1
|
Dhere VR, Schuster DM, Goyal S, Schreibmann E, Hershatter BW, Patel SA, Shelton JW, Hanasoge S, Patel PR, Sebastian NT, Adediran OA, Lawal IO, Jani AB. Biochemical Relapse-Free Survival in Postprostatectomy Patients Receiving 18F-Fluciclovine-Guided Prostate Bed-Only Radiation: Post Hoc Analysis of a Prospective Randomized Trial. Pract Radiat Oncol 2024; 14:e492-e499. [PMID: 39032598 DOI: 10.1016/j.prro.2024.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/17/2024] [Accepted: 05/31/2024] [Indexed: 07/23/2024]
Abstract
PURPOSE Whole-pelvis (WP) radiation therapy (radiation) improved biochemical relapse-free survival (bRFS) compared with prostate bed (PB)-only radiation in the Radiation Therapy Oncology Group 0534, but was performed in an era prior to positron emission tomography (PET) staging. Separately, 18F-fluciclovine PET/CT-guided postprostatectomy radiation improved 3-year bRFS versus radiation guided by conventional imaging alone. We hypothesized that patients who were changed from WP to PB-only radiation after PET would have bRFS that was: (a) no higher than patients initially planned for PB-only radiation; and (b) lower than patients planned for WP radiation without PET guidance. METHODS AND MATERIALS We conducted a post hoc analysis of a prospective, randomized trial comparing conventional (arm 1) versus PET-guided (arm 2) postprostatectomy radiation. In arm 2, pre-PET treatment field decisions were recorded and post-PET fields were defined per protocol; pathologic node negative (pN0) without pelvic or extrapelvic PET uptake received PB-only radiation. Three-year bRFS was compared in patients planned for WP with change to PB-only radiation (arm 2 [WP:PB]) vs arm 2 patients planned for PB-only with final radiation to PB-only (arm 2 [PB:PB]) and arm 1 pN0 patients treated with WP radiation (arm 1 [WP]) using the Z test and log-rank test. Demographics were compared using the chi-square test, Fisher exact test, or analysis of variance, as appropriate. RESULTS We identified 10 arm 2 (WP:PB), 31 arm 2 (PB:PB) and 11 arm 1 (WP) patients. Androgen deprivation was used in 50.0% of arm 2 (WP:PB) and 3.2% of arm 2 (PB:PB) patients, P < .01. Median preradiation prostate-specific antigen was higher in arm 2 (WP:PB) vs arm 2 (PB:PB) patients (0.4 vs 0.2 ng/mL, P = .03); however, there were no significant differences in T stage, Gleason score, or margin positivity. Three-year bRFS was 80% in arm 2 (WP:PB) vs 87.4% in arm 2 (PB:PB), P = .47, respectively. Arm 1(WP) patients had significantly worse 3-year (23%) bRFS vs arm 2 (WP:PB), P < .01. CONCLUSIONS Patients initially planned for WP radiation with field decision change to PB-only radiation after PET showed (1) no significant difference in 3-year bRFS compared with patients initially planned for PB-only radiation; and (2) improved bRFS compared with patients receiving WP radiation without PET guidance. PET-guided volume de-escalation in selected patients may be 1 approach to mitigating toxicity without compromising outcomes.
Collapse
Affiliation(s)
- Vishal R Dhere
- Department of Radiation Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia.
| | - David M Schuster
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia
| | - Subir Goyal
- Biostatistics Shared Resource, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Eduard Schreibmann
- Department of Radiation Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Bruce W Hershatter
- Department of Radiation Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Sagar A Patel
- Department of Radiation Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Joseph W Shelton
- Department of Radiation Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Sheela Hanasoge
- Department of Radiation Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Pretesh R Patel
- Department of Radiation Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Nikhil T Sebastian
- Department of Radiation Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| | - Omotayo A Adediran
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia
| | - Ismaheel O Lawal
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, Georgia
| | - Ashesh B Jani
- Department of Radiation Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia
| |
Collapse
|
2
|
Alberts IL, Seifert R, Werner RA, Rowe SP, Afshar-Oromieh A. Prostate-specific Membrane Antigen: Diagnostics. PET Clin 2024; 19:351-362. [PMID: 38702228 DOI: 10.1016/j.cpet.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2024]
Abstract
Since its clinical introduction in May 2011, prostate-specific membrane antigen (PSMA)-PET/computed tomography has quickly gained worldwide recognition as a significant breakthrough in prostate cancer diagnostics. In the meantime, several new PSMA radioligands for PET imaging have been introduced into routine clinical practice. This article aims to introduce the most commonly used tracers and their key areas of application.
Collapse
Affiliation(s)
- Ian L Alberts
- Molecular Imaging and Therapy, BC Cancer - Vancouver, 600 West 10th Avenue, Vancouver, British Columbia V5Z 1H5, Canada
| | - Robert Seifert
- University Clinic for Nuclear Medicine, Inselspital, University Hospital of Bern, Bern, Switzerland
| | - Rudolf A Werner
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacherstr. 6, 97080 Würzburg, Germany; Division of Nuclear Medicine, Department of Diagnostic and Interventional Radiology and Nuclear Medicine, Goethe University Frankfurt, University Hospital, Germany
| | - Steven P Rowe
- Molecular Imaging and Therapeutics, Department of Radiology, University of North Carolina, Chapel Hill, NC, USA
| | - Ali Afshar-Oromieh
- University Clinic for Nuclear Medicine, Inselspital, University Hospital of Bern, Bern, Switzerland.
| |
Collapse
|
3
|
Tateishi U, Kimura K, Tsuchiya J, Kano D, Watabe T, Nonomura N, Saito K, Yokoyama K, Yamagiwa K, Adachi T, Kojima Y, Yoshida S, Fujii Y. Phase I/IIa trial of 18F-prostate specific membrane antigen (PSMA) 1007 PET/CT in healthy volunteers and prostate cancer patients. Jpn J Clin Oncol 2024; 54:282-291. [PMID: 38066703 DOI: 10.1093/jjco/hyad166] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 11/15/2023] [Indexed: 03/12/2024] Open
Abstract
OBJECTIVE 18F-PSMA 1007 is a promising PET tracer for prostate cancer. We aimed to examine the safety, biodistribution, radiation dosimetry, and clinical effectiveness in Japanese healthy volunteers and patients with prostate cancer. METHODS Part A evaluated the pharmacokinetics and exposure doses in three healthy volunteers. Part B evaluated the diagnostic accuracy in patients with untreated preoperative prostate cancer (Cohort 1, n = 7) and patients with biochemical recurrence (Cohort 2, n = 3). All subjects received a single dose of 3.7 MBq/kg 18F-PSMA 1007. Results: 18F-PSMA 1007 was found to be safe and well tolerated in all subjects. No serous AEs or drug-related AEs were identified during the present study. The average blood radioactivity concentration reached a maximum of 47.87 ± 1.05 (percentage of injected dose [%ID]/ml) at 5 min and then decreased to 1.60 ± 0.78 in 6 h. The systemic radioactivity reached a maximum of 211.05 ± 6.77 (%ID$\times$103) at 5 min and decreased to 7.18 ± 3.91 in 6 h. The sensitivity and positive predictive value were 100% and 100% based on both pathologic and imaging confirmation as gold standard. In Cohort 1, 15 primary foci (11.9%) were >5 mm in the largest diameter and identified in 39 of 126 segments (30.1%). The sensitivity, specificity, positive predictive value, negative predictive value, and accuracy for 60 min uptake time acquisition were 80.0, 96.5, 91.4, 91.2 and 91.3%, respectively. CONCLUSIONS Our study revealed that 18F-PSMA 1007 was safe, well tolerated and showed high accuracy in the diagnosis of prostate cancer.
Collapse
Affiliation(s)
- Ukihide Tateishi
- Department of Diagnostic Radiology and Nuclear Medicine, Tokyo Medical and Dental University, Tokyo
| | - Koichiro Kimura
- Department of Diagnostic Radiology and Nuclear Medicine, Tokyo Medical and Dental University, Tokyo
| | - Junichi Tsuchiya
- Department of Diagnostic Radiology and Nuclear Medicine, Tokyo Medical and Dental University, Tokyo
| | - Daisuke Kano
- Department of Diagnostic Radiology and Nuclear Medicine, Tokyo Medical and Dental University, Tokyo
- Pharmaceutical Department, National Cancer Center Hospital East, Chiba
| | - Tadashi Watabe
- Department of Nuclear Medicine and Tracer Kinetics, Graduate School of Medicine, Osaka University, Osaka
| | - Norio Nonomura
- Department of Urology, Graduate School of Medicine, Osaka University, Osaka
| | - Katsuhiko Saito
- Medical & Advanced Equipment Unit, Industrial Equipment Division, Sumitomo Heavy Industries. Ltd., Tokyo
| | - Kota Yokoyama
- Department of Diagnostic Radiology and Nuclear Medicine, Tokyo Medical and Dental University, Tokyo
| | - Ken Yamagiwa
- Department of Diagnostic Radiology and Nuclear Medicine, Tokyo Medical and Dental University, Tokyo
| | - Takuya Adachi
- Department of Diagnostic Radiology and Nuclear Medicine, Tokyo Medical and Dental University, Tokyo
| | - Yuji Kojima
- Department of Diagnostic Radiology and Nuclear Medicine, Tokyo Medical and Dental University, Tokyo
| | - Soichiro Yoshida
- Department of Urology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yasuhisa Fujii
- Department of Urology, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
4
|
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
|
5
|
Awiwi MO, Gjoni M, Vikram R, Altinmakas E, Dogan H, Bathala TK, Naik S, Ravizzini G, Kandemirli SG, Elsayes KM, Salem UI. MRI and PSMA PET/CT of Biochemical Recurrence of Prostate Cancer. Radiographics 2023; 43:e230112. [PMID: 37999983 DOI: 10.1148/rg.230112] [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: 11/26/2023]
Abstract
Prostate cancer may recur several years after definitive treatment, such as prostatectomy or radiation therapy. A rise in serum prostate-specific antigen (PSA) level is the first sign of disease recurrence, and this is termed biochemical recurrence. Patients with biochemical recurrence have worse survival outcomes. Radiologic localization of recurrent disease helps in directing patient management, which may vary from active surveillance to salvage radiation therapy, androgen-deprivation therapy, or other forms of systemic and local therapy. The likelihood of detecting the site of recurrence increases with higher serum PSA level. MRI provides optimal diagnostic performance for evaluation of the prostatectomy bed. Prostate-specific membrane antigen (PSMA) PET radiotracers currently approved by the U.S. Food and Drug Administration demonstrate physiologic urinary excretion, which can obscure recurrence at the vesicourethral junction. However, MRI and PSMA PET/CT have comparable diagnostic performance for evaluation of local recurrence after external-beam radiation therapy or brachytherapy. PSMA PET/CT outperforms MRI in identifying recurrence involving the lymph nodes and bones. Caveats for use of both PSMA PET/CT and MRI do exist and may cause false-positive or false-negative results. Hence, these techniques have complementary roles and should be interpreted in conjunction with each other, taking the patient history and results of any additional prior imaging studies into account. Novel PSMA agents at various stages of investigation are being developed, and preliminary data show promising results; these agents may revolutionize the landscape of prostate cancer recurrence imaging in the future. ©RSNA, 2023 Quiz questions for this article are available through the Online Learning Center. See the invited commentary by Turkbey in this issue. The slide presentation from the RSNA Annual Meeting is available for this article.
Collapse
Affiliation(s)
- Muhammad O Awiwi
- From the Division of Diagnostic Imaging, University of Texas Health Science Center at Houston, 6431 Fannin St, MSB 2.132, Houston, TX 77030 (M.O.A.); Department of Medicine, Istanbul University-Cerrahpasa Hospital, Istanbul, Turkey (M.G.); Departments of Abdominal Imaging (R.V., T.K.B., S.N., K.M.E., U.I.S.) and Nuclear Medicine (G.R.), Division of Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, Tex; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (E.A.); Department of Radiology, Koç University School of Medicine, Istanbul, Turkey (E.A., H.D.); and Department of Nuclear Medicine, Division of Diagnostic Imaging, University of Iowa Hospitals and Clinics, Iowa City, Iowa (S.G.K.)
| | - Migena Gjoni
- From the Division of Diagnostic Imaging, University of Texas Health Science Center at Houston, 6431 Fannin St, MSB 2.132, Houston, TX 77030 (M.O.A.); Department of Medicine, Istanbul University-Cerrahpasa Hospital, Istanbul, Turkey (M.G.); Departments of Abdominal Imaging (R.V., T.K.B., S.N., K.M.E., U.I.S.) and Nuclear Medicine (G.R.), Division of Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, Tex; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (E.A.); Department of Radiology, Koç University School of Medicine, Istanbul, Turkey (E.A., H.D.); and Department of Nuclear Medicine, Division of Diagnostic Imaging, University of Iowa Hospitals and Clinics, Iowa City, Iowa (S.G.K.)
| | - Raghunandan Vikram
- From the Division of Diagnostic Imaging, University of Texas Health Science Center at Houston, 6431 Fannin St, MSB 2.132, Houston, TX 77030 (M.O.A.); Department of Medicine, Istanbul University-Cerrahpasa Hospital, Istanbul, Turkey (M.G.); Departments of Abdominal Imaging (R.V., T.K.B., S.N., K.M.E., U.I.S.) and Nuclear Medicine (G.R.), Division of Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, Tex; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (E.A.); Department of Radiology, Koç University School of Medicine, Istanbul, Turkey (E.A., H.D.); and Department of Nuclear Medicine, Division of Diagnostic Imaging, University of Iowa Hospitals and Clinics, Iowa City, Iowa (S.G.K.)
| | - Emre Altinmakas
- From the Division of Diagnostic Imaging, University of Texas Health Science Center at Houston, 6431 Fannin St, MSB 2.132, Houston, TX 77030 (M.O.A.); Department of Medicine, Istanbul University-Cerrahpasa Hospital, Istanbul, Turkey (M.G.); Departments of Abdominal Imaging (R.V., T.K.B., S.N., K.M.E., U.I.S.) and Nuclear Medicine (G.R.), Division of Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, Tex; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (E.A.); Department of Radiology, Koç University School of Medicine, Istanbul, Turkey (E.A., H.D.); and Department of Nuclear Medicine, Division of Diagnostic Imaging, University of Iowa Hospitals and Clinics, Iowa City, Iowa (S.G.K.)
| | - Hakan Dogan
- From the Division of Diagnostic Imaging, University of Texas Health Science Center at Houston, 6431 Fannin St, MSB 2.132, Houston, TX 77030 (M.O.A.); Department of Medicine, Istanbul University-Cerrahpasa Hospital, Istanbul, Turkey (M.G.); Departments of Abdominal Imaging (R.V., T.K.B., S.N., K.M.E., U.I.S.) and Nuclear Medicine (G.R.), Division of Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, Tex; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (E.A.); Department of Radiology, Koç University School of Medicine, Istanbul, Turkey (E.A., H.D.); and Department of Nuclear Medicine, Division of Diagnostic Imaging, University of Iowa Hospitals and Clinics, Iowa City, Iowa (S.G.K.)
| | - Tharakeswara K Bathala
- From the Division of Diagnostic Imaging, University of Texas Health Science Center at Houston, 6431 Fannin St, MSB 2.132, Houston, TX 77030 (M.O.A.); Department of Medicine, Istanbul University-Cerrahpasa Hospital, Istanbul, Turkey (M.G.); Departments of Abdominal Imaging (R.V., T.K.B., S.N., K.M.E., U.I.S.) and Nuclear Medicine (G.R.), Division of Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, Tex; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (E.A.); Department of Radiology, Koç University School of Medicine, Istanbul, Turkey (E.A., H.D.); and Department of Nuclear Medicine, Division of Diagnostic Imaging, University of Iowa Hospitals and Clinics, Iowa City, Iowa (S.G.K.)
| | - Sagar Naik
- From the Division of Diagnostic Imaging, University of Texas Health Science Center at Houston, 6431 Fannin St, MSB 2.132, Houston, TX 77030 (M.O.A.); Department of Medicine, Istanbul University-Cerrahpasa Hospital, Istanbul, Turkey (M.G.); Departments of Abdominal Imaging (R.V., T.K.B., S.N., K.M.E., U.I.S.) and Nuclear Medicine (G.R.), Division of Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, Tex; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (E.A.); Department of Radiology, Koç University School of Medicine, Istanbul, Turkey (E.A., H.D.); and Department of Nuclear Medicine, Division of Diagnostic Imaging, University of Iowa Hospitals and Clinics, Iowa City, Iowa (S.G.K.)
| | - Gregory Ravizzini
- From the Division of Diagnostic Imaging, University of Texas Health Science Center at Houston, 6431 Fannin St, MSB 2.132, Houston, TX 77030 (M.O.A.); Department of Medicine, Istanbul University-Cerrahpasa Hospital, Istanbul, Turkey (M.G.); Departments of Abdominal Imaging (R.V., T.K.B., S.N., K.M.E., U.I.S.) and Nuclear Medicine (G.R.), Division of Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, Tex; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (E.A.); Department of Radiology, Koç University School of Medicine, Istanbul, Turkey (E.A., H.D.); and Department of Nuclear Medicine, Division of Diagnostic Imaging, University of Iowa Hospitals and Clinics, Iowa City, Iowa (S.G.K.)
| | - Sedat Giray Kandemirli
- From the Division of Diagnostic Imaging, University of Texas Health Science Center at Houston, 6431 Fannin St, MSB 2.132, Houston, TX 77030 (M.O.A.); Department of Medicine, Istanbul University-Cerrahpasa Hospital, Istanbul, Turkey (M.G.); Departments of Abdominal Imaging (R.V., T.K.B., S.N., K.M.E., U.I.S.) and Nuclear Medicine (G.R.), Division of Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, Tex; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (E.A.); Department of Radiology, Koç University School of Medicine, Istanbul, Turkey (E.A., H.D.); and Department of Nuclear Medicine, Division of Diagnostic Imaging, University of Iowa Hospitals and Clinics, Iowa City, Iowa (S.G.K.)
| | - Khaled M Elsayes
- From the Division of Diagnostic Imaging, University of Texas Health Science Center at Houston, 6431 Fannin St, MSB 2.132, Houston, TX 77030 (M.O.A.); Department of Medicine, Istanbul University-Cerrahpasa Hospital, Istanbul, Turkey (M.G.); Departments of Abdominal Imaging (R.V., T.K.B., S.N., K.M.E., U.I.S.) and Nuclear Medicine (G.R.), Division of Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, Tex; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (E.A.); Department of Radiology, Koç University School of Medicine, Istanbul, Turkey (E.A., H.D.); and Department of Nuclear Medicine, Division of Diagnostic Imaging, University of Iowa Hospitals and Clinics, Iowa City, Iowa (S.G.K.)
| | - Usama I Salem
- From the Division of Diagnostic Imaging, University of Texas Health Science Center at Houston, 6431 Fannin St, MSB 2.132, Houston, TX 77030 (M.O.A.); Department of Medicine, Istanbul University-Cerrahpasa Hospital, Istanbul, Turkey (M.G.); Departments of Abdominal Imaging (R.V., T.K.B., S.N., K.M.E., U.I.S.) and Nuclear Medicine (G.R.), Division of Diagnostic Imaging, University of Texas MD Anderson Cancer Center, Houston, Tex; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY (E.A.); Department of Radiology, Koç University School of Medicine, Istanbul, Turkey (E.A., H.D.); and Department of Nuclear Medicine, Division of Diagnostic Imaging, University of Iowa Hospitals and Clinics, Iowa City, Iowa (S.G.K.)
| |
Collapse
|
6
|
Murray J, De Meerleer G. How High Should We Go for Para-aortic Lymph Node Radiation Therapy? Int J Radiat Oncol Biol Phys 2023; 117:641-643. [PMID: 37739609 DOI: 10.1016/j.ijrobp.2023.06.248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 06/21/2023] [Accepted: 06/23/2023] [Indexed: 09/24/2023]
Affiliation(s)
- Julia Murray
- Royal Marsden NHS Foundation Trust, Sutton, United Kingdom; Division of Radiotherapy and Imaging, Institute of Cancer Research, Sutton, United Kingdom.
| | - Gert De Meerleer
- Department of Radiotherapy, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
7
|
Falkenbach F, Knipper S, Koehler D, Ambrosini F, Steuber T, Graefen M, Budäus L, Eiber M, Lunger L, Lischewski F, Heck MM, Maurer T. Safety and efficiency of repeat salvage lymph node dissection for recurrence of prostate cancer using PSMA-radioguided surgery (RGS) after prior salvage lymph node dissection with or without initial RGS support. World J Urol 2023; 41:2343-2350. [PMID: 37515651 PMCID: PMC10465644 DOI: 10.1007/s00345-023-04534-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/06/2023] [Indexed: 07/31/2023] Open
Abstract
BACKGROUND AND OBJECTIVE Metastasis-directed therapy is a feasible option for low PSA, recurrent locoregional metastatic prostate cancer. After initial salvage surgery, patients with good response might consider a repeat salvage surgery in case of recurrent, isolated, and PSMA-positive metastases. This analysis aimed to evaluate the oncological outcome and safety of repeat PSMA-targeted radioguided surgery (RGS) after either prior RGS or "standard" salvage lymph node dissection (SLND). MATERIALS AND METHODS We identified 37 patients undergoing repeat RGS after prior SLND (n = 21) (SLND-RGS) or prior RGS (n = 16) (RGS-RGS) between 2014 and 2021 after initial radical prostatectomy with or without pelvic radiation therapy at two German tertiary referral centers. Kaplan-Meier analyses and uni-/multivariable Cox regression models were used to investigate factors associated with biochemical recurrence-free survival (BRFS) and treatment-free survival (TFS) after repeat salvage surgery. RESULTS AND LIMITATIONS Complete Biochemical Response (cBR, PSA < 0.2 ng/ml) was observed in 20/32 patients (5 NA). Median overall BRFS [95% confidence interval (CI)] after repeat salvage surgery was 10.8 months (mo) (5.3-22). On multivariable regression, only age (HR 1.09, 95% CI 1.01-1.17) and preoperative PSA (HR 1.23, 95% CI 1.01-1.50) were associated with shorter BRFS, although PSA (HR 1.16, 95% CI 0.99-1.36) did not achieve significant predictor status in univariable analysis before (p value = 0.07). Overall, one year after second salvage surgery, 89% of the patients (number at risk: 19) did not receive additional treatment and median TFS was not reached. Clavien-Dindo grade > 3a complications were observed in 8% (3/37 patients). Limitations are the retrospective evaluation, heterogeneous SLND procedures, lack of long-term follow-up data, and small cohort size. CONCLUSION In this study, repeat RGS was safe and provided clinically meaningful biochemical recurrence- and treatment-free intervals for selected cases. Patients having low preoperative PSA seemed to benefit most of repeat RGS, irrespective of prior SLND or RGS or the time from initial RP/first salvage surgery.
Collapse
Affiliation(s)
- Fabian Falkenbach
- Martini-Klinik Prostate Cancer Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany.
| | - Sophie Knipper
- Martini-Klinik Prostate Cancer Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Daniel Koehler
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Francesca Ambrosini
- Martini-Klinik Prostate Cancer Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
- Department of Urology, IRCCS Policlinico San Martino Hospital, University of Genova, Genoa, Italy
| | - Thomas Steuber
- Martini-Klinik Prostate Cancer Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
- Department of Urology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Graefen
- Martini-Klinik Prostate Cancer Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Lars Budäus
- Martini-Klinik Prostate Cancer Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
| | - Matthias Eiber
- Department of Nuclear Medicine, Rechts der Isar Medical Center, Technical University of Munich, Munich, Germany
| | - Lukas Lunger
- Department of Urology, Rechts der Isar Medical Center, Technical University of Munich, Munich, Germany
| | - Flemming Lischewski
- Department of Urology, Rechts der Isar Medical Center, Technical University of Munich, Munich, Germany
| | - Matthias M Heck
- Department of Urology, Rechts der Isar Medical Center, Technical University of Munich, Munich, Germany
| | - Tobias Maurer
- Martini-Klinik Prostate Cancer Center Hamburg-Eppendorf, Martinistr. 52, 20246, Hamburg, Germany
- Department of Urology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
8
|
Padayachee J, Chaudhary S, Shim B, So J, Lim R, Raman S. Utilizing clinical, pathological and radiological information to guide postoperative radiotherapy in prostate cancer. Expert Rev Anticancer Ther 2023; 23:293-305. [PMID: 36795862 DOI: 10.1080/14737140.2023.2181795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
INTRODUCTION A detectable and rising PSA following radical prostatectomy is indicative of recurrent prostate cancer. Salvage radiotherapy (SRT) with/without androgen deprivation therapy represents the main treatment option for these patients and has been historically associated with a biochemical control rate of ~70%. To determine the optimal timing, diagnostic workup, radiotherapy dosefractionation, treatment volume, and use of systemic therapy, several informative studies have been conducted in the last decade. AREAS COVERED This review examines the recent evidence to guide radiotherapy decision making in the SRT setting. Key topics include adjuvant vs salvage RT, utilization of molecular imaging and genomic classifiers, length of androgen deprivation therapy, inclusion of elective pelvic volume, and emerging role for hypofractionation. EXPERT OPINION Recently reported trials, conducted in an era prior to the routine use of molecular imaging and genomic classifiers, have been pivotal in establishing the current standard of care for SRT in prostate cancer. However, decisions about radiation treatment and systemic therapy may be tailored based on available prognostic and predictive biomarkers. Data from contemporary clinical trials are awaited to define and establish individualized, biomarker-driven approaches for SRT.
Collapse
Affiliation(s)
- Jerusha Padayachee
- Department of Radiation Oncology, Auckland City Hospital, Auckland, New Zealand
| | - Simone Chaudhary
- Princess Margaret Hospital Cancer Centre, Radiation Medicine Program, Toronto, ON, Canada
| | - Brian Shim
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jonathan So
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Remy Lim
- Mercy PET/CT Epsom, Auckland, New Zealand.,Department of Radiology, Auckland City Hospital, Auckland, New Zealand
| | - Srinivas Raman
- Princess Margaret Hospital Cancer Centre, Radiation Medicine Program, Toronto, ON, Canada
| |
Collapse
|
9
|
Variability of radiotherapy volume delineation: PSMA PET/MRI and MRI based clinical target volume and lymph node target volume for high-risk prostate cancer. Cancer Imaging 2023; 23:1. [PMID: 36600283 DOI: 10.1186/s40644-022-00518-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 12/25/2022] [Indexed: 01/06/2023] Open
Abstract
PURPOSE A comparative retrospective study to assess the impact of PSMA Ligand PET/MRI ([68 Ga]-Ga-PSMA-11 and [18F]-F-PSMA-1007 PET/MRI) as a new method of target delineation compared to conventional imaging on whole-pelvis radiotherapy for high-risk prostate cancer (PCa). PATIENTS AND METHODS Forty-nine patients with primary high-risk PCa completed the whole-pelvis radiotherapy plan based on PSMA PET/MRI and MRI. The primary endpoint compared the size and overlap of clinical target volume (CTV) and nodal gross tumour volume (GTVn) based on PSMA PET/MRI and MRI. The diagnostic performance of two methods for pelvic lymph node metastasis (PLNM) was evaluated. RESULTS In the radiotherapy planning for high-risk PCa patients, there was a significant correlation between MRI-CTV and PET/MRI-CTV (P = 0.005), as well as between MRI-GTVn and PET/MRI-GTVn (P < 0.001). There are non-significant differences in the CTV and GTVn based on MRI and PET/MRI images (P = 0.660, P = 0.650, respectively). The conformity index (CI), lesion coverage factor (LCF) and Dice similarity coefficient (DSC) of CTVs were 0.999, 0.953 and 0.954. The CI, LCF and DSC of GTVns were 0.927, 0.284, and 0.32. Based on pathological lymph node analysis of 463 lymph nodes from 37 patients, the sensitivity, specificity of PET/MRI in the diagnosis of PLNM were 77.78% and 99.76%, respectively, which were higher than those of MRI (P = 0.011). Eight high-risk PCa patients who finished PSMA PET/MRI changed their N or M stage. CONCLUSION The CTV delineated based on PET/MRI and MRI differ little. The GTVn delineated based on PET/MRI encompasses metastatic pelvic lymph nodes more accurately than MRI and avoids covering pelvic lymph nodes without metastasis. We emphasize the utility of PET/MRI fusion images in GTVn delineation in whole pelvic radiotherapy for PCa. The use of PSMA PET/MRI aids in the realization of more individual and precise radiotherapy for PCa.
Collapse
|
10
|
Kroenke M, Schweiger L, Horn T, Haller B, Schwamborn K, Wurzer A, Maurer T, Wester HJ, Eiber M, Rauscher I. Validation of 18F-rhPSMA-7 and 18F-rhPSMA-7.3 PET Imaging Results with Histopathology from Salvage Surgery in Patients with Biochemical Recurrence of Prostate Cancer. J Nucl Med 2022; 63:1809-1814. [PMID: 35393348 PMCID: PMC9730917 DOI: 10.2967/jnumed.121.263707] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 03/28/2022] [Indexed: 01/11/2023] Open
Abstract
18F-rhPSMA-7, and its single diastereoisomer form, 18F-rhPSMA-7.3, are prostate-specific membrane antigen (PSMA)-targeting radiopharmaceuticals. Here, we investigated their accuracy for the assessment of lymph node (LN) metastases validated by histopathology. Methods: Data from 58 patients with biochemical recurrence of prostate cancer after radical prostatectomy receiving salvage surgery after PET imaging with 18F-rhPSMA-7 or 18F-rhPSMA-7.3 were retrospectively reviewed. Two nuclear medicine physicians reviewed all PET scans and morphologic imaging in consensus. Readers were masked from the results of histopathology. PET and morphologic imaging were correlated with histopathology from resected LNs. Results: In 75 of 150 resected regions in 54 of 58 patients, tumor lesions were present in histopathology. The template-based specificity of PET (18F-rhPSMA-7 and 18F-rhPSMA-7.3 combined) and morphologic imaging was 93.3% and 100%, respectively. However, 18F-rhPSMA-7 and 18F-rhPSMA-7.3 PET detected metastases in 61 of 75 histopathologically proven metastatic LN fields (81.3%) whereas morphologic imaging was positive in only 9 of 75 (12.0%). The positive predictive value was 92.4% for 18F-rhPSMA-7 and 18F-rhPSMA-7.3 PET and 100% for morphologic imaging. 18F-rhPSMA-7 and 18F-rhPSMA-7.3 PET performance was significantly superior to morphologic imaging (difference in the areas under the receiver-operating-characteristic curves, 0.222; 95% CI, 0.147-0.298; P < 0.001). The mean size of PET-positive and histologically confirmed LN metastases was 6.3 ± 3.1 mm (range, 2-15 mm) compared with a mean size of 9.8 ± 2.5 mm (range, 7-15 mm) on morphologic imaging. Conclusion: 18F-rhPSMA-7 and 18F-rhPSMA-7.3 PET offer a high positive predictive value comparable to that reported for 68Ga-PSMA-11 and represent a valuable tool for guiding salvage lymphadenectomy.
Collapse
Affiliation(s)
- Markus Kroenke
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany;,Department of Radiology and Nuclear Medicine, German Heart Center Munich, Technical University of Munich, Munich, Germany
| | - Lilit Schweiger
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany;,Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Thomas Horn
- Department of Urology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Bernhard Haller
- Institute of Medical Informatics, Statistics and Epidemiology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Kristina Schwamborn
- Department of Pathology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Alexander Wurzer
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany;,Chair of Radiopharmacy, School of Medicine, Technical University of Munich, Munich, Germany; and
| | - Tobias Maurer
- Martini-Klinik and Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Hans-Jürgen Wester
- Chair of Radiopharmacy, School of Medicine, Technical University of Munich, Munich, Germany; and
| | - Matthias Eiber
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany;,Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Isabel Rauscher
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany;,Bavarian Cancer Research Center (BZKF), Munich, Germany
| |
Collapse
|
11
|
Glemser PA, Rotkopf LT, Ziener CH, Beuthien-Baumann B, Weru V, Kopp-Schneider A, Schlemmer HP, Dimitrakopoulou-Strauss A, Sachpekidis C. Hybrid imaging with [ 68Ga]PSMA-11 PET-CT and PET-MRI in biochemically recurrent prostate cancer. Cancer Imaging 2022; 22:53. [PMID: 36138437 PMCID: PMC9502876 DOI: 10.1186/s40644-022-00489-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 09/06/2022] [Indexed: 11/10/2022] Open
Abstract
AIM To compare [68Ga]PSMA-11 PET-CT, [68Ga]PSMA-11 PET-MRI and MRI in a cohort of prostate cancer (PCa) patients in biochemical recurrence after initial curative therapy. MATERIALS AND METHODS Fifty-three patients with biochemically recurrent PCa underwent whole-body [68Ga]PSMA-11 PET-CT 1 hour post-injection (p.i.) followed by [68Ga]PSMA-11 PET-MRI 2.5 hours p.i., including a multiparametric MRI pelvic protocol examination. Imaging data analysis consisted of visual (qualitative) evaluation of the PET-CT, PET-MRI and MRI scans, as well as semi-quantitative and quantitative analyses of the PET and MRI data, including calculation of the parameters standardized uptake value (SUV) and apparent diffusion coefficient (ADC) derived from the PCa lesions. Association analysis was performed between imaging and clinical data, including PSA level and Gleason score. The results were considered significant for p-values less than 0.05 (p < 0.05). RESULTS The hybrid imaging modalities [68Ga]PSMA-11 PET-CT and PET-MRI were positive in more patients than MRI alone. In particular, PET-CT detected lesions suggestive of PCa relapse in 34/53 (64.2%), PET-MRI in 36/53 (67.9%) and MRI in 23/53 patients (43.4%). While no significant differences in lesion detection rate were observed between PET-CT and PET-MRI, the latter was particularly efficient in detection of local recurrences in the prostate bed mainly due to the contribution of the MRI part of the modality. Association analysis revealed a statistically significant increase in the probability of a positive scan with increasing PSA levels for all imaging modalities. Accordingly, there was no significant association between scan positivity rate and Gleason score for any imaging modality. No significant correlation was observed between SUV and ADC values in lymph node metastases. CONCLUSION [68Ga]PSMA-11 PET-CT and PET-MRI provide equally good detection rates for PCa recurrence, both outperforming stand-alone MRI.
Collapse
Affiliation(s)
- P A Glemser
- Department of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - L T Rotkopf
- Department of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Medical Faculty, Ruprecht-Karls-University Heidelberg, 69120, Heidelberg, Germany
| | - C H Ziener
- Department of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - B Beuthien-Baumann
- Department of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - V Weru
- Department of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - A Kopp-Schneider
- Department of Biostatistics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - H P Schlemmer
- Department of Radiology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - A Dimitrakopoulou-Strauss
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69210, Heidelberg, Germany
| | - C Sachpekidis
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69210, Heidelberg, Germany.
| |
Collapse
|
12
|
Sorce G, Hoeh B, Flammia RS, Chierigo F, Hohenhorst L, Panunzio A, Nimer N, Tian Z, Gandaglia G, Tilki D, Terrone C, Gallucci M, Chun FKH, Antonelli A, Saad F, Shariat SF, Montorsi F, Briganti A, Karakiewicz PI. Rates of metastatic prostate cancer in newly diagnosed patients: Numbers needed to image according to risk level. Prostate 2022; 82:1210-1218. [PMID: 35652586 DOI: 10.1002/pros.24376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/07/2022] [Accepted: 05/13/2022] [Indexed: 12/31/2022]
Abstract
BACKGROUND The numbers needed to image to identify pelvic lymph node and/or distant metastases in newly diagnosed prostate cancer (PCa) patients according to risk level are unknown. METHODS Relying on Surveillance, Epidemiology, and End Results (2010-2016), we tabulated rates and proportions of patients with (a) lymph node or (b) distant metastases according to National Comprehensive Cancer Network (NCCN) risk level and calculated the number needed to image (NNI) for both endpoints. Multivariable logistic regression analyses were performed. RESULTS Of 145,939 newly diagnosed PCa patients assessable for analyses of pelvic lymph node metastases (cN1), 4559 (3.1%) harbored cN1 stage: 13 (0.02%), 18 (0.08%), 63 (0.3%), 512 (2.8%), and 3954 (14.9%) in low, intermediate favorable, intermediate unfavorable, high, and very high-risk levels. These resulted in NNI of 4619, 1182, 319, 35, and 7, respectively. Of 181,109 newly diagnosed PCa patients assessable for analyses of distant metastases (M1a-c ), 8920 (4.9%) harbored M1a-c stage: 50 (0.07%), 45 (0.1%), 161 (0.5%), 1290 (5.1%), and 7374 (22.0%) in low, intermediate favorable, intermediate unfavorable, high, and very high-risk. These resulted in NNI of 1347, 602, 174, 20, and 5, respectively. CONCLUSIONS Our observations perfectly validated the NCCN recommendations for imaging in newly diagnosed high and very high-risk PCa patients. However, in unfavorable intermediate-risk PCa patients, in whom bone and soft tissue imaging is recommended, the NNI might be somewhat elevated to support routine imaging in clinical practice.
Collapse
Affiliation(s)
- Gabriele Sorce
- Department of Urology and Division of Experimental Oncology, URI, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Québec, Canada
| | - Benedikt Hoeh
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Québec, Canada
- Department of Urology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Rocco S Flammia
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Québec, Canada
- Department of Maternal-Child and Urological Sciences, Policlinico Umberto I Hospital, Sapienza University Rome, Rome, Italy
| | - Francesco Chierigo
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Québec, Canada
- Department of Surgical and Diagnostic Integrated Sciences (DISC), University of Genova, Genova, Italy
| | - Lukas Hohenhorst
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Québec, Canada
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Andrea Panunzio
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Québec, Canada
- Department of Urology, Azienda Ospedaliera Universitaria Integrata di Verona, University of Verona, Verona, Italy
| | - Nancy Nimer
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Québec, Canada
| | - Zhe Tian
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Québec, Canada
| | - Giorgio Gandaglia
- Department of Urology and Division of Experimental Oncology, URI, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Derya Tilki
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany
- Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
- Department of Urology, Koc University Hospital, Istanbul, Turkey
| | - Carlo Terrone
- Department of Surgical and Diagnostic Integrated Sciences (DISC), University of Genova, Genova, Italy
| | - Michele Gallucci
- Department of Maternal-Child and Urological Sciences, Policlinico Umberto I Hospital, Sapienza University Rome, Rome, Italy
| | - Felix K H Chun
- Department of Urology, University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Alessandro Antonelli
- Department of Urology, Azienda Ospedaliera Universitaria Integrata di Verona, University of Verona, Verona, Italy
| | - Fred Saad
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Québec, Canada
| | - Shahrokh F Shariat
- Departments of Urology, Weill Cornell Medical College, New York, New York, USA
- Department of Urology, Second Faculty of Medicine, Charles University, Praga, Czech Republic
- Department of Urology, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
- Institute for Urology and Reproductive Health, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
- Division of Urology, Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman, Jordan
- Department of Urology, University of Texas Southwestern, Dallas, Texas, USA
| | - Francesco Montorsi
- Department of Urology and Division of Experimental Oncology, URI, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alberto Briganti
- Department of Urology and Division of Experimental Oncology, URI, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Pierre I Karakiewicz
- Cancer Prognostics and Health Outcomes Unit, Division of Urology, University of Montréal Health Center, Montréal, Québec, Canada
| |
Collapse
|
13
|
Bogdanova NV, Radmanesh H, Ramachandran D, Knoechelmann AC, Christiansen H, Derlin T, von Klot CAJ, Merten R, Henkenberens C. The Prognostic Value of Liquid Biopsies for Benefit of Salvage Radiotherapy in Relapsed Oligometastatic Prostate Cancer. Cancers (Basel) 2022; 14:cancers14174095. [PMID: 36077632 PMCID: PMC9454496 DOI: 10.3390/cancers14174095] [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: 07/16/2022] [Revised: 08/15/2022] [Accepted: 08/22/2022] [Indexed: 11/28/2022] Open
Abstract
Simple Summary Around 30% of patients with oligometastatic prostate cancer relapse will benefit from local PET/CT-guided ablative radiotherapy (RT) with improved progression-free and ADT (Androgene Deprivation Therapy)-free survivals. Therefore, there is an urgent need for predictive testing for therapeutic benefits prior to initiation. Various tests have already been established on tumor specimens for the prediction of prostate cancer’s behavior or therapy outcome. However, in imaging-proven relapse tumor tissue from the local recurrence or metastases is often not available. Hence, there is a need for a liquid biopsy-based testing. We aimed to assess the prognostic value of CTCs- associated mRNA and blood-derived RNA for the benefit of PSMA PET-guided salvage RT in oligometastatic prostate cancer relapses. Significant correlations were found between the relative transcript levels of several investigated genes and clinicopathological parameters. Furthermore, distinct “transcriptional signatures” were found in patients with temporary and long-term benefits from RT. Abstract To assess the prognostic value of “liquid biopsies” for the benefit of salvage RT in oligometastatic prostate cancer relapse, we enrolled 44 patients in the study between the years 2016 and 2020. All the patients were diagnosed as having an oligometastatic prostate cancer relapse on prostate-specific membrane antigen (PSMA)-targeted PET-CT and underwent irradiation at the Department of Radiotherapy at the Hannover Medical School. Tumor cells and total RNA, enriched from the liquid biopsies of patients, were processed for the subsequent quantification analysis of relative transcript levels in real-time PCR. In total, 54 gene transcripts known or suggested to be associated with prostate cancer or treatment outcome were prioritized for analysis. We found significant correlations between the relative transcript levels of several investigated genes and the Gleason score, PSA (prostate-specific antigen) value, or UICC stage (tumor node metastasis -TNM classification of malignant tumors from Union for International Cancer Control). Furthermore, a significant association of MTCO2, FOXM1, SREBF1, HOXB7, FDXR, and MTRNR transcript profiles was found with a temporary and/or long-term benefit from RT. Further studies on larger patients cohorts are necessary to prove our preliminary findings for establishing liquid biopsy tests as a predictive examination method prior to salvage RT.
Collapse
Affiliation(s)
- Natalia V. Bogdanova
- Department of Radiation Oncology, Hannover Medical School, 30625 Hannover, Germany
| | - Hoda Radmanesh
- Department of Radiation Oncology, Hannover Medical School, 30625 Hannover, Germany
| | - Dhanya Ramachandran
- Gynecology Research Unit, Clinics of Obstetrics and Gynaecology, Hannover Medical School, 30625 Hannover, Germany
| | | | - Hans Christiansen
- Department of Radiation Oncology, Hannover Medical School, 30625 Hannover, Germany
| | - Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany
| | | | - Roland Merten
- Department of Radiation Oncology, Hannover Medical School, 30625 Hannover, Germany
- Correspondence: ; Tel.: +49-(0)-511-532-3590
| | | |
Collapse
|
14
|
The Role of PSMA PET/CT in the Primary Diagnosis and Follow-Up of Prostate Cancer-A Practical Clinical Review. Cancers (Basel) 2022; 14:cancers14153638. [PMID: 35892897 PMCID: PMC9367536 DOI: 10.3390/cancers14153638] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary The combination of positron emission tomography (PET)-diagnostics with ligands binding to the prostate-specific membrane antigen (PSMA) has been a diagnostic milestone in the situation of biochemical recurrence of prostate cancer and is gaining importance in primary diagnostics, providing a highly specific and sensitive diagnostic method in various clinical situations. However, the clinical application of this method requires a comprehensive knowledge of its advantages and disadvantages, potential pitfalls and influencing factors. This review aims to provide a practical clinical review of the currently available background data on PSMA PET/CT, as well as the clinical implications. Although a large amount of data already exist, a thorough analysis is complicated by study heterogeneity, showing the need for future systematic and prospective research. Abstract The importance of PSMA PET/CT in both primary diagnostics and prostate cancer recurrence has grown steadily since its introduction more than a decade ago. Over the past years, a vast amount of data have been published on the diagnostic accuracy and the impact of PSMA PET/CT on patient management. Nevertheless, a large heterogeneity between studies has made reaching a consensus difficult; this review aims to provide a comprehensive clinical review of the available scientific literature, covering the currently known data on physiological and pathological PSMA expression, influencing factors, the differences and pitfalls of various tracers, as well as the clinical implications in initial TNM-staging and in the situation of biochemical recurrence. This review has the objective of providing a practical clinical overview of the advantages and disadvantages of the examination in various clinical situations and the body of knowledge available, as well as open questions still requiring further research.
Collapse
|
15
|
Liu FY, Sheng TW, Tseng JR, Yu KJ, Tsui KH, Pang ST, Wang LJ, Lin G. Prostate-specific membrane antigen (PSMA) fusion imaging in prostate cancer: PET-CT vs PET-MRI. Br J Radiol 2022; 95:20210728. [PMID: 34767482 PMCID: PMC8978229 DOI: 10.1259/bjr.20210728] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVES To investigate whether PET-CT or PET-MRI is more appropriate for imaging prostate cancer, in terms of primary tumor detection, local staging and recurrence, as well as lymph nodes and distant metastases. METHODS A systematic literature search was conducted on Embase, PubMed/MEDLINE, and the Cochrane Library database. Studies evaluating the diagnostic performance of PET-CT vs PET-MRI in prostate cancer patients were emphasized. RESULTS We reviewed 57 original research articles during the period 2016-2021: 14 articles regarding the radiotracer PSMA; 18 articles regarding the primary tumor detection, local tumor staging, managing local recurrence; 17 articles for managing lymph node metastases; and eight articles for managing bone and other distant metastases. PSMA PET could be complementary to mpMRI for primary prostate cancer localization and is particularly valuable for PI-RADS three lesions. PET-MRI is better than PET-CT in local tumor staging due to its specific benefit in predicting extracapsular extension in MRI-occult prostate cancer patients. PET-MRI is likely superior as compared with PET-CT in detecting local recurrence, and has slightly higher detection rates than PET-CT in lymph node recurrence. PET-CT and PET-MRI seem to have equivalent performance in detecting distant bony or visceral metastases. CONCLUSION In conclusion, PET-MRI is suitable for local and regional disease, either primary staging or restaging, whereas PET-CT is valuable for managing distant bony or visceral metastasis. ADVANCES IN KNOWLEDGE We reviewed the emerging applications of PET-MRI and PET-CT in clinical aspects. Readers will gain an objective overview on the strength and shortfalls of PET-MRI or PET-CT in the management of prostate cancer.
Collapse
Affiliation(s)
- Feng-Yuan Liu
- Department of Nuclear Medicine, Chang Gung Memorial Hospital at Linkou and Chang Gung University, Taoyuan, Taiwan
| | - Ting-Wen Sheng
- Department of Medical Imaging and Intervention, New Taipei Municipal TuCheng Hospital, Chang Gung Medical Foundation, New Taipei City, Taiwan
| | - Jing-Ren Tseng
- Department of Nuclear Medicine, New Taipei Municipal TuCheng Hospital, Chang Gung Medical Foundation, New Taipei City, Taiwan
| | - Kai-Jie Yu
- Department of Urology, Chang Gung Memorial Hospital at Linkou and Chang Gung University, Taoyuan, Taiwan
| | - Ke-Hong Tsui
- Department of Urology, Chang Gung Memorial Hospital at Linkou and Chang Gung University, Taoyuan, Taiwan
| | - Se-Tong Pang
- Department of Urology, Chang Gung Memorial Hospital at Linkou and Chang Gung University, Taoyuan, Taiwan
| | - Li-Jen Wang
- Department of Medical Imaging and Intervention, New Taipei Municipal TuCheng Hospital, Chang Gung Medical Foundation, New Taipei City, Taiwan
| | - Gigin Lin
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital at Linkou and Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
16
|
Cegla P, Wojewódzka M, Gorczewska I, Chalewska W, Łapińska G, Ochman P, Sackiewicz A, Dedecjus M. Identification of the Optimal Cut-Off Value of PSA for Assessing Severity of Disease in [68Ga]Ga-PSMA-11 PET/CT Study in Prostate Cancer Patients after Radical Prostatectomy. Diagnostics (Basel) 2022; 12:diagnostics12020349. [PMID: 35204440 PMCID: PMC8871181 DOI: 10.3390/diagnostics12020349] [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: 12/14/2021] [Revised: 01/22/2022] [Accepted: 01/27/2022] [Indexed: 12/24/2022] Open
Abstract
Objective: The objective of this study was to identify the optimal cut-off value of prostate specific antigen (PSA) to assess the extent of the disease in [68Ga]Ga-PSMA-11 PET/CT study in patients after radical prostatectomy. Materials and Methods: Retrospective analysis was performed on a group of 215 patients who underwent a [68Ga]Ga-PSMA-11 PET/CT examination because of suspected recurrence after radical prostatectomy. Patients were divided into four groups: 1, no active lesions suggesting recurrence (n = 92); 2, suspected isolated local recurrence (n = 19); 3, oligometastatic disease (n = 82); and 4, polymetastatic disease (n = 22). Results: In group 1, the mean PSA level was 0.962 ng/mL (median: 0.376; min: 0.004; max: 25 ng/mL); in group 2, it was 4.970 ng/mL (median 1.320; min: 0.003; max: 40.350 ng/mL); in group 3, it was 2.802 ng/mL (median: 1.270; min: 0.020; max: 59.670 ng/mL); and in group 4, it was 4.997 ng/mL (median: 3.795; min: 0.007; max 21.110 ng/mL). Statistically significant differences were shown in PSA levels when comparing groups 1 and 2 (p = 0.0025) and groups 3 and 4 (p = 0.0474). The PSA cut-off point for discriminating groups 1 and 2 was 0.831 (sensitivity: 0.684; specificity: 0.772; area under the curve (AUC): 0.775), and for groups 3 and 4, it was 2.51 (sensitivity: 0.682; specificity: 0.780; AUC: 0.720). Conclusions: Our preliminary data suggested that the PSA level has an essential influence on determining the extent of disease in a [68Ga]Ga-PSMA-11 PET/CT study in patients after radical prostatectomy. Identification of the optimal cut-off values for the oligo- and polymetastatic diseases might be helpful in stratifying these patients.
Collapse
Affiliation(s)
- Paulina Cegla
- Department of Endocrine Oncology and Nuclear Medicine, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (M.W.); (W.C.); (G.Ł.); (P.O.); (A.S.); (M.D.)
- Correspondence:
| | - Marta Wojewódzka
- Department of Endocrine Oncology and Nuclear Medicine, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (M.W.); (W.C.); (G.Ł.); (P.O.); (A.S.); (M.D.)
| | - Izabela Gorczewska
- Department of Nuclear Medicine and Endocrine Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 44-102 Gliwice, Poland;
| | - Wioletta Chalewska
- Department of Endocrine Oncology and Nuclear Medicine, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (M.W.); (W.C.); (G.Ł.); (P.O.); (A.S.); (M.D.)
| | - Grażyna Łapińska
- Department of Endocrine Oncology and Nuclear Medicine, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (M.W.); (W.C.); (G.Ł.); (P.O.); (A.S.); (M.D.)
| | - Paweł Ochman
- Department of Endocrine Oncology and Nuclear Medicine, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (M.W.); (W.C.); (G.Ł.); (P.O.); (A.S.); (M.D.)
| | - Agata Sackiewicz
- Department of Endocrine Oncology and Nuclear Medicine, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (M.W.); (W.C.); (G.Ł.); (P.O.); (A.S.); (M.D.)
| | - Marek Dedecjus
- Department of Endocrine Oncology and Nuclear Medicine, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (M.W.); (W.C.); (G.Ł.); (P.O.); (A.S.); (M.D.)
| |
Collapse
|
17
|
Changing Threshold-Based Segmentation Has No Relevant Impact on Semi-Quantification in the Context of Structured Reporting for PSMA-PET/CT. Cancers (Basel) 2022; 14:cancers14020270. [PMID: 35053434 PMCID: PMC8773894 DOI: 10.3390/cancers14020270] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/02/2022] [Accepted: 01/03/2022] [Indexed: 02/05/2023] Open
Abstract
Simple Summary Molecular imaging of patients with prostate cancer is widely utilized. We aimed to determine whether changes in post-processing parameters, such as maximum intensity thresholds, can significantly alter results. We investigated 623 lesions that were positive on a molecular imaging scan and could not find any relevant impact on results when certain parameters were changed, in particular in lesions indicative for metastases of prostate cancer. Abstract Prostate-specific membrane antigen (PSMA)-directed positron emission tomography/computed tomography (PET/CT) is increasingly utilized for staging of men with prostate cancer (PC). To increase interpretive certainty, the standardized PSMA reporting and data system (RADS) has been proposed. Using PSMA-RADS, we characterized lesions in 18 patients imaged with 18F-PSMA-1007 PET/CT for primary staging and determined the stability of semi-quantitative parameters. Six hundred twenty-three lesions were categorized according to PSMA-RADS and manually segmented. In this context, PSMA-RADS-3A (soft-tissue) or -3B (bone) lesions are defined as being indeterminate for the presence of PC. For PMSA-RADS-4 and -5 lesions; however, PC is highly likely or almost certainly present [with further distinction based on absence (PSMA-RADS-4) or presence (PSMA-RADS-5) of correlative findings on CT]. Standardized uptake values (SUVmax, SUVpeak, SUVmean) were recorded, and volumetric parameters [PSMA-derived tumor volume (PSMA-TV); total lesion PSMA (TL-PSMA)] were determined using different maximum intensity thresholds (MIT) (40 vs. 45 vs. 50%). SUVmax was significantly higher in PSMA-RADS-5 lesions compared to all other PSMA-RADS categories (p ≤ 0.0322). In particular, the clinically challenging PSMA-RADS-3A lesions showed significantly lower SUVmax and SUVpeak compared to the entire PSMA-RADS-4 or -5 cohort (p < 0.0001), while for PSMA-RADS-3B this only applies when compared to the entire PSMA-RADS-5 cohort (p < 0.0001), but not to the PSMA-RADS-4 cohort (SUVmax, p = 0.07; SUVpeak, p = 0.08). SUVmean (p = 0.30) and TL-PSMA (p = 0.16) in PSMA-RADS-5 lesions were not influenced by changing the MIT, while PSMA-TV showed significant differences when comparing 40 vs. 50% MIT (p = 0.0066), which was driven by lymph nodes (p = 0.0239), but not bone lesions (p = 0.15). SUVmax was significantly higher in PSMA-RADS-5 lesions compared to all other PSMA-RADS categories in 18F-PSMA-1007 PET/CT. As such, the latter parameter may assist the interpreting molecular imaging specialist in assigning the correct PSMA-RADS score to sites of disease, thereby increasing diagnostic certainty. In addition, changes of the MIT in PSMA-RADS-5 lesions had no significant impact on SUVmean and TL-PSMA in contrast to PSMA-TV.
Collapse
|
18
|
Jiang J, Tang X, Pu Y, Yang Y, Yang C, Yang F, Tian Y, Li J, Sun H, Zhao S, Chen L. The Value of Multimodality PET/CT Imaging in Detecting Prostate Cancer Biochemical Recurrence. Front Endocrinol (Lausanne) 2022; 13:897513. [PMID: 35712249 PMCID: PMC9197252 DOI: 10.3389/fendo.2022.897513] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 04/26/2022] [Indexed: 11/13/2022] Open
Abstract
Prostate cancer (PCa) induced death is the predominant cause of cancer-related death among men in 48 countries. After radical treatment, biochemical recurrence has become an important factor for prognosis. The early detection and diagnosis of recurrent lesions are very helpful in guiding treatment and improving the prognosis. PET/CT is a promising method for early detection of lesions in patients with biochemical recurrence of prostate cancer. This article reviews the progress of the research on PET/CT in the PCa biochemical recurrence and aims to introduce new technologies and provide more direction for future research.
Collapse
Affiliation(s)
- Jie Jiang
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Yunnan, China
| | - Xiaoxia Tang
- Department of Pharmacy, The Second Affiliated Hospital of Kunming Medical University, Yunnan, China
| | - Yongzhu Pu
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Yunnan, China
| | - Yong Yang
- Department of Urology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Yunnan, China
| | - Conghui Yang
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Yunnan, China
| | - Fake Yang
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Yunnan, China
| | - Yadong Tian
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Yunnan, China
| | - Jindan Li
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Yunnan, China
| | - Hua Sun
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Yunnan, China
- *Correspondence: Long Chen, ; Hua Sun, ; Sheng Zhao,
| | - Sheng Zhao
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Yunnan, China
- *Correspondence: Long Chen, ; Hua Sun, ; Sheng Zhao,
| | - Long Chen
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Yunnan, China
- *Correspondence: Long Chen, ; Hua Sun, ; Sheng Zhao,
| |
Collapse
|
19
|
Hong H, Wang G, Ploessl K, Zha Z, Zang J, Zhu Z, Zhu L, Kung HF. Kit-based preparation of [ 68Ga]Ga-P16-093 (PSMA-093) using different commercial 68Ge/ 68Ga generators. Nucl Med Biol 2021; 106-107:1-9. [PMID: 34952347 DOI: 10.1016/j.nucmedbio.2021.12.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/29/2021] [Accepted: 12/02/2021] [Indexed: 12/31/2022]
Abstract
PURPOSE Prostate-specific membrane antigen (PSMA) is an important biomarker for molecular imaging and a target for radionuclide therapy of prostate cancer. Recently, U.S. Food and Drug Administration (FDA) has approved [68Ga]Ga-PSMA-11 as a PSMA-targeted positron emission tomography (PET) imaging agent for the diagnosis of prostate cancer. As an alternative PSMA imaging agent, [68Ga]Ga-P16-093 ([68Ga]Ga-PSMA-093) showed excellent blood clearance and rapid tumor uptake, desirable in vivo properties for avidly detecting primary tumor and metastatic lesions in patients. To improve the availability and test the robustness of radiolabeling reaction, eluents of 68Ga/HCl from different sources of generators were evaluated. PROCEDURES Commercially available 68Ge/68Ga generators from Eckert & Ziegler, ITG and iThemba were eluted with varying molarities of hydrochloric acid (0.05-0.6 M, as recommended by each company) and reacted with P16-093 kits. Radiolabeling yields, in vitro stabilities, in vitro cell uptakes and drug release criteria of different preparations were investigated. PET/computed tomography (CT) imaging of prostate cancer patients with [68Ga]Ga-P16-093 produced by using different sources of 68Ga were performed. RESULTS Optimized P16-093 kit containing 15 μg of P16-093 (precursor) and 68 mg of sodium acetate trihydrate (buffer), a formulation previously tested in humans, was successfully labeled with eluents from Eckert & Ziegler, ITG and iThemba's generators. In vitro cell uptake studies showed that [68Ga]Ga-P16-093, formulated with ITG and iThemba's generators, exhibited equivalent PSMA-specific uptakes. Clinical studies in prostate cancer patients exhibited exceedingly comparable maximum standardized uptake value (SUVmax) for each lesion regardless of source of the generator used in preparation. CONCLUSION Using different vendors' generator and lyophilized P16-093 kits, [68Ga]Ga-P16-093 could be conveniently and reliably prepared by a simple one-step reaction with excellent yields. Clinically useful doses of [68Ga]Ga-P16-093 imaging tracer could be made available using different 68Ge/68Ga generators.
Collapse
Affiliation(s)
- Haiyan Hong
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Guochang Wang
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Karl Ploessl
- Five Eleven Pharma Inc., Philadelphia, PA 19104, USA
| | - Zhihao Zha
- Five Eleven Pharma Inc., Philadelphia, PA 19104, USA
| | - Jie Zang
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Zhaohui Zhu
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China
| | - Lin Zhu
- Key Laboratory of Radiopharmaceuticals, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Hank F Kung
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA; Five Eleven Pharma Inc., Philadelphia, PA 19104, USA.
| |
Collapse
|
20
|
Okudan B, Coşkun N, Seven B, Atalay MA, Yildirim A, Görtan FA. Assessment of volumetric parameters derived from 68Ga-PSMA PET/CT in prostate cancer patients with biochemical recurrence: an institutional experience. Nucl Med Commun 2021; 42:1254-1260. [PMID: 34284438 DOI: 10.1097/mnm.0000000000001459] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
OBJECTIVE The aim of this study was to evaluate the relationship between volumetric parameters calculated using semiautomatic quantification of lesions detected in 68Ga-labeled prostate-specific membrane antigen PET-computed tomography (68Ga-PSMA PET/CT) and clinical characteristics in prostate cancer (PCa) patients with biochemical recurrence. METHODS A total of 85 consecutive PCa patients with biochemical recurrence who underwent 68Ga-PSMA PET/CT at our institution from January 2019 to March 2020 were retrospectively assessed. 68Ga-PSMA PET/CT-derived volumetric parameters, including whole-body PSMA tumor volume (wbPSMA-TV) and whole-body total lesion PSMA (wbTL-PSMA), as well as the established maximum and mean standardized uptake value (SUVmax and SUVmean), were calculated for each patient. All PET-derived parameters were analyzed for correlation with prostate-specific antigen (PSA) levels and for association with Gleason scores. RESULTS Eighty-five patients with a mean age of 68.9 ± 7.8 years (range, 47-83 years) and a mean PSA level of 40.9 ± 92.1 ng/ml (range, 0.2-533.2 ng/ml) were analyzed. Volumetric parameters, that is, wbPSMA-TV and wbTL-PSMA, demonstrated a statistically significant correlation with PSA levels (r = 0.403 and r = 0.556, respectively, all at P < 0.001) and only the means of wbTL-PSMA were significantly different between the Gleason score groups (P < 0.05). CONCLUSIONS The results of our study indicate that 68Ga-PSMA PET/CT might be a valuable tool for the detection and follow-up of recurrence in PCa patients. 68Ga-PSMA PET/CT-derived quantitative volumetric parameters demonstrated a highly significant correlation with changes in PSA levels. Larger prospective studies are needed to help reveal the full potential of parameters such as PSMA-TV and TL-PSMA derived from PET imaging with 68Ga-PSMA.
Collapse
Affiliation(s)
- Berna Okudan
- Department of Nuclear Medicine, Ankara City Hospital, University of Health Sciences
| | - Nazim Coşkun
- Department of Nuclear Medicine, Ankara City Hospital, Ankara
| | - Bedri Seven
- Department of Nuclear Medicine, Sabuncuoğlu Şerefeddin Training and Research Hospital, University of Amasya, Amasya, Turkey
| | | | | | | |
Collapse
|
21
|
Winiger A, Pérez Lago MDS, Lehnick D, Roos JE, Strobel K. The value of intravenous contrast medium in PSMA PET/CT imaging in patients with biochemical recurrence of prostate cancer. Nucl Med Commun 2021; 42:1239-1246. [PMID: 34132237 DOI: 10.1097/mnm.0000000000001453] [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: 12/24/2022]
Abstract
PURPOSE To investigate the added value of diagnostic abdominal contrast-enhanced computed tomography (ceCT) in Ga-68 PSMA PET/CT in prostate cancer patients with biochemical recurrence (BCR). METHODS Eighty-two consecutive patients (median age, 69 years; range, 45-86 years) with BCR underwent Ga-68 PSMA PET/CT with low-dose nonenhanced (ne) whole-body CT and diagnostic ceCT. Imaging results were retrospectively reviewed by two readers (R1, R2) for diagnostic certainty, local recurrence, lymph node metastasis (LNM) and bone metastasis. Interobserver agreement was assessed. Histopathology served as reference standard in 7, imaging and clinical follow-up in 65 and clinical follow-up alone in 10 patients. RESULTS Certain local recurrence, LNM and bone metastasis diagnoses increased substantially from ceCT (R1: 5%/18%/32%; R2: 37%/50%/82%) to nePET/CT (R1: 78%/87%/93%; R2: 81%/87%/95%) for both readers, but the difference between nePET/CT and cePET/CT (R1: 77%/96%/92%; R2: 89%/94%/96%) was marginal. Interobserver agreement was minimal with ceCT (Krippendorff's alpha: 0.04-0.26), substantial with nePET/CT (0.60-0.86) and best with cePET/CT (0.76-0.86). The areas under the receiver operating characteristic curve (AUCs) for local recurrence for R1/R2 were 0.60/0.65 for ceCT, 0.81/0.79 for nePET/CT and 0.81/0.82 for cePET/CT. AUCs for LNMs for R1/R2 were 0.67/0.77 for ceCT, 0.91/0.82 for nePET/CT and 0.92/0.87 for cePET/CT. AUCs for BMs for R1/R2 were 0.60/0.53 for ceCT, 0.93/0.84 for nePET/CT and 0.93/0.86 for cePET/CT. CONCLUSION Diagnostic abdominal ceCT increases the diagnostic certainty and interobserver agreement in Ga-68 PSMA PET/CT in BCR of prostate cancer. The diagnostic performance of cePET/CT is significantly better than ceCT alone but not nePET/CT.
Collapse
Affiliation(s)
- Alain Winiger
- Department of Radiology and Nuclear Medicine, Lucerne Cantonal Hospital, Spitalstrasse
| | | | - Dirk Lehnick
- Department of Health Sciences and Medicine, University of Lucerne, Frohburgstrasse 3, Lucerne, Switzerland
| | - Justus E Roos
- Department of Radiology and Nuclear Medicine, Lucerne Cantonal Hospital, Spitalstrasse
| | - Klaus Strobel
- Department of Radiology and Nuclear Medicine, Lucerne Cantonal Hospital, Spitalstrasse
| |
Collapse
|
22
|
Martinez J, Subramanian K, Margolis D, O'Dwyer E, Osborne J, Jhanwar Y, Nagar H, Williams N, RoyChoudhury A, Madera G, Babich J, Huicochea Castellanos S. 68Ga-PSMA-HBED-CC PET/MRI is superior to multiparametric magnetic resonance imaging in men with biochemical recurrent prostate cancer: A prospective single-institutional study. Transl Oncol 2021; 15:101242. [PMID: 34649151 PMCID: PMC8517922 DOI: 10.1016/j.tranon.2021.101242] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/20/2021] [Accepted: 10/04/2021] [Indexed: 02/07/2023] Open
Abstract
PSMA PET/MRI has a higher true positive rate and sensitivity than mpMRI in patients with biochemically recurrent prostate cancer. The true positive rate for PSMA PET/MRI was significantly greater in patients who were post prostatectomy for primary treatment. 40% of patients with a PSA value less than 0.2 ng/mL had a positive PSMA PET/MRI read compared to 9% on MRI.
Background The primary objective was to compare the overall diagnostic performance, presented as detection rate of 68Ga-PSMA-HBED-CC positron emission tomography/magnetic resonance imaging (PSMA PET/MRI) versus conventional, multiparametric MRI (mpMRI) in a population of patients with biochemically recurrent prostate cancer. In conjunction with this analysis, secondary objectives included the evaluation of the detection rate stratified by PSA levels and primary treatment modality. Methods A total of 165 PSMA PET MRI were performed from April 2018 to May 2021, of whom 108 were presenting for biochemical recurrent disease. The PSMA PET vertex to thigh were read by two different board-certified nuclear medicine physicians while the MRI head and neck, chest, abdomen, and pelvis (with dedicated, PI-RADS compliant multiparametric prostate MRI) were read by two board certified diagnostic radiologists. Analysis PSMA PET/MRI had a higher detection rate than mpMRI when evaluating patients with biochemical recurrence (BCR) with similar results demonstrated when sub-analysis was performed using PSA levels, primary treatment modality, and time since androgen deprivation therapy. Our study also showed PSMA PET/MRI had a higher sensitivity than mpMRI. Discussion Our findings demonstrate that PSMA PET/MRI is a better imaging modality in the detection of disease in the setting of BCR when compared to MRI alone. Combined utility with PSMA PET/MRI is a powerful tool which can aid in not only the detection of disease, but also guide in treatment planning for prostate cancer patients.
Collapse
Affiliation(s)
- Juana Martinez
- Division of Molecular Imaging and Therapeutics, Department of Radiology, Weill Cornell Medicine, 525 E 68th Street, New York, NY 10065, USA
| | - Kritika Subramanian
- Division of Molecular Imaging and Therapeutics, Department of Radiology, Weill Cornell Medicine, 525 E 68th Street, New York, NY 10065, USA.
| | - Daniel Margolis
- Division of Body Imaging, Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Elisabeth O'Dwyer
- Division of Molecular Imaging and Therapeutics, Department of Radiology, Weill Cornell Medicine, 525 E 68th Street, New York, NY 10065, USA; Division of Body Imaging, Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Joseph Osborne
- Division of Molecular Imaging and Therapeutics, Department of Radiology, Weill Cornell Medicine, 525 E 68th Street, New York, NY 10065, USA
| | - Yuliya Jhanwar
- Department of Radiology, CareMount Medical, Mount Kisco, NY, USA
| | - Himanshu Nagar
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Nicholas Williams
- Division of Biostatistics, Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Arindam RoyChoudhury
- Division of Biostatistics, Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Gabriela Madera
- Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - John Babich
- Division of Radiopharmaceutical Sciences, Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Sandra Huicochea Castellanos
- Division of Molecular Imaging and Therapeutics, Department of Radiology, Weill Cornell Medicine, 525 E 68th Street, New York, NY 10065, USA
| |
Collapse
|
23
|
Mirzaei S, Lipp R, Zandieh S, Leisser A. Single-Center Comparison of [ 64Cu]-DOTAGA-PSMA and [ 18F]-PSMA PET-CT for Imaging Prostate Cancer. Curr Oncol 2021; 28:4167-4173. [PMID: 34677271 PMCID: PMC8534892 DOI: 10.3390/curroncol28050353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/04/2021] [Accepted: 10/13/2021] [Indexed: 11/16/2022] Open
Abstract
Introduction: the diagnostic performance of [64Cu]-DOTAGA-PSMA PET–CT imaging was compared retrospectively to [18F]-PSMA PET–CT in prostate cancer patients with recurrent disease and in the primary staging of selected patients with advanced local and possible metastatic disease. Methods: We retrospectively selected a total of 100 patients, who were consecutively examined in our department, with biochemical recurrence after radical prostatectomy or who had progressive local and possible metastatic disease in the last 3 months prior to this investigation. All patients were examined with a dedicated PET–CT scanner (Biograph; Siemens Healthineers). A total of 250 MBq (3.5 MBq per kg bodyweight, range 230–290 MBq) of [64Cu]-DOTAGA-PSMA or [18-F]-PSMA was applied intravenously. PET images were performed 1 h post-injection (skull base to mid-thigh). The maximum standardized uptake values (SUVmax) of PSMA-positive lesions and the mean standardized uptake value (SUVmean) of the right liver lobe were measured. Results: All but 9/50 of the patients (18%; PSA range: 0.01–0.7 µg/L) studied with [64Cu]-DOTAGA-PSMA and 6/50 of the ones (12%; PSA range: 0.01–4.2) studied with [18F]-PSMA had at least one positive PSMA lesion shown by PET–CT. The total number of lesions was higher with [64Cu]-DOTAGA-PSMA (209 vs. 191); however, the median number of lesions was one for [64Cu]-DOTAGA-PSMA and two for [18F]-PSMA. Interestingly, the median SUVmean of the right liver lobe was slightly higher for [18F]-PSMA (11.8 vs. 8.9). Conclusions: [64Cu]-DOTAGA-PSMA and [18F]-PSMA have comparable detection rates for the assessment of residual disease in patients with recurrent or primary progressive prostate cancer. The uptake in the liver is moderately different, and therefore at least the SUVs of the lesions in both studies would not be comparable.
Collapse
Affiliation(s)
- Siroos Mirzaei
- Department of Nuclear Medicine with PET-Center, Clinic Ottakring (Wilhelminenspital), 1160 Vienna, Austria;
- Correspondence:
| | - Rainer Lipp
- Department of Internal Medicine, Medical University of Graz, 8036 Graz, Austria;
| | - Shahin Zandieh
- Department of Radiology and Nuclear Medicine, Hanusch Hospital, 1160 Vienna, Austria;
| | - Asha Leisser
- Department of Nuclear Medicine with PET-Center, Clinic Ottakring (Wilhelminenspital), 1160 Vienna, Austria;
| |
Collapse
|
24
|
Kroenke M, Mirzoyan L, Horn T, Peeken JC, Wurzer A, Wester HJ, Makowski M, Weber WA, Eiber M, Rauscher I. Matched-Pair Comparison of 68Ga-PSMA-11 and 18F-rhPSMA-7 PET/CT in Patients with Primary and Biochemical Recurrence of Prostate Cancer: Frequency of Non-Tumor-Related Uptake and Tumor Positivity. J Nucl Med 2021; 62:1082-1088. [PMID: 33277394 DOI: 10.2967/jnumed.120.251447] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 11/12/2020] [Indexed: 11/16/2022] Open
Abstract
Radiohybrid prostate-specific membrane antigen (rhPSMA) ligands are a new class of prostate cancer theranostic agents. 18F-rhPSMA-7 offers the advantages of 18F labeling and low urinary excretion compared with 68Ga-PSMA-11. Here, we compare the frequency of non-tumor-related uptake and tumor positivity with 68Ga-PSMA-11 and 18F-rhPSMA-7 in patients with primary or recurrent prostate cancer. Methods: This retrospective matched-pair comparison matched 160 18F-rhPSMA-7 with 160 68Ga-PSMA-11 PET/CT studies for primary staging (n = 33) and biochemical recurrence (n = 127) according to clinical characteristics. Two nuclear medicine physicians reviewed all scans, first identifying all PET-positive lesions and then differentiating lesions suggestive of prostate cancer from those that were benign, on the basis of known pitfalls and ancillary information from CT. For each region, the SUVmax of the lesion with the highest PSMA ligand uptake was noted. Tumor positivity rates were determined, and SUVmax was compared separately for each tracer. Results:18F-rhPSMA-7 and 68Ga-PSMA-11 PET revealed 566 and 289 PSMA ligand-positive lesions, respectively. Of these, 379 and 100 lesions, equaling 67.0% and 34.6%, respectively, of all PSMA-positive lesions, were considered benign. The distribution of their etiology was similar (42%, 24%, and 25% with 18F-rhPSMA-7 vs. 32%, 24%, and 38% with 68Ga-PSMA-11 for ganglia, bone, and unspecific lymph nodes, respectively). All primary tumors were positive with both agents (n = 33 each), whereas slightly more metastatic lesions were observed with 68Ga-PSMA-11 in both disease stages (113 for 18F-rhPSMA-7 and 124 for 68Ga-PSMA-11). The SUVmax of 18F-rhPSMA-7 and 68Ga-PSMA-11 did not differ (P > 0.05) in local recurrence or primary prostate cancer; however, the tumor-to-bladder ratio was significantly higher with 18F-rhPSMA-7 (4.9 ± 5.3 vs. 2.2 ± 3.7, P = 0.02, for local recurrence; 9.8 ± 9.7 vs. 2.3 ± 2.6, P < 0.001, for primary prostate cancer). Conclusion: The tumor positivity rate was consistently high for 68Ga-PSMA-11 and 18F-rhPSMA-7. Both tracers revealed a considerable number of areas of uptake that were reliably identified as benign by trained physicians making use of corresponding morphologic imaging and known PSMA pitfalls. These were more frequent with 18F-rhPSMA-7. However, the matched-pair comparison could have introduced a source of bias. Adequate reader training can allow physicians to differentiate benign uptake from disease and be able to benefit from the logistical and clinical advantages of 18F-rhPSMA-7.
Collapse
Affiliation(s)
- Markus Kroenke
- Institute of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, Munich, Germany
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
| | - Lilit Mirzoyan
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
| | - Thomas Horn
- Department of Urology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Jan C Peeken
- Department of Radiation Oncology, School of Medicine, Technical University of Munich, Munich, Germany
- Institute of Radiation Medicine, Department of Radiation Sciences, Helmholtz Zentrum München, Neuherberg, Germany; and
| | - Alexander Wurzer
- Pharmaceutical Radiochemistry, School of Medicine, Technical University of Munich. Munich, Germany
| | - Hans-Jürgen Wester
- Pharmaceutical Radiochemistry, School of Medicine, Technical University of Munich. Munich, Germany
| | - Marcus Makowski
- Institute of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Wolfgang A Weber
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
| | - Matthias Eiber
- 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;
| |
Collapse
|
25
|
Capobianco N, Sibille L, Chantadisai M, Gafita A, Langbein T, Platsch G, Solari EL, Shah V, Spottiswoode B, Eiber M, Weber WA, Navab N, Nekolla SG. Whole-body uptake classification and prostate cancer staging in 68Ga-PSMA-11 PET/CT using dual-tracer learning. Eur J Nucl Med Mol Imaging 2021; 49:517-526. [PMID: 34232350 PMCID: PMC8803695 DOI: 10.1007/s00259-021-05473-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 06/17/2021] [Indexed: 01/16/2023]
Abstract
Purpose In PSMA-ligand PET/CT imaging, standardized evaluation frameworks and image-derived parameters are increasingly used to support prostate cancer staging. Clinical applicability remains challenging wherever manual measurements of numerous suspected lesions are required. Deep learning methods are promising for automated image analysis, typically requiring extensive expert-annotated image datasets to reach sufficient accuracy. We developed a deep learning method to support image-based staging, investigating the use of training information from two radiotracers. Methods In 173 subjects imaged with 68Ga-PSMA-11 PET/CT, divided into development (121) and test (52) sets, we trained and evaluated a convolutional neural network to both classify sites of elevated tracer uptake as nonsuspicious or suspicious for cancer and assign them an anatomical location. We evaluated training strategies to leverage information from a larger dataset of 18F-FDG PET/CT images and expert annotations, including transfer learning and combined training encoding the tracer type as input to the network. We assessed the agreement between the N and M stage assigned based on the network annotations and expert annotations, according to the PROMISE miTNM framework. Results In the development set, including 18F-FDG training data improved classification performance in four-fold cross validation. In the test set, compared to expert assessment, training with 18F-FDG data and the development set yielded 80.4% average precision [confidence interval (CI): 71.1–87.8] for identification of suspicious uptake sites, 77% (CI: 70.0–83.4) accuracy for anatomical location classification of suspicious findings, 81% agreement for identification of regional lymph node involvement, and 77% agreement for identification of metastatic stage. Conclusion The evaluated algorithm showed good agreement with expert assessment for identification and anatomical location classification of suspicious uptake sites in whole-body 68Ga-PSMA-11 PET/CT. With restricted PSMA-ligand data available, the use of training examples from a different radiotracer improved performance. The investigated methods are promising for enabling efficient assessment of cancer stage and tumor burden. Supplementary Information The online version contains supplementary material available at 10.1007/s00259-021-05473-2.
Collapse
Affiliation(s)
- Nicolò Capobianco
- Technische Universität München, Munich, Germany. .,Siemens Healthcare GmbH, Erlangen, Germany.
| | | | - Maythinee Chantadisai
- School of Medicine, Department of Nuclear Medicine, Technische Universität München, Munich, Germany.,Faculty of Medicine, King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Chulalongkorn University, Bangkok, Thailand
| | - Andrei Gafita
- School of Medicine, Department of Nuclear Medicine, Technische Universität München, Munich, Germany
| | - Thomas Langbein
- School of Medicine, Department of Nuclear Medicine, Technische Universität München, Munich, Germany
| | | | - Esteban Lucas Solari
- School of Medicine, Department of Nuclear Medicine, Technische Universität München, Munich, Germany
| | - Vijay Shah
- Siemens Medical Solutions USA, Inc., Knoxville, TN, USA
| | | | - Matthias Eiber
- School of Medicine, Department of Nuclear Medicine, Technische Universität München, Munich, Germany
| | - Wolfgang A Weber
- School of Medicine, Department of Nuclear Medicine, Technische Universität München, Munich, Germany
| | - Nassir Navab
- Computer Aided Medical Procedures (CAMP), Technische Universität München, Munich, Germany
| | - Stephan G Nekolla
- School of Medicine, Department of Nuclear Medicine, Technische Universität München, Munich, Germany
| |
Collapse
|
26
|
Hopland OA, Fosså SD, Ottosson F, Brennhovd B, Svindland A, Hole KH, Hernes E, Eri LM, Diep LM, Berge V. Robotic salvage pelvic lymph node dissection for locoregional recurrence after radical prostatectomy: a single institution experience. Scand J Urol 2021; 55:287-292. [PMID: 34196594 DOI: 10.1080/21681805.2021.1946135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVES To assess treatment response (PSA < 0.2 ng/ml), need for additional therapy and complication rate after robot assisted salvage pelvic lymph node dissection (sPLND). MATERIAL AND METHODS Analysis of outcomes data from radical prostatectomy (RP) patients consecutively operated with robot assisted sPLND due to biochemical recurrence and positron-emission tomography (PET)/computed tomography (CT)-detected nodal recurrence of pelvic lymph nodes. RESULTS Sixty-nine patients underwent robotic sPLND after a median time of 47 months post- RP. Sixty-four patients (93%) had malignant lymph nodes upon histological assessment of sPLND specimen. Twenty patients (29%) achieved PSA < 0.2 ng/ml 6 weeks postoperatively. After median (IQR) follow-up of 15 months (10-27), fourteen patients (20%) still had PSA < 0.2 ng/ml without additional therapy and forty-one patients (59%) had started additional therapy. No significant predictor for treatment response was found. Postoperative complications occurred in 14 patients (20%). Eleven of these complications were classified as Clavien-Dindo grade 1. CONCLUSION Oncological benefit of sPLND as the only salvage procedure seems to be limited, though almost one third of patients achieved treatment response. Clinical trials are needed to determine if sPLND as part of a multimodal treatment may improve outcome.
Collapse
Affiliation(s)
| | - Sophie D Fosså
- National Advisory Unit on Late Effects after Cancer Treatment, Radiumhospitalet, Oslo University, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Fredrik Ottosson
- Department of Urology, Radiumhospital, Oslo University Hospital, Oslo, Norway
| | - Bjørn Brennhovd
- Department of Urology, Radiumhospital, Oslo University Hospital, Oslo, Norway
| | - Aud Svindland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Department of Pathology, Radiumhospitalet, Oslo University Hospital, Oslo, Norway
| | - Knut Håkon Hole
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.,Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Eivor Hernes
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Lars Magne Eri
- Department of Urology, Radiumhospital, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Lien My Diep
- Oslo Centre for Biostatistics and Epidemiology (OCBE), Oslo University Hospital, Oslo, Norway
| | - Viktor Berge
- Department of Urology, Radiumhospital, Oslo University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
27
|
Has Simsek D, Kuyumcu S, Karadogan S, Oflas M, Isik EG, Ozkan ZG, Paksoy N, Ekmekcioglu O, Ekenel M, Sanli Y. Can PSMA-based tumor burden predict response to docetaxel treatment in metastatic castration-resistant prostate cancer? Ann Nucl Med 2021; 35:680-690. [PMID: 33783752 DOI: 10.1007/s12149-021-01610-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/16/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE We investigated the role of PSMA-derived tumor burden in predicting docetaxel (DTX) therapy response in metastatic castration-resistant prostate cancer (mCRPC). METHODS Fifty-two mCRPC patients who received at least six cycles of DTX as the first-line treatment following 68Ga-PSMA PET/CT were enrolled in this retrospective study. Total PSMA-derived tumor volume (TV-PSMA) and total lesion PSMA activity (TL-PSMA) were derived from metastatic lesions. A ≥ 50% decline in PSA was defined as a response; a ≥ 25% increase in PSA was defined as progression. Univariate/multivariate logistic and cox regression analyses were performed to predict PSA response, OS, and TTP. RESULTS Twelve (23%) patients had PSA progression after chemotherapy, while 40 patients (77%) achieved a PSA response. On univariate analysis, a significant association was found between TV-PSMA (p = 0.001), TL-PSMA (p = 0.001), pre-PSA (p = 0.012), LDH (p = 0.003), Hg (p = 0.035), and PSA response to DTX. High TV-PSMA (> 107 cm3) (p = 0.04) and high LDH (> 234 U/L) (p = 0.017) were 8.2 times and 12.2 times more likely for DTX failure in multivariate regression analyses. The median TTP was 16 months, and the median OS was not reached. Patients with high TV-PSMA (p = 0.017), high TL-PSMA (> 1013 cm3) (p = 0.042), high age (> 70 years) (p = 0.016), and high LDH (p ≤ 0.001) had significantly shorter OS, while only high TV-PSMA (p = 0.038) and high age (p = 0.006) were significantly related with shorter TTP. High TV-PSMA (p = 0.017) and high age (p = 0.01) were significant predictors for shorter OS, while only high age (p = 0.006) was a significant predictor for shorter TTP in multivariate analysis. CONCLUSION Patients with high TV-PSMA had a significantly higher risk for DTX failure. PSMA-based tumor burden prior to DTX therapy seems to be a reliable predictive tool for survival in mCRPC patients.
Collapse
Affiliation(s)
- Duygu Has Simsek
- Department of Nuclear Medicine, Istanbul Faculty of Medicine, Istanbul University, Fatih, 34093, Istanbul, Turkey.
| | - Serkan Kuyumcu
- Department of Nuclear Medicine, Istanbul Faculty of Medicine, Istanbul University, Fatih, 34093, Istanbul, Turkey
| | - Seyfullah Karadogan
- Department of Nuclear Medicine, Istanbul Faculty of Medicine, Istanbul University, Fatih, 34093, Istanbul, Turkey
| | - Melis Oflas
- Department of Nuclear Medicine, Istanbul Faculty of Medicine, Istanbul University, Fatih, 34093, Istanbul, Turkey
| | - Emine Goknur Isik
- Department of Nuclear Medicine, Istanbul Faculty of Medicine, Istanbul University, Fatih, 34093, Istanbul, Turkey
| | - Zeynep Gozde Ozkan
- Department of Nuclear Medicine, Istanbul Faculty of Medicine, Istanbul University, Fatih, 34093, Istanbul, Turkey
| | - Nail Paksoy
- Department of Medical Oncology, Institute of Oncology, Istanbul University, Istanbul, Turkey
| | - Ozgul Ekmekcioglu
- Department of Nuclear Medicine, Istanbul Sisli Hamidiye Etfal Training and Research Hospital, Istanbul, Turkey
| | - Meltem Ekenel
- Department of Medical Oncology, Institute of Oncology, Istanbul University, Istanbul, Turkey
| | - Yasemin Sanli
- Department of Nuclear Medicine, Istanbul Faculty of Medicine, Istanbul University, Fatih, 34093, Istanbul, Turkey
| |
Collapse
|
28
|
Ahmed ME, Phillips RM, Sharma V, Davis BJ, Karnes RJ. Oligometastatic prostatic cancer recurrence: role of salvage lymph node dissection (sLND) and radiation therapy-stereotactic body radiation therapy (RT-SBRT). Curr Opin Urol 2021; 31:199-205. [PMID: 33742974 DOI: 10.1097/mou.0000000000000865] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Metastases directed therapy (MDT) is an increasingly utilized modality in patients with oligometastatic prostate cancer (OMPC) recurrence. The purpose of our review is to discuss the recent literature on the safety and oncologic outcomes of this treatment approach. RECENT FINDINGS Metastases directed therapy, in particular, stereotactic body radiation therapy (SBRT) and salvage lymph node dissection (sLND), has shown promising efficacy in patients with OMPC. Many case series report favorable outcomes with MDT as compared to hormonal deprivation therapy alone or surveillance. Of the few case series investigating the use of MDT as part of a multimodality approach in castrate-resistant OMPC, more favorable outcomes in comparison to the use of systemic treatment alone are reported. SUMMARY With the recent advances in imaging techniques, particularly molecular imaging, management of OMPC has progressed rapidly in the last few years. The feasibility and benefits of MDT in OMPC have been demonstrated in prospective and retrospective series. Further prospective studies investigating the role of MDT to define optimal patient subgroups and management strategies are warranted.
Collapse
Affiliation(s)
| | - Ryan M Phillips
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Brian J Davis
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | | |
Collapse
|
29
|
Calais J, Zhu S, Hirmas N, Eiber M, Hadaschik B, Stuschke M, Herrmann K, Czernin J, Kishan AU, Nickols NG, Elashoff D, Fendler WP. Phase 3 multicenter randomized trial of PSMA PET/CT prior to definitive radiation therapy for unfavorable intermediate-risk or high-risk prostate cancer [PSMA dRT]: study protocol. BMC Cancer 2021; 21:512. [PMID: 33962579 PMCID: PMC8103642 DOI: 10.1186/s12885-021-08026-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 03/12/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Definitive radiation therapy (dRT) is an effective initial treatment of intermediate-risk (IR) and high-risk (HR) prostate cancer (PCa). PSMA PET/CT is superior to standard of care imaging (CT, MRI, bone scan) for detecting regional and distant metastatic PCa. PSMA PET/CT thus has the potential to guide patient selection and the planning for dRT and improve patient outcomes. METHODS This is a multicenter randomized phase 3 trial (NCT04457245). We will randomize 312 patients to proceed with standard dRT (control Arm, n = 150), or undergo a PSMA PET/CT scan at the study site (both 18F-DCFPyL and 68Ga-PSMA-11 can be used) prior to dRT planning (intervention arm, n = 162). dRT will be performed at the treating radiation oncologist facility. In the control arm, dRT will be performed as routinely planned. In the intervention arm, the treating radiation oncologist can incorporate PSMA PET/CT findings into the RT planning. Androgen deprivation therapy (ADT) is administered per discretion of the treating radiation oncologist and may be modified as a result of the PSMA PET/CT results. We assume that approximately 8% of subjects randomized to the PSMA PET arm will be found to have M1 disease and thus will be more appropriate candidates for long-term systemic or multimodal therapy, rather than curative intent dRT. PET M1 patients will thus not be included in the primary endpoint analysis. The primary endpoint is the success rate of patients with unfavorable IR and HR PCa after standard dRT versus PSMA PET-based dRT. Secondary Endpoints (whole cohort) include progression free survival (PFS), metastasis-free survival after initiation of RT, overall survival (OS), % of change in initial treatment intent and Safety. DISCUSSION This is the first randomized phase 3 prospective trial designed to determine whether PSMA PET/CT molecular imaging can improve outcomes in patients with PCa who receive dRT. In this trial the incorporation of PSMA PET/CT may improve the success rate of curative intent radiotherapy in two ways: to optimize patient selection as a biomarker and to personalizes the radiotherapy plan. CLINICAL TRIAL REGISTRATION UCLA IND#147591 ○ Submission: 02.27.2020 ○ Safe-to-proceed letter issued by FDA: 04.01.2020 UCLA IRB #20-000378 ClinicalTrials.gov Identifier NCT04457245 . Date of Registry: 07.07.2020. Essen EudraCT 2020-003526-23.
Collapse
Affiliation(s)
- Jeremie Calais
- Ahmanson Translational Theranostics Division, Department of Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California, Peter Norton Medical Building, 200 Medical Plaza, Suite B-114-51, Los Angeles, CA 90095-7370 USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA USA
- Institute of Urologic Oncology, University of California Los Angeles, Los Angeles, CA USA
| | - Shaojun Zhu
- Ahmanson Translational Theranostics Division, Department of Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California, Peter Norton Medical Building, 200 Medical Plaza, Suite B-114-51, Los Angeles, CA 90095-7370 USA
| | - Nader Hirmas
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Hufelandstraße 55, 45131 Essen, Germany
| | - Matthias Eiber
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Boris Hadaschik
- Department of Urology, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | - Martin Stuschke
- Department of Radiotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Hufelandstraße 55, 45131 Essen, Germany
| | - Johannes Czernin
- Ahmanson Translational Theranostics Division, Department of Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California, Peter Norton Medical Building, 200 Medical Plaza, Suite B-114-51, Los Angeles, CA 90095-7370 USA
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA USA
- Institute of Urologic Oncology, University of California Los Angeles, Los Angeles, CA USA
| | - Amar U. Kishan
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA USA
- Institute of Urologic Oncology, University of California Los Angeles, Los Angeles, CA USA
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Nicholas G. Nickols
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA USA
- Department of Radiation Oncology, David Geffen School of Medicine, University of California, Los Angeles, USA
- Department of Radiation Oncology, VA Greater Los Angeles Healthcare System, Los Angeles, California USA
- Department of Urology, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - David Elashoff
- Institute of Urologic Oncology, University of California Los Angeles, Los Angeles, CA USA
- Department of Medicine Statistics Core (DOMStat), UCLA CTSI Biostatistics and Computational Biology, University of California, Los Angeles, USA
| | - Wolfgang P. Fendler
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Hufelandstraße 55, 45131 Essen, Germany
| |
Collapse
|
30
|
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
|
31
|
Dosimetric Analysis of the Short-Ranged Particle Emitter 161Tb for Radionuclide Therapy of Metastatic Prostate Cancer. Cancers (Basel) 2021; 13:cancers13092011. [PMID: 33921956 PMCID: PMC8122331 DOI: 10.3390/cancers13092011] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/18/2021] [Accepted: 04/20/2021] [Indexed: 12/25/2022] Open
Abstract
Simple Summary A tremendous effort and rapid development of the prostate-specific membrane antigen (PSMA)-targeting radio ligands for radionuclide therapy has resulted in encouraging response rates for advanced prostate cancer. Different radionuclides have been utilized or suggested as suitable candidates. In this study, a dynamic model of metastatic progress was developed and utilized to estimate a radiopharmaceutical’s potential of obtaining metastatic control of advanced prostate cancer. The simulations performed demonstrated the advantage of utilizing radionuclides with short-range particle emission, i.e., alpha-emitters and low-energy electrons. The recently-proposed beta-emitting radionuclide terbium-161 demonstrates great potential of being a future candidate towards targeted radionuclide therapy of advanced prostate cancer. This is in line with recent encouraging preclinical results and development of upscaling the product quality. Recently, the first in-human application with a [161Tb]Tb-DOTATOC also demonstrated good SPECT image quality, which can enable dosimetry calculations for new 161Tb-based radiopharmaceuticals. Abstract The aim of this study was to analyze the required absorbed doses to detectable metastases (Dreq) when using radionuclides with prostate specific membrane antigen (PSMA)-targeting radioligands to achieve a high probability for metastatic control. The Monte Carlo based analysis was performed for the clinically-used radionuclides yttrium-90, iodine-131, lutetium-177, and actinium-225, and the newly-proposed low-energy electron emitter terbium-161. It was demonstrated that metastatic formation rate highly influenced the metastatic distribution. Lower values generated few large detectable metastases, as in the case with oligo metastases, while high values generated a distribution of multiple small detectable metastases, as observed in patients with diffused visualized metastases. With equal number of detectable metastases, the total metastatic volume burden was 4–6 times higher in the oligo metastatic scenario compared to the diffusely visualized scenario. The Dreq was around 30% higher for the situations with 20 detectable metastases compared to one detectable metastasis. The Dreq for iodine-131 and yttrium-90 was high (920–3300 Gy). The Dreq for lutetium-177 was between 560 and 780 Gy and considerably lower Dreq were obtained for actinium-225 and terbium-161, with 240–330 Gy and 210–280 Gy, respectively. In conclusion, the simulations demonstrated that terbium-161 has the potential for being a more effective targeted radionuclide therapy for metastases using PSMA ligands.
Collapse
|
32
|
Jiao J, Quan Z, Zhang J, Wen W, Qin J, Yang L, Meng P, Jing Y, Ma S, Wu P, Han D, Davis AA, Ren J, Yang X, Kang F, Zhang Q, Wang J, Qin W. The Establishment of New Thresholds for PLND-Validated Clinical Nomograms to Predict Non-Regional Lymph Node Metastases: Using 68Ga-PSMA PET/CT as References. Front Oncol 2021; 11:658669. [PMID: 33937073 PMCID: PMC8082014 DOI: 10.3389/fonc.2021.658669] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/09/2021] [Indexed: 12/31/2022] Open
Abstract
Purpose PLND (pelvic lymph node dissection)-validated nomograms are widely accepted clinical tools to determine the necessity of PLND by predicting the metastasis of lymph nodes (LNMs) in pelvic region. However, these nomograms are in lacking of a threshold to predict the metastasis of extrareolar lymph nodes beyond pelvic region, which is not suitable for PLND. The aim of this study is to evaluate a threshold can be set for current clinical PLND-validated nomograms to predict extrareolar LN metastases beyond pelvic region in high-risk prostate cancer patients, by using 68Ga-PSMA PET/CT as a reference to determine LN metastases (LNMs). Experimental Design We performed a retrospective analysis of 57 high-risk treatment-naïve PC patients in a large tertiary care hospital in China who underwent 68Ga-PSMA-617 PET/CT imaging. LNMs was detected by 68Ga-PSMA-617 PET/CT and further determined by imaging follow-up after anti-androgen therapy. The pattern of LN metastatic spread of PC patients were evaluated and analyzed. The impact of 68Ga-PSMA PET/CT on clinical decisions based on three clinical PLND-validated nomograms (Briganti, Memorial Sloan Kettering Cancer Center, Winter) were evaluated by a multidisciplinary prostate cancer therapy team. The diagnostic performance and the threshold of these nomograms in predicting extrareolar LNMs metastasis were evaluated via receiver operating characteristic (ROC) curve analysis. Results LNMs were observed in 49.1% of the patients by 68Ga-PSMA PET/CT, among which 65.5% of LNMs were pelvic-regional and 34.5% of LNMs were observed in extrareolar sites (52.1% of these were located above the diaphragm). The Briganti, MSKCC and Winter nomograms showed that 70.2%-71.9% of the patients in this study need to receive ePLND according to the EAU and NCCN guidelines. The LN staging information obtained from 68Ga-PSMA PET/CT would have led to changes of planned management in 70.2% of these patients, including therapy modality changes in 21.1% of the patients, which were mainly due to newly detected non-regional LNMs. The thresholds of nomograms to predict non-regional LNMs were between 64% and 75%. The PC patients with a score >64% in Briganti nomogram, a score >75% in MSKCC nomogram and a score >67% in Winter nomogram were more likely to have non-regional LNMs. The AUCs (Area under curves) of the clinical nomograms (Briganti, MSKCC and Winter) in predicting non-regional LNMs were 0.816, 0.830 and 0.793, respectively. Conclusions By using 68Ga-PSMA PET/CT as reference of LNM, the PLND-validated clinical nomograms can not only predict regional LNMs, but also predict non-regional LNMs. The additional information from 68Ga-PSMA PET/CT may provide added benefit to nomograms-based clinical decision-making in more than two-thirds of patients for reducing unnecessary PLND. We focused on that a threshold can be set for current clinical PLND-validated nomograms to predict extrareolar LN metastases with an AUC accuracy of about 80% after optimizing the simple nomograms which may help to improve the efficiency for PC therapy significantly in clinical practice.
Collapse
Affiliation(s)
- Jianhua Jiao
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhiyong Quan
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jingliang Zhang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Weihong Wen
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, China
| | - Jun Qin
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Lijun Yang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Ping Meng
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yuming Jing
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Shuaijun Ma
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Peng Wu
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Donghui Han
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Andrew A Davis
- Department of Medicine, Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jing Ren
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiaojian Yang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Fei Kang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Qiang Zhang
- Department of Medicine, Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States
| | - Jing Wang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Weijun Qin
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
33
|
Fourquet A, Lahmi L, Rusu T, Belkacemi Y, Créhange G, de la Taille A, Fournier G, Cussenot O, Gauthé M. Restaging the Biochemical Recurrence of Prostate Cancer with [ 68Ga]Ga-PSMA-11 PET/CT: Diagnostic Performance and Impact on Patient Disease Management. Cancers (Basel) 2021; 13:cancers13071594. [PMID: 33808301 PMCID: PMC8038030 DOI: 10.3390/cancers13071594] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/21/2021] [Accepted: 03/26/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary We aimed to evaluate the diagnostic performance, impact on patient disease management, and therapy efficacy prediction of [68Ga]Ga-PSMA-11 PET/CT on 294 patients with biochemical recurrence of prostate cancer. We established a composite standard of truth for the imaging based on all clinical data available collected during the follow-up period with a median duration of follow-up of 17 months. Using this methodology, we found that the overall per-patient sensitivity and specificity were both 70%, the patient disease management was changed in 68% of patients, and that [68Ga]Ga-PSMA-11 PET/CT impacted this change in 86% of patients. The treatment carried out on the patient was considered effective in 78% of patients; in 89% of patients when guided by [68Ga]Ga-PSMA-11 PET/CT versus 61% of patients when not guided by [68Ga]Ga-PSMA-11 PET/CT. Abstract Background: Detection rates of [68Ga]Ga-PSMA-11 PET/CT on the restaging of prostate cancer (PCa) patients presenting with biochemical recurrence (BCR) have been well documented, but its performance and impact on patient management have not been evaluated as extensively. Methods: Retrospective analysis of PCa patients presenting with BCR and referred for [68Ga]Ga-PSMA-11 PET/CT. Pathological foci were classified according to six anatomical sites and evaluated with a three-point scale according to the uptake intensity. The impact of [68Ga]Ga-PSMA-11 PET/CT was defined as any change in management that was triggered by [68Ga]Ga-PSMA-11 PET/CT. The existence of a PCa lesion was established according to a composite standard of truth based on all clinical data available collected during the follow-up period. Results: We included 294 patients. The detection rate was 69%. Per-patient sensitivity and specificity were both 70%. Patient disease management was changed in 68% of patients, and [68Ga]Ga-PSMA-11 PET/CT impacted this change in 86% of patients. The treatment carried out on patient was considered effective in 89% of patients when guided by [68Ga]Ga-PSMA-11 PET/CT versus 61% of patients when not guided by [68Ga]Ga-PSMA-11 PET/CT (p < 0.001). Conclusions: [68Ga]Ga-PSMA-11 PET/CT demonstrated high performance in locating PCa recurrence sites and impacted therapeutic management in nearly two out of three patients.
Collapse
Affiliation(s)
- Aloÿse Fourquet
- Department of Nuclear Medicine, Hôpital Tenon-AP-HP, Sorbonne Université, 75020 Paris, France; (A.F.); (T.R.)
| | - Lucien Lahmi
- Department of Radiation Oncology, Hôpital Tenon-AP-HP, Sorbonne Université, 75020 Paris, France;
| | - Timofei Rusu
- Department of Nuclear Medicine, Hôpital Tenon-AP-HP, Sorbonne Université, 75020 Paris, France; (A.F.); (T.R.)
| | - Yazid Belkacemi
- Department of Radiation Oncology and Henri Mondor Breast Center, Hôpitaux Universitaires Henri Mondor, Université Paris-Est Créteil (UEPC) et IMRB—INSERM U955 Team 21, 94000 Créteil, France;
| | - Gilles Créhange
- Department of Radiation Oncology, Institut Curie, 75005 Paris, France;
| | - Alexandre de la Taille
- Department of Urology, Hôpitaux Universitaires Henri Mondor, Université Paris-Est Créteil (UEPC), 94000 Créteil, France;
| | - Georges Fournier
- Department of Urology, Hôpital de la Cavale Blanche, Université de Brest, 29200 Brest, France;
| | - Olivier Cussenot
- Department of Urology, Hôpital Tenon-AP-HP, Sorbonne Université, 75020 Paris, France;
| | - Mathieu Gauthé
- Department of Nuclear Medicine, Hôpital Tenon-AP-HP, Sorbonne Université, 75020 Paris, France; (A.F.); (T.R.)
- AP-HP Health Economics Research Unit, INSERM-UMR1153, 75004 Paris, France
- Correspondence: author: ; Tel.: +33-156017842; Fax: +33-156016171
| |
Collapse
|
34
|
Rauscher I, Karimzadeh A, Schiller K, Horn T, D’Alessandria C, Franz C, Wörther H, Nguyen N, Combs SE, Weber WA, Eiber M. Detection efficacy of 18F-rhPSMA-7.3 PET/CT and impact on patient management in patients with biochemical recurrence of prostate cancer after radical prostatectomy and prior to potential salvage treatment. J Nucl Med 2021; 62:jnumed.120.260091. [PMID: 33712531 PMCID: PMC8612184 DOI: 10.2967/jnumed.120.260091] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 03/02/2021] [Accepted: 03/02/2021] [Indexed: 01/22/2023] Open
Abstract
Purpose: Radiohybrid prostate-specific membrane antigen (rhPSMA) ligands are a new class of 18F-labeled PSMA-targeting agents. 18F-rhPSMA-7.3 is a lead compound which is currently under investigation in two multicenter phase III trials for PET-imaging. Here, we report the first retrospective data on its detection efficacy and potential impact on clinical management in a homogeneous cohort of patients with biochemical recurrence after radical prostatectomy, and prior to any salvage therapy. Methods: 242 patients (median [range] PSA, 0.60 [0.2-60.8] ng/mL) who underwent 18F-rhPSMA-7.3 PET/CT were retrospectively selected from the institutions' database. Images were re-read by an experienced nuclear medicine physician. Lesion detection rates were stratified by PSA. Further, potential management before and after PET was assessed by an interdisciplinary simulated tumor board and categorized (major vs. minor vs. no therapeutic change). The distribution of management change identified in each PSA subgroup was determined. Results: In total, 176/242 (72.7%) patients showed PSMA-ligand positive findings. 18F-rhPSMA-7.3 detection rates were 61.8% (63/102), 67.9% (38/56), 81.1% (30/37) and 95.7% (45/47) for PSA-levels of 0.2-<0.5 ng/mL, 0.5-<1 ng/mL, 1-<2 ng/mL and ≥2 ng/mL, respectively. 18F-rhPSMA-7.3 PET/CT revealed local recurrence, pelvic lymph node metastases, retroperitoneal lymph nodes metastases, supradiaphragmatic lymph nodes, bone metastases, and visceral metastases in 48.8% (n = 118), 28.9% (n = 70), 6.6% (n = 16), 1.2% (n = 3), 13.2% (n = 32) and 1.2% (n = 3) of patients, respectively. Notably, bone lesions were identified in 8.8% of patients (9/102) with PSA <0.5 ng/mL. Results from the interdisciplinary simulated tumor board indicated change of therapeutic management in 153/242 patients (63.2%) with 54/242 (22.3%) considered major and 99/242 (40.9%) minor, respectively. 18F-rhPSMA-7.3 PET/CT did not prompt any therapeutic changes in 64/242 patients (26.4%). Conclusion: 18F-rhPSMA-7.3 PET offers high detection efficacy in patients with biochemical recurrence after radical prostatectomy, and prior to potential salvage therapy, and results in a potential change in treatment plans in nearly 2/3 of patients. Keywords: Biochemical recurrence; hybrid imaging; positron emission tomography; prostate cancer; prostate-specific membrane antigen.
Collapse
Affiliation(s)
- Isabel Rauscher
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
| | - Amir Karimzadeh
- 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; and
| | - Thomas Horn
- Department of Urology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Calogero D’Alessandria
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
| | - Charlott Franz
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
| | - Hannah Wörther
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
| | - Noemi Nguyen
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
| | - Stephanie E. Combs
- Department of Radiation Oncology, School of Medicine, Technical University of Munich, Munich, Germany; and
| | - Wolfgang A. Weber
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
| | - Matthias Eiber
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
35
|
Hernes E, Revheim ME, Hole KH, Tulipan AJ, Strømme H, Lilleby W, Seierstad T. Prostate-Specific Membrane Antigen PET for Assessment of Primary and Recurrent Prostate Cancer with Histopathology as Reference Standard: A Systematic Review and Meta-Analysis. PET Clin 2021; 16:147-165. [PMID: 33648661 DOI: 10.1016/j.cpet.2020.12.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Prostate-specific membrane antigen PET is a promising diagnostic tool in prostate cancer. The gold standard for the detection of prostate tumor and lymph node metastases is histopathology. The aim of the present review was to investigate accuracy measures of 68Ga/18F-labeled prostate-specific membrane antigen PET tracers in primary and recurrent prostate cancer with systematic sector-based histopathology as the reference standard. A systematic literature search was performed and 34 studies were included. Overall, prostate-specific membrane antigen PET showed high specificity, but variable sensitivity to localize known prostate cancer and detect pelvic lymph node metastases.
Collapse
Affiliation(s)
- Eivor Hernes
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, P.O. Box 4956 Nydalen, 0424 Oslo, Norway.
| | - Mona-Elisabeth Revheim
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, P.O. Box 4956 Nydalen, 0424 Oslo, Norway; Institute of Clinical Medicine, University of Oslo, P.O. Box 1171 Blindern, 0318 Oslo, Norway
| | - Knut Håkon Hole
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, P.O. Box 4956 Nydalen, 0424 Oslo, Norway; Institute of Clinical Medicine, University of Oslo, P.O. Box 1171 Blindern, 0318 Oslo, Norway
| | - Andreas Julius Tulipan
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, P.O. Box 4956 Nydalen, 0424 Oslo, Norway; Institute of Clinical Medicine, University of Oslo, P.O. Box 1171 Blindern, 0318 Oslo, Norway
| | - Hilde Strømme
- Library of Medicine and Science, University of Oslo, Sognsvannsveien 20, 0372 Oslo, Norway
| | - Wolfgang Lilleby
- Department of Oncology, Oslo University Hospital, P.O. Box 4953 Nydalen, 0424 Oslo, Norway
| | - Therese Seierstad
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, P.O. Box 4956 Nydalen, 0424 Oslo, Norway
| |
Collapse
|
36
|
Leitsmann C, Schmid M, Sahlmann CO, Trojan L, Strauss A. Mesorectal Lymph Node Metastases as Index Lesion in 68Ga-PSMA-PET/CT Imaging for Recurrent Prostate Cancer. Front Surg 2021; 8:637134. [PMID: 33732729 PMCID: PMC7957000 DOI: 10.3389/fsurg.2021.637134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 01/22/2021] [Indexed: 12/04/2022] Open
Abstract
Purpose: Several studies have demonstrated an advantage of 68Ga-PSMA-PET/CT as staging modality for detection of prostate cancer (PCa) metastases. Data concerning metastatic manifestation and impact on PCa development of mesorectal lymph nodes (MLN) is limited. Our investigation describes MLN metastases as index lesion in 68Ga-PSMA PET/CT imaging for recurrent PCa. Methods: Twelve PCa patients with biochemical recurrence (BCR) after primary therapy who prospectively underwent a baseline 68Ga-PSMA-PET/CT initially showed MLN metastases. Eight of these patients received a follow-up 68Ga-PSMA-PET/CT to evaluate treatment response and further evolution. Prostate-specific antigen (PSA)-levels, changes in PSMA-uptake of MLN metastases and further 68Ga-PSMA PET/CT findings were recorded. Results: Median PSA at the first 68Ga-PSMA-PET/CT was 5.39 ng/ml. In all patients therapeutic management changed after the first 68Ga-PSMA-PET/CT. Androgen deprivation therapy (ADT) was initiated in seven of eight patients, one patient restarted initial ADT. Three patients additionally received salvage radiation therapy (sRT) including the prostatic lodge and docetaxel chemotherapy was started in one case. At follow-up, a decrease of PSA-level was detected in all patients (median 2.05 ng/ml) after median 10 months. In six of eight patients we observed a decrease or complete regress of PSMA-uptake in MLN in the follow-up 68Ga-PSMA-PET/CT. Conclusion: MLN metastases detected by 68Ga-PSMA-PET/CT seem to be a relevant localization of tumor manifestation and may serve as index lesion in the treatment of recurrent PCa. Besides the known oncological benefits of ADT and sRT, in case of sole MLN metastases individualized therapy like salvage lymphadenectomy or RT with a defined radiation field could be options for these patients.
Collapse
Affiliation(s)
- Conrad Leitsmann
- Department of Urology, University Medical Center Goettingen, Goettingen, Germany
| | - Marianne Schmid
- Department of Urology, University Medical Center Goettingen, Goettingen, Germany
| | | | - Lutz Trojan
- Department of Urology, University Medical Center Goettingen, Goettingen, Germany
| | - Arne Strauss
- Department of Urology, University Medical Center Goettingen, Goettingen, Germany
| |
Collapse
|
37
|
Rowe LS, Harmon S, Horn A, Shankavaram U, Roy S, Ning H, Lindenberg L, Mena E, Citrin DE, Choyke P, Turkbey B. Pattern of failure in prostate cancer previously treated with radical prostatectomy and post-operative radiotherapy: a secondary analysis of two prospective studies using novel molecular imaging techniques. Radiat Oncol 2021; 16:32. [PMID: 33568190 PMCID: PMC7874470 DOI: 10.1186/s13014-020-01733-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 12/17/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Prostate Membrane Specific Antigen (PSMA) positron emission tomography (PET) and multiparametric MRI (mpMRI) have shown high accuracy in identifying recurrent lesions after definitive treatment in prostate cancer (PCa). In this study, we aimed to outline patterns of failure in a group of post-prostatectomy patients who received adjuvant or salvage radiation therapy (PORT) and subsequently experienced biochemical recurrence, using 18F-PSMA PET/CT and mpMRI. METHODS PCa patients with biochemical failure post-prostatectomy, and no evident site of recurrence on conventional imaging, were enrolled on two prospective trials of first and second generation 18F-PSMA PET agents (18F-DCFBC and 18F-DCFPyL) in combination with MRI between October 2014 and December 2018. The primary aim of our study is to characterize these lesions with respect to their location relative to previous PORT field and received dose. RESULTS A total of 34 participants underwent 18F-PSMA PET imaging for biochemical recurrence after radical prostatectomy and PORT, with 32/34 found to have 18F-PSMA avid lesions. On 18F-PSMA, 17/32 patients (53.1%) had metastatic disease, 8/32 (25.0%) patients had locoregional recurrences, and 7/32 (21.9%) had local failure in the prostate fossa. On further exploration, we noted 6/7 (86%) of prostate fossa recurrences were in-field and were encompassed by 100% isodose lines, receiving 64.8-72 Gy. One patient had marginal failure encompassed by the 49 Gy isodose. CONCLUSIONS 18F-PSMA PET imaging demonstrates promise in identifying occult PCa recurrence after PORT. Although distant recurrence was the predominant pattern of failure, in-field recurrence was noted in approximately 1/5th of patients. This should be considered in tailoring radiotherapy practice after prostatectomy. Trial registration www.clinicaltrials.gov , NCT02190279 and NCT03181867. Registered July 12, 2014, https://clinicaltrials.gov/ct2/show/NCT02190279 and June 8 2017, https://clinicaltrials.gov/ct2/show/NCT03181867 .
Collapse
Affiliation(s)
- Lindsay S. Rowe
- Radiation Oncology Branch, National Cancer Institute, 10 Center Drive Magnuson Clinical Center, Room B2-3500, Bethesda, MD 20892 USA
- Department of Radiation Oncology, Cross Cancer Institute, 11560 University Avenue, Edmonton, AB T6G 1Z2 Canada
| | - Stephanie Harmon
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research, 10 Center Drive Magnuson Clinical Center, Room B3B69F, Bethesda, MD 20892 USA
| | - Adam Horn
- Walter Reed National Military Medical Center, Bethesda, MD 8901 Rockville Pike USA
| | - Uma Shankavaram
- Radiation Oncology Branch, National Cancer Institute, 10 Center Drive Magnuson Clinical Center, Room 1002, Bethesda, MD 20892 USA
| | - Soumyajit Roy
- Radiation Oncology Branch, National Cancer Institute, 10 Center Drive Magnuson Clinical Center, Room B2-3500, Bethesda, MD 20892 USA
| | - Holly Ning
- Radiation Oncology Branch, National Cancer Institute, 10 Center Drive Magnuson Clinical Center, Room B2-3500, Bethesda, MD 20892 USA
| | - Liza Lindenberg
- Molecular Imaging Program, National Cancer Institute, 10 Center Drive Magnuson Clinical Center, Room B3B69F, Bethesda, MD 20892 USA
| | - Esther Mena
- Molecular Imaging Program, National Cancer Institute, 10 Center Drive Magnuson Clinical Center, Room B3B69F, Bethesda, MD 20892 USA
| | - Deborah E. Citrin
- Radiation Oncology Branch, National Cancer Institute, 10 Center Drive Magnuson Clinical Center, Room B2-3500, Bethesda, MD 20892 USA
| | - Peter Choyke
- Molecular Imaging Program, National Cancer Institute, 10 Center Drive Magnuson Clinical Center, Room B3B69F, Bethesda, MD 20892 USA
| | - Baris Turkbey
- Molecular Imaging Program, National Cancer Institute, 10 Center Drive Magnuson Clinical Center, Room B3B69F, Bethesda, MD 20892 USA
| |
Collapse
|
38
|
Li R, Liu X, Yang B, Qiu J. External beam radiotherapy for prostate cancer: What are the current research trends and hotspots? Cancer Med 2021; 10:772-782. [PMID: 33480190 PMCID: PMC7877352 DOI: 10.1002/cam4.3700] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/11/2020] [Accepted: 12/12/2020] [Indexed: 01/03/2023] Open
Abstract
Background The external beam radiotherapy (EBRT) applied for prostate cancer (PCa) has been one of the most important and hottest research fields over recent decades. This study aimed to explore the research hotspots of EBRT in PCa and help the researchers have a clear and intuitive reference basis for later researches. Methods The literature scientometric analysis related to “EBRT applied for PCa” was conducted via the Web of Science Core Collection from 2010 to 2019. The Microsoft Office Excel 2019 and CiteSpace V. 5.7.R1 software were introduced for visualizing and analyzing the data. Results A total of 7860 relevant papers were extracted and downloaded. A total of 7828 papers were extracted and analyzed after data cleansing by CiteSpace. The tendency of published papers was comprehensively increasing from 2010 to 2019. Among all 73 countries/regions, USA published the most papers, accounting for 39%, which was the most active contributor with most publications. Australia (Centrality: 0.18), England (Centrality: 0.12) were cooperating most cohesively with other countries. Univ Toronto was the most productive institute (229), while Harvard Univ (Centrality: 0.67) had extensive collaborations with other institutes. The International journal of Radiation Oncology Biology Physics had the largest number of publications and the highest number of co‐citations. Briganti A had the largest volume of publications. D'Amico AV had the highest number of co‐citations. Four latest and largest clusters were identified as oligometastases, salvage therapy (SRT), prostate‐specific membrane antigen (PSMA), and hypofractionation. Thirteen references became strongest burst citations lasting until 2019. The studies of “oligometastases,” “SRT,” “PSMA,” “hypofractionation,” “postoperative radiotherapy,” and “dose and fraction regimen changes” were prevailing in the recent years. Conclusion The “oligometastases,” “SRT,” “PSMA,” “hypofractionation,” “postoperative radiotherapy,” and “dose and fraction regimen changes” may be the state‐of‐art research frontiers, and related studies will advance in this field over time.
Collapse
Affiliation(s)
- Rui Li
- Department of Radiation Oncology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.,Graduate School of Peking Union Medical College, Beijing, China
| | - Xia Liu
- Department of Radiation Oncology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.,Graduate School of Peking Union Medical College, Beijing, China
| | - Bo Yang
- Department of Radiation Oncology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.,Graduate School of Peking Union Medical College, Beijing, China
| | - Jie Qiu
- Department of Radiation Oncology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China.,Graduate School of Peking Union Medical College, Beijing, China
| |
Collapse
|
39
|
Kuten J, Dekalo S, Mintz I, Yossepowitch O, Mano R, Even-Sapir E. The significance of equivocal bone findings in staging PSMA imaging in the preoperative setting: validation of the PSMA-RADS version 1.0. EJNMMI Res 2021; 11:3. [PMID: 33409930 PMCID: PMC7788112 DOI: 10.1186/s13550-020-00745-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 12/22/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Assessing the extent of disease in newly diagnosed prostate cancer (PC) patients is crucial for tailoring an appropriate treatment approach. Prostate-specific membrane antigen (PSMA)-targeted positron emission tomography/computed tomography (PET/CT) reportedly has greater accuracy than conventional imaging for staging PC. As with any imaging modality, pitfalls and nonspecific findings do occur. The PSMA reporting and data system (PSMA-RADS) version 1.0 offers structured interpretation of PSMA-targeted studies and classifies lesions by likelihood of clinical significance. The aim of this retrospective study was to evaluate the clinical significance of equivocal bone findings on staging PSMA-targeted imaging, as defined by PSMA-RADS version 1.0, in the preoperative setting. Fifteen of 406 consecutive patients staged by PET/CT prior to radical prostatectomy had equivocal bone lesions. The scans were retrospectively scored with the PSMA-RADS version 1.0 system, blinded to disease course and follow-up data. Postoperative persistence of prostate-specific antigen levels supported by imaging and histological findings was used as the reference standard for the true significance of equivocal imaging findings. RESULTS Thirteen of the 15 patients had an overall PSMA-RADS score of 3B, of whom only two had true metastatic disease. The remaining patients had scores of 4 (n = 1) or 5 (n = 1), all confirmed as true positive prostate-related malignant lesions. A per-lesion analysis identified 29 bone lesions, of which 27 were scored PSMA-RADS 3B, and only three of them were true metastases. Thus, debatable lesions proved to have no clinical significance in 84.6% of cases, and only 11% of equivocal PSMA-RADS 3B bone lesions were true positive. CONCLUSIONS In intermediate and high-risk patients staged prior to radical prostatectomy, the majority of PSMA-RADS 3B lesions are of no clinical relevance. Bone lesions judged as being highly suspicious for metastases (PSMA-RADS 4/5) were all validated as true positives.
Collapse
Affiliation(s)
- Jonathan Kuten
- Departments of Nuclear Medicine, Tel-Aviv Sourasky Medical Center, 6 Weizmann St, 6423906, Tel-Aviv, Israel. .,Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| | - Snir Dekalo
- Departments of Urology, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Ishai Mintz
- Departments of Urology, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Ofer Yossepowitch
- Departments of Urology, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Roy Mano
- Departments of Urology, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Einat Even-Sapir
- Departments of Nuclear Medicine, Tel-Aviv Sourasky Medical Center, 6 Weizmann St, 6423906, Tel-Aviv, Israel.,Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
40
|
Salvage Pelvic Lymph Node Dissection and Current State of Imaging for Recurrent Prostate Cancer: Does a Standard Exist? Curr Urol Rep 2020; 21:62. [PMID: 33159608 DOI: 10.1007/s11934-020-01011-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2020] [Indexed: 10/23/2022]
Abstract
PURPOSE OF REVIEW We aim to evaluate the efficacy of salvage lymph node dissection (SLND) for nodal recurrent prostate cancer after primary treatment. We also provide a review of the diagnostic performance of next-generation sequencing (next-generation imaging (NGI)) radiotracers in the salvage setting. RECENT FINDINGS Most studies evaluating SLND include a heterogeneous population with a small sample size and are retrospective in design. The 5-year clinical recurrence-free and cancer-specific survival following SLND are 26-52% and 57-89%, respectively, among prospective studies. NGI improves accuracy in detecting nodal recurrence compared to conventional CT, with PMSA PET-CT showing the most promise. However, limited studies exist comparing imaging modalities and performance is variable at low PSA values. SLND is a promising treatment option, but more prospective data are needed to determine the ideal surgical candidate and long-term oncologic outcomes. More studies comparing different NGI are needed to determine the best imaging modality in patients who may be candidates for salvage treatment.
Collapse
|
41
|
Preisser F, Bandini M, Nazzani S, Mazzone E, Marchioni M, Tian Z, Chun FK, Saad F, Briganti A, Haese A, Montorsi F, Huland H, Graefen M, Tilki D, Karakiewicz PI. Development and Validation of a Lookup Table for the Prediction of Metastatic Prostate Cancer According to Prostatic-specific Antigen Value, Clinical Tumor Stage, and Gleason Grade Groups. Eur Urol Oncol 2020; 3:631-639. [DOI: 10.1016/j.euo.2019.03.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 02/15/2019] [Accepted: 03/08/2019] [Indexed: 10/27/2022]
|
42
|
Lindenberg L, Mena E, Turkbey B, Shih JH, Reese SE, Harmon SA, Lim I, Lin F, Ton A, McKinney YL, Eclarinal P, Citrin DE, Dahut W, Madan R, Wood BJ, Krishnasamy V, Chang R, Levy E, Pinto P, Eary JF, Choyke PL. Evaluating Biochemically Recurrent Prostate Cancer: Histologic Validation of 18F-DCFPyL PET/CT with Comparison to Multiparametric MRI. Radiology 2020; 296:564-572. [PMID: 32633674 PMCID: PMC7457947 DOI: 10.1148/radiol.2020192018] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 04/29/2020] [Accepted: 05/12/2020] [Indexed: 12/24/2022]
Abstract
Background Prostate cancer recurrence is found in up to 40% of men with prior definitive (total prostatectomy or whole-prostate radiation) treatment. Prostate-specific membrane antigen PET agents such as 2-(3-{1-carboxy-5-[(6-[18F]fluoro-pyridine 3-carbonyl)-amino]-pentyl}-ureido)-pentanedioic acid (18F-DCFPyL) may improve detection of recurrence compared with multiparametric MRI; however, histopathologic validation is lacking. Purpose To determine the sensitivity, specificity, and positive predictive value (PPV) of 18F-DCFPyL PET/CT based on histologic analysis and to compare with pelvic multiparametric MRI in men with biochemically recurrent prostate cancer. Materials and Methods Men were prospectively recruited after prostatectomy and/or radiation therapy with rising prostate-specific antigen level (median, 2.27 ng/mL; range, 0.2-27.45 ng/mL) and a negative result at conventional imaging (bone scan and/or CT). Participants underwent 18F-DCFPyL PET/CT imaging and 3.0-T pelvic multiparametric MRI. Statistical analysis included Wald and modified χ2 tests. Results A total of 323 lesions were visualized in 77 men by using 18F-DCFPyL or multiparametric MRI, with imaging detection concordance of 25% (82 of 323) when including all lesions in the MRI field of view and 53% (52 of 99) when only assessing prostate bed lesions. 18F-DCFPyL depicted more pelvic lymph nodes than did MRI (128 vs 23 nodes). Histologic validation was obtained in 80 locations with sensitivity, specificity, and PPV of 69% (25 of 36; 95% confidence interval [CI]: 51%, 88%), 91% (40 of 44; 95% CI: 74%, 98%), and 86% (25 of 29; 95% CI: 73%, 97%) for 18F-DCFPyL and 69% (24 of 35; 95% CI: 50%, 86%), 74% (31 of 42; 95% CI: 42%, 89%), and 69% (24 of 35; 95% CI: 50%, 88%) for multiparametric MRI (P = .95, P = .14, and P = .07, respectively). In the prostate bed, sensitivity, specificity, and PPV were 57% (13 of 23; 95% CI: 32%, 81%), 86% (18 of 21; 95% CI: 73%, 100%), and 81% (13 of 16; 95% CI: 59%, 100%) for 18F-DCFPyL and 83% (19 of 23; 95% CI: 59%, 100%), 52% (11 of 21; 95% CI: 29%, 74%), and 66% (19 of 29; 95% CI: 44%, 86%) for multiparametric MRI (P = .19, P = .02, and P = .17, respectively). The addition of 18F-DCFPyL to multiparametric MRI improved PPV by 38% overall (P = .02) and by 30% (P = .09) in the prostate bed. Conclusion Findings with 2-(3-{1-carboxy-5-[(6-[18F]fluoro-pyridine 3-carbonyl)-amino]-pentyl}-ureido)-pentanedioic acid (18F-DCFPyL) were histologically validated and demonstrated high specificity and positive predictive value. In the pelvis, 18F-DCFPyL depicted more lymph nodes and improved positive predictive value and specificity when added to multiparametric MRI. © RSNA, 2020 Online supplemental material is available for this article. See also the editorial by Zukotynski and Rowe in this issue.
Collapse
Affiliation(s)
- Liza Lindenberg
- From the Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B47A, Bethesda, MD 20892 (L.L., E.M., B.T., I.L., F.L., A.T., Y.L.M., P.E., P.L.C.); Division of Cancer Treatment and Diagnosis: Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, Md (J.H.S.); National Cancer Institute Biometrics Research Program Contract, General Dynamics Information Technology, Falls Church, Va (S.E.R.); Clinical Research Directorate, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Bethesda, Md (S.A.H.); Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md (D.E.C.); Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (W.D., R.M.); Center of Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Md (B.J.W., V.K., R.C., E.L.); Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (P.P.); and Cancer Imaging Program, National Cancer Institute, Bethesda, Md (J.F.E.)
| | - Esther Mena
- From the Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B47A, Bethesda, MD 20892 (L.L., E.M., B.T., I.L., F.L., A.T., Y.L.M., P.E., P.L.C.); Division of Cancer Treatment and Diagnosis: Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, Md (J.H.S.); National Cancer Institute Biometrics Research Program Contract, General Dynamics Information Technology, Falls Church, Va (S.E.R.); Clinical Research Directorate, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Bethesda, Md (S.A.H.); Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md (D.E.C.); Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (W.D., R.M.); Center of Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Md (B.J.W., V.K., R.C., E.L.); Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (P.P.); and Cancer Imaging Program, National Cancer Institute, Bethesda, Md (J.F.E.)
| | - Baris Turkbey
- From the Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B47A, Bethesda, MD 20892 (L.L., E.M., B.T., I.L., F.L., A.T., Y.L.M., P.E., P.L.C.); Division of Cancer Treatment and Diagnosis: Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, Md (J.H.S.); National Cancer Institute Biometrics Research Program Contract, General Dynamics Information Technology, Falls Church, Va (S.E.R.); Clinical Research Directorate, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Bethesda, Md (S.A.H.); Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md (D.E.C.); Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (W.D., R.M.); Center of Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Md (B.J.W., V.K., R.C., E.L.); Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (P.P.); and Cancer Imaging Program, National Cancer Institute, Bethesda, Md (J.F.E.)
| | - Joanna H. Shih
- From the Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B47A, Bethesda, MD 20892 (L.L., E.M., B.T., I.L., F.L., A.T., Y.L.M., P.E., P.L.C.); Division of Cancer Treatment and Diagnosis: Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, Md (J.H.S.); National Cancer Institute Biometrics Research Program Contract, General Dynamics Information Technology, Falls Church, Va (S.E.R.); Clinical Research Directorate, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Bethesda, Md (S.A.H.); Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md (D.E.C.); Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (W.D., R.M.); Center of Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Md (B.J.W., V.K., R.C., E.L.); Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (P.P.); and Cancer Imaging Program, National Cancer Institute, Bethesda, Md (J.F.E.)
| | - Sarah E. Reese
- From the Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B47A, Bethesda, MD 20892 (L.L., E.M., B.T., I.L., F.L., A.T., Y.L.M., P.E., P.L.C.); Division of Cancer Treatment and Diagnosis: Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, Md (J.H.S.); National Cancer Institute Biometrics Research Program Contract, General Dynamics Information Technology, Falls Church, Va (S.E.R.); Clinical Research Directorate, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Bethesda, Md (S.A.H.); Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md (D.E.C.); Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (W.D., R.M.); Center of Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Md (B.J.W., V.K., R.C., E.L.); Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (P.P.); and Cancer Imaging Program, National Cancer Institute, Bethesda, Md (J.F.E.)
| | - Stephanie A. Harmon
- From the Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B47A, Bethesda, MD 20892 (L.L., E.M., B.T., I.L., F.L., A.T., Y.L.M., P.E., P.L.C.); Division of Cancer Treatment and Diagnosis: Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, Md (J.H.S.); National Cancer Institute Biometrics Research Program Contract, General Dynamics Information Technology, Falls Church, Va (S.E.R.); Clinical Research Directorate, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Bethesda, Md (S.A.H.); Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md (D.E.C.); Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (W.D., R.M.); Center of Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Md (B.J.W., V.K., R.C., E.L.); Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (P.P.); and Cancer Imaging Program, National Cancer Institute, Bethesda, Md (J.F.E.)
| | - Ilhan Lim
- From the Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B47A, Bethesda, MD 20892 (L.L., E.M., B.T., I.L., F.L., A.T., Y.L.M., P.E., P.L.C.); Division of Cancer Treatment and Diagnosis: Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, Md (J.H.S.); National Cancer Institute Biometrics Research Program Contract, General Dynamics Information Technology, Falls Church, Va (S.E.R.); Clinical Research Directorate, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Bethesda, Md (S.A.H.); Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md (D.E.C.); Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (W.D., R.M.); Center of Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Md (B.J.W., V.K., R.C., E.L.); Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (P.P.); and Cancer Imaging Program, National Cancer Institute, Bethesda, Md (J.F.E.)
| | - Frank Lin
- From the Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B47A, Bethesda, MD 20892 (L.L., E.M., B.T., I.L., F.L., A.T., Y.L.M., P.E., P.L.C.); Division of Cancer Treatment and Diagnosis: Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, Md (J.H.S.); National Cancer Institute Biometrics Research Program Contract, General Dynamics Information Technology, Falls Church, Va (S.E.R.); Clinical Research Directorate, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Bethesda, Md (S.A.H.); Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md (D.E.C.); Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (W.D., R.M.); Center of Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Md (B.J.W., V.K., R.C., E.L.); Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (P.P.); and Cancer Imaging Program, National Cancer Institute, Bethesda, Md (J.F.E.)
| | - Anita Ton
- From the Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B47A, Bethesda, MD 20892 (L.L., E.M., B.T., I.L., F.L., A.T., Y.L.M., P.E., P.L.C.); Division of Cancer Treatment and Diagnosis: Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, Md (J.H.S.); National Cancer Institute Biometrics Research Program Contract, General Dynamics Information Technology, Falls Church, Va (S.E.R.); Clinical Research Directorate, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Bethesda, Md (S.A.H.); Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md (D.E.C.); Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (W.D., R.M.); Center of Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Md (B.J.W., V.K., R.C., E.L.); Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (P.P.); and Cancer Imaging Program, National Cancer Institute, Bethesda, Md (J.F.E.)
| | - Yolanda L. McKinney
- From the Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B47A, Bethesda, MD 20892 (L.L., E.M., B.T., I.L., F.L., A.T., Y.L.M., P.E., P.L.C.); Division of Cancer Treatment and Diagnosis: Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, Md (J.H.S.); National Cancer Institute Biometrics Research Program Contract, General Dynamics Information Technology, Falls Church, Va (S.E.R.); Clinical Research Directorate, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Bethesda, Md (S.A.H.); Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md (D.E.C.); Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (W.D., R.M.); Center of Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Md (B.J.W., V.K., R.C., E.L.); Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (P.P.); and Cancer Imaging Program, National Cancer Institute, Bethesda, Md (J.F.E.)
| | - Philip Eclarinal
- From the Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B47A, Bethesda, MD 20892 (L.L., E.M., B.T., I.L., F.L., A.T., Y.L.M., P.E., P.L.C.); Division of Cancer Treatment and Diagnosis: Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, Md (J.H.S.); National Cancer Institute Biometrics Research Program Contract, General Dynamics Information Technology, Falls Church, Va (S.E.R.); Clinical Research Directorate, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Bethesda, Md (S.A.H.); Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md (D.E.C.); Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (W.D., R.M.); Center of Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Md (B.J.W., V.K., R.C., E.L.); Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (P.P.); and Cancer Imaging Program, National Cancer Institute, Bethesda, Md (J.F.E.)
| | - Deborah E. Citrin
- From the Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B47A, Bethesda, MD 20892 (L.L., E.M., B.T., I.L., F.L., A.T., Y.L.M., P.E., P.L.C.); Division of Cancer Treatment and Diagnosis: Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, Md (J.H.S.); National Cancer Institute Biometrics Research Program Contract, General Dynamics Information Technology, Falls Church, Va (S.E.R.); Clinical Research Directorate, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Bethesda, Md (S.A.H.); Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md (D.E.C.); Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (W.D., R.M.); Center of Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Md (B.J.W., V.K., R.C., E.L.); Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (P.P.); and Cancer Imaging Program, National Cancer Institute, Bethesda, Md (J.F.E.)
| | - William Dahut
- From the Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B47A, Bethesda, MD 20892 (L.L., E.M., B.T., I.L., F.L., A.T., Y.L.M., P.E., P.L.C.); Division of Cancer Treatment and Diagnosis: Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, Md (J.H.S.); National Cancer Institute Biometrics Research Program Contract, General Dynamics Information Technology, Falls Church, Va (S.E.R.); Clinical Research Directorate, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Bethesda, Md (S.A.H.); Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md (D.E.C.); Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (W.D., R.M.); Center of Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Md (B.J.W., V.K., R.C., E.L.); Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (P.P.); and Cancer Imaging Program, National Cancer Institute, Bethesda, Md (J.F.E.)
| | - Ravi Madan
- From the Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B47A, Bethesda, MD 20892 (L.L., E.M., B.T., I.L., F.L., A.T., Y.L.M., P.E., P.L.C.); Division of Cancer Treatment and Diagnosis: Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, Md (J.H.S.); National Cancer Institute Biometrics Research Program Contract, General Dynamics Information Technology, Falls Church, Va (S.E.R.); Clinical Research Directorate, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Bethesda, Md (S.A.H.); Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md (D.E.C.); Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (W.D., R.M.); Center of Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Md (B.J.W., V.K., R.C., E.L.); Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (P.P.); and Cancer Imaging Program, National Cancer Institute, Bethesda, Md (J.F.E.)
| | - Bradford J. Wood
- From the Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B47A, Bethesda, MD 20892 (L.L., E.M., B.T., I.L., F.L., A.T., Y.L.M., P.E., P.L.C.); Division of Cancer Treatment and Diagnosis: Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, Md (J.H.S.); National Cancer Institute Biometrics Research Program Contract, General Dynamics Information Technology, Falls Church, Va (S.E.R.); Clinical Research Directorate, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Bethesda, Md (S.A.H.); Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md (D.E.C.); Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (W.D., R.M.); Center of Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Md (B.J.W., V.K., R.C., E.L.); Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (P.P.); and Cancer Imaging Program, National Cancer Institute, Bethesda, Md (J.F.E.)
| | - Venkatesh Krishnasamy
- From the Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B47A, Bethesda, MD 20892 (L.L., E.M., B.T., I.L., F.L., A.T., Y.L.M., P.E., P.L.C.); Division of Cancer Treatment and Diagnosis: Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, Md (J.H.S.); National Cancer Institute Biometrics Research Program Contract, General Dynamics Information Technology, Falls Church, Va (S.E.R.); Clinical Research Directorate, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Bethesda, Md (S.A.H.); Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md (D.E.C.); Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (W.D., R.M.); Center of Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Md (B.J.W., V.K., R.C., E.L.); Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (P.P.); and Cancer Imaging Program, National Cancer Institute, Bethesda, Md (J.F.E.)
| | - Richard Chang
- From the Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B47A, Bethesda, MD 20892 (L.L., E.M., B.T., I.L., F.L., A.T., Y.L.M., P.E., P.L.C.); Division of Cancer Treatment and Diagnosis: Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, Md (J.H.S.); National Cancer Institute Biometrics Research Program Contract, General Dynamics Information Technology, Falls Church, Va (S.E.R.); Clinical Research Directorate, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Bethesda, Md (S.A.H.); Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md (D.E.C.); Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (W.D., R.M.); Center of Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Md (B.J.W., V.K., R.C., E.L.); Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (P.P.); and Cancer Imaging Program, National Cancer Institute, Bethesda, Md (J.F.E.)
| | - Elliot Levy
- From the Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B47A, Bethesda, MD 20892 (L.L., E.M., B.T., I.L., F.L., A.T., Y.L.M., P.E., P.L.C.); Division of Cancer Treatment and Diagnosis: Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, Md (J.H.S.); National Cancer Institute Biometrics Research Program Contract, General Dynamics Information Technology, Falls Church, Va (S.E.R.); Clinical Research Directorate, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Bethesda, Md (S.A.H.); Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md (D.E.C.); Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (W.D., R.M.); Center of Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Md (B.J.W., V.K., R.C., E.L.); Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (P.P.); and Cancer Imaging Program, National Cancer Institute, Bethesda, Md (J.F.E.)
| | - Peter Pinto
- From the Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B47A, Bethesda, MD 20892 (L.L., E.M., B.T., I.L., F.L., A.T., Y.L.M., P.E., P.L.C.); Division of Cancer Treatment and Diagnosis: Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, Md (J.H.S.); National Cancer Institute Biometrics Research Program Contract, General Dynamics Information Technology, Falls Church, Va (S.E.R.); Clinical Research Directorate, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Bethesda, Md (S.A.H.); Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md (D.E.C.); Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (W.D., R.M.); Center of Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Md (B.J.W., V.K., R.C., E.L.); Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (P.P.); and Cancer Imaging Program, National Cancer Institute, Bethesda, Md (J.F.E.)
| | - Janet F. Eary
- From the Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B47A, Bethesda, MD 20892 (L.L., E.M., B.T., I.L., F.L., A.T., Y.L.M., P.E., P.L.C.); Division of Cancer Treatment and Diagnosis: Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, Md (J.H.S.); National Cancer Institute Biometrics Research Program Contract, General Dynamics Information Technology, Falls Church, Va (S.E.R.); Clinical Research Directorate, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Bethesda, Md (S.A.H.); Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md (D.E.C.); Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (W.D., R.M.); Center of Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Md (B.J.W., V.K., R.C., E.L.); Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (P.P.); and Cancer Imaging Program, National Cancer Institute, Bethesda, Md (J.F.E.)
| | - Peter L. Choyke
- From the Molecular Imaging Program, National Cancer Institute, Building 10, Room B3B47A, Bethesda, MD 20892 (L.L., E.M., B.T., I.L., F.L., A.T., Y.L.M., P.E., P.L.C.); Division of Cancer Treatment and Diagnosis: Biometric Research Program, National Cancer Institute, National Institutes of Health, Bethesda, Md (J.H.S.); National Cancer Institute Biometrics Research Program Contract, General Dynamics Information Technology, Falls Church, Va (S.E.R.); Clinical Research Directorate, Frederick National Laboratory for Cancer Research Sponsored by the National Cancer Institute, Bethesda, Md (S.A.H.); Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Md (D.E.C.); Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (W.D., R.M.); Center of Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Md (B.J.W., V.K., R.C., E.L.); Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Md (P.P.); and Cancer Imaging Program, National Cancer Institute, Bethesda, Md (J.F.E.)
| |
Collapse
|
43
|
Donswijk ML, van Leeuwen PJ, Vegt E, Cheung Z, Heijmink SWTPJ, van der Poel HG, Stokkel MPM. Clinical impact of PSMA PET/CT in primary prostate cancer compared to conventional nodal and distant staging: a retrospective single center study. BMC Cancer 2020; 20:723. [PMID: 32758168 PMCID: PMC7409439 DOI: 10.1186/s12885-020-07192-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 07/17/2020] [Indexed: 01/01/2023] Open
Abstract
Background To evaluate the impact of Gallium-68 [68Ga] labeled prostate specific membrane antigen (PSMA) positron emission tomography (PET)/X-ray computed tomography (CT) compared with conventional imaging on staging and clinical management of men evaluated for primary prostate cancer (PCa). Methods Men with newly diagnosed biopsy-proven PCa who had been staged with a conventional staging protocol including bone scintigraphy (BS) and additionally underwent [68Ga]PSMA PET/CT, were evaluated retrospectively. Imaging findings from BS, magnetic resonance imaging (MRI) and/or CT were categorized regarding locoregional nodal (N) and distant metastasis (M) status as negative, positive or equivocal before and after addition of the information of PET/CT. Also, the imaging-based level of confidence (LoC) in correct assessment of N and M status was scored. Impact of PET/CT on clinical management was evaluated by the percentage of treatment category changes after PET/CT as determined in the multidisciplinary tumour board. Results Sixty-four men with intermediate and high-risk PCa were evaluated. With additional information of PET/CT, N status was upstaged in 23%, and downstaged in 9%. M status was upstaged in 13%, and downstaged in 23%. A net increase in LoC of 20% was noted, mainly regarding M status. Treatment category changed from palliative to curative in 9%, and from curative to palliative in 3%. An undecided treatment plan changed to curative in 14%, as well as to palliative in another 9%. In total, a 36% treatment category change was noted. High negative predictive value of PET/CT for M status was indicated by 27 patients that underwent robot-assisted radical prostatectomy and reached postoperative biochemical disease-free status or had a likely other site of disease recurrence. Conclusions PSMA PET/CT can cause considerable changes in N and M staging, as well as in management compared to conventional staging. Findings of this study support the replacement of BS and CT by PSMA PET/CT in staging primary PCa.
Collapse
Affiliation(s)
- Maarten L Donswijk
- Department of Nuclear Medicine, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, the Netherlands, 1066, CX.
| | - Pim J van Leeuwen
- Department of Urology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Erik Vegt
- Department of Nuclear Medicine, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, the Netherlands, 1066, CX.,Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, Netherlands
| | - Zing Cheung
- Department of Nuclear Medicine, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, the Netherlands, 1066, CX
| | | | - Henk G van der Poel
- Department of Urology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Marcel P M Stokkel
- Department of Nuclear Medicine, The Netherlands Cancer Institute, Plesmanlaan 121, Amsterdam, the Netherlands, 1066, CX
| |
Collapse
|
44
|
Pfister D, Haidl F, Nestler T, Verburg F, Schmidt M, Wittersheim M, Steib F, Heidenreich A. 68 Ga-PSMA-PET/CT helps to select patients for salvage radical prostatectomy with local recurrence after primary radiotherapy for prostate cancer. BJU Int 2020; 126:679-683. [PMID: 32531840 DOI: 10.1111/bju.15135] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2020] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To investigate the diagnostic performance of gallium-68 prostate-specific membrane antigen positron emission tomography/computed tomography (68 Ga-PSMA PET/CT) in patients with recurrent prostate cancer with regard to the presence of lymph node metastases (LNM) and local recurrences after primary radiotherapy. PATIENTS AND METHODS We retrospectively reviewed 142 patients following salvage radical prostatectomy (sRP), 50 of which had a 68 Ga-PSMA PET/CT performed as a preoperative staging module. Predictive clinical parameters were analysed in a multivariate Cox regression analysis. Sensitivity, specificity, positive (PPV) and negative predictive values (NPV) and the accuracy of 68 Ga-PSMA PET/CT were analysed with regard to LNM and local recurrence. RESULTS In all, 613 lymph nodes were resected in 40 patients and 23 lymph nodes had metastatic deposits in 14 patients. In all patients local recurrence could have been found with 68 Ga-PSMA PET/CT. Sensitivity, specificity, PPV and NPV and accuracy on a per lymph node basis were 34.78% (16.38-57.2%), 100% (99.38-100%), 100%, 97.52% (96.69-98.15%) and 97.55% (96.00-98.62%). For detecting local recurrence, the sensitivity and PPV were both 100% with an accuracy of 100% (92.89-100%). CONCLUSION 68 Ga-PSMA PET/CT should be the standard imaging in biochemical recurrent prostate cancer. With this imaging module one detects first local recurrence and can detect locoregional and distant metastases more precisely than standard CT and bone scan.
Collapse
Affiliation(s)
- David Pfister
- Department of Urology, Uro-Oncology and Robot Assisted Surgery, University Hospital of Cologne, Cologne, Germany
| | - Friederike Haidl
- Department of Urology, Uro-Oncology and Robot Assisted Surgery, University Hospital of Cologne, Cologne, Germany
| | - Tim Nestler
- Department of Urology, Uro-Oncology and Robot Assisted Surgery, University Hospital of Cologne, Cologne, Germany
| | - Frederik Verburg
- Department of Radiation and Nuclear Medicine, ErasmusMC, Rotterdam, Netherlands
| | - Matthias Schmidt
- Department of Nuclear Medicine, University of Cologne, Köln, Germany
| | - Maike Wittersheim
- Institute for Pathology, University Hospital of Cologne, Cologne, Germany
| | - Florain Steib
- Institute of Pathologie, University Hospital of Aachen, Aachen, Germany
| | - Axel Heidenreich
- Department of Urology, Uro-Oncology and Robot Assisted Surgery, University Hospital of Cologne, Cologne, Germany
| |
Collapse
|
45
|
Abstract
Prostate cancer is the commonest malignancy to affect men in the United Kingdom. Extraprostatic disease detection at staging and in the setting of biochemical recurrence is essential in determining treatment strategy. Conventional imaging including computed tomography and bone scintigraphy are limited in their ability to detect sites of loco-regional nodal and metastatic bone disease, particularly at clinically relevant low prostate-specific antigen levels. The use of positron emission tomography-computed tomography has helped overcome these deficiencies and is leading a paradigm shift in the management of prostate cancer using a wide range of radiopharmaceuticals. Their mechanisms of action, utility in both staging and biochemical recurrence, and comparative strengths and weaknesses will be covered in this article.
Collapse
Affiliation(s)
- Manil Subesinghe
- King's College London & Guy's & St. Thomas' PET Centre, St. Thomas' Hospital, London, UK; Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK.
| | - Meghana Kulkarni
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Gary J Cook
- King's College London & Guy's & St. Thomas' PET Centre, St. Thomas' Hospital, London, UK; Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| |
Collapse
|
46
|
|
47
|
Mena E, Black PC, Rais-Bahrami S, Gorin M, Allaf M, Choyke P. Novel PET imaging methods for prostate cancer. World J Urol 2020; 39:687-699. [PMID: 32671604 DOI: 10.1007/s00345-020-03344-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/02/2020] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Prostate cancer is a common neoplasm but conventional imaging methods such as CT and bone scan are often insensitive. A new class of PET agents have emerged to diagnose and manage prostate cancer. METHODS The relevant literature on PET imaging agents for prostate cancer was reviewed. RESULTS This review shows a broad range of PET imaging agents, the most successful of which is prostate specific membrane antigen (PSMA) PET. Other agents either lack the sensitivity or specificity of PSMA PET. CONCLUSION Among the available PET agents for prostate cancer, PSMA PET has emerged as the leader. It is likely to have great impact on the diagnosis, staging and management of prostate cancer patients.
Collapse
Affiliation(s)
- Esther Mena
- Molecular Imaging Program, National Cancer Institute, 10 Center Dr, Bldg 10, Room B3B69F, Bethesda, MD, 20892-1088, USA
| | - Peter C Black
- University of British Columbia, Vancouver, BC, Canada
| | | | - Michael Gorin
- Department of Urology, Johns Hopkins University, Baltimore, MD, USA
| | - Mohamad Allaf
- Department of Urology, Johns Hopkins University, Baltimore, MD, USA
| | - Peter Choyke
- Molecular Imaging Program, National Cancer Institute, 10 Center Dr, Bldg 10, Room B3B69F, Bethesda, MD, 20892-1088, USA.
| |
Collapse
|
48
|
Devos G, Witters M, Moris L, Van den Broeck T, Berghen C, Devlies W, De Meerleer G, Goffin K, Jentjens S, Albersen M, Van Poppel H, Everaerts W, Joniau S. Site-specific relapse patterns of patients with biochemical recurrence following radical prostatectomy assessed by 68Ga-PSMA-11 PET/CT or 11C-Choline PET/CT: impact of postoperative treatments. World J Urol 2020; 39:399-406. [PMID: 32417995 DOI: 10.1007/s00345-020-03220-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 04/21/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Salvage radiotherapy (RT) (± androgen deprivation therapy (ADT)) is often used as a treatment in patients with biochemical recurrence (BCR) following radical prostatectomy (RP). Unfortunately, even after RT ± ADT, a significant number of patients will develop 'second' BCR. The aim of this study was to investigate the impact of postoperative treatments (adjuvant/salvage radiotherapy (RT) ± androgen deprivation therapy) on the recurrence pattern in patients with BCR following RP assessed by 11C-Choline PET/CT or 68 Ga-PSMA PET/CT. METHODS Patients who developed BCR following RP and who had at least one positive lesion on PET/CT were retrospectively assessed. Positive spots were mapped as local, lymph node (LN), skeletal or visceral recurrence. A distinction was made between locoregional (prostate bed and pelvic LN) and extrapelvic recurrence (skeletal, visceral and/or extrapelvic LN). Patients were categorized according to postoperative treatment received in three subgroups (RT, ADT and RT + ADT) and compared with the reference group (RP only). The impact of the radiation field was also investigated. RESULTS We identified 200 patients assessed by 68Ga-PSMA-11 (80%) or 11C-Choline PET/CT (20%). Patients who received postoperative RT + ADT had less LN recurrence distal to the common iliac bifurcation (26.7% vs 66.6%; p = 0.0004), but more recurrence to retroperitoneal LN than the reference group (38% vs. 14.4%, p = 0.02). Moreover, the RT + ADT subgroup had more extrapelvic recurrence compared to the reference group (66.2% vs 40.8%, p = 0.02). Patients who received RT to the prostate bed had more recurrence distal to the common iliac bifurcation compared to those who received RT to the prostate bed + pelvic LN (51.6% vs 26.1%, p = 0.0069). CONCLUSION Post-prostatectomy treatments (ADT and/or RT) and the postoperative radiation field (prostate bed vs. prostate bed + pelvis) have a significant impact on the recurrence pattern. This knowledge can help clinicians to counsel their patients on their chances of being eligible for (locoregional) metastasis-directed therapies.
Collapse
Affiliation(s)
- Gaëtan Devos
- Department of Urology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium.
| | - Manuel Witters
- Department of Urology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Lisa Moris
- Department of Urology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium.,Laboratory of Molecular Endocrinology, KU Leuven, Leuven, Belgium
| | - Thomas Van den Broeck
- Department of Urology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium.,Laboratory of Molecular Endocrinology, KU Leuven, Leuven, Belgium
| | - Charlien Berghen
- Departement of Radiation Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Wout Devlies
- Department of Urology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium.,Laboratory of Molecular Endocrinology, KU Leuven, Leuven, Belgium
| | - Gert De Meerleer
- Departement of Radiation Oncology, University Hospitals Leuven, Leuven, Belgium
| | - Karolien Goffin
- Departement of Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Sander Jentjens
- Departement of Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Maarten Albersen
- Department of Urology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Hendrik Van Poppel
- Department of Urology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Wouter Everaerts
- Department of Urology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Steven Joniau
- Department of Urology, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium
| |
Collapse
|
49
|
Chodyla MK, Eiber M, Wetter A, Rauscher I. Hybridbildgebung beim Prostatakarzinom. Radiologe 2020; 60:386-393. [DOI: 10.1007/s00117-020-00642-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
50
|
Jokar N, Assadi M, Yordanova A, Ahmadzadehfar H. Bench-to-Bedside Theranostics in Nuclear Medicine. Curr Pharm Des 2020; 26:3804-3811. [PMID: 32067609 DOI: 10.2174/1381612826666200218104313] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 12/11/2019] [Indexed: 11/22/2022]
Abstract
The optimum selection of the appropriate radiolabelled probe for the right target and the right patient is the foundation of theranostics in personalised medicine. In nuclear medicine, this process is realised through the appropriate choice of radiopharmaceuticals based on molecular biomarkers regarding molecular imaging. Theranostics is developing a strategy that can be used to implement accepted tools for individual molecular targeting, including diagnostics, and advances in genomic molecular knowledge, which has led to identifying theranostics biomaterials that have the potency to diagnose and treat malignancies. Today, numerous studies have reported on the discovery and execution of these radiotracers in personalised medicine. In this review, we presented our point of view of the most important theranostics agents that can be used to treat several types of malignancies. Molecular targeted radionuclide treatment methods based on theranostics are excellent paradigms of the relationship between molecular imaging and therapy that has been used to provide individualised or personalised patient care. Toward that end, a precise planned prospective examination of theranostics must be done to compare this approach to more standard therapies.
Collapse
Affiliation(s)
- Narges Jokar
- The Persian Gulf Nuclear Medicine Research Center, Department of Molecular Imaging and Radionuclide Therapy (MIRT), Bushehr Medical University Hospital, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Majid Assadi
- The Persian Gulf Nuclear Medicine Research Center, Department of Molecular Imaging and Radionuclide Therapy (MIRT), Bushehr Medical University Hospital, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Anna Yordanova
- Department of Nuclear Medicine, University Hospital Bonn, Bonn, Germany
| | | |
Collapse
|