1
|
Pio F, Murdock A, Fuller RE, Whalen MJ. The Role of Whole-Gland and Focal Cryotherapy in Recurrent Prostate Cancer. Cancers (Basel) 2024; 16:3225. [PMID: 39335196 PMCID: PMC11430134 DOI: 10.3390/cancers16183225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/11/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
Prostate cancer is the most common non-cutaneous malignancy in men, with the majority of newly diagnosed patients eligible for active surveillance. Despite definitive treatment, a considerable percentage of men will experience biochemical recurrence and even regional and distant metastatic recurrence after radiation therapy or radical prostatectomy. Salvage prostatectomy, while oncologically effective, poses significant morbidity with poor functional outcomes. Salvage cryotherapy has emerged as a promising alternative for localized recurrence, demonstrating safety and efficacy. This review examines the oncologic and functional outcomes of whole-gland and focal salvage cryotherapy, including disease-free survival, cancer-specific survival, and overall survival. The crucial role of multiparametric prostate MRI and evolving role of next-generation PSMA-targeted PET imaging are also examined. The comparison of outcomes of cryotherapy to other salvage ablation modalities, such as high-intensity focused ultrasound (HIFU), is also explored.
Collapse
Affiliation(s)
- Faozia Pio
- Department of Urology, George Washington University School of Medicine, 2300 I St NW, Washington, DC 20052, USA
| | - Andeulazia Murdock
- Department of Urology, George Washington University School of Medicine, 2300 I St NW, Washington, DC 20052, USA
| | - Renee E Fuller
- Department of Urology, George Washington University School of Medicine, 2300 I St NW, Washington, DC 20052, USA
| | - Michael J Whalen
- Department of Urology, George Washington University School of Medicine, 2300 I St NW, Washington, DC 20052, USA
| |
Collapse
|
2
|
Saripalli AL, Venkatesulu BP, Nickols NG, Valle LF, Harkenrider MM, Kishan AU, Solanki AA. Systematic review and recommendations for re-irradiation for intraprostatic radiorecurrent prostate cancer after definitive radiation therapy. World J Urol 2024; 42:520. [PMID: 39264453 DOI: 10.1007/s00345-024-05205-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 07/17/2024] [Indexed: 09/13/2024] Open
Abstract
PURPOSE Intraprostatic recurrence (IRR) of prostate cancer after radiation therapy is increasingly identified. Our objective was to review the literature to determine the optimal workup for identifying IRR, the management options, and practical considerations for the delivery of re-irradiation as salvage local therapy. METHODS We performed a systematic review of available publications and ongoing studies on the topics of IRR, with a focus on salvage re-irradiation. RESULTS Work up of biochemically recurrent prostate cancer includes PSMA PET/CT and multiparametric MRI, followed by biopsy to confirm IRR. Management options include continued surveillance, palliative hormonal therapy, and salvage local therapy. Salvage local therapy can be delivered using re-irradiation with low dose rate brachytherapy, high dose rate (HDR) brachytherapy, and stereotactic body radiotherapy (SBRT), as well as non-radiation modalities, such as cryotherapy, high-intensity focused ultrasound, irreversible electroporation and radical prostatectomy. Data demonstrate that HDR brachytherapy and SBRT have similar efficacy compared to the other salvage local therapy modalities, while having more favorable side effect profiles. Recommendations for radiation therapy planning and delivery using HDR and SBRT based on the available literature are discussed. CONCLUSION Salvage re-irradiation is safe and effective and should be considered in patients with IRR.
Collapse
Affiliation(s)
- Anjali L Saripalli
- Department of Radiation Oncology, Stritch School of Medicine, Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL, USA
| | - Bhanu Prasad Venkatesulu
- Department of Radiation Oncology, Stritch School of Medicine, Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL, USA
| | - Nicholas G Nickols
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, USA
- Department of Radiation Oncology, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, USA
| | - Luca F Valle
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, USA
- Department of Radiation Oncology, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, USA
| | - Matthew M Harkenrider
- Department of Radiation Oncology, Stritch School of Medicine, Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL, USA
| | - Amar U Kishan
- Department of Radiation Oncology, University of California Los Angeles, Los Angeles, CA, USA
| | - Abhishek A Solanki
- Department of Radiation Oncology, Stritch School of Medicine, Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL, USA.
| |
Collapse
|
3
|
Beyhan E, Turan Bektaş C, Baloğlu MC, Tosunoğlu Z, Çermik TF. Bilateral Adrenal Metastasis of Prostatic Adenocarcinoma on 68 Ga-PSMA PET/CT Imaging. Clin Nucl Med 2024; 49:e324-e326. [PMID: 38689449 DOI: 10.1097/rlu.0000000000005248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
ABSTRACT An 84-year-old man with prostate adenocarcinoma underwent 68 Ga-PSMA PET/CT due to PSA recurrence. Foci of 68 Ga-PSMA uptake were observed in bilateral adrenal glands. Adrenal MRI showed metastasis only in the left adrenal gland. Metastatic 68 Ga-PSMA uptake was also observed in the mediastinum and bone. Enzalutamide treatment was started. Follow-up 68 Ga-PSMA PET/CT scan showed regression in both adrenal gland metastases and other metastases.
Collapse
Affiliation(s)
- Ediz Beyhan
- From the Clinic of Nuclear Medicine, Istanbul Training and Research Hospital
| | - Ceyda Turan Bektaş
- Clinic of Radiology, Istanbul Training and Research Hospital, Istanbul, Turkey
| | - Mehmet Can Baloğlu
- From the Clinic of Nuclear Medicine, Istanbul Training and Research Hospital
| | - Zehranur Tosunoğlu
- From the Clinic of Nuclear Medicine, Istanbul Training and Research Hospital
| | | |
Collapse
|
4
|
Moul JW, Shore ND, Pienta KJ, Czernin J, King MT, Freedland SJ. Application of next-generation imaging in biochemically recurrent prostate cancer. Prostate Cancer Prostatic Dis 2024; 27:202-211. [PMID: 37679601 PMCID: PMC11096127 DOI: 10.1038/s41391-023-00711-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 09/09/2023]
Abstract
BACKGROUND Biochemical recurrence (BCR) following primary interventional treatment occurs in approximately one-third of patients with prostate cancer (PCa). Next-generation imaging (NGI) can identify local and metastatic recurrence with greater sensitivity than conventional imaging, potentially allowing for more effective interventions. This narrative review examines the current clinical evidence on the utility of NGI for patients with BCR. METHODS A search of PubMed was conducted to identify relevant publications on NGI applied to BCR. Given other relevant recent reviews on the topic, this review focused on papers published between January 2018 to May 2023. RESULTS NGI technologies, including positron emission tomography (PET) radiotracers and multiparametric magnetic resonance imaging, have demonstrated increased sensitivity and selectivity for diagnosing BCR at prostate-specific antigen (PSA) concentrations <2.0 ng/ml. Detection rates range between 46% and 50%, with decreasing PSA levels for choline (1-3 ng/ml), fluciclovine (0.5-1 ng/ml), and prostate-specific membrane antigen (0.2-0.49 ng/ml) PET radiotracers. Expert working groups and European and US medical societies recommend NGI for patients with BCR. CONCLUSIONS Available data support the improved detection performance and selectivity of NGI modalities versus conventional imaging techniques; however, limited clinical evidence exists demonstrating the application of NGI to treatment decision-making and its impact on patient outcomes. The emergence of NGI and displacement of conventional imaging may require a reexamination of the current definitions of BCR, altering our understanding of early recurrence. Redefining the BCR disease state by formalizing the role of NGI in patient management decisions will facilitate greater alignment across research efforts and better reflect the published literature.
Collapse
Affiliation(s)
- Judd W Moul
- Duke Cancer Institute and Division of Urology, Duke University, Durham, NC, USA
| | - Neal D Shore
- Carolina Urologic Research Center, Myrtle Beach, SC, USA
| | | | - Johannes Czernin
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Martin T King
- Brigham and Women's Hospital and Dana-Farber Cancer Institute, Boston, MA, USA
| | - Stephen J Freedland
- Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Veterans Affairs Medical Center, Durham, NC, USA.
| |
Collapse
|
5
|
Wang H, Zhu H, Li G, Dai J, Huang H, Jia Q. Effect of 18F-DCFPyL PET on changes in management of patients with prostate cancer: a systematic review and meta-analysis. Front Med (Lausanne) 2024; 11:1355236. [PMID: 38725467 PMCID: PMC11079165 DOI: 10.3389/fmed.2024.1355236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/15/2024] [Indexed: 05/12/2024] Open
Abstract
Purpose Prostate-specific membrane antigen (PSMA)-targeted imaging has gained increasing interest in its application in prostate cancer lesion detection. Compared with 68Galium (68Ga), 18Fluoride (18F)-labeled imaging agent has easier syntheses, lower price, and a longer half-time. 2-(3-{1-Carboxy-5-[(6-[18F]fluoro-pyridine-3-carbonyl)-amino]-pentyl}-ureido)-pentanedioic acid positron emission tomography (18F-DCFPyL PET) has been recently approved by the U.S. Food and Drug Administration. Several studies have proven its superiority to conventional imaging techniques in detecting prostate cancer lesions. However, the impact of 18F-DCFPyL PET on the management of patients with prostate cancer is not well established. Thus, we performed a systematic review and meta-analysis of available data to evaluate the impact of 18F-DCFPyL PET on the management of patients with prostate cancer. Methods The PubMed, Embase, Scopus, and Cochrane databases were searched up to April 2024. Studies that reported the proportion of changes in management after 18F-DCFPyL PET was performed in patients with prostate cancer were included. The Grading of Recommendations Assessment, Development, and Evaluation system was used for the quality evaluation of the included studies. The proportion of changes in management was pooled using a random effects model. Meta-regression analyses were performed to assess the potential correlation between the PET positivity and management changes. Results Fourteen studies (3,078 patients with prostate cancer) were included in our review and analysis. The pooled percentage of management changes was 43.5% (95% confidence interval [CI]: 33-54%). In patients with biochemical recurrent and for primary staging, the pooled percentage was 50% (95% CI: 39-60%) and 22% (95% CI: 15-29%), respectively. In the meta-regression analyses, PET positivity was detected as a significant predictor of management change (p = 0.0023). Conclusion 18F-DCFPyL PET significantly affects the management of patients with prostate cancer. Higher PET positivity rate significantly correlated with a higher proportion of management changes in patients with prostate cancer. However, more studies are still needed to confirm the important role of 18F-DCFPyL PET in the management of prostate cancer. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/#myprospero, CRD42022339178.
Collapse
Affiliation(s)
- Hui Wang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - HongMei Zhu
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - GuanNan Li
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - JiaoNa Dai
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - HeXiao Huang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Qiong Jia
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| |
Collapse
|
6
|
Sciarra A, Santarelli V, Salciccia S, Moriconi M, Basile G, Santodirocco L, Carino D, Frisenda M, Di Pierro G, Del Giudice F, Gentilucci A, Bevilacqua G. How the Management of Biochemical Recurrence in Prostate Cancer Will Be Modified by the Concept of Anticipation and Incrementation of Therapy. Cancers (Basel) 2024; 16:764. [PMID: 38398155 PMCID: PMC10886975 DOI: 10.3390/cancers16040764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/06/2024] [Accepted: 02/10/2024] [Indexed: 02/25/2024] Open
Abstract
Biochemical recurrence (BCR) after primary treatments for prostate cancer (PC) is an extremely heterogeneous phase and at least a stratification into low- and high-risk cases for early progression in metastatic disease is necessary. At present, PSA-DT represents the best parameter to define low- and high-risk BCR PC, but real precision medicine is strongly suggested to define tailored management for patients with BCR. Before defining management, it is necessary to exclude the presence of low-volume metastasis associated with PSA progression using new-generation imaging, preferably with PSMA PET/CT. Low-risk BCR cases should be actively observed without early systemic therapies. Early treatment of low-risk BCR with continuous androgen deprivation therapy (ADT) can produce disadvantages such as the development of castration resistance before the appearance of metastases (non-metastatic castration-resistant PC). Patients with high-risk BCR benefit from early systemic therapy. Even with overall survival (OS) as the primary treatment endpoint, metastasis-free survival (MFS) should be used as a surrogate endpoint in clinical trials, especially in long survival stages of the disease. The EMBARK study has greatly influenced the management of high-risk BCR, by introducing the concept of anticipation and intensification through the use of androgen receptor signaling inhibitors (ARSIs) and ADT combination therapy. In high-risk (PSA-DT ≤ 9 months) BCR cases, the combination of enzalutamide with leuprolide significantly improves MFS when compared to leuprolide alone, maintaining an unchanged quality of life in the asymptomatic phase of the disease. The possibility of using ARSIs alone in this early disease setting is suggested by the EMBARK study (arm with enzalutamide alone) with less evidence than with the intensification of the combination therapy. Continued use versus discontinuation of enzalutamide plus leuprolide intensified therapy upon reaching undetectable PSA levels needs to be better defined with further analysis. Real-world analysis must verify the significant results obtained in the context of a phase 3 study.
Collapse
Affiliation(s)
- Alessandro Sciarra
- Department Materno Infantile e Scienze Urologiche, Sapienza University, Viale Policlinico 155, 00161 Rome, Italy; (V.S.); (S.S.); (M.M.); (G.B.); (L.S.); (D.C.); (M.F.); (G.D.P.); (F.D.G.); (A.G.); (G.B.)
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Costa LB, Moreira R, Gaspar PR, de Galiza Barbosa F. Prostate-Specific Membrane Antigen PET/Computed Tomography: Pearls and Pitfalls. Radiol Clin North Am 2024; 62:161-175. [PMID: 37973240 DOI: 10.1016/j.rcl.2023.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2023]
Abstract
Prostate-specific membrane antigen PET (PSMA-PET) has emerged as a powerful imaging tool for prostate cancer primary staging, biochemical recurrence, and advanced disease assessment. This article offers a concise overview of the benefits and challenges associated with PSMA-PET for prostate cancer evaluation. The article highlights the advantages of PSMA-PET over conventional imaging, such as its higher sensitivity and specificity for detecting metastases, and the potential for guiding personalized treatment decisions. However, it also explores the limitations and potential pitfalls for interpretation. Overall, the article aims to provide valuable insights for clinicians and diagnostic imaging physicians in clinical practice.
Collapse
Affiliation(s)
- Larissa Bastos Costa
- Radiology and Nuclear Medicine Department, Hospital Sirio Libanes, Rua Adma Jafet 91, São Paulo, Brazil; Radiology and Nuclear Medicine Department, Americas Group, Rua Tupi 535, São Paulo, Brazil
| | - Renata Moreira
- Radiology and Nuclear Medicine Department, Casa de Saúde São José, R. Macedo Sobrinho, 21 - Humaitá, Rio de Janeiro 22271-080, Brazil
| | - Priscilla Romano Gaspar
- Nuclear Medicine Department, Hospital Vitória (Americas Group) and Hospital de Força Aérea do Galeão, Avenida Jorge Curry 550, Rio de Janeiro, Brazil
| | - Felipe de Galiza Barbosa
- Radiology and Nuclear Medicine Department, Hospital Sirio Libanes, Rua Adma Jafet 91, São Paulo, Brazil; Radiology and Nuclear Medicine Department, Americas Group, Rua Tupi 535, São Paulo, Brazil.
| |
Collapse
|
8
|
Pant V, Vinjamuri S, Zanial AZ, Naeem F. Lessons from a 3-Year Review of PSMA PET-CT in a Tertiary Setting: Can We Fine Tune Referral Criteria by Identifying Factors Predicting Positivity and Negativity? Diagnostics (Basel) 2023; 13:2542. [PMID: 37568904 PMCID: PMC10417573 DOI: 10.3390/diagnostics13152542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
AIM OF THE STUDY To draw inferences from a retrospective evaluation of PSMA PET CT scans performed for the evaluation of biochemical recurrence. MATERIAL AND METHODS A retrospective analysis of 295 PSMA PET CT scans spanning 3 years between 2020 and 2022 was undertaken. RESULTS Of 295 PET CT scans, 179 were positive, 66 were negative and 50 had indeterminate findings. In the positive group, 67 had radical prostatectomy and PSMA avid lesions were seen most commonly in pelvic lymph nodes. The remaining 112 positive scans were in the non-radical prostatectomy group; 25 had recurrence only in the prostate, 17 had recurrence involving the prostate bed; 28 had no recurrence in the prostate gland, while 42 had recurrence in the prostate as well as in extra-prostatic sites. Overall, in the non-prostatectomy group, 75% of the population was harboring a PSMA avid lesion in the prostate gland while in the remaining 25% of the population, recurrence did not involve the prostate gland. The majority of indeterminate findings were seen in small pelvic or retroperitoneal lymph nodes or skeletal regions (ribs/others) and in nine patients indeterminate focus was seen in the prostate bed only. Follow-up PSMA PET CT was helpful in prior indeterminate findings and unexplained PSA rise. CONCLUSION A higher recurrence in the prostate bed while evaluating biochemical recurrence prompts the following: question: should prostatectomy be offered more proactively? Follow-up PSMA PET CT is helpful for indeterminate findings; a PSA rise of 0.7 ng/mL in 6 months can result in positive PSMA PET CT while negative scans can be seen up to a 2 ng/mL PSA rise in 6 months.
Collapse
Affiliation(s)
- Vineet Pant
- Department of Nuclear Medicine, Royal Liverpool University Hospital, Liverpool L78XP, UK; (S.V.); (F.N.)
| | - Sobhan Vinjamuri
- Department of Nuclear Medicine, Royal Liverpool University Hospital, Liverpool L78XP, UK; (S.V.); (F.N.)
| | - Ahmad Zaid Zanial
- Department of Nuclear Medicine, General Hospital, Kuala Lumpur 50586, Malaysia;
| | - Faisal Naeem
- Department of Nuclear Medicine, Royal Liverpool University Hospital, Liverpool L78XP, UK; (S.V.); (F.N.)
| |
Collapse
|
9
|
Beyer T, Czernin J, Freudenberg L, Giesel F, Hacker M, Hicks RJ, Krause BJ. A 2022 International Survey on the Status of Prostate Cancer Theranostics. J Nucl Med 2023; 64:47-53. [PMID: 35953304 DOI: 10.2967/jnumed.122.264298] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/29/2022] [Accepted: 06/29/2022] [Indexed: 01/07/2023] Open
Abstract
Growing interest in PSMA imaging using [68Ga]- or [18F]-labeled ligands and PSMA-based radioligand therapy (RLT) of prostate cancer (PCa) prompted us to survey the global community on their experiences and expectations. Methods: A web-based survey was composed to interrogate areas specific to PET imaging, the clinical value chain, and RLT applications. International responses were collected in early 2022. In total, over 300 valid responses were received and evaluated. Results: Most responses (83%) were given by nuclear medicine specialists with extensive experience in PET. At 22% of sites, PCa ranked "top" in cancer-type-specific PET indications, with an average and median of 15% and 10% of all cases, respectively. The most frequently used PSMA PET tracers were [68Ga]PSMA (32%) and [18F]PSMA-1007 (31%). Users reported a steady growth in PSMA PET and RLT over the past 5 y, averaging 50% and 82%, respectively, with a further 100% median growth projected over the next 5 y. Of note, more respondents indicated cognizance of personalized dosimetry than actually used it routinely. The most commonly identified barriers to future growth in PCa theranostics were radiopharmaceutical supply, reimbursement, staff availability, and buy-in of medical oncologists. Conclusion: Despite enthusiasm, this survey indicates variable adoption of PSMA imaging and RLT globally. Several challenges need to be addressed by the medical community, authorities, and patient advocacy groups in integrating PSMA-targeted theranostics into personalized medicine.
Collapse
Affiliation(s)
- Thomas Beyer
- QIMP Team, Centre Medical Physics and Biomedical Engineering, Medical University Vienna, Vienna, Austria
| | - Johannes Czernin
- Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California
| | | | - Frederik Giesel
- Department of Nuclear Medicine, Medical Faculty, Heinrich-Heine-University and University Hospital Duesseldorf, Duesseldorf, Germany; Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Marcus Hacker
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University Vienna, Vienna, Austria
| | - Rodney J Hicks
- The Department of Medicine, St Vincent's Hospital, the University of Melbourne, Melbourne, Australia; and
| | - Bernd J Krause
- Rostock University Medical Centre, Department of Nuclear Medicine, Rostock, Germany
| |
Collapse
|
10
|
Alghazo O, O'Callaghan M. PSMA-PET to detect prostate cancer recurrence after radiotherapy. Nat Rev Urol 2023; 20:5-6. [PMID: 36316467 DOI: 10.1038/s41585-022-00672-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Omar Alghazo
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia.
- Royal Darwin Hospital, Tiwi, Northern Territory, Australia.
| | - Michael O'Callaghan
- College of Medicine and Public Health, Flinders University, Adelaide, South Australia, Australia
- Urology Unit, Flinders Medical Centre, Bedford Park, South Australia, Australia
- Discipline of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
11
|
Roberts MJ, Maurer T, Perera M, Eiber M, Hope TA, Ost P, Siva S, Hofman MS, Murphy DG, Emmett L, Fendler WP. Using PSMA imaging for prognostication in localized and advanced prostate cancer. Nat Rev Urol 2023; 20:23-47. [PMID: 36473945 DOI: 10.1038/s41585-022-00670-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2022] [Indexed: 12/12/2022]
Abstract
The use of prostate-specific membrane antigen (PSMA)-directed applications in modern prostate cancer management has evolved rapidly over the past few years, helping to establish new treatment pathways and provide further insights into prostate cancer biology. However, the prognostic implications of PSMA-PET have not been studied systematically, owing to rapid clinical implementation without long follow-up periods to determine intermediate-term and long-term oncological outcomes. Currently available data suggest that traditional prognostic factors and survival outcomes are associated with high PSMA expression (both according to immunohistochemistry and PET uptake) in men with localized and biochemically recurrent disease. Treatment with curative intent (primary and/or salvage) often fails when PSMA-positive metastases are present; however, the sensitivity of PSMA-PET in detecting all metastases is poor. Low PSMA-PET uptake in recurrent disease is a favourable prognostic factor; however, it can be associated with poor prognosis in conjunction with high 18F-fluorodeoxyglucose uptake in metastatic castration-resistant prostate cancer. Clinical trials embedding PSMA-PET for guiding management with reliable oncological outcomes are needed to support ongoing clinical use.
Collapse
Affiliation(s)
- Matthew J Roberts
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia.
- University of Queensland Centre for Clinical Research, Faculty of Medicine, Brisbane, Queensland, Australia.
- Department of Urology, Redcliffe Hospital, Brisbane, Queensland, Australia.
| | - Tobias Maurer
- Martini-Klinik Prostate Cancer Center, Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Marlon Perera
- Department of Surgery, Austin Health, Heidelberg, Victoria, Australia
| | - Matthias Eiber
- Department of Nuclear Medicine, Technical University of Munich, Munich, Germany
| | - Thomas A Hope
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Piet Ost
- Department of Radiation Oncology, Iridium Network, GZA Ziekenhuizen, Antwerp, Belgium
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Shankar Siva
- Peter MacCallum Cancer Centre, Radiation Oncology, Parkville, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, Melbourne University, Parkville, Victoria, Australia
| | - Michael S Hofman
- Sir Peter MacCallum Department of Oncology, Melbourne University, Parkville, Victoria, Australia
- Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Declan G Murphy
- Sir Peter MacCallum Department of Oncology, Melbourne University, Parkville, Victoria, Australia
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Louise Emmett
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, New South Wales, Australia
- Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University of Duisburg-Essen, Essen, Germany
- PET Committee of the German Society of Nuclear Medicine, Goettingen, Germany
| |
Collapse
|
12
|
Li B, Duan L, Shi J, Han Y, Wei W, Cheng X, Cao Y, Kader A, Ding D, Wu X, Gao Y. Diagnostic performance of 99mTc-HYNIC-PSMA SPECT/CT for biochemically recurrent prostate cancer after radical prostatectomy. Front Oncol 2022; 12:1072437. [PMID: 36568205 PMCID: PMC9768541 DOI: 10.3389/fonc.2022.1072437] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022] Open
Abstract
Objectives 99mTc-HYNIC-PSMA is a novel technetium-99m-labeled small-molecule inhibitor of prostate-specific membrane antigen (PSMA) for detection of prostate cancer. The present study investigated the diagnostic yield of 99mTc-HYNIC-PSMA Single photon emission computed tomography (SPECT)/CT in 147 patients with biochemically recurrent prostate cancer after radical prostatectomy. Methods 147 patients with biochemical relapse after radical prostatectomy were finally eligible for this retrospective analysis. The median prostate-specific antigen (PSA) level was 8.26 ng/mL (range, 0.22-187.40 ng/mL). Of the 147 patients, 72 patients received androgen deprivation therapy (ADT) at least 6 months before the 99mTc-HYNIC-PSMA SPECT/CT. All patients underwent planar whole-body scans and subsequent SPECT/CT of the thoracic and abdominal regions after intravenous injection of 705 ± 70 MBq of 99mTc-HYNIC-PSMA. Images were evaluated for the presence and location of PSMA-positive lesions, in which SUVmax were also measured. Detection rates were stratified according to PSA levels, ADT and Gleason scores. The relationships between SUVmax and clinical characteristics were analyzed using univariate and multivariable linear regression models for patients with positive findings. Results Of the 147 patients, 99mTc-HYNIC-PSMA SPECT/CT revealed at least one positive lesion in 118 patients with a high detection rate (80.3%). The detection rates were 48.6% (17/35), 85.1% (40/47), 92.1% (35/38), and 96.3% (26/27) at PSA levels of greater than 0.2 to 2, greater than 2 to 5, greater than 5 to 10, and greater than 10 ng/mL, respectively. PSMA SPECT/CT indicated local recurrence, lymph node metastases, bone metastases, and visceral metastases in 14 (9.5%), 73 (49.7%), 48 (32.7%) and 3 (2.0%) patients. The detection rates of local recurrence and metastasis increased with increasing PSA levels. The detection rate was higher in patients treated with ADT than those without (90.3% vs. 70.7%; P =0.0029). In patients with Gleason scores ≥8, detection rate was slightly higher than those with ≤7 (81.7% vs. 78.5%), but not statistically significant (P = 0.6265). Multivariable linear regression analysis showed a significant correlation of PSA levels and ADT with SUVmax (P=0.0005 and P=0.0397). Conclusions 99mTc-HYNIC-PSMA SPECT/CT offers high detection rates for biochemically recurrent prostate cancer after radical prostatectomy. The detection rate and SUVmax were positively correlated with PSA levels and ADT.
Collapse
Affiliation(s)
- Bo Li
- Henan Key Laboratory for Molecular Nuclear Medicine and Translational Medicine, Department of Nuclear Medicine, Henan Provincial People’s Hospital & Zhengzhou University People’s Hospital, Zhengzhou, China,Department of Medical Imaging, Xinjiang Production and Construction Corps 13 division Red Star Hospital, Hami, China
| | - Lili Duan
- Henan Key Laboratory for Molecular Nuclear Medicine and Translational Medicine, Department of Nuclear Medicine, Henan Provincial People’s Hospital & Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Jingqi Shi
- Henan Key Laboratory for Molecular Nuclear Medicine and Translational Medicine, Department of Nuclear Medicine, Henan Provincial People’s Hospital & Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Yunyun Han
- Henan Key Laboratory for Molecular Nuclear Medicine and Translational Medicine, Department of Nuclear Medicine, Henan Provincial People’s Hospital & Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Wei Wei
- Department of Urology Surgery, Henan Provincial People’s Hospital & Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Xiaoliang Cheng
- Department of Pharmacy, The First Affiliated Hospital of Xi'an Jiaotong Universityl, Xi’an, China
| | - Yong Cao
- Department of Medical Imaging, Xinjiang Production and Construction Corps 13 division Red Star Hospital, Hami, China
| | - Akeban Kader
- Department of Medical Imaging, Xinjiang Production and Construction Corps 13 division Red Star Hospital, Hami, China,*Correspondence: Yongju Gao, ; Xinyu Wu, ; Degang Ding, ; Akeban Kader,
| | - Degang Ding
- Department of Urology Surgery, Henan Provincial People’s Hospital & Zhengzhou University People’s Hospital, Zhengzhou, China,*Correspondence: Yongju Gao, ; Xinyu Wu, ; Degang Ding, ; Akeban Kader,
| | - Xinyu Wu
- Henan Key Laboratory for Molecular Nuclear Medicine and Translational Medicine, Department of Nuclear Medicine, Henan Provincial People’s Hospital & Zhengzhou University People’s Hospital, Zhengzhou, China,*Correspondence: Yongju Gao, ; Xinyu Wu, ; Degang Ding, ; Akeban Kader,
| | - Yongju Gao
- Henan Key Laboratory for Molecular Nuclear Medicine and Translational Medicine, Department of Nuclear Medicine, Henan Provincial People’s Hospital & Zhengzhou University People’s Hospital, Zhengzhou, China,*Correspondence: Yongju Gao, ; Xinyu Wu, ; Degang Ding, ; Akeban Kader,
| |
Collapse
|
13
|
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
|
14
|
Ilhan H, Kroenke M, Wurzer A, Unterrainer M, Heck M, Belka C, Knorr K, Langbein T, Rauscher I, Schmidt-Hegemann NS, Schiller K, Bartenstein P, Wester HJ, Eiber M. 18F-rhPSMA-7 PET for the Detection of Biochemical Recurrence of Prostate Cancer After Curative-Intent Radiation Therapy: A Bicentric Retrospective Study. J Nucl Med 2022; 63:1208-1214. [PMID: 35273094 PMCID: PMC9364349 DOI: 10.2967/jnumed.121.262861] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 12/02/2021] [Indexed: 02/03/2023] Open
Abstract
This bicentric, retrospective analysis investigated the efficacy of PET/CT with a novel theranostic prostate-specific membrane antigen (PSMA)--targeting ligand, 18F-rhPSMA-7, in patients with biochemical recurrence (BCR) of prostate cancer after curative-intent primary radiotherapy. Methods: Datasets from patients with BCR of prostate cancer after external-beam radiation therapy or brachytherapy who underwent 18F-rhPSMA-7 PET/CT at either Technical University Munich or Ludwig-Maximilians-University Munich were retrospectively reviewed by experienced nuclear medicine physicians and radiologists at both centers. The median injected activity was 299 MBq (range, 204-420 MBq), and the median uptake time was 77 min (range, 46-120 min). All lesions suggestive of recurrent prostate cancer were noted. Detection rates were correlated with patients' prostate-specific antigen (PSA) level, primary Gleason score, and prior use of androgen-deprivation therapy (ADT). Results: Ninety-seven patients were included (65 at Technical University Munich and 32 at Ludwig-Maximilians-University Munich). The median prescan PSA was 4.19 ng/mL (range, 0.1-159 ng/mL). The primary Gleason score was ≤6 in 19 patients, 7 in 25, ≥8 in 33, and unknown in 20. Thirty patients received ADT in the 6 mo preceding PET/CT. 18F-rhPSMA-7 identified lesions in 91 of 97 (94%) patients. Detection rates stratified by PSA were 88% (22/25), 97% (30/31), 90% (19/21), and 100% (20/20) for a PSA of <2, 2-<5, 5-<10, and ≥10 ng/mL, respectively. Detection rates in the subgroup of patients not meeting the Phoenix criteria for BCR were 80% (4/5), 90% (9/10), 100% (4/4), and 83% (5/6) for a PSA of <0.5, 0.5-<1, 1-<1.5, and 1.5-2 ng/mL, respectively. There were no significant differences in detection rates between patients with and without prior ADT (100% vs. 91%, P = 0.173) or patients with a Gleason score of ≤7 and a Gleason score of ≥8 (98% vs. 91%, P = 0.316).18F-rhPSMA-7 revealed local recurrence in 80% (78/97); pelvic lymph node metastases in 38% (37/97); retroperitoneal and supradiaphragmatic lymph node metastases in 9% (9/97) and 4% (4/97), respectively; bone metastases in 27% (26/97); and visceral metastases in 3% (3/97). In the subgroup of patients with a PSA of <2 ng/mL above nadir, local recurrence occurred in 76% (19/25) and pelvic lymph node metastases in 36% (9/25). Conclusion:18F-rhPSMA-7 PET/CT demonstrates high detection rates in prostate cancer patients with BCR after primary radiation therapy, even at low PSA values. Its diagnostic efficacy is comparable to published data for other PSMA ligands.
Collapse
Affiliation(s)
- Harun Ilhan
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany;,Die Radiologie, Munich, Germany
| | - Markus Kroenke
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
| | - Alexander Wurzer
- Chair of Pharmaceutical Radiochemistry, Technical University of Munich, Garching, Germany
| | - Marcus Unterrainer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany;,Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Matthias Heck
- Department of Urology, Technical University of Munich, Munich, Germany
| | - Claus Belka
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany; and
| | - Karina Knorr
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
| | - Thomas Langbein
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
| | - Isabel Rauscher
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
| | | | - Kilian Schiller
- Department of Radiation Oncology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Hans-Jürgen Wester
- Chair of Pharmaceutical Radiochemistry, Technical University of Munich, Garching, Germany
| | - Matthias Eiber
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
15
|
Basuli F, Phelps TE, Zhang X, Woodroofe CC, Roy J, Choyke PL, Swenson RE, Jagoda EM. Fluorine-18 Labeled Urea-Based Ligands Targeting Prostate-Specific Membrane Antigen (PSMA) with Increased Tumor and Decreased Renal Uptake. Pharmaceuticals (Basel) 2022; 15:597. [PMID: 35631423 PMCID: PMC9144807 DOI: 10.3390/ph15050597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/07/2022] [Accepted: 05/11/2022] [Indexed: 12/22/2022] Open
Abstract
High expression of prostate-specific membrane antigen (PSMA) in prostate cancers prompted the development of the PSMA-targeted PET-imaging agent [18F]DCFPyL, which was recently approved by the FDA. Fluorine-18-labeled Lys-Urea-Glu-based oxime derivatives of [18F]DCFPyL were prepared for the comparison of their in vitro and in vivo properties to potentially improve kidney clearance and tumor targeting. The oxime radiotracers were produced by condensation of an aminooxy functionalized PSMA-inhibitor Lys-Urea-Glu scaffold with fluorine-18-labeled aldehydes. The radiochemical yields were between 15-42% (decay uncorrected) in 50-60 min. In vitro saturation and competition binding assays with human prostate cancer cells transfected with PSMA, PC3(+), indicated similar high nM binding affinities to PSMA for all radiotracers. In vivo biodistribution studies with positive control PC3(+) tumor xenografts showed that the kidneys had the highest uptake followed by tumors at 60 min. The PC3(+) tumor uptake was blocked with non-radioactive DCFPyL, and PC3(-) tumor xenograft (negative control) tumor uptake was negligible indicating that PSMA targeting was preserved. The most lipophilic tracer, [18F]2a, displayed comparable tumor-targeting to [18F]DCFPyL and a desirable alteration in pharmacokinetics and metabolism, resulting in significantly lower kidney uptake with a shift towards hepatobiliary clearance and increased liver uptake.
Collapse
Affiliation(s)
- Falguni Basuli
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; (X.Z.); (C.C.W.); (R.E.S.)
| | - Tim E. Phelps
- Molecular Imaging Branch, National Cancer Institute, Bethesda, MD 20892, USA; (T.E.P.); (J.R.); (P.L.C.); (E.M.J.)
| | - Xiang Zhang
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; (X.Z.); (C.C.W.); (R.E.S.)
| | - Carolyn C. Woodroofe
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; (X.Z.); (C.C.W.); (R.E.S.)
| | - Jyoti Roy
- Molecular Imaging Branch, National Cancer Institute, Bethesda, MD 20892, USA; (T.E.P.); (J.R.); (P.L.C.); (E.M.J.)
| | - Peter L. Choyke
- Molecular Imaging Branch, National Cancer Institute, Bethesda, MD 20892, USA; (T.E.P.); (J.R.); (P.L.C.); (E.M.J.)
| | - Rolf E. Swenson
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; (X.Z.); (C.C.W.); (R.E.S.)
| | - Elaine M. Jagoda
- Molecular Imaging Branch, National Cancer Institute, Bethesda, MD 20892, USA; (T.E.P.); (J.R.); (P.L.C.); (E.M.J.)
| |
Collapse
|
16
|
Feasibility of biology-guided radiotherapy using PSMA-PET to boost to dominant intraprostatic tumour. Clin Transl Radiat Oncol 2022; 35:84-89. [PMID: 35662883 PMCID: PMC9156937 DOI: 10.1016/j.ctro.2022.05.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 11/23/2022] Open
Abstract
Biology-guided radiation therapy (BGRT) uses PET imaging for online image guidance. PSMA PET uptake is abundant in the dominant intraprostatic lesion (DIL). BgRT boost to PSMA-avid subvolume in the prostate region may be feasible. Suitable targets for BgRT were identified in the ProPSMA clinical trial.
Background Methods Results Conclusions
Collapse
|
17
|
Zhang-Yin J, Montravers F, Montagne S, Hennequin C, Renard-Penna R. Diagnosis of early biochemical recurrence after radical prostatectomy or radiation therapy in patients with prostate cancer: State of the art. Diagn Interv Imaging 2022; 103:191-199. [DOI: 10.1016/j.diii.2022.02.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/11/2022] [Accepted: 02/13/2022] [Indexed: 12/30/2022]
|
18
|
Maitre P, Sood S, Pathare P, Krishnatry R, Agarwal A, Rangarajan V, Murthy V. Timing of Ga68-PSMA PETCT and patterns of recurrence after prostate radiotherapy: Implications for potential salvage. Radiother Oncol 2022; 169:71-76. [DOI: 10.1016/j.radonc.2022.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/12/2022] [Accepted: 02/14/2022] [Indexed: 10/19/2022]
|
19
|
Evaluation of Predictors of Biochemical Recurrence in Prostate Cancer Patients, as Detected by 68Ga-PSMA PET/CT. Diagnostics (Basel) 2022; 12:diagnostics12010195. [PMID: 35054362 PMCID: PMC8774699 DOI: 10.3390/diagnostics12010195] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 01/11/2022] [Accepted: 01/11/2022] [Indexed: 12/30/2022] Open
Abstract
Objectives: To explore the existence of new predictors of the 68Ga-Prostate-Specific Membrane Antigen (PSMA) PET/CT detection rate at biochemical recurrence (BCR) and to determine the detection rate of 68Ga-PSMA PET/CT dependent of prostate-specific antigen (PSA) levels. Materials and methods: In total, 189 PCa patients scanned with 68Ga-PSMA PET/CT for detection of BCR after curatively intended treatment with either radical prostatectomy (n = 153) or radiotherapy (n = 36) were included. Clinicopathological information at the time of diagnosis (PSA, clinical tumor-stage, International Society of Urological Pathology Grade Group and whether 68Ga-PSMA PET/CT was used for primary staging), treatment (RT/RP and histopathology of the prostatectomies), and pre-PET PSA were collected from medical records. Results: Of the 189 68Ga-PSMA PET/CT scans, 103 (54.5%) were positive for BCR of PCa. No significant coherency was observed between detection rate and any clinicopathological variables at diagnosis. Detection rates significantly increased with rising PSA: <0.5 ng/mL = 28%, 0.5 ≤ 1 ng/mL = 39%, 1 ≤ 2 ng/mL = 64%, 2 ≤ 5 ng/mL = 87.5% and ≥5 ng/mL = 97%. Conclusions: The detection rate of PCa recurrence was strongly dependent of pre-PET PSA levels. None of the additional clinical variables acquired during primary staging, prostatectomy pathology reports, nor primary staging imaging modality affected the detection rate.
Collapse
|
20
|
Paterson F, Nottage M, Kitchener M, Jarvis M, Reid J, Oakden-Rayner L. Assessing the accuracy of 68 Ga-PSMA PET/CT compared with MRI in the initial diagnosis of prostate malignancy: A cohort analysis of 114 consecutive patients. J Med Imaging Radiat Oncol 2021; 66:319-323. [PMID: 34250746 DOI: 10.1111/1754-9485.13284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 06/22/2021] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Prostate cancer diagnosis is shifting towards a minimally invasive approach, maintaining accuracy and efficacy while reducing morbidity. We aimed to assess if PSMA-Ga68 PET/CT can accurately grade and localise prostatic malignancy using objective methods, compared with pathology and MRI. METHODS Retrospective analysis on 114 consecutive patients undergoing staging PSMA PET/CT scans over 12 months was carried out. The SUVmax and site of highest PSMA activity within the prostate gland were recorded. Pathology/biopsy review assessed maximum Gleason score (and location). MRI analysis assessed the highest PIRADS score and location. The grade, location and size of malignant tissue on biopsy, and PSA, were correlated with the SUVmax and the PIRADS score. RESULTS SUVmax was significantly elevated in cases with PSA ≥10 (P = 0.003) and Gleason score ≥8 (P = 0.0002). SUVmax demonstrated equivalent sensitivity to MRI-PIRADS in predicting Gleason ≥8 disease, with higher specificity when tested under a high-specificity regime (SUVmax ≥10, PIRADS = 5, P = 0.002). Furthermore, the region of highest SUVmax was superior to MRI-PIRADS for localising the highest grade tumour region, correctly identifying 71% of highest grade regions compared to 54% with MRI (P = 0.015). CONCLUSION PSMA PET/CT is as effective as MRI in identifying high-grade prostate malignancy. Our findings also support previous studies in showing a significant relationship between SUVmax and Gleason grade. These benefits, along with the known advantage in identifying distant metastases and the reduced cost, further support the argument that PSMA PET/CT should be offered as an initial investigation in the workup of prostate cancer.
Collapse
Affiliation(s)
- Felix Paterson
- Dr Jones and Partners Medical Imaging, Adelaide, South Australia, Australia
| | - Michelle Nottage
- Dr Jones and Partners Medical Imaging, Adelaide, South Australia, Australia
| | - Michael Kitchener
- Dr Jones and Partners Medical Imaging, Adelaide, South Australia, Australia
| | - Matthew Jarvis
- South Australian Medical Imaging, Adelaide, South Australia, Australia
| | - Jessica Reid
- Department of Surgery, The University of Adelaide, The Queen Elizabeth Hospital, Adelaide, South Australia, Australia
| | - Luke Oakden-Rayner
- Dr Jones and Partners Medical Imaging, Adelaide, South Australia, Australia
| |
Collapse
|
21
|
Murthy V, Maitre P. Reply to A.C. Olson et al, R.T. Dess et al, D.D. Yang et al, and R.J. Brenneman et al. J Clin Oncol 2021; 39:2848-2849. [PMID: 34086489 DOI: 10.1200/jco.21.00810] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Vedang Murthy
- Vedang Murthy, MD, and Priyamvada Maitre, MD, Tata Memorial Hospital and Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Homi Bhabha National Institute (HBNI), Mumbai, India
| | - Priyamvada Maitre
- Vedang Murthy, MD, and Priyamvada Maitre, MD, Tata Memorial Hospital and Advanced Centre for Treatment Research and Education in Cancer (ACTREC), Homi Bhabha National Institute (HBNI), Mumbai, India
| |
Collapse
|
22
|
Wu SY, Wong AC, Shinohara K, Roach M, Cunha JAM, Valdes G, Hsu IC. Salvage High-Dose-Rate Brachytherapy for Recurrent Prostate Cancer After Definitive Radiation. Pract Radiat Oncol 2021; 11:515-526. [PMID: 34077809 DOI: 10.1016/j.prro.2021.04.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE Salvage high-dose-rate brachytherapy (sHDRBT) for locally recurrent prostate cancer after definitive radiation is associated with biochemical control in approximately half of patients at 3 to 5 years. Given potential toxicity, patient selection is critical. We present our institutional experience with sHDRBT and validate a recursive partitioning machines model for biochemical control. MATERIALS AND METHODS We performed a retrospective analysis of 129 patients who underwent whole-gland sHDRBT between 1998 and 2016. We evaluated clinical factors associated with biochemical control as well as toxicity. RESULTS At diagnosis the median prostate-specific antigen (PSA) was 7.77 ng/mL. A majority of patients had T1-2 (73%) and Gleason 6-7 (82%) disease; 71% received external beam radiation therapy (RT) alone, and 22% received permanent prostate implants. The median disease-free interval (DFI) was 56 months, and median presalvage PSA was 4.95 ng/mL. At sHDRBT, 46% had T3 disease and 51% had Gleason 8 to 10 disease. At a median of 68 months after sHDRBT, 3- and 5-year disease-free survival were 85% (95% CI, 79-91) and 71% (95% CI, 62-79), respectively. Median PSA nadir was 0.18 ng/mL, achieved a median of 10 months after sHDRBT. Patients with ≥35%+ cores and a DFI <4.1 years had worse biochemical control (19% vs 50%, P = .02). Local failure (with or without regional/distant failure) was seen in 11% of patients (14/129), and 14 patients (11%) developed acute urinary obstruction requiring Foley placement and 19 patients (15%) developed strictures requiring dilation. CONCLUSIONS sHDRBT is a reasonable option for patients with locally recurrent prostate cancer after definitive RT. Those with <35%+ cores or an initial DFI of ≥4.1 years may be more likely to achieve long-term disease control after sHDRBT.
Collapse
Affiliation(s)
- Susan Y Wu
- Department of Radiation Oncology, University of California, San Francisco, California
| | - Anthony C Wong
- Department of Radiation Oncology, University of California, San Francisco, California
| | - Katsuto Shinohara
- Department of Urology, University of California, San Francisco, California
| | - Mack Roach
- Department of Radiation Oncology, University of California, San Francisco, California
| | - J Adam M Cunha
- Department of Radiation Oncology, University of California, San Francisco, California
| | - Gilmer Valdes
- Department of Radiation Oncology, University of California, San Francisco, California
| | - I-Chow Hsu
- Department of Radiation Oncology, University of California, San Francisco, California.
| |
Collapse
|
23
|
Matushita CS, da Silva AMM, Schuck PN, Bardisserotto M, Piant DB, Pereira JL, Cerci JJ, Coura-Filho GB, Esteves FP, Amorim BJ, Gomes GV, Brito AET, Bernardo WM, Mundstock E, Fanti S, Macedo B, Roman DH, Tem-Pass CS, Hochhegger B. 68Ga-Prostate-specific membrane antigen (psma) positron emission tomography (pet) in prostate cancer: a systematic review and meta-analysis. Int Braz J Urol 2021; 47:705-729. [PMID: 33566470 PMCID: PMC8321470 DOI: 10.1590/s1677-5538.ibju.2019.0817] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 07/14/2020] [Indexed: 12/02/2022] Open
Abstract
Introduction: Prostate cancer (PC) is the second most commonly diagnosed cancer in males. 68Ga-PSMA PET/CT, a non-invasive diagnostic tool to evaluate PC with prostate-specific membrane antigen (PSMA) expression, has emerged as a more accurate alternative to assess disease staging. We aimed to identify predictors of positive 68Ga-PSMA PET and the accuracy of this technique. Materials and methods: Diagnostic accuracy cross-sectional study with prospective and retrospective approaches. We performed a comprehensive literature search on PubMed, Cochrane Library, and Embase database in search of studies including PC patients submitted to radical prostatectomy or radiotherapy with curative intent and presented biochemical recurrence following ASTRO 1996 criteria. A total of 35 studies involving 3910 patients submitted to 68-Ga-PSMA PET were included and independently assessed by two authors: 8 studies on diagnosis, four on staging, and 23 studies on restaging purposes. The significance level was α=0.05. Results: pooled sensitivity and specificity were 0.90 (0.86-0.93) and 0.90 (0.82-0.96), respectively, for diagnostic purposes; as for staging, pooled sensitivity and specificity were 0.93 (0.86-0.98) and 0.96 (0.92-0.99), respectively. In the restaging scenario, pooled sensitivity and specificity were 0.76 (0.74-0.78) and 0.45 (0.27-0.58), respectively, considering the identification of prostate cancer in each described situation. We also obtained specificity and sensitivity results for PSA subdivisions. Conclusion: 68Ga-PSMA PET provides higher sensitivity and specificity than traditional imaging for prostate cancer.
Collapse
Affiliation(s)
- Cristina S Matushita
- Instituto do Cérebro do Rio Grande do Sul, Pontifícia Universidade Católica do Rio Grande do Sul - PUCRS, Porto Alegre, RS, Brasil
| | - Ana M Marques da Silva
- Instituto do Cérebro do Rio Grande do Sul, Pontifícia Universidade Católica do Rio Grande do Sul - PUCRS, Porto Alegre, RS, Brasil.,Laboratório de Imagens Médicas, Faculdade de Ciências, Pontifícia Universidade Católica do Rio Grande do Sul - PUCRS, Porto Alegre, RS, Brasil
| | - Phelipi N Schuck
- Laboratório de Imagens Médicas, Faculdade de Ciências, Pontifícia Universidade Católica do Rio Grande do Sul - PUCRS, Porto Alegre, RS, Brasil
| | | | - Diego B Piant
- Instituto do Cérebro do Rio Grande do Sul, Pontifícia Universidade Católica do Rio Grande do Sul - PUCRS, Porto Alegre, RS, Brasil
| | | | | | - George B Coura-Filho
- Departamento de Medicina Nuclear, Instituto do Câncer de São Paulo, São Paulo, SP, Brasil
| | | | - Barbara J Amorim
- Departamento de Medicina Nuclear, Universidade Estadual de Campinas - UNICAMP, Campinas, SP, Brasil
| | | | | | - Wanderley M Bernardo
- Programa de Pós-Graduação em Medicina, Faculdade de Medicina - USP, São Paulo, SP, Brasil
| | - Eduardo Mundstock
- Programa de Pós-Graduação em Saúde da Criança, Faculdade de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul - PUCRS, Porto Alegre, RS, Brasil
| | - Stefano Fanti
- Department of Experimental, Diagnostic and Specialized Medicine-DIMES, University of Bologna, Bologna, Italy
| | - Bruna Macedo
- Instituto do Cérebro do Rio Grande do Sul, Pontifícia Universidade Católica do Rio Grande do Sul - PUCRS, Porto Alegre, RS, Brasil
| | - Diego H Roman
- Instituto do Cérebro do Rio Grande do Sul, Pontifícia Universidade Católica do Rio Grande do Sul - PUCRS, Porto Alegre, RS, Brasil
| | - Cinthia Scatolin Tem-Pass
- Programa de Pós-Graduação em Saúde da Criança, Faculdade de Medicina, Pontifícia Universidade Católica do Rio Grande do Sul - PUCRS, Porto Alegre, RS, Brasil
| | - Bruno Hochhegger
- Instituto do Cérebro do Rio Grande do Sul, Pontifícia Universidade Católica do Rio Grande do Sul - PUCRS, Porto Alegre, RS, Brasil
| |
Collapse
|
24
|
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
|
25
|
Young S, Liu W, Zukotynski K, Bauman G. Prostate-specific membrane antigen targeted PET/CT for recurrent prostate cancer: a clinician's guide. Expert Rev Anticancer Ther 2021; 21:641-655. [PMID: 33476253 DOI: 10.1080/14737140.2021.1878883] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION PSMA-targeted PET/CT is a 'Next Generation Imaging' technique with superior sensitivity and specificity for detecting recurrent prostate cancer compared with conventional imaging, allowing more accurate staging and re-staging. AREAS COVERED This article reviews the role of PSMA-targeted PET/CT in clinical management of men with recurrent prostate cancer. EXPERT OPINION Through enhanced spatial characterization of recurrent prostate cancer, PSMA-targeted PET/CT has shown significant impact on management decisions. In particular, by identifying men with recurrence confined to the prostate or pelvic nodes, PSMA-targeted PET/CT enables selective deployment of localized salvage therapies for management of biochemical failure after primary treatment with prostatectomy or radiotherapy. In oligometastatic disease, PSMA-targeted PET/CT may improve patient selection and treatment accuracy for metastasis-directed therapy and early phase II studies show encouraging results in delaying the need for systemic therapy. Further, quantitative PSMA-targeted PET/CT for monitoring response and therapeutic PSMA-targeted radiopharmaceuticals are emerging as encouraging treatment options in the setting of castrate-resistant disease.
Collapse
Affiliation(s)
- Sympascho Young
- Department of Radiation Oncology, London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada
| | - Wei Liu
- Department of Radiation Oncology, London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada
| | | | - Glenn Bauman
- Department of Radiation Oncology, London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada
| |
Collapse
|
26
|
Das C, Kubihal V, Sharma S, Kumar R, Seth A, Kumar R, Kaushal S, Sarangi J, Gupta R. Multiparametric magnetic resonance imaging, 68Ga prostate-specific membrane antigen positron emission tomography–Computed tomography, and respective quantitative parameters in detection and localization of clinically significant prostate cancer in intermediate- and high-risk group patients: An Indian demographic study. Indian J Nucl Med 2021; 36:362-370. [PMID: 35125753 PMCID: PMC8771078 DOI: 10.4103/ijnm.ijnm_80_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 09/12/2021] [Accepted: 09/14/2021] [Indexed: 11/09/2022] Open
Abstract
Objectives: The objective of this study was to evaluate the diagnostic accuracy of multiparametric magnetic resonance imaging (mpMRI) and 68Ga prostate-specific membrane antigen positron emission tomography–computed tomography (PSMA PET-CT) and respective quantitative parameters (Ktrans – influx rate contrast, Kep – efflux rate constant, ADC – apparent diffusion coefficient, and SUVmax ratio – prostate SUVmax to background SUVmax ratio) in detection and localization of clinically significant prostate cancer (CSPCa) in D’Amico intermediate- and high-risk group patients (prostate-specific antigen [PSA] >10 ng/ml). Methodology: The study included thirty-three consecutive adult men with serum prostate specific antigen >10ng/ml, and systematic 12 core prostate biopsy proven prostate cancer. All the 33 patients, were evaluated with mpMRI, and 68Ga PSMA PET-CT. The biopsy specimens and imaging were evaluated for 12 sectors per prostate by a predetermined scheme. Results: MpMRI Prostate Imaging Reporting and Data System version 2 (PI-RADS v2) score ≥3 showed higher sensitivity than 68Ga PSMA PET-CT (96.3% vs. 82.4%), with similar specificity (54.5% vs. 54.5%) (n = 33 patients, 396 sectors). Combined use of MRI and 68Ga PSMA PET-CT in parallel increased sensitivity (99.5%) and NPV (98.7%) for detection of CSPCa and combined use of MRI and 68Ga PSMA PET-CT in series increased specificity (71.8%) and PPV (71.5%) (n = 33 patients, 396 sectors). ADC showed a strong negative correlation with Gleason score (r = −0.77), and the highest discriminative ability for detection and localization of CSPCa (area under curve [AUC]: 0.91), followed by Ktrans (r = 0.74; AUC: 0.89), PI-RADS (0.73; 0.86), SUVmax ratio (0.49; 0.74), and Kep (0.24; 0.66). Conclusion: MpMRI PI-RADS v2 score and 68Ga PSMA PET-CT (individually as well as in combination) are reliable tool for detection and localization of CSPCa. Quantitative MRI and 68Ga PSMA PET-CT parameters have potential to predict Gleason score and detect CSPCa.
Collapse
|
27
|
Kalinauskaite G, Senger C, Kluge A, Furth C, Kufeld M, Tinhofer I, Budach V, Beck M, Hochreiter A, Grün A, Stromberger C. 68Ga-PSMA-PET/CT-based radiosurgery and stereotactic body radiotherapy for oligometastatic prostate cancer. PLoS One 2020; 15:e0240892. [PMID: 33085712 PMCID: PMC7577453 DOI: 10.1371/journal.pone.0240892] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 10/05/2020] [Indexed: 02/07/2023] Open
Abstract
Background Androgen deprivation therapy (ADT) remains the standard therapy for patients with oligometastatic prostate cancer (OMPC). Prostate-specific membrane antigen positron emission tomography/computed tomography (PSMA-PET/CT)-based stereotactic body radiotherapy (SBRT) is emerging as an alternative option to postpone starting ADT and its associated side effects including the development of drug resistance. The aim of this study was to determine progression free-survival (PFS) and treatment failure free-survival (TFFS) after PSMA-PET/CT-based SBRT in OMPC patients. The efficacy and safety of single fraction radiosurgery (SFRS) and ADT delay were investigated. Methods Patients with ≤5 metastases from OMPC, with/without ADT treated with PSMA-PET/CT-based SBRT were retrospectively analyzed. PFS and TFFS were primary endpoints. Secondary endpoints were local control (LC), overall survival (OS) and ADT-free survival (ADTFS). Results Fifty patients with a total of 75 metastases detected by PSMA-PET/CT were analyzed. At the time of SBRT, 70% of patients were castration-sensitive. Overall, 80% of metastases were treated with SFRS (median dose 20 Gy, range: 16–25). After median follow-up of 34 months (range: 5–70) median PFS and TFFS were 12 months (range: 2–63) and 14 months (range: 2–70), respectively. Thirty-two (64%) patients had repeat oligometastatic disease. Twenty-four (48%) patients with progression underwent second SBRT course. Two-year LC after SFRS was 96%. Grade 1 and 2 toxicity occurred in 3 (6%) and 1 (2%) patients, respectively. ADTFS and OS rates at 2-years were 60.5% and 100%, respectively. In multivariate analysis, TFFS significantly improved in patients with time to first metastasis (TTM) >36 months (p = 0.01) and PSA before SBRT ≤1 ng/ml (p = 0.03). Conclusion For patients with OMPC, SBRT might be used as an alternative to ADT. This way, the start/escalation of palliative ADT and its side effects can be deferred. Metastases treated with PSMA-PET/CT-based SFRS reached excellent LC with minimal toxicity. Low PSA levels and longer TTM predict elongated TFFS.
Collapse
Affiliation(s)
- Goda Kalinauskaite
- Charité –Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Radiation Oncology and Radiotherapy, Berlin, Germany
- Charité CyberKnife Center, Departments of Radiation Oncology and Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
- * E-mail:
| | - Carolin Senger
- Charité –Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Radiation Oncology and Radiotherapy, Berlin, Germany
- Charité CyberKnife Center, Departments of Radiation Oncology and Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Anne Kluge
- Charité –Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Radiation Oncology and Radiotherapy, Berlin, Germany
- Charité CyberKnife Center, Departments of Radiation Oncology and Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Furth
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Nuclear Medicine, Berlin, Germany
| | - Markus Kufeld
- Charité CyberKnife Center, Departments of Radiation Oncology and Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ingeborg Tinhofer
- Charité –Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Radiation Oncology and Radiotherapy, Berlin, Germany
- The Translational Radiooncology and Radiobiology Research Laboratory, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Volker Budach
- Charité –Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Radiation Oncology and Radiotherapy, Berlin, Germany
- Charité CyberKnife Center, Departments of Radiation Oncology and Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Marcus Beck
- Charité –Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Radiation Oncology and Radiotherapy, Berlin, Germany
- Charité CyberKnife Center, Departments of Radiation Oncology and Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Alexandra Hochreiter
- Charité –Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Radiation Oncology and Radiotherapy, Berlin, Germany
| | - Arne Grün
- Charité –Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Radiation Oncology and Radiotherapy, Berlin, Germany
- Charité CyberKnife Center, Departments of Radiation Oncology and Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Carmen Stromberger
- Charité –Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Radiation Oncology and Radiotherapy, Berlin, Germany
- Charité CyberKnife Center, Departments of Radiation Oncology and Neurosurgery, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
28
|
Miksch J, Bottke D, Krohn T, Thamm R, Bartkowiak D, Solbach C, Bolenz C, Beer M, Wiegel T, Beer AJ, Prasad V. Interobserver variability, detection rate, and lesion patterns of 68Ga-PSMA-11-PET/CT in early-stage biochemical recurrence of prostate cancer after radical prostatectomy. Eur J Nucl Med Mol Imaging 2020; 47:2339-2347. [PMID: 32157427 PMCID: PMC7396406 DOI: 10.1007/s00259-020-04718-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 02/03/2020] [Indexed: 12/17/2022]
Abstract
PURPOSE 68Ga-PSMA-11-PET/CT is increasingly used in early-stage biochemical recurrence of prostate cancer to detect potential lesions for an individualized radiotherapy concept. However, subtle findings especially concerning small local recurrences can still be challenging to interpret and are prone to variability between different readers. Thus, we analyzed interobserver variability, detection rate, and lesion patterns systematically in a homogeneous patient population with low-level biochemical recurrence. METHODS We analyzed 68Ga-PSMA-11-PET/CTs in 116 patients with status post-prostatectomy and PSA levels up to 0.6 ng/ml. None of them received ADT or radiotherapy beforehand. Images were interpreted and blinded by two nuclear medicine physicians (R1 and R2). Findings were rated using a 5-point scale concerning local recurrence, lymph nodes, bone lesions, and other findings (1: definitely benign, 2: probably benign, 3: equivocal, 4: probably malignant, 5: definitely malignant). In findings with substantial discrepancies of 2 or more categories and/or potentially leading to differences in further patient management, a consensus reading was done with a third reader (R3). Interobserver agreement was measured by Cohens Kappa analysis after sub-categorizing our classification system to benign (1 + 2), equivocal (3), and malignant (4 + 5). Time course of PSA levels after salvage treatment of patients rated as positive (4 + 5) was analyzed. RESULTS The overall detection rate (categories 4 and 5) was 50% (R1/R2, 49%/51%) and in the PSA subgroups 0-0.2 ng/ml, 0.21-0.3 ng/ml, and 0.31-0.6 ng/ml 24%/27%, 57%/57%, and 65%/68%, respectively. Local recurrence was the most common lesion manifestation followed by lymphatic and bone metastases. The overall agreement in the Cohens Kappa analysis was 0.74 between R1 and R2. For local, lymphatic, and bone sites, the agreement was 0.76, 0.73, and 0.58, respectively. PSA levels of PSMA PET/CT-positive patients after salvage treatment decreased in 75% (27/36) and increased in 25% (9/36). A decrease of PSA, although more frequent in patients with imaging suggesting only local tumor recurrence (86%, 18/21), was also observed in 67% (10/15) of patients with findings of metastatic disease. CONCLUSIONS In a highly homogeneous group of prostate cancer patients with early-stage biochemical recurrence after radical prostatectomy, we could show that 68Ga-PSMA-11-PET/CT has a good detection rate of 50% which is in accordance with literature, with clinically relevant findings even in patients with PSA < 0.21 ng/ml. The interobserver variability is low, particularly concerning assessment of local recurrences and lymph nodes. Therefore, PSMA-PET/CT is a robust diagnostic modality in this patient group for therapy planning.
Collapse
Affiliation(s)
- Jonathan Miksch
- Department of Nuclear Medicine, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany.
| | - Dirk Bottke
- Department of Radiation Oncology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany
- Department of Radiation Oncology, Esslingen Hospital, Hirschlandstraße 97, 73730, Esslingen, Germany
| | - Thomas Krohn
- Department of Nuclear Medicine, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany
- Radiologie Aachen Land, Bahnhofstraße 17, 52146, Würselen, Germany
| | - Reinhard Thamm
- Department of Radiation Oncology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Detlef Bartkowiak
- Department of Radiation Oncology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Christoph Solbach
- Department of Nuclear Medicine, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Christian Bolenz
- Department of Urology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Meinrad Beer
- Department of Radiology, University Hospital of Ulm , Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Thomas Wiegel
- Department of Radiation Oncology, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Ambros J Beer
- Department of Nuclear Medicine, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| | - Vikas Prasad
- Department of Nuclear Medicine, University Hospital of Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Germany
| |
Collapse
|
29
|
Karahissarlian V, Sargos P, Blais E, Cazeau AL, Fontes F, Petitpierre F, Rigou G, De Vaugelade C, Mariey A, Hindié E, Thomas L. Diagnostic performance of imaging techniques for detecting of local recurrence after prostate brachytherapy. Cancer Radiother 2020; 24:323-331. [PMID: 32532578 DOI: 10.1016/j.canrad.2020.03.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/04/2020] [Accepted: 03/10/2020] [Indexed: 10/24/2022]
Abstract
PURPOSE The purpose of this study was to evaluate MRI and fluorocholine PET/CT diagnostic performances for the detection of local recurrence following prostate brachytherapy for localised prostate cancer. MATERIAL AND METHODS In this single-centre study, we retrospectively reviewed data from 21 patients treated by brachytherapy for localised prostate cancer and diagnosed with biochemical recurrence according to Phoenix Criteria, who underwent MRI and fluorocholine PET/CT. We included patients with local relapse suspicion according to imaging exams, with biopsy for the final assessment of local recurrence. Patient analysis data were supplemented by segment analysis using an 8-segment model. RESULTS The fluorocholine PET/CT was positive for 81% and negative for 19% of patients. The sensitivity and specificity were 92% and 33% with diagnosis accuracy of 67%. The MRI was positive for 57% and negative for 43% of patients. The sensitivity and specificity were 67% and 56% with diagnosis accuracy of 62%. There was no statistically significant difference between fluorocholine PET/CT and MRI accuracy (P=0.63). On a segment-based analysis, the sensitivity and specificity were 44% and 82% for fluorocholine PET/CT with diagnosis accuracy of 78%. For MRI, specificity was 91% diagnosis accuracy was 82%. CONCLUSION Both MRI and fluorocholine PET/CT permit to highlight local recurrence sites after prostate brachytherapy. Confirmation biopsies are, however, necessary since this accuracy is insufficient.
Collapse
Affiliation(s)
- V Karahissarlian
- Department of Radiation Oncology, institut Bergonié, 229, cours de l'Argonne, 33076 Bordeaux cedex, France
| | - P Sargos
- Department of Radiation Oncology, Jewish General Hospital, 3755, Côte-Sainte-Catherine road, Montreal, Quebec H3T 1E2, Canada.
| | - E Blais
- Department of Radiation Oncology, institut Bergonié, 229, cours de l'Argonne, 33076 Bordeaux cedex, France
| | - A-L Cazeau
- Department of Nuclear Medicine, institut Bergonié, 229, cours de l'Argonne, 33076 Bordeaux cedex, France
| | - F Fontes
- Interventional Radiology, nouvelle clinique Bel-Air, 138, avenue de la République, 33000 Bordeaux, France
| | - F Petitpierre
- Interventional Radiology, service d'imagerie diagnostique et thérapeutique de l'adulte, hôpital Pellegrin, place Amélie-Raba-Léon, 33076 Bordeaux cedex, France
| | - G Rigou
- IRMBA, centre de radiologie et d'imagerie médicale, hôpital d'Arcachon, avenue Jean-Hameau, 33164 La Teste-de-Buch, France
| | - C De Vaugelade
- Department of Nuclear Medicine, institut Bergonié, 229, cours de l'Argonne, 33076 Bordeaux cedex, France
| | - A Mariey
- Department of Nuclear Medicine, institut Bergonié, 229, cours de l'Argonne, 33076 Bordeaux cedex, France
| | - E Hindié
- Department of Nuclear Medicine, hôpital Haut-Lévêque, CHU de Bordeaux, avenue de Magellan, 33600 Pessac, France
| | - L Thomas
- Department of Radiation Oncology, institut Bergonié, 229, cours de l'Argonne, 33076 Bordeaux cedex, France
| |
Collapse
|
30
|
Tan N, Oyoyo U, Bavadian N, Ferguson N, Mukkamala A, Calais J, Davenport MS. PSMA-targeted Radiotracers versus 18F Fluciclovine for the Detection of Prostate Cancer Biochemical Recurrence after Definitive Therapy: A Systematic Review and Meta-Analysis. Radiology 2020; 296:44-55. [PMID: 32396045 DOI: 10.1148/radiol.2020191689] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background National guidelines endorse fluorine 18 (18F) fluciclovine PET/CT for the detection of prostate cancer (PCa) in men with biochemically recurrent PCa. The comparative performance between fluciclovine and gallium 68 or 18F prostate-specific membrane antigen (PSMA) PET/CT, a newer examination, is unclear. Purpose To compare the detection of biochemical recurrence using fluciclovine versus PSMA-targeted radiotracers in patients with a prostate-specific antigen (PSA) level less than 2 ng/mL. Materials and Methods With use of the Preferred Reporting Items for a Systematic Review and Meta-Analysis of Diagnostic Test Accuracy, or PRISMA-DTA, guidelines, a systematic review of PubMed and EMBASE databases between 2012 and 2019 was performed. Studies of fluciclovine PET/CT or PSMA PET/CT in biochemical recurrence were identified. PSA levels, clinical data, and reference standards were obtained when available. A random-effects model was applied to pooled estimates and 95% confidence intervals (CIs) around the prevalence of a positive examination, stratified according to PSA tier. Results Quantitative analysis included 482 patients (median age, 67 years; interquartile range, 67-67 years) in six fluciclovine studies and 3217 patients (median age, 68 years; interquartile range, 67-70 years) in 38 PSMA studies. Pooled detection rates for PSMA and fluciclovine were 45% (95% CI: 38%, 52%) and 37% (95% CI: 25%, 49%), respectively, for a PSA level less than 0.5 ng/mL (P = .46); 59% (95% CI: 52%, 66%) and 48% (95% CI: 34%, 61%) for a PSA level of 0.5-0.9 ng/mL (P = .19); and 80% (95% CI: 75%, 85%) and 62% (95% CI: 54%, 70%) for a PSA level of 1.0-1.9 ng/mL (P = .01). A reference standard was positive in 703 of 735 patients (96%) in the PSMA cohort and 247of 256 (97%) in the fluciclovine cohort. Conclusion Patient-level detection rates for biochemically recurrent prostate cancer were greater for prostate-specific membrane antigen-targeted radiotracers than fluciclovine for prostate specific antigen levels of 1.0-1.9 ng/mL. © RSNA, 2020 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Nelly Tan
- From the Department of Radiology, Loma Linda University Medical Center, 11234 Anderson St, Suite MC-2605E, Loma Linda, CA 92354 (N.T., U.O., N.F.); Riverside School of Medicine, University of California, Riverside, Calif (N.T., N.B.); Departments of Radiology and Urology, Michigan Medicine, Ann Arbor, Mich (A.M., M.S.D.); and Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Calif (J.C.)
| | - Udochukwu Oyoyo
- From the Department of Radiology, Loma Linda University Medical Center, 11234 Anderson St, Suite MC-2605E, Loma Linda, CA 92354 (N.T., U.O., N.F.); Riverside School of Medicine, University of California, Riverside, Calif (N.T., N.B.); Departments of Radiology and Urology, Michigan Medicine, Ann Arbor, Mich (A.M., M.S.D.); and Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Calif (J.C.)
| | - Niusha Bavadian
- From the Department of Radiology, Loma Linda University Medical Center, 11234 Anderson St, Suite MC-2605E, Loma Linda, CA 92354 (N.T., U.O., N.F.); Riverside School of Medicine, University of California, Riverside, Calif (N.T., N.B.); Departments of Radiology and Urology, Michigan Medicine, Ann Arbor, Mich (A.M., M.S.D.); and Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Calif (J.C.)
| | - Nicholas Ferguson
- From the Department of Radiology, Loma Linda University Medical Center, 11234 Anderson St, Suite MC-2605E, Loma Linda, CA 92354 (N.T., U.O., N.F.); Riverside School of Medicine, University of California, Riverside, Calif (N.T., N.B.); Departments of Radiology and Urology, Michigan Medicine, Ann Arbor, Mich (A.M., M.S.D.); and Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Calif (J.C.)
| | - Anudeep Mukkamala
- From the Department of Radiology, Loma Linda University Medical Center, 11234 Anderson St, Suite MC-2605E, Loma Linda, CA 92354 (N.T., U.O., N.F.); Riverside School of Medicine, University of California, Riverside, Calif (N.T., N.B.); Departments of Radiology and Urology, Michigan Medicine, Ann Arbor, Mich (A.M., M.S.D.); and Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Calif (J.C.)
| | - Jeremie Calais
- From the Department of Radiology, Loma Linda University Medical Center, 11234 Anderson St, Suite MC-2605E, Loma Linda, CA 92354 (N.T., U.O., N.F.); Riverside School of Medicine, University of California, Riverside, Calif (N.T., N.B.); Departments of Radiology and Urology, Michigan Medicine, Ann Arbor, Mich (A.M., M.S.D.); and Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Calif (J.C.)
| | - Matthew S Davenport
- From the Department of Radiology, Loma Linda University Medical Center, 11234 Anderson St, Suite MC-2605E, Loma Linda, CA 92354 (N.T., U.O., N.F.); Riverside School of Medicine, University of California, Riverside, Calif (N.T., N.B.); Departments of Radiology and Urology, Michigan Medicine, Ann Arbor, Mich (A.M., M.S.D.); and Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Calif (J.C.)
| |
Collapse
|
31
|
Fendler WP, Calais J, Eiber M, Flavell RR, Mishoe A, Feng FY, Nguyen HG, Reiter RE, Rettig MB, Okamoto S, Emmett L, Zacho HD, Ilhan H, Wetter A, Rischpler C, Schoder H, Burger IA, Gartmann J, Smith R, Small EJ, Slavik R, Carroll PR, Herrmann K, Czernin J, Hope TA. Assessment of 68Ga-PSMA-11 PET Accuracy in Localizing Recurrent Prostate Cancer: A Prospective Single-Arm Clinical Trial. JAMA Oncol 2020; 5:856-863. [PMID: 30920593 DOI: 10.1001/jamaoncol.2019.0096] [Citation(s) in RCA: 479] [Impact Index Per Article: 119.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Importance In retrospective studies, 68Ga-PSMA-11 positron emission tomographic (PET) imaging improves detection of biochemically recurrent prostate cancer compared with conventional imaging. Objective To assess 68Ga-PSMA-11 PET accuracy in a prospective multicenter trial. Design, Setting, and Participants In this single-arm prospective trial conducted at University of California, San Francisco and University of California, Los Angeles, 635 patients with biochemically recurrent prostate cancer after prostatectomy (n = 262, 41%), radiation therapy (n = 169, 27%), or both (n = 204, 32%) underwent 68Ga-PSMA-11 PET. Presence of prostate cancer was recorded by 3 blinded readers on a per-patient and per-region base. Lesions were validated by histopathologic analysis and a composite reference standard. Main Outcomes and Measures Endpoints were positive predictive value (PPV), detection rate, interreader reproducibility, and safety. Results A total of 635 men were enrolled with a median age of 69 years (range, 44-95 years). On a per-patient basis, PPV was 0.84 (95% CI, 0.75-0.90) by histopathologic validation (primary endpoint, n = 87) and 0.92 (95% CI, 0.88-0.95) by the composite reference standard (n = 217). 68Ga-PSMA-11 PET localized recurrent prostate cancer in 475 of 635 (75%) patients; detection rates significantly increased with prostate-specific antigen (PSA): 38% for <0.5 ng/mL (n = 136), 57% for 0.5 to <1.0 ng/mL (n = 79), 84% for 1.0 to <2.0 ng/mL (n = 89), 86% for 2.0 to <5.0 ng/mL (n = 158), and 97% for ≥5.0 ng/mL (n = 173, P < .001). Interreader reproducibility was substantial (Fleiss κ, 0.65-0.78). There were no serious adverse events associated with 68Ga-PSMA-11 administration. PET-directed focal therapy alone led to a PSA drop of 50% or more in 31 of 39 (80%) patients. Conclusions and Relevance Using blinded reads and independent lesion validation, we establish high PPV for 68Ga-PSMA-11 PET, detection rate and interreader agreement for localization of recurrent prostate cancer. Trial Registration ClinicalTrials.gov identifiers: NCT02940262 and NCT03353740.
Collapse
Affiliation(s)
- Wolfgang P Fendler
- Ahmanson Translational Imaging Division, Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles.,Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Jeremie Calais
- Ahmanson Translational Imaging Division, Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles
| | - Matthias Eiber
- Ahmanson Translational Imaging Division, Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles.,Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Robert R Flavell
- Departments of Radiology and Biomedical Imaging and Pharmaceutical Chemistry, University of California San Francisco, San Francisco
| | - Ashley Mishoe
- Departments of Radiology and Biomedical Imaging and Pharmaceutical Chemistry, University of California San Francisco, San Francisco
| | - Felix Y Feng
- Department of Urology, University of California San Francisco, San Francisco
| | - Hao G Nguyen
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco
| | - Robert E Reiter
- Department of Urology, UCLA Medical Center, University of California Los Angeles, Los Angeles
| | - Matthew B Rettig
- Department of Urology, UCLA Medical Center, University of California Los Angeles, Los Angeles.,Division of Hematology/Oncology, Department of Medicine, University of California Los Angeles, Los Angeles.,Division of Hematology/Oncology, Department of Medicine, VA Greater Los Angeles, Los Angeles, California
| | - Shozo Okamoto
- Department of Radiology, Obihiro Kosei Hospital, Obihiro, Japan.,Department of Nuclear Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Louise Emmett
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, Australia
| | - Helle D Zacho
- Department of Nuclear Medicine, Aalborg University Hospital, Aalborg, Denmark
| | - Harun Ilhan
- Department of Nuclear Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Axel Wetter
- Department of Diagnostic and Interventional Radiology and Neuroradiology, University of Duisburg-Essen, Essen, Germany
| | - Christoph Rischpler
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Heiko Schoder
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Irene A Burger
- Department of Nuclear Medicine, University Hospital Zürich, University of Zürich, Switzerland
| | - Jeannine Gartmann
- Ahmanson Translational Imaging Division, Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles
| | - Raven Smith
- Departments of Radiology and Biomedical Imaging and Pharmaceutical Chemistry, University of California San Francisco, San Francisco
| | - Eric J Small
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco.,Division of Hematology/Oncology, Department of Medicine, University of California San Francisco
| | - Roger Slavik
- Ahmanson Translational Imaging Division, Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles
| | - Peter R Carroll
- Department of Urology, University of California San Francisco, San Francisco
| | - Ken Herrmann
- Ahmanson Translational Imaging Division, Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles.,Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Johannes Czernin
- Ahmanson Translational Imaging Division, Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles
| | - Thomas A Hope
- Departments of Radiology and Biomedical Imaging and Pharmaceutical Chemistry, University of California San Francisco, San Francisco
| |
Collapse
|
32
|
Trabulsi EJ, Rumble RB, Jadvar H, Hope T, Pomper M, Turkbey B, Rosenkrantz AB, Verma S, Margolis DJ, Froemming A, Oto A, Purysko A, Milowsky MI, Schlemmer HP, Eiber M, Morris MJ, Choyke PL, Padhani A, Oldan J, Fanti S, Jain S, Pinto PA, Keegan KA, Porter CR, Coleman JA, Bauman GS, Jani AB, Kamradt JM, Sholes W, Vargas HA. Optimum Imaging Strategies for Advanced Prostate Cancer: ASCO Guideline. J Clin Oncol 2020; 38:1963-1996. [PMID: 31940221 DOI: 10.1200/jco.19.02757] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Provide evidence- and expert-based recommendations for optimal use of imaging in advanced prostate cancer. Due to increases in research and utilization of novel imaging for advanced prostate cancer, this guideline is intended to outline techniques available and provide recommendations on appropriate use of imaging for specified patient subgroups. METHODS An Expert Panel was convened with members from ASCO and the Society of Abdominal Radiology, American College of Radiology, Society of Nuclear Medicine and Molecular Imaging, American Urological Association, American Society for Radiation Oncology, and Society of Urologic Oncology to conduct a systematic review of the literature and develop an evidence-based guideline on the optimal use of imaging for advanced prostate cancer. Representative index cases of various prostate cancer disease states are presented, including suspected high-risk disease, newly diagnosed treatment-naïve metastatic disease, suspected recurrent disease after local treatment, and progressive disease while undergoing systemic treatment. A systematic review of the literature from 2013 to August 2018 identified fully published English-language systematic reviews with or without meta-analyses, reports of rigorously conducted phase III randomized controlled trials that compared ≥ 2 imaging modalities, and noncomparative studies that reported on the efficacy of a single imaging modality. RESULTS A total of 35 studies met inclusion criteria and form the evidence base, including 17 systematic reviews with or without meta-analysis and 18 primary research articles. RECOMMENDATIONS One or more of these imaging modalities should be used for patients with advanced prostate cancer: conventional imaging (defined as computed tomography [CT], bone scan, and/or prostate magnetic resonance imaging [MRI]) and/or next-generation imaging (NGI), positron emission tomography [PET], PET/CT, PET/MRI, or whole-body MRI) according to the clinical scenario.
Collapse
Affiliation(s)
- Edouard J Trabulsi
- Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA
| | | | | | - Thomas Hope
- University of California, San Francisco, San Francisco, CA
| | | | | | | | - Sadhna Verma
- University of Cincinnati Medical Center, Cincinnati, OH
| | | | | | | | | | | | | | | | | | | | - Anwar Padhani
- Paul Strickland Scanner Centre, Mount Vernon Cancer Centre, Northwood, United Kingdom
| | - Jorge Oldan
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC
| | | | - Suneil Jain
- Queen's University Belfast, Belfast, Northern Ireland
| | | | | | | | | | | | | | | | - Westley Sholes
- Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA
| | | |
Collapse
|
33
|
Dadgar H, Emami F, Norouzbeigi N, Vafaee MS, Jafari E, Gholamrezanezhad A, Assadi M, Ahmadzadehfar H. Application of [68Ga]PSMA PET/CT in Diagnosis and Management of Prostate Cancer Patients. Mol Imaging Biol 2019; 22:1062-1069. [DOI: 10.1007/s11307-019-01445-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
34
|
Brachytherapy for the urologist: A multidisciplinary team update for 2019. JOURNAL OF CLINICAL UROLOGY 2019. [DOI: 10.1177/2051415819841703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Brachytherapy is a well-established treatment for localised prostate cancer. Urologists are often tasked with discussing all available treatment options with the newly diagnosed patient. Unlike radical prostatectomy and external beam radiotherapy, knowledge of brachytherapy may be limited. The aim of this article is to provide an up-to-date guide on patient selection, modern brachytherapy techniques and the management of side effects, such that the core urologist can be more confident in both discussing initial treatment options and the long-term management of brachytherapy patients. Level of Evidence: Level 5 - review article
Collapse
|
35
|
The use of 68Ga-PET/CT PSMA to determine patterns of disease for biochemically recurrent prostate cancer following primary radiotherapy. Prostate Cancer Prostatic Dis 2019; 22:385-390. [DOI: 10.1038/s41391-019-0163-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/15/2019] [Accepted: 06/06/2019] [Indexed: 02/04/2023]
|
36
|
Feasibility and Initial Results: Fluciclovine Positron Emission Tomography/Ultrasound Fusion Targeted Biopsy of Recurrent Prostate Cancer. J Urol 2019; 202:413-421. [PMID: 30817240 DOI: 10.1097/ju.0000000000000200] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE We assessed the feasibility and cancer detection rate of fluciclovine (18F) positron emission tomography-ultrasound fusion targeted biopsy vs standard template biopsy in the same patient with biochemical failure after nonsurgical therapy for prostate cancer. MATERIALS AND METHODS A total of 21 patients with a mean ± SD prostate specific antigen of 7.4 ± 6.8 ng/ml and biochemical failure after nonoperative prostate cancer treatment underwent fluciclovine (18F) positron emission tomography-computerized tomography (mean 364.1 ± 37.7 MBq) and planning transrectal prostate ultrasound with 3-dimensional image reconstruction. Focal prostatic activity on positron emission tomography was delineated and co-registered with planning ultrasound. During the subsequent biopsy session computer generated 12-core template biopsies were performed and then fluciclovine defined targets were revealed and biopsied. Histological analysis of template and targeted cores were completed. RESULTS Template biopsy was positive for malignancy in 6 of 21 patients (28.6%), including 10 of 124 regions and 11 of 246 cores, vs targeted biopsy in 10 of 21 (47.6%), including 17 of 50 regions and 40 of 125 cores. Five of 21 patients had positive findings on targeted biopsy only and 1 of 21 had positive findings on template biopsy only. An additional case was upgraded from Grade Group 2 to 3 on targeted biopsy. Extraprostatic disease was detected in 8 of 21 men (38.1%) with histological confirmation in all 3 who underwent lesion biopsy. CONCLUSIONS Fluciclovine positron emission tomography real-time ultrasound fusion guidance for biopsy is feasible in patients with biochemical failure after nonsurgical therapy for prostate cancer. It identifies more recurrent prostate cancer using fewer cores compared with template biopsy in the same patient. Further study is required to determine in what manner targeted biopsy may augment template biopsy of recurrent prostate cancer.
Collapse
|
37
|
Gallium-68-Labeled Prostate-Specific Membrane Antigen-11 PET/CT of Prostate and Nonprostate Cancers. AJR Am J Roentgenol 2019; 213:286-299. [PMID: 31166760 DOI: 10.2214/ajr.19.21084] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE. The purpose of this study is to provide a concise summary of the current experience with 68Ga-labeled prostate-specific membrane antigen (PSMA)-11 imaging of prostate and nonprostate malignancies and benign conditions. CONCLUSION. PSMA is overexpressed in prostate cancer and in the neovasculature of many other malignancies. The relevance of PSMA as a biologic target, coupled with advances in the design, synthesis, and evaluation of PSMA-based radionuclides for imaging and therapy, is anticipated to play a major role in patient care.
Collapse
|
38
|
Ferreira G, Iravani A, Hofman MS, Hicks RJ. Intra-individual comparison of 68Ga-PSMA-11 and 18F-DCFPyL normal-organ biodistribution. Cancer Imaging 2019; 19:23. [PMID: 31092293 PMCID: PMC6521415 DOI: 10.1186/s40644-019-0211-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 05/01/2019] [Indexed: 01/08/2023] Open
Abstract
Purpose Detailed data comparing the biodistribution of PSMA radioligands is still scarce, raising concerns regarding the comparability of different compounds. We investigated differences in normal-organ biodistribution and uptake variability between the two most commonly PSMA tracers in clinical use, 68Ga-PSMA-11 and 18F-DCFPyL. Methods This retrospective analysis included 34 patients with low tumor burden referred for PET/CT imaging with 68Ga-PSMA-11 and subsequently 18F-DCFPyL. Images were acquired with 4 cross-calibrated PET/CT systems. Volumes of interest were placed on major salivary and lacrimal glands, liver, spleen, duodenum, kidneys, bladder, blood-pool and muscle. Normal-organ biodistribution of both tracers was then quantified as SUVpeak and compared using paired tests, linear regression and Bland-Altman analysis. Between-patient variability was also assessed. Clinical and protocol variables were investigated for possible interference. Results For both tracers the highest uptake was found in the kidneys and bladder and low background activity was noted across all scans. In the quantitative analysis there was significantly higher uptake of 68Ga-PSMA-11 in the kidneys, spleen and major salivary glands (p < 0.001), while the liver exhibited slightly higher 18F-DCFPyL uptake (p = 0.001, mean bias 0.79 ± 1.30). The lowest solid-organ uptake variability was found in the liver (COV 21.9% for 68Ga-PSMA-11, 22.5% for 18F-DCFPyL). There was a weak correlation between 18F-DCFPyL uptake time and liver SUVpeak (r = 0.488, p = 0.003) and, accordingly, patients scanned at later time-points had a larger mean bias between the two tracers’ liver uptake values (0.05 vs 1.46, p = 0.001). Conclusion Normal tissue biodistribution patterns of 68Ga-PSMA-11 and 18F-DCFPyL were similar, despite subtle differences in quantitative values. Liver uptake showed an acceptable intra-patient agreement and low inter-patient variability between the two tracers, allowing its use as a reference organ for thresholding scans in the qualitative comparison of PSMA expression using these different tracers. Electronic supplementary material The online version of this article (10.1186/s40644-019-0211-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Gonçalo Ferreira
- Nuclear Medicine Department, Instituto Português de Oncologia do Porto, Rua Dr. António Bernardino de Almeida, 4200-072, Porto, Portugal. .,Centre for Molecular Imaging, Department of Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.
| | - Amir Iravani
- Centre for Molecular Imaging, Department of Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Michael S Hofman
- Centre for Molecular Imaging, Department of Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Rodney J Hicks
- Centre for Molecular Imaging, Department of Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
39
|
Abstract
Following definitive treatment with curative intent a subset of patients with prostate cancer experience biochemical recurrence. In these patients clinical parameters are mostly used to decide if a local or systemic disease recurrence is present. While salvage radiation treatment is advocated for local recurrence after radical prostatectomy, no standard recommendations exist in cases of local recurrence after primary radiation therapy although salvage prostatectomy may be considered. Imaging procedures have traditionally not routinely been recommended for the onset of prostate-specific antigen (PSA) relapse; however, prostate-specific membrane antigen (PSMA) positron emission tomography (PET) computed tomography (CT) exhibits high detection rates even at low PSA values. Thus, the current German guidelines state that PSMA PET/CT can be considered if this could result in a decisive change in further treatment management. Currently, a positive influence on oncological long-term outcome, however, has not yet been proven.
Collapse
|
40
|
Hamed MAG, Basha MAA, Ahmed H, Obaya AA, Afifi AHM, Abdelbary EH. 68Ga-PSMA PET/CT in Patients with Rising Prostatic-Specific Antigen After Definitive Treatment of Prostate Cancer: Detection Efficacy and Diagnostic accuracy. Acad Radiol 2019; 26:450-460. [PMID: 29935970 DOI: 10.1016/j.acra.2018.05.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 05/20/2018] [Accepted: 05/22/2018] [Indexed: 02/03/2023]
Abstract
RATIONALE AND OBJECTIVES 68Ga-prostate-specific membrane antigen-11 (68Ga-PSMA-11) is a recently developed positron emission tomography (PET) tracer that can detect prostate cancer (PC) relapses and metastases with high contrast resolution. The aim of this study was to assess the detection efficacy and diagnostic accuracy of 68Ga-PSMA PET/CT image in patients with rising prostatic-specific antigen (PSA) after treatment of PC. MATERIALS AND METHODS The present prospective study included 188 patients who exhibited rising of PSA level on a routine follow-up examination after definitive treatment of PC. All patients underwent a 68Ga-PSMA PET/CT examination. For each patient, we determined the disease stage, the Gleason score, and the maximum standardized uptake value of the local recurrence and extraprostatic metastases. The detection efficacy and diagnostic accuracy of 68Ga-PSMA PET/CT were established by histopathology and clinical and imaging follow-up as the reference standards. RESULTS 68Ga-PSMA PET/CT detected tumour relapse in 165 patients (35 patients had local recurrence, 106 patients had extraprostatic metastases, and 24 patients had combined lesions). The sensitivity, specificity, and accuracy values of 68Ga-PSMA PET/CT examination in the detection of PC recurrence were 98.8%, 100%, and 98.8%, respectively. 68Ga-PSMA PET/CT revealed an overall detection rate of 87.8% (165/188) in patients with rising PSA (median of 2.2 ng/mL, and range of 0.01-70 ng/mL). CONCLUSION 68Ga-PSMA PET/CT is a valuable tool for the detection of PC local recurrence or extraprostatic metastases following rising PSA levels after primary definitive therapy and should be incorporated during routine work-up.
Collapse
|
41
|
Ekmekcioglu Ö, Busstra M, Klass ND, Verzijlbergen F. Bridging the Imaging Gap: PSMA PET/CT Has a High Impact on Treatment Planning in Prostate Cancer Patients with Biochemical Recurrence—A Narrative Review of the Literature. J Nucl Med 2019; 60:1394-1398. [DOI: 10.2967/jnumed.118.222885] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Accepted: 03/04/2019] [Indexed: 11/16/2022] Open
|
42
|
Krimphove MJ, Theissen LH, Cole AP, Preisser F, Mandel PC, Chun FKH. Performance and Impact of Prostate Specific Membrane Antigen-Based Diagnostics in the Management of Men with Biochemical Recurrence of Prostate Cancer and its Role in Salvage Lymph Node Dissection. World J Mens Health 2019; 38:32-47. [PMID: 30929322 PMCID: PMC6920066 DOI: 10.5534/wjmh.180133] [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: 12/31/2018] [Accepted: 01/20/2019] [Indexed: 02/04/2023] Open
Abstract
Up to 50% of patients initially treated for prostate cancer in a curative intent experience biochemical recurrence, possibly requiring adjuvant treatment. However, salvage treatment decisions, such as lymph node dissection or radiation therapy, are typically based on prostate specific antigen (PSA) recurrence. Importantly, common imaging modalities (e.g., computed tomography [CT], magnetic resonance imaging, and bone scan) are limited and the detection of recurrent disease is particularly challenging if PSA is low. Prostate specific membrane antigen (PSMA) positron-emission tomography/computed tomography (PET/CT) is a novel and promising imaging modality which aims to overcome the incapability of early identification of distant and regional metastases. Within this review, we summarize the current evidence related to PSMA-PET/CT in prostate cancer men diagnosed with biochemical recurrence after local treatment with curative intent. We discuss detection rates of PSMA-PET/CT stratified by PSA-levels and its impact on clinical decision making. Furthermore, we compare different image-fusion techniques such as PSMA-PET vs. F-/C-Choline-PET scans vs. PSMA-single photon emission computed tomography/CT. Finally, we touch upon the contemporary role of radio-guided-PSMA salvage lymphadenectomy.
Collapse
Affiliation(s)
- Marieke J Krimphove
- Department of Urology, University Hospital Frankfurt, Frankfurt am Main, Germany.,Division of Urological Surgery and Center for Surgery and Public Health, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Lena H Theissen
- Department of Urology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Alexander P Cole
- Division of Urological Surgery and Center for Surgery and Public Health, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Felix Preisser
- Department of Urology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Philipp C Mandel
- Department of Urology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Felix K H Chun
- Department of Urology, University Hospital Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
43
|
Perera M, Papa N, Roberts M, Williams M, Udovicich C, Vela I, Christidis D, Bolton D, Hofman MS, Lawrentschuk N, Murphy DG. Gallium-68 Prostate-specific Membrane Antigen Positron Emission Tomography in Advanced Prostate Cancer-Updated Diagnostic Utility, Sensitivity, Specificity, and Distribution of Prostate-specific Membrane Antigen-avid Lesions: A Systematic Review and Meta-analysis. Eur Urol 2019; 77:403-417. [PMID: 30773328 DOI: 10.1016/j.eururo.2019.01.049] [Citation(s) in RCA: 586] [Impact Index Per Article: 117.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 01/31/2019] [Indexed: 12/16/2022]
Abstract
CONTEXT Accurate staging of high-risk localised, advanced, and metastatic prostate cancer is becoming increasingly more important in guiding local and systemic treatment. Gallium-68 prostate-specific membrane antigen (PSMA) positron emission tomography (PET) has increasingly been utilised globally to assess the local and metastatic burden of prostate cancer, typically in biochemically recurrent or advanced disease. Following our previous meta-analysis, a high-volume series has been reported highlighting the utility of 68Ga-PSMA PET in this setting. OBJECTIVE To perform a systematic review and meta-analysis to update reported predictors of positive 68Ga-PSMA PET according to prior therapy and proportion of positivity in various anatomical locations with sensitivity and specificity profiles. EVIDENCE ACQUISITION We performed critical reviews of MEDLINE, EMBASE, ScienceDirect, Cochrane Libraries, and Web of Science databases in July 2018 according to the Preferred Reporting Items for Systematic Review and Meta-analysis (PRISMA) statement. Quality assessment was performed using Quality Assessment if Diagnostic Accuracy Studies-2 tool. Meta-analyses of proportions were performed using a random-effect model. Summary sensitivity and specificity values were obtained by fitting bivariate hierarchical regression models. EVIDENCE SYNTHESIS A total of 37 articles including 4790 patients were analysed. For patients with biochemical recurrence, positive 68Ga-PSMA PET scans increased with higher pre-PET prostate-specific antigen (PSA) levels. For PSA categories 0-0.19, 0.2-0.49, 0.5-0.99, 1-1.99, and ≥2ng/ml, the percentages of positive scans were 33%, 45%, 59%, 75%, and 95%, respectively. No significant differences in positivity were noted between Gleason sums ≤7 and ≥8. Significant differences in positivity after biochemical recurrence in the prostate bed were noted between radical prostatectomy (22%) and radiotherapy (52%) patients. On per-node analysis, high sensitivity (75%) and specificity (99%) were observed. CONCLUSIONS Ga-68-PSMA PET improves detection of metastases with biochemical recurrence, particularly at low pre-PET PSA levels of >0.2ng/ml (33%) and 0.2-0.5ng/ml (45%). Ga-68-PSMA-PET produces favourable sensitivity and specificity profiles on meta-analysis of pooled data. This analysis highlights different anatomic patterns of metastatic spread according to PSMA PET in the primary and biochemically recurrent settings. PATIENT SUMMARY Gallium-68 prostate-specific membrane antigen positron emission tomography is now an established imaging technique that has been developed in response to inadequacies in standard of care imaging modalities to improve the detection of metastatic disease in prostate cancer, particularly in the setting of disease recurrence. To date, this imaging modality in the setting of primary staging is controversial, given the paucity of data. In light of the growing body of evidence, we summarised the data to date to provide clinicians with an overview of this imaging modality.
Collapse
Affiliation(s)
- Marlon Perera
- Department of Surgery, Austin Health, The University of Melbourne, Victoria, Australia; Department of Urology, Princess Alexandra Hospital, Brisbane, Queensland, Australia; Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia.
| | - Nathan Papa
- Department of Surgery, Austin Health, The University of Melbourne, Victoria, Australia
| | - Matthew Roberts
- Department of Urology, Princess Alexandra Hospital, Brisbane, Queensland, Australia; Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Michael Williams
- Department of Urology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Cristian Udovicich
- Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Ian Vela
- Department of Urology, Princess Alexandra Hospital, Brisbane, Queensland, Australia; Australian Prostate Cancer Research Center QLD, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Daniel Christidis
- Department of Surgery, Austin Health, The University of Melbourne, Victoria, Australia
| | - Damien Bolton
- Department of Surgery, Austin Health, The University of Melbourne, Victoria, Australia; Olivia Newton-John Cancer and Wellness Centre, Austin Health, Heidelberg, Victoria, Australia
| | - Michael S Hofman
- Centre for Molecular Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Nathan Lawrentschuk
- Department of Surgery, Austin Health, The University of Melbourne, Victoria, Australia; Olivia Newton-John Cancer and Wellness Centre, Austin Health, Heidelberg, Victoria, Australia; Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| | - Declan G Murphy
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
44
|
Brito AET, Mourato FA, de Oliveira RPM, Leal ALG, Filho PJA, de Filho JLL. Evaluation of whole-body tumor burden with 68Ga-PSMA PET/CT in the biochemical recurrence of prostate cancer. Ann Nucl Med 2019; 33:344-350. [DOI: 10.1007/s12149-019-01342-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 02/04/2019] [Indexed: 12/12/2022]
|
45
|
De Visschere PJ, Standaert C, Fütterer JJ, Villeirs GM, Panebianco V, Walz J, Maurer T, Hadaschik BA, Lecouvet FE, Giannarini G, Fanti S. A Systematic Review on the Role of Imaging in Early Recurrent Prostate Cancer. Eur Urol Oncol 2019; 2:47-76. [DOI: 10.1016/j.euo.2018.09.010] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 09/17/2018] [Accepted: 09/24/2018] [Indexed: 12/20/2022]
|
46
|
Barbosa FG, Queiroz MA, Nunes RF, Viana PCC, Marin JFG, Cerri GG, Buchpiguel CA. Revisiting Prostate Cancer Recurrence with PSMA PET: Atlas of Typical and Atypical Patterns of Spread. Radiographics 2019; 39:186-212. [DOI: 10.1148/rg.2019180079] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Felipe G. Barbosa
- From the Department of Radiology, Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, CEP 01308-060, São Paulo, SP, Brazil; and Department of Radiology and Oncology, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil (M.A.Q., P.C.C.V., J.F.G.M., G.G.C., C.A.B.)
| | - Marcelo A. Queiroz
- From the Department of Radiology, Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, CEP 01308-060, São Paulo, SP, Brazil; and Department of Radiology and Oncology, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil (M.A.Q., P.C.C.V., J.F.G.M., G.G.C., C.A.B.)
| | - Rafael F. Nunes
- From the Department of Radiology, Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, CEP 01308-060, São Paulo, SP, Brazil; and Department of Radiology and Oncology, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil (M.A.Q., P.C.C.V., J.F.G.M., G.G.C., C.A.B.)
| | - Publio C. C. Viana
- From the Department of Radiology, Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, CEP 01308-060, São Paulo, SP, Brazil; and Department of Radiology and Oncology, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil (M.A.Q., P.C.C.V., J.F.G.M., G.G.C., C.A.B.)
| | - José Flávio G. Marin
- From the Department of Radiology, Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, CEP 01308-060, São Paulo, SP, Brazil; and Department of Radiology and Oncology, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil (M.A.Q., P.C.C.V., J.F.G.M., G.G.C., C.A.B.)
| | - Giovanni G. Cerri
- From the Department of Radiology, Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, CEP 01308-060, São Paulo, SP, Brazil; and Department of Radiology and Oncology, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil (M.A.Q., P.C.C.V., J.F.G.M., G.G.C., C.A.B.)
| | - Carlos A. Buchpiguel
- From the Department of Radiology, Hospital Sírio-Libanês, Rua Dona Adma Jafet 115, CEP 01308-060, São Paulo, SP, Brazil; and Department of Radiology and Oncology, Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil (M.A.Q., P.C.C.V., J.F.G.M., G.G.C., C.A.B.)
| |
Collapse
|
47
|
Natarajan A, Agrawal A, Murthy V, Bakshi G, Joshi A, Purandare N, Shah S, Puranik A, Rangarajan V. Initial experience of Ga-68 prostate-specific membrane antigen positron emission tomography/computed tomography imaging in evaluation of biochemical recurrence in prostate cancer patients. World J Nucl Med 2019; 18:244-250. [PMID: 31516367 PMCID: PMC6714163 DOI: 10.4103/wjnm.wjnm_47_18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Gallium-68 labeled prostate-specific membrane antigen (Ga-68 PSMA) ligand (HBED-CC) is a novel tracer used for prostate cancer imaging. The aim of the study was to investigate the performance of Ga-68 PSMA positron emission tomography/computed tomography (PET/CT) in patients with biochemical recurrence (BCR) after definitive treatment. Scans of 96 consecutive patients were analyzed. Sixty-two patients received external beam radiotherapy, 34 underwent radical prostatectomy (RP), and 20 patients were on androgen deprivation therapy. Patients with prostate-specific antigen (PSA) level ≥>0.2 ng/mL following RP and PSA rise by 2 ng/mL or more above the nadir PSA following RT (Phoenix criteria) was considered as BCR, respectively. All patients underwent contrast-enhanced PET/CT after injection of 67–111 MBq Ga-68 PSMA ligand. Detection rates were correlated with serum PSA level. Detection rate for nodal metastases was compared with CT. Results of the scan were validated by using either biopsy or follow-up imaging or clinical follow-up. Seventy-four (77%) patients showed abnormal finding in Ga-68 PSMA PET/CT. The median serum PSA level of the population was 5.5 ng/ml (range 0.2–123 ng/ml). The median PSA of the positive scans was higher than that of the negative scans (6 vs. 1.7 ng/ml) and was statistically significant (P = 0.001 by Mann–Whitney U-test). In post-RP group, the detection rates were 23%, 50%, and 82% for PSA <1, 1–2, and >2 ng/ml, respectively. For post-RT, the detection was 86%, 85%, and 95% for PSA 2–5, 5.1–10, and >10 ng/ml, respectively. PSMA PET/CT revealed nodal metastases in 52 (54%) patients while CT showed pathological nodes only in 27 (28%) patients. Overall PSMA PET/CT revealed more number of nodes than CT (111 vs. 48 nodal station). PSMA PET/CT showed relapse in prostate/prostatic bed in 26 (27%) patients, nodal metastases in 50 (52%), skeletal metastases in 20 (21%), and other sites in 4 (4%) patients. Ga-68 PSMA PET/CT has high detection rate for localizing the site of recurrence in patients with biochemical failure and is superior to CT scan in the detection of nodal disease.
Collapse
Affiliation(s)
- Aravintho Natarajan
- Department of Nuclear Medicine and Molecular Imaging, Uro-Oncology Disease Management Group, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Archi Agrawal
- Department of Nuclear Medicine and Molecular Imaging, Uro-Oncology Disease Management Group, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Vedang Murthy
- Department of Radiation Oncology, Uro-Oncology Disease Management Group, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Ganesh Bakshi
- Department of Surgical Oncology, Uro-Oncology Disease Management Group, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Amit Joshi
- Department of Medical Oncology, Uro-Oncology Disease Management Group, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Nilendu Purandare
- Department of Nuclear Medicine and Molecular Imaging, Uro-Oncology Disease Management Group, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Sneha Shah
- Department of Nuclear Medicine and Molecular Imaging, Uro-Oncology Disease Management Group, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Ameya Puranik
- Department of Nuclear Medicine and Molecular Imaging, Uro-Oncology Disease Management Group, Tata Memorial Hospital, Mumbai, Maharashtra, India
| | - Venkatesh Rangarajan
- Department of Nuclear Medicine and Molecular Imaging, Uro-Oncology Disease Management Group, Tata Memorial Hospital, Mumbai, Maharashtra, India
| |
Collapse
|
48
|
Hope TA, Goodman JZ, Allen IE, Calais J, Fendler WP, Carroll PR. Metaanalysis of 68Ga-PSMA-11 PET Accuracy for the Detection of Prostate Cancer Validated by Histopathology. J Nucl Med 2018; 60:786-793. [PMID: 30530831 DOI: 10.2967/jnumed.118.219501] [Citation(s) in RCA: 165] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 11/01/2018] [Indexed: 12/17/2022] Open
Abstract
68Ga-PSMA-11 PET is used to stage patients with prostate cancer. We performed an updated metaanalysis that separates imaging at the time of diagnosis and at the time of biochemical recurrence and focuses on pathology correlation in both populations. Methods: We searched the MEDLINE and EMBASE databases using the PRISMA statement. Quality was assessed using the Quality Assessment of Diagnostic Accuracy Studies tool 2. In total, 1,811 studies were screened, 58 were analyzed, 41 were included for qualitative synthesis, and 29 were included for quantitative analysis. A random-effect model and a hierarchical summary receiver-operating-characteristic model were used to summarize the sensitivity, specificity, positive predictive value (PPV), negative predictive value, and accuracy for pelvic lymph nodes in initial staging compared with pathology at prostatectomy and the PPV for lesions with pathologic correlation in those with biochemical recurrence. We also summarized the detection rate of 68Ga-PSMA-11 in those with biochemical recurrence stratified by prostate-specific antigen (PSA) at the time of imaging. Results: The metaanalysis of 68Ga-PSMA-11 at initial staging demonstrated a sensitivity and specificity of 0.74 (95% confidence interval [95% CI], 0.51-0.89) and 0.96 (95% CI, 0.85-0.99), respectively, using nodal pathology at prostatectomy as a gold standard. At biochemical recurrence, the PPV was 0.99 (95% CI, 0.96-1.00). The detection rate was 0.63 (95% CI, 0.55-0.70), with a PSA of less than 2.0 and 0.94 (95% CI, 0.91-0.96) with a PSA of more than 2.0. Conclusion: 68Ga-PSMA-11 performed well for the localization of metastatic prostate cancer at initial staging and at the time of biochemical recurrence.
Collapse
Affiliation(s)
- Thomas A Hope
- Department of Radiology and Biomedical Imaging, UCSF, San Francisco, California .,Department of Radiology, San Francisco VA Medical Center, San Francisco, California.,UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California.,Department of Urology, UCSF, San Francisco, California
| | | | - Isabel E Allen
- Department of Epidemiology and Biostatistics, UCSF, San Francisco, California
| | - Jeremie Calais
- Ahmanson Translational Imaging Division, Department of Molecular and Medical Pharmacology, UCLA, Los Angeles, California; and
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
| | - Peter R Carroll
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California.,Department of Urology, UCSF, San Francisco, California
| |
Collapse
|
49
|
Afaq A, Bomanji J. Prostate-specific membrane antigen positron emission tomography in the management of recurrent prostate cancer. Br Med Bull 2018; 128:37-48. [PMID: 30272121 DOI: 10.1093/bmb/ldy032] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 08/30/2018] [Indexed: 11/12/2022]
Abstract
INTRODUCTION There is an unmet clinical need for early, accurate imaging of recurrent prostate cancer to improve patient outcomes. Staging, by conventional bone scintigraphy and CT have become outdated. 68Ga-PSMA PET/CT imaging in this setting has developed rapidly, with widespread International adoption in line with evidence-based guidelines in this group of patients. SOURCES OF DATA A PubMed search of English language articles was performed using following keywords: PSMA, PET/CT, biochemical recurrence, prostate cancer. The search revealed 85 articles, of which 75 were original; 70 of these involved use of the most widely available type of PSMA tracer (HBED). The review also relied on the clinical experience of reporting over 1000 PSMA PET/CT studies at a major tertiary referral centre for uro-oncology, with the majority of cases having been performed in the biochemical recurrence setting from 2015 to 2018. AREAS OF AGREEMENT 68Ga-PSMA PET is a game changer and superior to choline PET and other established tracers which have been used in prostate cancer evaluation. Detection of recurrence at the prostate bed remains challenging due to bladder and urethral tracer accumulation. The main strength of PSMA PET/CT is its ability to identify small (<8 mm) pathological lymph nodes, upstaging nodal status in up to two-thirds of cases. Additionally, PSMA PET/CT, detects bone and bone marrow metastases missed by conventional bone and CT imaging. Thus, PSMA PET/CT has major impact on patient management, with studies reporting overall changes in 39-76% of cases. AREAS OF CONTROVERSY Controversy exists regarding patient access and NHS affordability of PSMA PET/CT imaging. Currently, no reimbursement is available under the NHS tariff system. The cost outlay for tertiary hospital linked PET centres ranges from £150-170 K. Large referral volumes, and technical advances in manufacturing process will make this tracer cost neutral and similar to the current funded, but less sensitive, choline PET. Current NICE guidelines for prostate cancer management do not include a recommendation on when PSMA PET/CT should be used and this is likely to remain the case in the next revision, due in 2019. GROWING POINTS Although PSMA PET/CT imaging results in significant management change, there is a need for high quality economic evaluation and cost analysis for this modality. Lack of this data will result in poor adoption of this technique and thus limit patient access. Furthermore, it is hoped that future tracers will become even more sensitive and identify disease at earlier thresholds. AREAS TIMELY FOR DEVELOPING RESEARCH Well-designed clinical trials with consideration of the health economic benefit of using PSMA PET/CT will be essential to provide a basis for entry into guidelines such as NICE and to provide a rationale for reimbursement.
Collapse
Affiliation(s)
- Asim Afaq
- Institute of Nuclear Medicine, University College London Hospitals NHS Foundation Trust, London, UK
| | - Jamshed Bomanji
- Institute of Nuclear Medicine, University College London Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
50
|
Das CJ, Razik A, Sharma S. Positron emission tomography in prostate cancer: An update on state of the art. Indian J Urol 2018; 34:172-179. [PMID: 30034126 PMCID: PMC6034413 DOI: 10.4103/iju.iju_320_17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Prostate cancer (PCa), one of the most common cancers in males, is a topic of active interest in imaging research. Positron emission tomography/computed tomography (PET/CT) and PET/magnetic resonance imaging (PET/MRI) have enabled the combination of morphologic and functional imaging with the promise of providing better information in guiding therapy. 18F-fluorodeoxyglucose, the workhorse radiopharmaceutical in PET imaging, has not found preference in PCa since these tumors show poor glucose uptake and can be obscured by the normal urinary excretion of the radiotracer. Hence, the last two decades have seen the development of multiple newer radiotracers and better optimization of the technical aspects of PET imaging. The combination of functional imaging and MRI holds great promise. We searched PubMed, Scopus, and Google Scholar for peer-reviewed literature concerning the advances and newer developments in the imaging of PCa between the years 2005 and 2017. This review aims at summarizing current evidence on the role of PET imaging in PCa and its impact on the diagnosis, staging, prognostication, response assessment, and restaging of this malignancy.
Collapse
Affiliation(s)
- Chandan Jyoti Das
- Department of Radio-Diagnosis, All India Institute of Medical Sciences, New Delhi, India
| | - Abdul Razik
- Department of Radio-Diagnosis, All India Institute of Medical Sciences, New Delhi, India
| | - Sanjay Sharma
- Department of Radio-Diagnosis, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|