre, University of Duisburg-Essen, Essen, Germany
| | - Claus M Rödel
- German Cancer Consortium (DKTK), Partner Site Frankfurt, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiotherapy and Oncology, Goethe-University Frankfurt, Frankfurt, Germany
| | - Imke Schatka
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | | | - Andrei S Todica
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Wolfgang Weber
- Department of Nuclear Medicine, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - Simone Wegen
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Thomas Wiegel
- Department of Radiation Oncology, Ulm University Hospital, Ulm, Germany
| | - Constantinos Zamboglou
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Daniel Zips
- German Cancer Consortium (DKTK), Partner Site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Klaus Zöphel
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, Helmholtz Association/Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Department of Nuclear Medicine, Klinikum Chemnitz gGmbH, Chemnitz, Germany
| | - Sebastian Zschaeck
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Daniela Thorwarth
- German Cancer Consortium (DKTK), Partner Site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Section for Biomedical Physics, Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Esther G C Troost
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.
- National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany, Helmholtz Association/Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.
- German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, Dresden, Germany.
| |
Collapse
|
13
|
Lapa C, Nestle U, Albert NL, Baues C, Beer A, Buck A, Budach V, Bütof R, Combs SE, Derlin T, Eiber M, Fendler WP, Furth C, Gani C, Gkika E, Grosu AL, Henkenberens C, Ilhan H, Löck S, Marnitz-Schulze S, Miederer M, Mix M, Nicolay NH, Niyazi M, Pöttgen C, Rödel CM, Schatka I, Schwarzenboeck SM, Todica AS, Weber W, Wegen S, Wiegel T, Zamboglou C, Zips D, Zöphel K, Zschaeck S, Thorwarth D, Troost EGC. Value of PET imaging for radiation therapy. Nuklearmedizin 2021; 60:326-343. [PMID: 34261141 DOI: 10.1055/a-1525-7029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This comprehensive review written by experts in their field gives an overview on the current status of incorporating positron emission tomography (PET) into radiation treatment planning. Moreover, it highlights ongoing studies for treatment individualisation and per-treatment tumour response monitoring for various primary tumours. Novel tracers and image analysis methods are discussed. The authors believe this contribution to be of crucial value for experts in the field as well as for policy makers deciding on the reimbursement of this powerful imaging modality.
Collapse
Affiliation(s)
- Constantin Lapa
- Nuclear Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Ursula Nestle
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany.,Department of Radiation Oncology, Kliniken Maria Hilf, Mönchengladbach, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Christian Baues
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Ambros Beer
- Department of Nuclear Medicine, Ulm University Hospital, Ulm, Germany
| | - Andreas Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Volker Budach
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Berlin, Germany
| | - Rebecca Bütof
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Stephanie E Combs
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany.,Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany.,Department of Radiation Sciences (DRS), Institute of Radiation Medicine (IRM), Neuherberg, Germany
| | - Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Germany
| | - Matthias Eiber
- Department of Nuclear Medicine, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | - Christian Furth
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Cihan Gani
- German Cancer Consortium (DKTK), Partner Site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Eleni Gkika
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany
| | - Anca L Grosu
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | | | - Harun Ilhan
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Steffen Löck
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Simone Marnitz-Schulze
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Matthias Miederer
- Department of Nuclear Medicine, University Hospital Mainz, Mainz, Germany
| | - Michael Mix
- Department of Nuclear Medicine, Faculty of Medicine, Medical Center, University of Freiburg, Freiburg, Germany
| | - Nils H Nicolay
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Maximilian Niyazi
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Christoph Pöttgen
- Department of Radiation Oncology, West German Cancer Centre, University of Duisburg-Essen, Essen, Germany
| | - Claus M Rödel
- German Cancer Consortium (DKTK), Partner Site Frankfurt, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Radiotherapy and Oncology, Goethe University Frankfurt, Frankfurt, Germany
| | - Imke Schatka
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | | | - Andrei S Todica
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Wolfgang Weber
- Department of Nuclear Medicine, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - Simone Wegen
- Department of Radiation Oncology, Cyberknife and Radiotherapy, Medical Faculty, University Hospital Cologne, Cologne, Germany
| | - Thomas Wiegel
- Department of Radiation Oncology, Ulm University Hospital, Ulm, Germany
| | - Constantinos Zamboglou
- Department of Radiation Oncology, Faculty of Medicine, University Medical Center Freiburg, Freiburg, Germany.,German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Daniel Zips
- German Cancer Consortium (DKTK), Partner Site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Klaus Zöphel
- OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz Association/Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Nuclear Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Department of Nuclear Medicine, Klinikum Chemnitz gGmbH, Chemnitz, Germany
| | - Sebastian Zschaeck
- Department of Radiation Oncology, Charité-Universitätsmedizin Berlin, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Daniela Thorwarth
- German Cancer Consortium (DKTK), Partner Site Tübingen, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Section for Biomedical Physics, Department of Radiation Oncology, University of Tübingen, Tübingen, Germany
| | - Esther G C Troost
- Department of Radiotherapy and Radiation Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,OncoRay - National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany; Helmholtz Association/Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.,German Cancer Consortium (DKTK), Partner Site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.,Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology - OncoRay, Dresden, Germany
| | | |
Collapse
|
14
|
Lawhn-Heath C, Salavati A, Behr SC, Rowe SP, Calais J, Fendler WP, Eiber M, Emmett L, Hofman MS, Hope TA. Prostate-specific Membrane Antigen PET in Prostate Cancer. Radiology 2021; 299:248-260. [PMID: 33787338 DOI: 10.1148/radiol.2021202771] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Prostate-specific membrane antigen (PSMA)-targeted radiopharmaceuticals are playing a large role at the time of initial staging and biochemical recurrence for localizing prostate cancer, as well as in other emerging clinical settings. PSMA PET has demonstrated increased detection rate compared with conventional imaging and has been shown to change management plans in a substantial percentage of cases. The aims of this narrative review are to highlight the development and clinical impact of PSMA PET radiopharmaceuticals, to compare PSMA to other agents such as fluorine 18 fluciclovine and carbon 11 choline, and to highlight some of the individual PSMA PET agents that have contributed to the advancement of prostate cancer imaging.
Collapse
Affiliation(s)
- Courtney Lawhn-Heath
- From the Department of Radiology and Biomedical Imaging (C.L.H., S.C.B., T.A.H.) and Helen Diller Family Comprehensive Cancer Center (S.C.B., T.A.H.), University of California San Francisco, 505 Parnassus Ave, M391, San Francisco, CA 94143; Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Md (A.S., S.P.R.); Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, Calif (J.C.); Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany (W.P.F.); Department of Nuclear Medicine, Technical University of Munich, Munich, Germany (M.E.); Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital, Sydney, Australia (L.E.); Prostate Theranostics and Imaging Centre of Excellence (ProsTIC), Peter MacCallum Cancer Centre, Melbourne, Australia (M.S.H.); and Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia (M.S.H.)
| | - Ali Salavati
- From the Department of Radiology and Biomedical Imaging (C.L.H., S.C.B., T.A.H.) and Helen Diller Family Comprehensive Cancer Center (S.C.B., T.A.H.), University of California San Francisco, 505 Parnassus Ave, M391, San Francisco, CA 94143; Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Md (A.S., S.P.R.); Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, Calif (J.C.); Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany (W.P.F.); Department of Nuclear Medicine, Technical University of Munich, Munich, Germany (M.E.); Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital, Sydney, Australia (L.E.); Prostate Theranostics and Imaging Centre of Excellence (ProsTIC), Peter MacCallum Cancer Centre, Melbourne, Australia (M.S.H.); and Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia (M.S.H.)
| | - Spencer C Behr
- From the Department of Radiology and Biomedical Imaging (C.L.H., S.C.B., T.A.H.) and Helen Diller Family Comprehensive Cancer Center (S.C.B., T.A.H.), University of California San Francisco, 505 Parnassus Ave, M391, San Francisco, CA 94143; Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Md (A.S., S.P.R.); Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, Calif (J.C.); Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany (W.P.F.); Department of Nuclear Medicine, Technical University of Munich, Munich, Germany (M.E.); Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital, Sydney, Australia (L.E.); Prostate Theranostics and Imaging Centre of Excellence (ProsTIC), Peter MacCallum Cancer Centre, Melbourne, Australia (M.S.H.); and Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia (M.S.H.)
| | - Steven P Rowe
- From the Department of Radiology and Biomedical Imaging (C.L.H., S.C.B., T.A.H.) and Helen Diller Family Comprehensive Cancer Center (S.C.B., T.A.H.), University of California San Francisco, 505 Parnassus Ave, M391, San Francisco, CA 94143; Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Md (A.S., S.P.R.); Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, Calif (J.C.); Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany (W.P.F.); Department of Nuclear Medicine, Technical University of Munich, Munich, Germany (M.E.); Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital, Sydney, Australia (L.E.); Prostate Theranostics and Imaging Centre of Excellence (ProsTIC), Peter MacCallum Cancer Centre, Melbourne, Australia (M.S.H.); and Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia (M.S.H.)
| | - Jeremie Calais
- From the Department of Radiology and Biomedical Imaging (C.L.H., S.C.B., T.A.H.) and Helen Diller Family Comprehensive Cancer Center (S.C.B., T.A.H.), University of California San Francisco, 505 Parnassus Ave, M391, San Francisco, CA 94143; Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Md (A.S., S.P.R.); Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, Calif (J.C.); Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany (W.P.F.); Department of Nuclear Medicine, Technical University of Munich, Munich, Germany (M.E.); Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital, Sydney, Australia (L.E.); Prostate Theranostics and Imaging Centre of Excellence (ProsTIC), Peter MacCallum Cancer Centre, Melbourne, Australia (M.S.H.); and Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia (M.S.H.)
| | - Wolfgang P Fendler
- From the Department of Radiology and Biomedical Imaging (C.L.H., S.C.B., T.A.H.) and Helen Diller Family Comprehensive Cancer Center (S.C.B., T.A.H.), University of California San Francisco, 505 Parnassus Ave, M391, San Francisco, CA 94143; Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Md (A.S., S.P.R.); Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, Calif (J.C.); Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany (W.P.F.); Department of Nuclear Medicine, Technical University of Munich, Munich, Germany (M.E.); Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital, Sydney, Australia (L.E.); Prostate Theranostics and Imaging Centre of Excellence (ProsTIC), Peter MacCallum Cancer Centre, Melbourne, Australia (M.S.H.); and Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia (M.S.H.)
| | - Mattias Eiber
- From the Department of Radiology and Biomedical Imaging (C.L.H., S.C.B., T.A.H.) and Helen Diller Family Comprehensive Cancer Center (S.C.B., T.A.H.), University of California San Francisco, 505 Parnassus Ave, M391, San Francisco, CA 94143; Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Md (A.S., S.P.R.); Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, Calif (J.C.); Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany (W.P.F.); Department of Nuclear Medicine, Technical University of Munich, Munich, Germany (M.E.); Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital, Sydney, Australia (L.E.); Prostate Theranostics and Imaging Centre of Excellence (ProsTIC), Peter MacCallum Cancer Centre, Melbourne, Australia (M.S.H.); and Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia (M.S.H.)
| | - Louise Emmett
- From the Department of Radiology and Biomedical Imaging (C.L.H., S.C.B., T.A.H.) and Helen Diller Family Comprehensive Cancer Center (S.C.B., T.A.H.), University of California San Francisco, 505 Parnassus Ave, M391, San Francisco, CA 94143; Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Md (A.S., S.P.R.); Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, Calif (J.C.); Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany (W.P.F.); Department of Nuclear Medicine, Technical University of Munich, Munich, Germany (M.E.); Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital, Sydney, Australia (L.E.); Prostate Theranostics and Imaging Centre of Excellence (ProsTIC), Peter MacCallum Cancer Centre, Melbourne, Australia (M.S.H.); and Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia (M.S.H.)
| | - Michael S Hofman
- From the Department of Radiology and Biomedical Imaging (C.L.H., S.C.B., T.A.H.) and Helen Diller Family Comprehensive Cancer Center (S.C.B., T.A.H.), University of California San Francisco, 505 Parnassus Ave, M391, San Francisco, CA 94143; Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Md (A.S., S.P.R.); Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, Calif (J.C.); Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany (W.P.F.); Department of Nuclear Medicine, Technical University of Munich, Munich, Germany (M.E.); Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital, Sydney, Australia (L.E.); Prostate Theranostics and Imaging Centre of Excellence (ProsTIC), Peter MacCallum Cancer Centre, Melbourne, Australia (M.S.H.); and Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia (M.S.H.)
| | - Thomas A Hope
- From the Department of Radiology and Biomedical Imaging (C.L.H., S.C.B., T.A.H.) and Helen Diller Family Comprehensive Cancer Center (S.C.B., T.A.H.), University of California San Francisco, 505 Parnassus Ave, M391, San Francisco, CA 94143; Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, Md (A.S., S.P.R.); Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, Calif (J.C.); Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany (W.P.F.); Department of Nuclear Medicine, Technical University of Munich, Munich, Germany (M.E.); Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital, Sydney, Australia (L.E.); Prostate Theranostics and Imaging Centre of Excellence (ProsTIC), Peter MacCallum Cancer Centre, Melbourne, Australia (M.S.H.); and Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia (M.S.H.)
| |
Collapse
|
15
|
Role of 18F-Fluciclovine and Prostate-Specific Membrane Antigen PET/CT in Guiding Management of Oligometastatic Prostate Cancer: AJR Expert Panel Narrative Review. AJR Am J Roentgenol 2021; 216:851-859. [PMID: 33206564 DOI: 10.2214/ajr.20.24711] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Twenty-five years ago, oligometastatic disease was proposed as an intermediary clinical state of cancer with unique implications for therapies that may impact cancer evolution and patient outcome. Identification of limited metastases that are potentially amenable to targeted therapies fundamentally depends on the sensitivity of diagnostic tools, including new-generation imaging methods. For men with biochemical recurrence after definitive therapy of the primary prostate cancer, PET/CT using either the FDA-approved radiolabeled amino acid analogue 18F-fluciclovine or investigational radiolabeled agents targeting prostate-specific membrane antigen (PSMA) enables identification of early metastases at lower serum PSA levels than was previously feasible using conventional imaging. Evidence supports PSMA PET/CT as the most sensitive imaging modality available for identifying disease sites in oligometastatic prostate cancer. PSMA PET/CT will likely become the modality of choice after regulatory approval and will drive the development of trials of emerging metastasis-directed therapies such as stereotactic ablative body radiation and radioguided surgery. Indeed, numerous ongoing or planned clinical trials are studying advances in management of oligometastatic prostate cancer based on this heightened diagnostic capacity. In this rapidly evolving clinical environment, radiologists and nuclear medicine physicians will play major roles in facilitating clinical decision making and management of patients with oligometastatic prostate cancer.
Collapse
|
16
|
Impact of PSMA PET on management of biochemical recurrent prostate cancer: a systematic review and meta-analysis of prospective studies. Clin Transl Imaging 2021. [DOI: 10.1007/s40336-020-00406-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
17
|
Rowe SP, Johnson GB, Pomper MG, Gorin MA, Behr SC. Recent updates and developments in PET imaging of prostate cancer. Abdom Radiol (NY) 2020; 45:4063-4072. [PMID: 32417934 DOI: 10.1007/s00261-020-02570-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A number of positron emission tomography (PET) radiotracers have been developed to improve the sensitivity and specificity of imaging for prostate cancer. These radiotracers include the bone-seeking agent Na18F as well as more tumor-specific compounds such as 11C-choline and 18F-fluciclovine. In this review, we will discuss the advantages and disadvantages of these PET radiotracers for the imaging of men with prostate cancer across a range of clinical contexts. We will also touch upon radiotracers in late clinical development that have not gained regulatory approval, including those targeted against prostate-specific membrane antigen (PSMA) and gastrin-releasing peptide receptor (GRPR).
Collapse
Affiliation(s)
- 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, 21287, USA.
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Geoffrey B Johnson
- Division of Nuclear Medicine, Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Martin G Pomper
- 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, 21287, USA
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael A Gorin
- 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, 21287, USA
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Spencer C Behr
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
18
|
Pastorino S, Riondato M, Uccelli L, Giovacchini G, Giovannini E, Duce V, Ciarmiello A. Toward the Discovery and Development of PSMA Targeted Inhibitors for Nuclear Medicine Applications. Curr Radiopharm 2020; 13:63-79. [PMID: 31362683 PMCID: PMC7509769 DOI: 10.2174/1874471012666190729151540] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 07/15/2019] [Accepted: 07/15/2019] [Indexed: 11/22/2022]
Abstract
BACKGROUND The rising incidence rate of prostate cancer (PCa) has promoted the development of new diagnostic and therapeutic radiopharmaceuticals during the last decades. Promising improvements have been achieved in clinical practice using prostate specific membrane antigen (PSMA) labeled agents, including specific antibodies and small molecular weight inhibitors. Focusing on molecular docking studies, this review aims to highlight the progress in the design of PSMA targeted agents for a potential use in nuclear medicine. RESULTS Although the first development of radiopharmaceuticals able to specifically recognize PSMA was exclusively oriented to macromolecule protein structure such as radiolabeled monoclonal antibodies and derivatives, the isolation of the crystal structure of PSMA served as the trigger for the synthesis and the further evaluation of a variety of low molecular weight inhibitors. Among the nuclear imaging probes and radiotherapeutics that have been developed and tested till today, labeled Glutamate-ureido inhibitors are the most prevalent PSMA-targeting agents for nuclear medicine applications. CONCLUSION PSMA represents for researchers the most attractive target for the detection and treatment of patients affected by PCa using nuclear medicine modalities. [99mTc]MIP-1404 is considered the tracer of choice for SPECT imaging and [68Ga]PSMA-11 is the leading diagnostic for PET imaging by general consensus. [18F]DCFPyL and [18F]PSMA-1007 are clearly the emerging PET PSMA candidates for their great potential for a widespread commercial distribution. After paving the way with new imaging tools, academic and industrial R&Ds are now focusing on the development of PSMA inhibitors labeled with alpha or beta minus emitters for a theragnostic application.
Collapse
Affiliation(s)
- Sara Pastorino
- Nuclear Medicine Department, S. Andrea Hospital, Via Vittorio Veneto 197, 19124 La Spezia, Italy
| | - Mattia Riondato
- Nuclear Medicine Department, S. Andrea Hospital, Via Vittorio Veneto 197, 19124 La Spezia, Italy
| | - Licia Uccelli
- Morphology, Surgery and Experimental Medicine Department, University of Ferrara, Via L. Borsari 46, 44121 Ferrara, Italy.,Nuclear Medicine Unit, University Hospital, Via Aldo Moro 8, 44124 Ferrara, Italy
| | - Giampiero Giovacchini
- Nuclear Medicine Department, S. Andrea Hospital, Via Vittorio Veneto 197, 19124 La Spezia, Italy
| | - Elisabetta Giovannini
- Nuclear Medicine Department, S. Andrea Hospital, Via Vittorio Veneto 197, 19124 La Spezia, Italy
| | - Valerio Duce
- Nuclear Medicine Department, S. Andrea Hospital, Via Vittorio Veneto 197, 19124 La Spezia, Italy
| | - Andrea Ciarmiello
- Nuclear Medicine Department, S. Andrea Hospital, Via Vittorio Veneto 197, 19124 La Spezia, Italy
| |
Collapse
|
19
|
Bone scintigraphy as a gatekeeper for the detection of bone metastases in patients with prostate cancer: comparison with Ga-68 PSMA PET/CT. Ann Nucl Med 2020; 34:932-941. [PMID: 32975741 DOI: 10.1007/s12149-020-01529-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 09/08/2020] [Indexed: 10/23/2022]
Abstract
OBJECTIVES Gallium-68-labeled prostate-specific membrane antigen (Ga-PSMA) positron emission tomography (PET)/computed tomography (CT) is a valuable diagnostic tool for the detection of bone metastases in patients with prostate cancer (PCa). However, bone scintigraphy (BS) with technetium-labeled diphosphonates is cheap and widely available for the same patient population. PSMA PET comes with a cost, and financial constraints in the present economic environment may require its more selective use. In this study, we aimed to compare the diagnostic performance of BS with Ga-PSMA PET/CT for the detection of bone metastases in patients with PCa and correlate the results with various clinical and biochemical variables. MATERIALS AND METHODS Ninety-five patients who underwent Ga-PSMA PET/CT and BS within 3 months for newly diagnosed or recurrent PCa were extracted from our database. Lesion, region and patient-based analyses were performed. Clinical and imaging follow-up was used as the reference test. Results were compared with tumor grade, serum prostate-specific antigen (PSA), and alkaline phosphatase (ALP) values. RESULTS On the patient-based analysis, 75% (42/56) and 98.2% (55/56) of the patients with bone metastases were correctly diagnosed by BS and Ga-PSMA PET, respectively. In 26/95 patients with equivocal lesions on BS, Ga-PSMA PET correctly reclassified skeletal involvement in 11 and excluded metastases in 15 patients BS missed bone metastases in 3 patients. The true-positive rate of BS in patients with serum ALP ≥ 120U/L and PSA ≥ 50 ng/ml was 95.8% and 87.5 respectively. CONCLUSION Ga-PSMA is superior to BS for the evaluation of metastatic disease in patients with PCa. However, BS can also detect bone metastases in patients with PCa with a minimum sensitivity of 75%. Biochemical data are helpful to select patients with a high pretest probability who should undergo BS first as a part of the initial workup from an economic point of view. Due to its higher cost, Ga-PSMA PET should be performed in a selective group of patients when BS results are inconclusive or metastasis-directed therapy is planned.
Collapse
|
20
|
Petersen LJ, Johansen MN, Strandberg J, Stenholt L, Zacho HD. Reporting and handling of equivocal imaging findings in diagnostic studies of bone metastasis in prostate cancer. Acta Radiol 2020; 61:1096-1104. [PMID: 31821767 DOI: 10.1177/0284185119890087] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Equivocal scanning results occur. It remains unclear how these results are presented and their management influence diagnostic characteristics. PURPOSE To investigate the reporting and handling of equivocal imaging findings in diagnostic studies of bone metastases, and to assess the impact on diagnostic performance of the methods used to analyze equivocal findings. The conceptual issue was reified based on two actual observations. MATERIAL AND METHODS A recent meta-analysis of bone metastases in prostate cancer was conducted and data were obtained from a large clinical trial with a true reference of bone metastasis, where diagnostic characteristics were calculated with equivocal scans handled by: removal; considered malignant; considered benign; and intention-to-diagnose. RESULTS The meta-analysis included 18 trials where the median proportion of reported equivocal results was 27%. Eleven (61%) studies reported an equivocal option for the index test, 42% reported equivocal results and described how these were analyzed. The clinical trial included 583 prostate cancer patients with 20% equivocal results. The different methods of managing equivocal findings resulted in highly variable outcomes: sensitivity = 85%-100%; specificity = 78%-99%; and positive and negative predictive values = 44%-94% and 97%-100%, respectively. The diagnostic performances obtained using the four methods were differentially susceptible to the proportion of equivocal imaging findings and the prevalence of bone metastases. CONCLUSION Reporting of equivocal results was inadequate in bone imaging trials. The handling of equivocal findings strongly influenced diagnostic accuracy.
Collapse
Affiliation(s)
- Lars J Petersen
- Department of Nuclear Medicine, Clinical Cancer Research Centre, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | | | - Jesper Strandberg
- Department of Nuclear Medicine, Clinical Cancer Research Centre, Aalborg University Hospital, Aalborg, Denmark
| | - Louise Stenholt
- The Medical Library, Aalborg University Hospital, Aalborg, Denmark
| | - Helle D Zacho
- Department of Nuclear Medicine, Clinical Cancer Research Centre, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| |
Collapse
|
21
|
Walker SM, Lim I, Lindenberg L, Mena E, Choyke PL, Turkbey B. Positron emission tomography (PET) radiotracers for prostate cancer imaging. Abdom Radiol (NY) 2020; 45:2165-2175. [PMID: 32047993 DOI: 10.1007/s00261-020-02427-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Imaging plays an increasing role in prostate cancer diagnosis and staging. Accurate staging of prostate cancer is required for optimal treatment planning. In detecting extraprostatic cancer and sites of early recurrence, traditional imaging methods (computed tomography, magnetic resonance imaging, radionuclide bone scan) have suboptimal performance. This leaves a gap between known disease recurrence as indicated by rising prostate-specific antigen and the ability to localize the recurrence on imaging. Novel positron emission tomography (PET) agents including radiolabeled choline, fluciclovine (18F-FACBC), and agents targeting prostate-specific membrane antigen are being developed and tested to increase diagnostic performance of non-invasive prostate cancer localization. When combined with CT or MRI, these tracers offer a combination of functional information and anatomic localization that is superior to conventional imaging methods. These PET radiotracers have varying mechanisms and excretion patterns affecting their pharmacokinetics and diagnostic performance, which will be reviewed in this article.
Collapse
Affiliation(s)
- Stephanie M Walker
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ilhan Lim
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
- Department of Nuclear Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences (KIRAMS), Seoul, Korea
| | - Liza Lindenberg
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Esther Mena
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Peter L Choyke
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Baris Turkbey
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
22
|
Lin M, Ta RT, Kairemo K, Le DB, Ravizzini GC. Prostate-Specific Membrane Antigen-Targeted Radiopharmaceuticals in Diagnosis and Therapy of Prostate Cancer: Current Status and Future Perspectives. Cancer Biother Radiopharm 2020; 36:237-251. [PMID: 32589458 DOI: 10.1089/cbr.2020.3603] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Prostate cancer is the most common cancer to affect men in the United States and the second most common cancer in men worldwide. Prostate-specific membrane antigen (PSMA)-based positron emission tomography (PET) imaging has become increasingly popular as a novel molecular imaging technique capable of improving the clinical management of patients with prostate cancer. To date, several 68Ga and 18F-labeled PSMA-targeted molecules have shown promising results in imaging patients with recurrent prostate cancer using PET/computed tomography (PET/CT). Studies of involving PSMA-targeted radiopharmaceuticals also suggest a higher sensitivity and specificity, along with an improved detection rate over conventional imaging (CT scan and methylene diphosphonate bone scintigraphy) and 11C/18F-choline PET/CT. In addition, PSMA-617 and PSMA I&T ligands can be labeled with α- and β-emitters (e.g., 225Ac, 90Y, and 177Lu) and serve as a theranostic tool for patients with metastatic prostate cancer. While the clinical impact of such concept remains to be verified, the preliminary results of PSMA molecular radiotherapy are very encouraging. Herein, we highlighted the current status of development and future perspectives of PSMA-targeted radiopharmaceuticals and their clinical applications.
Collapse
Affiliation(s)
- Mai Lin
- Cyclotron Radiochemistry Facility, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Robert T Ta
- Cyclotron Radiochemistry Facility, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kalevi Kairemo
- Department of Nuclear Medicine and Molecular Radiotherapy, Docrates Cancer Center, Helsinki, Finland.,Department of Nuclear Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Dao B Le
- Cyclotron Radiochemistry Facility, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Nuclear Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Gregory C Ravizzini
- Department of Nuclear Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
23
|
Lebastchi AH, Gupta N, DiBianco JM, Piert M, Davenport MS, Ahdoot MA, Gurram S, Bloom JB, Gomella PT, Mehralivand S, Turkbey B, Pinto PA, George AK. Comparison of cross-sectional imaging techniques for the detection of prostate cancer lymph node metastasis: a critical review. Transl Androl Urol 2020; 9:1415-1427. [PMID: 32676426 PMCID: PMC7354341 DOI: 10.21037/tau.2020.03.20] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Conventional staging for prostate cancer (PCa) is performed for men diagnosed with unfavorable-intermediate or higher risk disease. Computed tomography (CT) of the abdomen and pelvis and whole body bone scan remains the standard of care for the detection of visceral, nodal, and bone metastasis. The implementation of the 2012 United States Preventive Services Task Force recommendation against routine prostate specific antigen (PSA) screening resulted in a rise of metastatic PCa at the time of diagnosis, emphasizing the importance of effective imaging modalities for evaluating metastatic disease. CT plays a major role in clinical staging at the time of PCa diagnosis, but multi-parametric magnetic resonance imaging (MRI) is now integrated into many prostate biopsy protocols for the detection of primary PCa, and may be a surrogate for CT for nodal staging. Current guidelines incorporate both CT and MRI as appropriate cross-sectional imaging modalities for the identification of nodal metastasis in indicated patients. There is an ongoing debate about the utility of traditional cross-sectional imaging modalities as well as advanced imaging modalities in detection of both organ-confined PCa detection and nodal involvement.
Collapse
Affiliation(s)
- Amir H Lebastchi
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nikhil Gupta
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - John M DiBianco
- Department of Urology, George Washington University Medical School, Washington D.C., USA
| | - Morand Piert
- Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | | | - Michael A Ahdoot
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sandeep Gurram
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jonathan B Bloom
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Patrick T Gomella
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Baris Turkbey
- Molecular Imaging Program, National Cancer Institute, Bethesda, MD, USA
| | - Peter A Pinto
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Arvin K George
- Department of Urology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
24
|
Torga G, Yin Y, Pomper MG, Pienta KJ, Gorin MA, Rowe SP. Uptake of prostate-specific membrane antigen-targeted 18F-DCFPyL in avascular necrosis of the femoral head. World J Nucl Med 2020; 18:416-419. [PMID: 31933560 PMCID: PMC6945356 DOI: 10.4103/wjnm.wjnm_106_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 01/24/2019] [Indexed: 01/10/2023] Open
Abstract
In recent years, the emergence of prostate-specific membrane antigen (PSMA)-targeted positron-emission tomography (PET) imaging has brought about a paradigm shift in the way that prostate cancer (PCa) is imaged in many parts of the world. Although PSMA-targeted PET imaging has been demonstrated to be a highly sensitive and specific imaging modality for the identification of sites of PCa, its clinical utility hinges on the ability of imaging specialists and their clinical colleagues to recognize potential false-positive sources of uptake and to tailor therapy based on that recognition. In this manuscript, we report the case of a 74-year-old male with a history of recurrent PCa who was referred for a restaging PSMA-targeted 18F-DCFPyL PET/computed tomography (PET/CT). PET images demonstrated low level but focal and definitive uptake in the left femoral head. This uptake corresponded to sclerotic changes on CT whose morphology was most compatible with avascular necrosis without femoral head collapse. In the presented case, the integrated assessment of the CT imaging together with the PET findings was fundamental to avoid misinterpretation of the left femur finding as metastatic disease, which would have ultimately altered the clinical management of the patient.
Collapse
Affiliation(s)
- Gonzalo Torga
- Department of Urology, The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Yafu Yin
- Department of Urology, The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Nuclear Medicine, The First Hospital of China Medical University, Shenyang, China.,Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Martin G Pomper
- Department of Urology, The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kenneth J Pienta
- Department of Urology, The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael A Gorin
- Department of Urology, The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Steven P Rowe
- Department of Urology, The James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
25
|
Current Status of PSMA-Radiotracers for Prostate Cancer: Data Analysis of Prospective Trials Listed on ClinicalTrials.gov. Pharmaceuticals (Basel) 2020; 13:ph13010012. [PMID: 31940969 PMCID: PMC7168903 DOI: 10.3390/ph13010012] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 12/16/2019] [Accepted: 12/31/2019] [Indexed: 12/11/2022] Open
Abstract
The recent development of dedicated prostate-specific membrane antigen (PSMA) targeted radioligands shows the potential to change and improve the diagnosis and therapy of prostate cancer. There is an increasing number of prospective trials to further establish these tracers in the clinical setting. We analyzed data from the ClinicalTrials.gov registry including all listed prospective trials with PSMA-ligands for prostate cancer as of October 2019 concerning the different tracers and study characteristics. We found n = 104 eligible studies with a total of n = 25 different tracers in use: most frequently [68Ga]Ga-PSMA-11 (32%), followed by [18F]DCFPyL (24%) and [177Lu]Lu-PSMA-617 (10%). 85% are single-center, 15% multi-center studies. 95% national and 5% international studies. 34% are phase-II, 24% phase-I, 13% phase-I/-II, 12% phase-II/-III and phase-III and 7% early-phase-I. The primary purpose was classified as diagnostic in 72% of cases and therapeutic in 23% of cases. Most studies were executed in the USA (70%), followed by Canada (13%) and France (6%). This quantitative descriptive registry analysis indicates the rapid and global clinical developments and current status of PSMA-radioligands with emphasis on radiopharmaceutical and organizational aspects. It will be very interesting to see which tracers will prevail in the clinical setting.
Collapse
|
26
|
Werner RA, Derlin T, Lapa C, Sheikbahaei S, Higuchi T, Giesel FL, Behr S, Drzezga A, Kimura H, Buck AK, Bengel FM, Pomper MG, Gorin MA, Rowe SP. 18F-Labeled, PSMA-Targeted Radiotracers: Leveraging the Advantages of Radiofluorination for Prostate Cancer Molecular Imaging. Theranostics 2020; 10:1-16. [PMID: 31903102 PMCID: PMC6929634 DOI: 10.7150/thno.37894] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 09/11/2019] [Indexed: 12/22/2022] Open
Abstract
Prostate-specific membrane antigen (PSMA)-targeted PET imaging for prostate cancer with 68Ga-labeled compounds has rapidly become adopted as part of routine clinical care in many parts of the world. However, recent years have witnessed the start of a shift from 68Ga- to 18F-labeled PSMA-targeted compounds. The latter imaging agents have several key advantages, which may lay the groundwork for an even more widespread adoption into the clinic. First, facilitated delivery from distant suppliers expands the availability of PET radiopharmaceuticals in smaller hospitals operating a PET center but lacking the patient volume to justify an onsite 68Ge/68Ga generator. Thus, such an approach meets the increasing demand for PSMA-targeted PET imaging in areas with lower population density and may even lead to cost-savings compared to in-house production. Moreover, 18F-labeled radiotracers have a higher positron yield and lower positron energy, which in turn decreases image noise, improves contrast resolution, and maximizes the likelihood of detecting subtle lesions. In addition, the longer half-life of 110 min allows for improved delayed imaging protocols and flexibility in study design, which may further increase diagnostic accuracy. Moreover, such compounds can be distributed to sites which are not allowed to produce radiotracers on-site due to regulatory issues or to centers without access to a cyclotron. In light of these advantageous characteristics, 18F-labeled PSMA-targeted PET radiotracers may play an important role in both optimizing this transformative imaging modality and making it widely available. We have aimed to provide a concise overview of emerging 18F-labeled PSMA-targeted radiotracers undergoing active clinical development. Given the wide array of available radiotracers, comparative studies are needed to firmly establish the role of the available 18F-labeled compounds in the field of molecular PCa imaging, preferably in different clinical scenarios.
Collapse
Affiliation(s)
- Rudolf A. Werner
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Constantin Lapa
- Department of Nuclear Medicine, University Hospital Würzburg, Germany
| | - Sara Sheikbahaei
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Takahiro Higuchi
- Department of Nuclear Medicine, University Hospital Würzburg, Germany
- Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Frederik L. Giesel
- Department of Nuclear Medicine, University Hospital Heidelberg, INF 400, 69120 Heidelberg, Germany
| | - Spencer Behr
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA
| | - Alexander Drzezga
- Department of Nuclear Medicine, University Hospital Cologne, Germany
| | - Hiroyuki Kimura
- Department of Analytical and Bioinorganic Chemistry, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Andreas K. Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Germany
| | - Frank M. Bengel
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Martin G. Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael A. Gorin
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Steven P. Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
27
|
Ribeiro AMB, Lima ENP, Rocha MM. Fibrous dysplasia as a possible false-positive finding in 68Ga-labeled prostate-specific membrane antigen positron emission tomography/computed tomography study in the follow-up of prostate cancer. World J Nucl Med 2019; 18:409-412. [PMID: 31933558 PMCID: PMC6945358 DOI: 10.4103/wjnm.wjnm_111_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 12/21/2018] [Indexed: 01/20/2023] Open
Abstract
Positron emission tomography/computed tomography (PET/CT) using 68Ga-labeled prostate-specific membrane antigen (68Ga-PSMA) has become an important tool in restaging patients with prostate cancer (PCa). Despite its high sensitivity and specificity, this method may produce false-positive findings, as indicated by previous studies. This case report aims to warn nuclear medicine physicians, oncologists, and urologists about the possibility of false-positive findings using this imaging modality, especially when the detected site is unusual for bone metastasis. A 68-year-old man with PCa underwent restaging tests after presenting with increased prostate-specific antigen. 68Ga-PSMA PET/CT imaging revealed abnormal uptake in the left humeral head, which anatomically corresponded to the intramedullary and cortical sclerotic area. A biopsy was performed, and the pathology showed a lesion consisting of hard bone tissue with a small focal spot of fibrous dysplasia. Diagnostic issues related to 68Ga-PSMA PET/CT imaging should be disseminated to help physicians make appropriate treatment choices for each patient.
Collapse
|
28
|
Mena E, Lindenberg ML, Turkbey IB, Shih JH, Harmon SA, Lim I, Lin F, Adler S, Eclarinal P, McKinney YL, Citrin D, Dahut W, Wood BJ, Krishnasamy V, Chang R, Levy E, Merino M, Pinto P, Eary JF, Choyke PL. 18F-DCFPyL PET/CT Imaging in Patients with Biochemically Recurrent Prostate Cancer After Primary Local Therapy. J Nucl Med 2019; 61:881-889. [PMID: 31676732 DOI: 10.2967/jnumed.119.234799] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 10/07/2019] [Indexed: 11/16/2022] Open
Abstract
Our objective was to investigate the lesion detection rate of 18F-DCFPyL PET/CT, a prostate-specific membrane antigen (PSMA)-targeted PET agent, in patients with biochemically relapsed prostate cancer after primary local therapy. Methods: This was a prospective institutional review board-approved study of 90 patients with documented biochemical recurrence (median prostate-specific antigen [PSA], 2.5 ng/mL; range, 0.21-35.5 ng/mL) and negative results on conventional imaging after primary local therapies, including radical prostatectomy (n = 38), radiation (n = 27), or a combination of the two (n = 25). Patients on androgen deprivation therapy were excluded. Patients underwent whole-body 18F-DCFPyL PET/CT (299.9 ± 15.5 MBq) at 2 h after injection. The PSMA PET lesion detection rate was correlated with PSA, PSA kinetics, and original primary tumor grade. Results: Seventy patients (77.8%) showed positive PSMA PET results, with a total of 287 lesions identified: 37 prostate bed foci, 208 lesions in lymph nodes, and 42 in distant sites in bones or organs, Eleven patients had negative results, and 9 patients showed indeterminate lesions, which were considered negative in this study. The detection rates were 47.6% (n = 10/21), 50% (n = 5/10), 88.9% (n = 8/9), and 94% (n = 47/50) for PSA levels of >0.2 to <0.5, 0.5 to <1.0, 1 to <2.0, and ≥2.0 ng/mL, respectively. In postsurgical patients, PSA, PSA doubling time, and PSA velocity correlated with PET results, but the same was not true for postradiation patients. These parameters also correlated with the extent of disease on PET (intrapelvic vs. extrapelvic). There was no significant difference in the rate of positive scans between patients with higher-grade and lower-grade primary tumors (Gleason score of ≥4 + 3 vs. <3 + 4). Tumor recurrence was histology-confirmed in 40% (28/70) of patients. On a per-patient basis, positive predictive value was 93.3% (95% confidence interval, 77.6%-99.2%) by histopathologic validation and 96.2% (95% confidence interval, 86.3%-99.7%) by the combination of histology and imaging/clinical follow-up. Conclusion: 18F-DCFPyL PET/CT imaging offers high detection rates in biochemically recurrent prostate cancer patients and is positive in about 50% of patients with a PSA level of less than 0.5 ng/mL, which could substantially impact clinical management. In postsurgical patients, 18F-DCFPyL PET/CT correlates with PSA, PSA doubling time, and PSA velocity, suggesting it may have prognostic value. 18F-DCFPyL PET/CT is highly promising for localizing sites of recurrent prostate cancer.
Collapse
Affiliation(s)
- Esther Mena
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Maria Liza Lindenberg
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Ismail Baris Turkbey
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Joanna H Shih
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Stephanie A Harmon
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Ilhan Lim
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Frank Lin
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Stephen Adler
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Philip Eclarinal
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Yolanda L McKinney
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Deborah Citrin
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - William Dahut
- Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Bradford J Wood
- Center of Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Venkatesh Krishnasamy
- Center of Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Richard Chang
- Center of Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Elliot Levy
- Center of Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Maria Merino
- Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Peter Pinto
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland; and
| | - Janet F Eary
- Cancer Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Peter L Choyke
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
29
|
Rowe SP, Li X, Trock BJ, Werner RA, Frey S, DiGianvittorio M, Bleiler JK, Reyes DK, Abdallah R, Pienta KJ, Gorin MA, Pomper MG. Prospective Comparison of PET Imaging with PSMA-Targeted 18F-DCFPyL Versus Na 18F for Bone Lesion Detection in Patients with Metastatic Prostate Cancer. J Nucl Med 2019; 61:183-188. [PMID: 31451492 DOI: 10.2967/jnumed.119.227793] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 07/12/2019] [Indexed: 12/12/2022] Open
Abstract
Bone metastases in prostate cancer (PCa) have important prognostic significance, and imaging modalities used for PCa staging should have high sensitivity for detecting such lesions. Prostate-specific membrane antigen (PSMA)-targeted PET radiotracers are promising new agents for imaging PCa. We undertook a head-to-head comparison of PSMA-targeted 2-(3-{1-carboxy-5-[(6-18F-fluoro-pyridine-3-carbonyl)-amino]-pentyl}-ureido)-pentanedioic acid (18F-DCFPyL) PET to Na18F PET to determine which modality was more sensitive for the detection of lesions suggestive of bone metastases in a group of patients with metastatic PCa. Methods: Patients with progressive, metastatic PCa were prospectively imaged with both 18F-DCFPyL and Na18F PET/CT, with both scans occurring within 24 h of each other. A consensus 2-reader central review was performed to identify all bone lesions suggestive of sites of PCa involvement on both scans, and maximized SUVs corrected for body weight (SUVmax) and lean body mass (SULmax) were recorded. Soft-tissue lesions were also noted on both scans, and SUVmax, SULmax, and PSMA reporting and data system (RADS) version 1.0 scores were recorded. Data from the 2 scans were compared using a generalized estimating equation. Results: In total, 16 patients meeting all inclusion criteria were enrolled in this study, and 15 of the 16 (93.8%) were imaged with both PET radiotracers. In total, 405 bone lesions suggestive of sites of PCa were identified on at least 1 scan. On 18F-DCFPyL PET/CT, 391 (96.5%) were definitively positive, 4 (1.0%) were equivocally positive, and 10 (2.5%) were negative. On Na18F PET/CT, the corresponding values were 388 (95.8%), 4 (1.0%), and 13 (3.2%). Of the definitively negative lesions on 18F-DCFPyL PET, 8 of 10 (80.0%) were sclerotic and 2 of 10 (20.0%) were infiltrative or marrow-based. Additionally, 12 of 13 (92.3%) of the definitively negative lesions on Na18F PET were infiltrative or marrow-based and 1 of 13 (7.7%) was lytic. Also identified were 78 PSMA-RADS-4, 17 PSMA-RADS-5, and 1 PSMA-RADS-3C soft-tissue lesions. Conclusion: PET/CT imaging using 18F-DCFPyL and Na18F PET had nearly identical sensitivities for the detection of bone lesions in patients with metastatic PCa. As would be expected, PSMA-targeted PET provides more information on soft-tissue disease. There may be little additional value to imaging PCa patients with Na18F after a PSMA-targeted PET scan has already been performed.
Collapse
Affiliation(s)
- Steven P Rowe
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland .,James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Xin Li
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Radiology, Shandong Provincial Hospital, Shandong University, Jinan City, Shandong Province, China
| | - Bruce J Trock
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Rudolf A Werner
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Nuclear Medicine, University Hospital Wurzburg, Wurzburg, Germany
| | - Sarah Frey
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael DiGianvittorio
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Spectrum Medical Group, South Portland, Maine; and
| | | | - Diane K Reyes
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Rehab Abdallah
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kenneth J Pienta
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael A Gorin
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland.,James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Martin G Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland.,James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
30
|
Banerjee SR, Kumar V, Lisok A, Chen J, Minn I, Brummet M, Boinapally S, Cole M, Ngen E, Wharram B, Brayton C, Hobbs RF, Pomper MG. 177Lu-labeled low-molecular-weight agents for PSMA-targeted radiopharmaceutical therapy. Eur J Nucl Med Mol Imaging 2019; 46:2545-2557. [PMID: 31399803 DOI: 10.1007/s00259-019-04434-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 07/09/2019] [Indexed: 11/29/2022]
Abstract
PURPOSE To develop a prostate-specific membrane antigen (PSMA)-targeted radiotherapeutic for metastatic castration-resistant prostate cancer (mCRPC) with optimized efficacy and minimized toxicity employing the β-particle radiation of 177Lu. METHODS We synthesized 14 new PSMA-targeted, 177Lu-labeled radioligands (177Lu-L1-177Lu-L14) using different chelating agents and linkers. We evaluated them in vitro using human prostate cancer PSMA(+) PC3 PIP and PSMA(-) PC3 flu cells and in corresponding flank tumor models. Efficacy and toxicity after 8 weeks were evaluated at a single administration of 111 MBq for 177Lu-L1, 177Lu-L3, 177Lu-L5 and 177Lu-PSMA-617. Efficacy of 177Lu-L1 was further investigated using different doses, and long-term toxicity was determined in healthy immunocompetent mice. RESULTS Radioligands were produced in high radiochemical yield and purity. Cell uptake and internalization indicated specific uptake only in PSMA(+) PC3 cells. 177Lu-L1, 177Lu-L3 and 177Lu-L5 demonstrated comparable uptake to 177Lu-PSMA-617 and 177Lu-PSMA-I&T in PSMA-expressing tumors up to 72 h post-injection. 177Lu-L1, 177Lu-L3 and 177Lu-L5 also demonstrated efficient tumor regression at 8 weeks. 177Lu-L1 enabled the highest survival rate. Necropsy studies of the treated group at 8 weeks revealed subacute damage to lacrimal glands and testes. No radiation nephropathy was observed 1 year post-treatment in healthy mice receiving 111 MBq of 177Lu-L1, most likely related to the fast renal clearance of this agent. CONCLUSIONS 177Lu-L1 is a viable clinical candidate for radionuclide therapy of PSMA-expressing malignancies because of its high tumor-targeting ability and low off-target radiotoxic effects.
Collapse
Affiliation(s)
- Sangeeta Ray Banerjee
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Vivek Kumar
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ala Lisok
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jian Chen
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Il Minn
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mary Brummet
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Srikanth Boinapally
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael Cole
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ethel Ngen
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bryan Wharram
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cory Brayton
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert F Hobbs
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Martin G Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA. .,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
31
|
Sultana S, Song DY, Lee J. Deformable registration of PET/CT and ultrasound for disease-targeted focal prostate brachytherapy. J Med Imaging (Bellingham) 2019; 6:035003. [PMID: 31528661 PMCID: PMC6739636 DOI: 10.1117/1.jmi.6.3.035003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 08/20/2019] [Indexed: 12/27/2022] Open
Abstract
We propose a deformable registration algorithm for prostate-specific membrane antigen (PSMA) PET/CT and transrectal ultrasound (TRUS) fusion. Accurate registration of PSMA PET to intraoperative TRUS will allow physicians to customize dose planning based on the regions involved. The inputs to the registration algorithm are the PET/CT and TRUS volumes as well as the prostate segmentations. PET/CT and TRUS volumes are first rigidly registered by maximizing the overlap between the segmented prostate binary masks. Three-dimensional anatomical landmarks are then automatically extracted from the boundary as well as within the prostate. Then, a deformable registration is performed using a regularized thin plate spline where the landmark localization error is optimized between the extracted landmarks that are in correspondence. The proposed algorithm was evaluated on 25 prostate cancer patients treated with low-dose-rate brachytherapy. We registered the postimplant CT to TRUS using the proposed algorithm and computed target registration errors (TREs) by comparing implanted seed locations. Our approach outperforms state-of-the-art methods, with significantly lower ( mean ± standard deviation ) TRE of 1.96 ± 1.29 mm while being computationally efficient (mean computation time of 38 s). The proposed landmark-based PET/CT-TRUS deformable registration algorithm is simple, computationally efficient, and capable of producing quality registration of the prostate boundary as well as the internal gland.
Collapse
Affiliation(s)
- Sharmin Sultana
- Johns Hopkins University, Department of Radiation Oncology and Molecular Radiation Sciences, Baltimore, Maryland, United States
| | - Daniel Y. Song
- Johns Hopkins University, Department of Radiation Oncology and Molecular Radiation Sciences, Baltimore, Maryland, United States
| | - Junghoon Lee
- Johns Hopkins University, Department of Radiation Oncology and Molecular Radiation Sciences, Baltimore, Maryland, United States
| |
Collapse
|
32
|
Ware RE, Williams S, Hicks RJ. Molecular Imaging of Recurrent and Metastatic Prostate Cancer. Semin Nucl Med 2019; 49:280-293. [DOI: 10.1053/j.semnuclmed.2019.02.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
33
|
Abstract
As described in more detail in other contributions in this issue of Seminars in Nuclear Medicine, prostate-specific membrane antigen (PSMA) has become one of the most promising molecular targets in nuclear medicine. Due to its overexpression on prostate cancer cells in proportion to the stage and grade of tumour progression, especially in androgen-independent, advanced and metastatic disease, various tracers for the detection and treatment of prostate cancer by means of radioligand imaging, radioligand therapy or radioguided surgery have been developed and transferred to clinical applications. Even though monoclonal antibodies were investigated and introduced as first PSMA-targeted probes, the inherent advantage of fast tumour uptake and rapid excretion of small molecules has shifted the research focus during the last decade to low molecular weight inhibitors with high affinity to PSMA, such as [18F]FDCFPyL, [18F]PSMA-1007, [68Ga]PSMA-HBED, [177Lu]PSMA-617, [177Lu]PSMA-I&T, [99mTc]MIP-1404 or [99mTc]PSMA I&S, to mention only a few. Due to the plethora of such PSMA probes described during the last years, this review aims to give an overview over the specific characteristics of those radiopharmaceuticals that have already found widespread clinical application. In addition, recently introduced concepts such as PSMA-tracers with increased plasma protein binding, are discussed.
Collapse
Affiliation(s)
- Hans-Jürgen Wester
- Chair of Pharmaceutical Radiochemistry, Walther-Meissner-Strasse 3, 85748 Garching, Germany.
| | - Margret Schottelius
- Chair of Pharmaceutical Radiochemistry, Walther-Meissner-Strasse 3, 85748 Garching, Germany
| |
Collapse
|
34
|
Diao W, Cai H, Chen L, Jin X, Liao X, Jia Z. Recent Advances in Prostate-Specific Membrane Antigen-Based Radiopharmaceuticals. Curr Top Med Chem 2019; 19:33-56. [PMID: 30706785 DOI: 10.2174/1568026619666190201100739] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 12/13/2018] [Accepted: 12/15/2018] [Indexed: 02/05/2023]
Abstract
BACKGROUND Prostate cancer (PCa) is the most common sex-related malignancy with high mortality in men worldwide. Prostate-specific membrane antigen (PSMA) is overexpressed on the surface of most prostate tumor cells and considered a valuable target for both diagnosis and therapy of prostate cancer. A series of radiolabeled agents have been developed based on the featured PSMA ligands in the previous decade and have demonstrated promising outcomes in clinical research of primary and recurrent PCa. Furthermore, the inspiring response and safety of lutetium-177-PSMA-617 (177Lu-PSMA-617) radiotherapy represent the potential for expanded therapeutic options for metastatic castration-resistant PCa. Retrospective cohort studies have revealed that radiolabeled PSMA agents are the mainstays of the current success, especially in detecting prostate cancer with metastasis and biochemical recurrence. OBJECTIVE This review is intended to present a comprehensive overview of the current literature on PSMA ligand-based agents for both radionuclide imaging and therapeutic approaches, with a focus on those that have been clinically adopted. CONCLUSION PSMA-based diagnosis and therapy hold great promise for improving the clinical management of prostate cancer.
Collapse
Affiliation(s)
- Wei Diao
- Department of Nuclear Medicine, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Huawei Cai
- Department of Nuclear Medicine, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Lihong Chen
- Department of Biochemistry & Molecular Biology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Xi Jin
- Institute of Urology, Department of Urology, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Xinyang Liao
- Institute of Urology, Department of Urology, West China Hospital, Sichuan University, 610041, Chengdu, China
| | - Zhiyun Jia
- Department of Nuclear Medicine, West China Hospital, Sichuan University, 610041, Chengdu, China
| |
Collapse
|
35
|
Can Local Ablative Radiotherapy Revert Castration-resistant Prostate Cancer to an Earlier Stage of Disease? Eur Urol 2019; 75:548-551. [DOI: 10.1016/j.eururo.2018.11.050] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 11/23/2018] [Indexed: 11/23/2022]
|
36
|
Smith CP, Laucis A, Harmon S, Mena E, Lindenberg L, Choyke PL, Turkbey B. Novel Imaging in Detection of Metastatic Prostate Cancer. Curr Oncol Rep 2019; 21:31. [DOI: 10.1007/s11912-019-0780-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
37
|
van der Toom EE, Axelrod HD, de la Rosette JJ, de Reijke TM, Pienta KJ, Valkenburg KC. Prostate-specific markers to identify rare prostate cancer cells in liquid biopsies. Nat Rev Urol 2019; 16:7-22. [PMID: 30479377 DOI: 10.1038/s41585-018-0119-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Despite improvements in early detection and advances in treatment, patients with prostate cancer continue to die from their disease. Minimal residual disease after primary definitive treatment can lead to relapse and distant metastases, and increasing evidence suggests that circulating tumour cells (CTCs) and bone marrow-derived disseminated tumour cells (BM-DTCs) can offer clinically relevant biological insights into prostate cancer dissemination and metastasis. Using epithelial markers to accurately detect CTCs and BM-DTCs is associated with difficulties, and prostate-specific markers are needed for the detection of these cells using rare cell assays. Putative prostate-specific markers have been identified, and an optimized strategy for staining rare cancer cells from liquid biopsies using these markers is required. The ideal prostate-specific marker will be expressed on every CTC or BM-DTC throughout disease progression (giving high sensitivity) and will not be expressed on non-prostate-cancer cells in the sample (giving high specificity). Some markers might not be specific enough to the prostate to be used as individual markers of prostate cancer cells, whereas others could be truly prostate-specific and would make ideal markers for use in rare cell assays. The goal of future studies is to use sensitive and specific prostate markers to consistently and reliably identify rare cancer cells.
Collapse
Affiliation(s)
| | - Haley D Axelrod
- The James Buchanan Brady Urological Institute, Baltimore, MD, USA.,Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | | | - Kenneth J Pienta
- The James Buchanan Brady Urological Institute, Baltimore, MD, USA
| | | |
Collapse
|
38
|
Rowe SP, Gorin MA, Pomper MG. Imaging of Prostate-Specific Membrane Antigen with Small-Molecule PET Radiotracers: From the Bench to Advanced Clinical Applications. Annu Rev Med 2019; 70:461-477. [DOI: 10.1146/annurev-med-062117-073027] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In recent years, small-molecule inhibitors of prostate-specific membrane antigen (PSMA) labeled with radionuclides that allow for positron emission tomography (PET) imaging have been extensively studied in many clinical contexts in men with prostate cancer (PCa). The high sensitivity and specificity of these agents for identifying sites of PCa has quickly led to their widespread adoption as a de facto clinical standard of care throughout much of the world. PSMA-targeted PET radiotracers have been particularly well-studied in preoperatively staging men with high-risk PCa, evaluating biochemical recurrence following definitive therapy, and guiding metastasis-directed therapy in patients suspected of having oligorecurrent/oligometastatic disease. Furthermore, the expression of PSMA on the tumor neovasculature of many nonprostate malignancies has enabled a burgeoning subfield concentrated on delineating the potential utility of PSMA-targeted PET agents for imaging other cancers. In this review, we highlight the preclinical development of key small molecules that are now being clinically utilized for PCa imaging, discuss the roles of PSMA-targeted agents in guiding patient management, and consider the role these compounds may play in imaging nonprostate cancers.
Collapse
Affiliation(s)
- Steven P. Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA;,
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| | - Michael A. Gorin
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA;,
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| | - Martin G. Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA;,
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21287, USA
| |
Collapse
|
39
|
|
40
|
Gennaro KH, Porter KK, Gordetsky JB, Galgano SJ, Rais-Bahrami S. Imaging as a Personalized Biomarker for Prostate Cancer Risk Stratification. Diagnostics (Basel) 2018; 8:diagnostics8040080. [PMID: 30513602 PMCID: PMC6316045 DOI: 10.3390/diagnostics8040080] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 11/13/2018] [Accepted: 11/15/2018] [Indexed: 02/07/2023] Open
Abstract
Biomarkers provide objective data to guide clinicians in disease management. Prostate-specific antigen serves as a biomarker for screening of prostate cancer but has come under scrutiny for detection of clinically indolent disease. Multiple imaging techniques demonstrate promising results for diagnosing, staging, and determining definitive management of prostate cancer. One such modality, multiparametric magnetic resonance imaging (mpMRI), detects more clinically significant disease while missing lower volume and clinically insignificant disease. It also provides valuable information regarding tumor characteristics such as location and extraprostatic extension to guide surgical planning. Information from mpMRI may also help patients avoid unnecessary biopsies in the future. It can also be incorporated into targeted biopsies as well as following patients on active surveillance. Other novel techniques have also been developed to detect metastatic disease with advantages over traditional computer tomography and magnetic resonance imaging, which primarily rely on defined size criteria. These new techniques take advantage of underlying biological changes in prostate cancer tissue to identify metastatic disease. The purpose of this review is to present literature on imaging as a personalized biomarker for prostate cancer risk stratification.
Collapse
Affiliation(s)
- Kyle H Gennaro
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Kristin K Porter
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Jennifer B Gordetsky
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Samuel J Galgano
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| | - Soroush Rais-Bahrami
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
41
|
Harmon SA, Bergvall E, Mena E, Shih JH, Adler S, McKinney Y, Mehralivand S, Citrin DE, Couvillon A, Madan RA, Gulley JL, Mease RC, Jacobs PM, Pomper MG, Turkbey B, Choyke PL, Lindenberg ML. A Prospective Comparison of 18F-Sodium Fluoride PET/CT and PSMA-Targeted 18F-DCFBC PET/CT in Metastatic Prostate Cancer. J Nucl Med 2018; 59:1665-1671. [PMID: 29602821 PMCID: PMC6225539 DOI: 10.2967/jnumed.117.207373] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 03/21/2018] [Indexed: 11/16/2022] Open
Abstract
The purpose of this study was to compare the diagnostic performance of 18F-DCFBC PET/CT, a first-generation 18F-labeled prostate-specific membrane antigen (PSMA)-targeted agent, and 18F-NaF PET/CT, a sensitive marker of osteoblastic activity, in a prospective cohort of patients with metastatic prostate cancer. Methods: Twenty-eight prostate cancer patients with metastatic disease on conventional imaging prospectively received up to 4 PET/CT scans. All patients completed baseline 18F-DCFBC PET/CT and 18F-NaF PET/CT scans, and 23 patients completed follow-up imaging, with a median follow-up interval of 5.7 mo (range, 4.2-12.6 mo). Lesion detection was compared across the 2 PET/CT agents at each time point. Detection and SUV characteristics of each PET/CT agent were compared with serum prostate-specific antigen (PSA) levels and treatment status at the time of baseline imaging using nonparametric statistical testing (Spearman correlation, Wilcoxon rank). Results: Twenty-six patients had metastatic disease detected on 18F-NaF or 18F-DCFBC at baseline, and 2 patients were negative on both scans. Three patients demonstrated soft tissue-only disease. Of 241 lesions detected at baseline, 56 were soft-tissue lesions identified by 18F-DCFBC only and 185 bone lesions detected on 18F-NaF or 18F-DCFBC. 18F-NaF detected significantly more bone lesions than 18F-DCFBC (P < 0.001). Correlation of PSA with patient-level SUV metrics was strong in 18F-DCFBC (ρ > 0.5, P < 0.01) and poor in 18F-NaF (ρ < 0.3, P > 0.1). When PSA levels were combined with treatment status, patients with below-median levels of PSA (<2 ng/mL) on androgen deprivation therapy (n = 11) demonstrated more lesions on 18F-NaF than 18F-DCFBC (P = 0.02). In PSA greater than 2 ng/mL, patients on androgen deprivation therapy (n = 8) showed equal to or more lesions on 18F-DCFBC than on 18F-NaF. Conclusion: The utility of PSMA-targeting imaging in metastatic prostate cancer appears to depend on patient disease course and treatment status. Compared with 18F-NaF PET/CT, 18F-DCFBC PET/CT detected significantly fewer bone lesions in the setting of early or metastatic castrate-sensitive disease on treatment. However, in advanced metastatic castrate-resistant prostate cancer, 18F-DCFBC PET/CT shows good concordance with NaF PET/CT.
Collapse
Affiliation(s)
- Stephanie A Harmon
- Clinical Research Directorate/Clinical Monitoring Research Program, Leidos Biomedical Research, Inc., National Cancer Institute, Campus at Frederick, Frederick, Maryland
| | - Ethan Bergvall
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Esther Mena
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Joanna H Shih
- Division of Cancer Treatment and Diagnosis: Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Stephen Adler
- Clinical Research Directorate/Clinical Monitoring Research Program, Leidos Biomedical Research, Inc., National Cancer Institute, Campus at Frederick, Frederick, Maryland
| | - Yolanda McKinney
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Sherif Mehralivand
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Deborah E Citrin
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Anna Couvillon
- Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Ravi A Madan
- Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - James L Gulley
- Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Ronnie C Mease
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Paula M Jacobs
- Cancer Imaging Program, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Martin G Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland; and
| | - Baris Turkbey
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Peter L Choyke
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - M Liza Lindenberg
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
42
|
Czarniecki M, Mena E, Lindenberg L, Cacko M, Harmon S, Radtke JP, Giesel F, Turkbey B, Choyke PL. Keeping up with the prostate-specific membrane antigens (PSMAs): an introduction to a new class of positron emission tomography (PET) imaging agents. Transl Androl Urol 2018; 7:831-843. [PMID: 30456186 PMCID: PMC6212618 DOI: 10.21037/tau.2018.08.03] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 07/31/2018] [Indexed: 12/11/2022] Open
Abstract
Prostate-specific membrane antigen (PSMA) targeted positron emission tomography (PET) is an emerging prostate cancer imaging method, which has been reported to have a higher sensitivity and specificity than the currently approved PET imaging agents. Multiple PSMA ligands are being investigated around the world and applications range from primary tumor characterization, to local staging, biochemical recurrence, metastasis, and image-guided interventions. The most investigated PET tracers are labelled with 68-Gallium or 18-Fluoride and are discussed in this review. Additionally, 99mTc labeled PSMA agents for single photon emission computed tomography (SPECT) imaging are elucidated as an alternative method of PSMA image acquisition.
Collapse
Affiliation(s)
- Marcin Czarniecki
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Esther Mena
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Liza Lindenberg
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Marek Cacko
- Department of Nuclear Medicine, Medical University Warsaw, Warsaw, Poland
| | - Stephanie Harmon
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Clinical Research Directorate/Clinical Monitoring Research Program, Frederick National Laboratory for Cancer Research sponsored by the National Cancer Institute, Frederick, MD, USA
| | - Jan Philipp Radtke
- Department of Urology, Heidelberg University Hospital, INF 400, 69120 Heidelberg, Germany
| | - Frederick Giesel
- Department of Nuclear Medicine, Heidelberg University Hospital, INF 400, 69120 Heidelberg, Germany
| | - Baris Turkbey
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Peter L. Choyke
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
43
|
Pesapane F, Czarniecki M, Suter MB, Turkbey B, Villeirs G. Imaging of distant metastases of prostate cancer. Med Oncol 2018; 35:148. [DOI: 10.1007/s12032-018-1208-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 09/06/2018] [Indexed: 02/06/2023]
|
44
|
Yin Y, Werner RA, Higuchi T, Lapa C, Pienta KJ, Pomper MG, Gorin MA, Rowe SP. Follow-up of Lesions with Equivocal Radiotracer Uptake on PSMA-Targeted PET in Patients with Prostate Cancer: Predictive Values of the PSMA-RADS-3A and PSMA-RADS-3B Categories. J Nucl Med 2018; 60:511-516. [PMID: 30190303 DOI: 10.2967/jnumed.118.217653] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 08/28/2018] [Indexed: 12/21/2022] Open
Abstract
Prostate-specific membrane antigen (PSMA)-targeted PET imaging has become commonly used in patients with prostate cancer (PCa). The PSMA reporting and data system version 1.0 (PSMA-RADS version 1.0) categorizes lesions on the basis of the likelihood of PCa involvement, with PSMA-RADS-3A (soft-tissue) and PSMA-RADS-3B (bone) lesions being indeterminate for the presence of disease. We retrospectively reviewed the imaging follow-up of such lesions to determine the rate at which they underwent changes suggestive of underlying PCa. Methods: PET/CT imaging with 18F-DCFPyL was performed in 110 patients with PCa, and lesions were categorized according to PSMA-RADS version 1.0. The study reported herein is a retrospective analysis of those patients. Fifty-six of 110 (50.9%) patients were determined to have indeterminate PSMA-RADS-3A or PSMA-RADS-3B lesions, and 22 of 56 (39.3%) patients had adequate follow-up to be included in the analysis (median follow-up time was 10 mo [range, 3-22 mo]). The SUVmax of the lesions was obtained, and the ratios of SUVmax of the lesions to SUVmean of blood pool (SUVmax-lesion/SUVmean-bloodpool) were calculated. Predetermined criteria were used to evaluate the PSMA-RADS-3A and PSMA-RADS-3B lesions on follow-up imaging to determine whether they demonstrated evidence of underlying malignancy. Results: A total of 46 lesions in 22 patients were considered indeterminate for PCa (i.e., PSMA-RADS-3A [32 lesions] or PSMA-RADS-3B [14 lesions]) and were evaluable on follow-up imaging. Twenty-seven of 46 (58.7%) lesions demonstrated changes suggesting they were true-positive for PCa. These lesions included 24 of 32 (75.0%) PSMA-RADS-3A lesions and 3 of 14 (21.4%) lesions categorized as PSMA-RADS-3B. The ranges of SUVmax and SUVmax-lesion/SUVmean-bloodpool overlapped between those lesions demonstrating changes consistent with malignancy on follow-up imaging and those lesions that remained unchanged on follow-up. The presence of additional definitive sites of PCa (PSMA-RADS-4 and PSMA-RADS-5) increases the likelihood that indeterminate lesions will manifest as true-positive on follow-up imaging. Conclusion: PSMA-RADS-3A and PSMA-RADS-3B lesions are truly indeterminate in that proportions of findings in both categories demonstrate evidence of malignancy on follow-up imaging. Overall, PSMA-RADS-3A lesions are more likely than PSMA-RADS-3B lesions to represent sites of PCa, and this information should be considered when guiding patient therapy.
Collapse
Affiliation(s)
- Yafu Yin
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Nuclear Medicine, The First Hospital of China Medical University, Shenyang, China.,Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rudolf A Werner
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland.,Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany; and
| | - Takahiro Higuchi
- Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany; and
| | - Constantin Lapa
- Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany; and
| | - Kenneth J Pienta
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Martin G Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland.,The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michael A Gorin
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland.,The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Steven P Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland .,The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
45
|
Bauman G, Martin P, Thiessen JD, Taylor R, Moussa M, Gaed M, Rachinsky I, Kassam Z, Chin J, Pautler S, Lee TY, Valliant JF, Ward A. [18F]-DCFPyL Positron Emission Tomography/Magnetic Resonance Imaging for Localization of Dominant Intraprostatic Foci: First Experience. Eur Urol Focus 2018; 4:702-706. [DOI: 10.1016/j.euf.2016.10.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 10/01/2016] [Accepted: 10/10/2016] [Indexed: 11/16/2022]
|
46
|
Galgano SJ, Valentin R, McConathy J. Role of PET imaging for biochemical recurrence following primary treatment for prostate cancer. Transl Androl Urol 2018; 7:S462-S476. [PMID: 30363475 PMCID: PMC6178324 DOI: 10.21037/tau.2018.06.09] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Prostate cancer is one of the most common cancers in men worldwide, and primary prostate cancer is typically treated with surgery, radiation, androgen deprivation, or a combination of these therapeutic modalities. Despite technical advances, approximately 30% of men will experience biochemical recurrent within 10 years of definitive treatment. Upon detection of a rise in serum prostate specific antigen (PSA), there is great need to accurately stage these patients to help guide further therapy. As a result, there are considerable efforts underway to establish the role of positron emission tomography (PET) in the diagnostic algorithm of biochemically recurrent prostate cancer. This manuscript provides an overview of PET tracers used for the detection and localization of prostate cancer in the setting of biochemical recurrence with a focus on PET tracers that are currently being used in clinical practice in the United States.
Collapse
Affiliation(s)
- Samuel J Galgano
- Department of Radiology, Section of Molecular Imaging and Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Roberto Valentin
- Department of Radiology, Section of Molecular Imaging and Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jonathan McConathy
- Department of Radiology, Section of Molecular Imaging and Therapeutics, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
47
|
Rauscher I, Eiber M, Weber WA, Gschwend JE, Horn T, Maurer T. Positron-emission tomography imaging in urological oncology: Current aspects and developments. Int J Urol 2018; 25:912-921. [DOI: 10.1111/iju.13779] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 07/17/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Isabel Rauscher
- Department of Nuclear Medicine; Klinikum rechts der Isar; Technical University of Munich; Munich Germany
| | - Matthias Eiber
- Department of Nuclear Medicine; Klinikum rechts der Isar; Technical University of Munich; Munich Germany
| | - Wolfgang A Weber
- Department of Nuclear Medicine; Klinikum rechts der Isar; Technical University of Munich; Munich Germany
| | - Jürgen E Gschwend
- Department of Urology; Klinikum rechts der Isar; Technical University of Munich; Munich Germany
| | - Thomas Horn
- Department of Urology; Klinikum rechts der Isar; Technical University of Munich; Munich Germany
| | - Tobias Maurer
- Department of Urology; Klinikum rechts der Isar; Technical University of Munich; Munich Germany
| |
Collapse
|
48
|
Current and potential future role of PSMA-PET in patients with castration-resistant prostate cancer. World J Urol 2018; 37:457-467. [PMID: 30030659 DOI: 10.1007/s00345-018-2408-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 07/12/2018] [Indexed: 12/13/2022] Open
Abstract
PURPOSE To review the current literature and discuss potential future roles of the novel positron emission tomography (PET) tracers targeting the prostate-specific membrane antigen (PSMA) in patients with castration-resistant prostate cancer (CRPC). METHODS A literature search on February 19th 2018 was conducted using the Medline database and www.clinicaltrials.gov . Additionally, illustrative cases of CRPC patients from our own institution who were restaged and treated based on PSMA-PET scan results are provided. RESULTS 11 Studies met the inclusion criteria. PSMA-PET detected more metastatic lesions compared to conventional bone scan. Several patients were up-staged from non-metastatic CRPC (nmCRPC) to metastatic CRPC (mCRPC). Currently, no clear consensus exists regarding treatment response assessment in PSMA-PET scans for mCRPC patients undergoing treatment. Also, the role of PSMA-PET as a gatekeeper for systemic therapy or radioligands is currently undefined. PSMA-guided metastasis-directed radiotherapy may not only alleviate local symptoms but has the potential to defer systemic treatment in patients with oligoprogressive CRPC. CONCLUSION Compared to bone scan, PSMA-PET is more sensitive and specific to detect metastases but the therapeutic consequences of PSMA-PET results in the setting of CRPC remain unclear. Until future studies define the role of PSMA-PET in patients with CRPC, the current standard for imaging remains bone scan and computerized tomography.
Collapse
|
49
|
Kulkarni HR, Singh A, Langbein T, Schuchardt C, Mueller D, Zhang J, Lehmann C, Baum RP. Theranostics of prostate cancer: from molecular imaging to precision molecular radiotherapy targeting the prostate specific membrane antigen. Br J Radiol 2018; 91:20180308. [PMID: 29762048 DOI: 10.1259/bjr.20180308] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Alterations at the molecular level are a hallmark of cancer. Prostate cancer is associated with the overexpression of prostate-specific membrane antigen (PSMA) in a majority of cases, predominantly in advanced tumors, increasing with the grade or Gleason's score. PSMA can be selectively targeted using radiolabeled PSMA ligands. These small molecules binding the PSMA can be radiolabeled with γ-emitters like 99mTc and 111In or positron emitters like 68Ga and 18F for diagnosis as well as with their theranostic pairs such as 177Lu (β-emitter) or 225Ac (α-emitter) for therapy. This review summarizes the theranostic role of PSMA ligands for molecular imaging and targeted molecular radiotherapy, moving towards precision oncology.
Collapse
Affiliation(s)
- Harshad R Kulkarni
- 1 Theranostics Center for Molecular Radiotherapy and Molecular Imaging, Zentralklinik Bad Berka , Bad Berka , Germany
| | - Aviral Singh
- 1 Theranostics Center for Molecular Radiotherapy and Molecular Imaging, Zentralklinik Bad Berka , Bad Berka , Germany
| | - Thomas Langbein
- 1 Theranostics Center for Molecular Radiotherapy and Molecular Imaging, Zentralklinik Bad Berka , Bad Berka , Germany
| | - Christiane Schuchardt
- 1 Theranostics Center for Molecular Radiotherapy and Molecular Imaging, Zentralklinik Bad Berka , Bad Berka , Germany
| | - Dirk Mueller
- 1 Theranostics Center for Molecular Radiotherapy and Molecular Imaging, Zentralklinik Bad Berka , Bad Berka , Germany
| | - Jingjing Zhang
- 1 Theranostics Center for Molecular Radiotherapy and Molecular Imaging, Zentralklinik Bad Berka , Bad Berka , Germany
| | - Coline Lehmann
- 1 Theranostics Center for Molecular Radiotherapy and Molecular Imaging, Zentralklinik Bad Berka , Bad Berka , Germany
| | - Richard P Baum
- 1 Theranostics Center for Molecular Radiotherapy and Molecular Imaging, Zentralklinik Bad Berka , Bad Berka , Germany
| |
Collapse
|
50
|
Heterogeneous Flare in Prostate-specific Membrane Antigen Positron Emission Tomography Tracer Uptake with Initiation of Androgen Pathway Blockade in Metastatic Prostate Cancer. Eur Urol Oncol 2018; 1:78-82. [DOI: 10.1016/j.euo.2018.03.010] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 03/15/2018] [Indexed: 11/22/2022]
|