151
|
Yadav D, Hwang H, Qiao W, Upadhyay R, Chapin BF, Tang C, Aparicio A, Lopez-Olivo MA, Kang SK, Macapinlac HA, Bathala TK, Surasi DS. 18F-Fluciclovine versus PSMA PET Imaging in Primary Tumor Detection during Initial Staging of High-Risk Prostate Cancer: A Systematic Review and Meta-Analysis. Radiol Imaging Cancer 2022; 4:e210091. [PMID: 35212559 PMCID: PMC8965534 DOI: 10.1148/rycan.210091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/28/2021] [Accepted: 01/20/2022] [Indexed: 06/14/2023]
Abstract
Purpose Fluorine 18 (18F)-fluciclovine and prostate-specific membrane antigen (PSMA) tracers are commonly used for localizing biochemical recurrence of prostate cancer, but their accuracy in primary tumor detection in the initial staging of high-risk prostate cancer has not been established. Materials and Methods A systematic review was performed of the electronic databases for original studies published between 2012 and 2020. Included studies were those in which 18F-fluciclovine or PSMA PET was used for initial staging of patients with high-risk prostate cancer. The diagnostic performance data were collected for primary tumor with histopathologic results as reference standard. The Quality Assessment of Diagnostic Accuracy Studies-2 tool was used for quality appraisal. A random-effects model was used to summarize the effect sizes and to evaluate the difference between two groups. Results Overall, 28 studies met the eligibility criteria, and 17 were included in the meta-analysis (18F-fluciclovine = 4, PSMA = 13). Of these 17 studies, 12 (70%) were judged to have high risk of bias in one of the evaluated domains, and nine studies were deemed to have applicability concerns. The pooled sensitivity, specificity, and diagnostic odds ratio for 18F-fluciclovine versus PSMA were 85% (95% CI: 73%, 92%) versus 84% (95% CI: 77%, 89%) (P = .78), 77% (95% CI: 60%, 88%) versus 83% (95% CI: 76%, 89%) (P = .40), and 18.88 (95% CI: 5.01, 71.20) versus 29.37 (95% CI: 13.35, 64.60) (P = .57), respectively, with no significant difference in diagnostic test accuracy. Conclusion 18F-fluciclovine and PSMA PET demonstrated no statistically significant difference in diagnostic accuracy in primary tumor detection during initial staging of high-risk prostate cancer. Keywords: PET, Prostate, Molecular Imaging-Cancer, Staging Supplemental material is available for this article. © RSNA, 2022.
Collapse
Affiliation(s)
- Divya Yadav
- From the Departments of Radiation Oncology (D.Y., R.U., C.T.),
Biostatistics (H.H., W.Q.), Urology (B.F.C.), Genitourinary Medical Oncology
(A.A.), Health Services Research (M.A.L.O.), Nuclear Medicine (H.A.M., D.S.S.),
and Abdominal Imaging (T.K.B.), The University of Texas MD Anderson Cancer
Center, 1515 Holcombe Blvd, Unit 483, Houston, TX 77030; and Department of
Radiology, NYU Langone Health, New York, NY (S.K.K.)
| | - Hyunsoo Hwang
- From the Departments of Radiation Oncology (D.Y., R.U., C.T.),
Biostatistics (H.H., W.Q.), Urology (B.F.C.), Genitourinary Medical Oncology
(A.A.), Health Services Research (M.A.L.O.), Nuclear Medicine (H.A.M., D.S.S.),
and Abdominal Imaging (T.K.B.), The University of Texas MD Anderson Cancer
Center, 1515 Holcombe Blvd, Unit 483, Houston, TX 77030; and Department of
Radiology, NYU Langone Health, New York, NY (S.K.K.)
| | - Wei Qiao
- From the Departments of Radiation Oncology (D.Y., R.U., C.T.),
Biostatistics (H.H., W.Q.), Urology (B.F.C.), Genitourinary Medical Oncology
(A.A.), Health Services Research (M.A.L.O.), Nuclear Medicine (H.A.M., D.S.S.),
and Abdominal Imaging (T.K.B.), The University of Texas MD Anderson Cancer
Center, 1515 Holcombe Blvd, Unit 483, Houston, TX 77030; and Department of
Radiology, NYU Langone Health, New York, NY (S.K.K.)
| | - Rituraj Upadhyay
- From the Departments of Radiation Oncology (D.Y., R.U., C.T.),
Biostatistics (H.H., W.Q.), Urology (B.F.C.), Genitourinary Medical Oncology
(A.A.), Health Services Research (M.A.L.O.), Nuclear Medicine (H.A.M., D.S.S.),
and Abdominal Imaging (T.K.B.), The University of Texas MD Anderson Cancer
Center, 1515 Holcombe Blvd, Unit 483, Houston, TX 77030; and Department of
Radiology, NYU Langone Health, New York, NY (S.K.K.)
| | - Brian F. Chapin
- From the Departments of Radiation Oncology (D.Y., R.U., C.T.),
Biostatistics (H.H., W.Q.), Urology (B.F.C.), Genitourinary Medical Oncology
(A.A.), Health Services Research (M.A.L.O.), Nuclear Medicine (H.A.M., D.S.S.),
and Abdominal Imaging (T.K.B.), The University of Texas MD Anderson Cancer
Center, 1515 Holcombe Blvd, Unit 483, Houston, TX 77030; and Department of
Radiology, NYU Langone Health, New York, NY (S.K.K.)
| | - Chad Tang
- From the Departments of Radiation Oncology (D.Y., R.U., C.T.),
Biostatistics (H.H., W.Q.), Urology (B.F.C.), Genitourinary Medical Oncology
(A.A.), Health Services Research (M.A.L.O.), Nuclear Medicine (H.A.M., D.S.S.),
and Abdominal Imaging (T.K.B.), The University of Texas MD Anderson Cancer
Center, 1515 Holcombe Blvd, Unit 483, Houston, TX 77030; and Department of
Radiology, NYU Langone Health, New York, NY (S.K.K.)
| | - Ana Aparicio
- From the Departments of Radiation Oncology (D.Y., R.U., C.T.),
Biostatistics (H.H., W.Q.), Urology (B.F.C.), Genitourinary Medical Oncology
(A.A.), Health Services Research (M.A.L.O.), Nuclear Medicine (H.A.M., D.S.S.),
and Abdominal Imaging (T.K.B.), The University of Texas MD Anderson Cancer
Center, 1515 Holcombe Blvd, Unit 483, Houston, TX 77030; and Department of
Radiology, NYU Langone Health, New York, NY (S.K.K.)
| | - Maria A. Lopez-Olivo
- From the Departments of Radiation Oncology (D.Y., R.U., C.T.),
Biostatistics (H.H., W.Q.), Urology (B.F.C.), Genitourinary Medical Oncology
(A.A.), Health Services Research (M.A.L.O.), Nuclear Medicine (H.A.M., D.S.S.),
and Abdominal Imaging (T.K.B.), The University of Texas MD Anderson Cancer
Center, 1515 Holcombe Blvd, Unit 483, Houston, TX 77030; and Department of
Radiology, NYU Langone Health, New York, NY (S.K.K.)
| | - Stella K. Kang
- From the Departments of Radiation Oncology (D.Y., R.U., C.T.),
Biostatistics (H.H., W.Q.), Urology (B.F.C.), Genitourinary Medical Oncology
(A.A.), Health Services Research (M.A.L.O.), Nuclear Medicine (H.A.M., D.S.S.),
and Abdominal Imaging (T.K.B.), The University of Texas MD Anderson Cancer
Center, 1515 Holcombe Blvd, Unit 483, Houston, TX 77030; and Department of
Radiology, NYU Langone Health, New York, NY (S.K.K.)
| | - Homer A. Macapinlac
- From the Departments of Radiation Oncology (D.Y., R.U., C.T.),
Biostatistics (H.H., W.Q.), Urology (B.F.C.), Genitourinary Medical Oncology
(A.A.), Health Services Research (M.A.L.O.), Nuclear Medicine (H.A.M., D.S.S.),
and Abdominal Imaging (T.K.B.), The University of Texas MD Anderson Cancer
Center, 1515 Holcombe Blvd, Unit 483, Houston, TX 77030; and Department of
Radiology, NYU Langone Health, New York, NY (S.K.K.)
| | - Tharakeswara K. Bathala
- From the Departments of Radiation Oncology (D.Y., R.U., C.T.),
Biostatistics (H.H., W.Q.), Urology (B.F.C.), Genitourinary Medical Oncology
(A.A.), Health Services Research (M.A.L.O.), Nuclear Medicine (H.A.M., D.S.S.),
and Abdominal Imaging (T.K.B.), The University of Texas MD Anderson Cancer
Center, 1515 Holcombe Blvd, Unit 483, Houston, TX 77030; and Department of
Radiology, NYU Langone Health, New York, NY (S.K.K.)
| | - Devaki Shilpa Surasi
- From the Departments of Radiation Oncology (D.Y., R.U., C.T.),
Biostatistics (H.H., W.Q.), Urology (B.F.C.), Genitourinary Medical Oncology
(A.A.), Health Services Research (M.A.L.O.), Nuclear Medicine (H.A.M., D.S.S.),
and Abdominal Imaging (T.K.B.), The University of Texas MD Anderson Cancer
Center, 1515 Holcombe Blvd, Unit 483, Houston, TX 77030; and Department of
Radiology, NYU Langone Health, New York, NY (S.K.K.)
| |
Collapse
|
152
|
Rodríguez-Fraile M, Tamayo Alonso P, Rosales Castillo JJ, de Arcocha-Torres M, Caresia-Aróztegui A, Puig Cózar-Santiago M, Orcajo-Rincon J, Simó Perdigó M, Delgado Bolton RC, Artigas Guix C. Utilidad de los radioligandos PSMA en el diagnóstico y tratamiento del carcinoma de próstata. Rev Esp Med Nucl Imagen Mol 2022. [DOI: 10.1016/j.remn.2021.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
153
|
Regula N, Kostaras V, Johansson S, Trampal C, Lindström E, Lubberink M, Iyer V, Velikyan I, Sörensen J. Comparison of 68Ga-PSMA PET/CT with fluoride PET/CT for detection of bone metastatic disease in prostate cancer. Eur J Hybrid Imaging 2022; 6:5. [PMID: 35229224 PMCID: PMC8885936 DOI: 10.1186/s41824-022-00127-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 02/01/2022] [Indexed: 12/12/2022] Open
Abstract
Background 18F-NaF positron emission tomography/computed tomography (fluoride PET/CT) is considered the most sensitive technique to detect bone metastasis in prostate cancer (PCa). 68Ga-PSMA-11 (PSMA) PET/CT is increasingly used for staging of PCa. This study primarily aimed to compare the diagnostic performance of fluoride PET/CT and gallium-based PSMA PET/CT in identifying bone metastasis followed by a comparison of PSMA PET/CT with contrast-enhanced CT (CE-CT) in identifying soft tissue lesions as a secondary objective. Methods Twenty-eight PCa patients with high suspicion of disseminated disease following curative treatment were prospectively evaluated. PET/CT examinations using fluoride and PSMA were performed. All suspicious bone lesions were counted, and the tracer uptake was measured as standardized uptake values (SUV) for both tracers. In patients with multiple findings, ten bone lesions with highest SUVmax were selected from which identical lesions from both scans were considered for direct comparison of SUVmax. Soft tissue findings of local and lymph node lesions from CE-CT were compared with PSMA PET/CT. Results Both scans were negative for bone lesions in 7 patients (25%). Of 699 lesions consistent with skeletal metastasis in 21 patients on fluoride PET/CT, PSMA PET/CT identified 579 lesions (83%). In 69 identical bone lesions fluoride PET/CT showed significantly higher uptake (mean SUVmax: 73.1 ± 36.8) compared to PSMA PET/CT (34.5 ± 31.4; p < 0.001). Compared to CE-CT, PSMA PET/CT showed better diagnostic performance in locating local (96% vs 61%, p = 0.004) and lymph node (94% vs 46%, p < 0.001) metastasis. Conclusion In this prospective comparative study, PSMA PET/CT detected the majority of bone lesions that were positive on fluoride PET/CT. Further, this study indicates better diagnostic performance of PSMA PET/CT to locate soft tissue lesions compared to CE-CT. Supplementary Information The online version contains supplementary material available at 10.1186/s41824-022-00127-4.
Collapse
|
154
|
Ye S, Li H, Hu K, Li L, Zhong J, Yan Q, Wang Q. Radiosynthesis and biological evaluation of 18F-labeled bispecific heterodimer targeted dual gastrin-releasing peptide receptor and prostate-specific membrane antigen for prostate cancer imaging. Nucl Med Commun 2022; 43:323-331. [PMID: 34919064 DOI: 10.1097/mnm.0000000000001520] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Approximately 5% of prostatic primary tumors and 15% of metastatic tumors were found to be prostate-specific membrane antigen (PSMA)-negative. Targeting gastrin-releasing peptide receptor (GRPR) has been shown to complement patients with PSMA-negative prostate cancer (PCa). Based on previous findings, simultaneously targeting PSMA and GRPR imaging may improve the diagnosis of PCa. In this study, we report the radiosynthesis and biological evaluation of a bispecific heterodimer of NOTA-GRPR-PSMA that targeted both PSMA and GRPR for extended PCa imaging. METHODS NOTA-GRPR-PSMA was labeled using the Al18F-chelating one-step method. The competitive combination experiment and specific binding assay were performed in vitro using 22Rv1 (PSMA+) and PC-3 (GRPR+) cells. To determine the distribution and specificity in vivo, biodistribution and micro-PET/computed tomography of [18F]AlF-GRPR-PSMA were performed on mice bearing 22Rv1 or PC-3 tumors. RESULTS [18F]AlF-GRPR-PSMA had a radiochemical purity of over 98% and demonstrated high stability in vivo and in vitro, with a LogD of -1.2 ± 0.05. Cell uptake and inhibition studies of [18F]AlF-GRPR-PSMA in 22Rv1 and PC-3 cells revealed bispecific GRPR and PSMA bindings. According to the biodistribution study and PET imaging, [18F]AlF-GRPR-PSMA was mainly excreted through the kidney. Tumor uptake was high in 22Rv1 tumor (10.1 ± 0.4 %ID/g) and moderate in PC-3 tumor (2.1 ± 0.6 %ID/g) 2 h p.i., whereas blocking studies significantly decreased the tumor uptake of 22Rv1 and PC-3. CONCLUSION [18F]AlF-GRPR-PSMA has the potential to simultaneously target PSMA and GRPR for PCa imaging.
Collapse
Affiliation(s)
- Shimin Ye
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou
| | - Hongsheng Li
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou
| | - Kongzhen Hu
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou
| | - Li Li
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou
| | - Jiawei Zhong
- Department of Chemistry, Jinan University, Guangzhou, China
| | - Qingsong Yan
- Department of Chemistry, Jinan University, Guangzhou, China
| | - Quanshi Wang
- Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou
| |
Collapse
|
155
|
Ceci F, Rovera G, Iorio GC, Guarneri A, Chiofalo V, Passera R, Oderda M, Dall'Armellina S, Liberini V, Grimaldi S, Bellò M, Gontero P, Ricardi U, Deandreis D. Event-free survival after 68 Ga-PSMA-11 PET/CT in recurrent hormone-sensitive prostate cancer (HSPC) patients eligible for salvage therapy. Eur J Nucl Med Mol Imaging 2022; 49:3257-3268. [PMID: 35217883 PMCID: PMC9250462 DOI: 10.1007/s00259-022-05741-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 02/17/2022] [Indexed: 11/30/2022]
Abstract
Background/aim Prostate-specific-membrane-antigen/positron emission tomography (PSMA-PET) detects with high accuracy disease-recurrence, leading to changes in the management of biochemically-recurrent (BCR) prostate cancer (PCa). However, data regarding the oncological outcomes of patients who performed PSMA-PET are needed. The aim of this study was to evaluate the incidence of clinically relevant events during follow-up in patients who performed PSMA-PET for BCR after radical treatment. Materials and methods This analysis included consecutive, hormone-sensitive, hormone-free, recurrent PCa patients (HSPC) enrolled through a prospective study. All patients were eligible for salvage therapy, having at least 24 months of follow-up after PSMA-PET. The primary endpoint was the Event-Free Survival (EFS), defined as the time between the PSMA-PET and the date of event/last follow-up. The Kaplan–Meier method was used to estimate the EFS curves. EFS was also investigated by Cox proportional hazards regression. Events were defined as death, radiological progression, or PSA recurrence after therapy. Results One-hundred and seventy-six (n = 176) patients were analyzed (median PSA 0.62 [IQR: 0.43–1.00] ng/mL; median follow-up of 35.4 [IQR: 26.5–40.3] months). The EFS was 78.8% at 1 year, 65.2% (2 years), and 52.2% (3 years). Patients experiencing events during study follow-up had a significantly higher median PSA (0.81 [IQR: 0.53–1.28] vs 0.51 [IQR: 0.36–0.80] ng/mL) and a lower PSA doubling time (PSAdt) (5.4 [IQR: 3.7–11.6] vs 12.7 [IQR: 6.6–24.3] months) (p < 0.001) compared to event-free patients. The Kaplan–Meier curves showed that PSA > 0.5 ng/mL, PSAdt ≤ 6 months, and a positive PSMA-PET result were associated with a higher event rate (p < 0.01). No significant differences of event rates were observed in patients who received changes in therapy management after PSMA-PET vs. patients who did not receive therapy changes. Finally, PSA > 0.5 ng/mL and PSAdt ≤ 6 months were statistically significant event-predictors in multivariate model (p < 0.001). Conclusion Low PSA and long PSAdt were significant predictors of longer EFS. A lower incidence of events was observed in patients having negative PSMA-PET, since longer EFS was significantly more probable in case of a negative scan. Supplementary Information The online version contains supplementary material available at 10.1007/s00259-022-05741-9.
Collapse
Affiliation(s)
- Francesco Ceci
- Nuclear Medicine, Department of Medical Sciences, AOU Città Della Salute E Della Scienza Di Torino, University of Turin, Turin, Italy. .,Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS, Milan, Italy. .,Department of Oncology and Hemato-Oncolology, University of Milan, Milan, Italy. .,Nuclear Medicine Department, S. Croce E Carle Hospital, Cuneo, Italy.
| | - Guido Rovera
- Nuclear Medicine, Department of Medical Sciences, AOU Città Della Salute E Della Scienza Di Torino, University of Turin, Turin, Italy.,Section of Nuclear Medicine, University Department of Radiological Sciences and Haematology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giuseppe Carlo Iorio
- Radiation Oncology, Department of Oncology, AOU Città Della Salute E Della Scienza Di Torino, Turin, Italy
| | - Alessia Guarneri
- Radiation Oncology, Department of Oncology, AOU Città Della Salute E Della Scienza Di Torino, Turin, Italy
| | - Valeria Chiofalo
- Radiation Oncology, Department of Oncology, AOU Città Della Salute E Della Scienza Di Torino, Turin, Italy
| | - Roberto Passera
- Nuclear Medicine, Department of Medical Sciences, AOU Città Della Salute E Della Scienza Di Torino, University of Turin, Turin, Italy
| | - Marco Oderda
- Urology, Department of Surgical Sciences, AOU Città Della Salute E Della Scienza Di Torino, University of Turin, Turin, Italy
| | - Sara Dall'Armellina
- Nuclear Medicine, Department of Medical Sciences, AOU Città Della Salute E Della Scienza Di Torino, University of Turin, Turin, Italy
| | - Virginia Liberini
- Nuclear Medicine, Department of Medical Sciences, AOU Città Della Salute E Della Scienza Di Torino, University of Turin, Turin, Italy
| | - Serena Grimaldi
- Nuclear Medicine, Department of Medical Sciences, AOU Città Della Salute E Della Scienza Di Torino, University of Turin, Turin, Italy
| | - Marilena Bellò
- Nuclear Medicine, Department of Medical Sciences, AOU Città Della Salute E Della Scienza Di Torino, University of Turin, Turin, Italy
| | - Paolo Gontero
- Urology, Department of Surgical Sciences, AOU Città Della Salute E Della Scienza Di Torino, University of Turin, Turin, Italy
| | - Umberto Ricardi
- Section of Nuclear Medicine, University Department of Radiological Sciences and Haematology, Università Cattolica del Sacro Cuore, Rome, Italy.,Department of Oncology, University of Turin, Turin, Italy
| | - Désirée Deandreis
- Nuclear Medicine, Department of Medical Sciences, AOU Città Della Salute E Della Scienza Di Torino, University of Turin, Turin, Italy
| |
Collapse
|
156
|
Luining WI, Cysouw MCF, Meijer D, Hendrikse NH, Boellaard R, Vis AN, Oprea-Lager DE. Targeting PSMA Revolutionizes the Role of Nuclear Medicine in Diagnosis and Treatment of Prostate Cancer. Cancers (Basel) 2022; 14:1169. [PMID: 35267481 PMCID: PMC8909566 DOI: 10.3390/cancers14051169] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 02/21/2022] [Indexed: 02/08/2023] Open
Abstract
Targeting the prostate-specific membrane antigen (PSMA) protein has become of great clinical value in prostate cancer (PCa) care. PSMA positron emission tomography/computed tomography (PET/CT) is increasingly used in initial staging and restaging at biochemical recurrence in patients with PCa, where it has shown superior detection rates compared to previous imaging modalities. Apart from targeting PSMA for diagnostic purposes, there is a growing interest in developing ligands to target the PSMA-protein for radioligand therapy (RLT). PSMA-based RLT is a novel treatment that couples a PSMA-antibody to (alpha or beta-emitting) radionuclide, such as Lutetium-177 (177Lu), to deliver high radiation doses to tumor cells locally. Treatment with 177Lu-PSMA RLT has demonstrated a superior overall survival rate within randomized clinical trials as compared to routine clinical care in patients with metastatic castration-resistant prostate cancer (mCRPC). The current review provides an overview of the literature regarding recent developments in nuclear medicine related to PSMA-targeted PET imaging and Theranostics.
Collapse
Affiliation(s)
- Wietske I. Luining
- Department of Urology, Prostate Cancer Network Netherlands, Amsterdam University Medical Center, VU University, 1081 HV Amsterdam, The Netherlands; (D.M.); (A.N.V.)
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Center, Location VUmc, 1081 HV Amsterdam, The Netherlands; (M.C.F.C.); (N.H.H.); (R.B.); (D.E.O.-L.)
| | - Matthijs C. F. Cysouw
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Center, Location VUmc, 1081 HV Amsterdam, The Netherlands; (M.C.F.C.); (N.H.H.); (R.B.); (D.E.O.-L.)
| | - Dennie Meijer
- Department of Urology, Prostate Cancer Network Netherlands, Amsterdam University Medical Center, VU University, 1081 HV Amsterdam, The Netherlands; (D.M.); (A.N.V.)
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Center, Location VUmc, 1081 HV Amsterdam, The Netherlands; (M.C.F.C.); (N.H.H.); (R.B.); (D.E.O.-L.)
| | - N. Harry Hendrikse
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Center, Location VUmc, 1081 HV Amsterdam, The Netherlands; (M.C.F.C.); (N.H.H.); (R.B.); (D.E.O.-L.)
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Center, Location VUmc, 1081 HV Amsterdam, The Netherlands; (M.C.F.C.); (N.H.H.); (R.B.); (D.E.O.-L.)
| | - André N. Vis
- Department of Urology, Prostate Cancer Network Netherlands, Amsterdam University Medical Center, VU University, 1081 HV Amsterdam, The Netherlands; (D.M.); (A.N.V.)
| | - Daniela E. Oprea-Lager
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Center, Location VUmc, 1081 HV Amsterdam, The Netherlands; (M.C.F.C.); (N.H.H.); (R.B.); (D.E.O.-L.)
| |
Collapse
|
157
|
Rodríguez-Fraile M, Tamayo Alonso P, Rosales JJ, de Arcocha-Torres M, Caresia-Aróztegui AP, Cózar-Santiago MP, Orcajo-Rincon J, Simó Perdigó M, Delgado Bolton RC, Artigas Guix C. The role of PSMA radioligands in the diagnosis and treatment of prostate carcinoma. Rev Esp Med Nucl Imagen Mol 2022; 41:126-135. [PMID: 35216940 DOI: 10.1016/j.remnie.2022.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/01/2021] [Indexed: 11/25/2022]
Abstract
Prostate cancer (PC) is the most common tumor in men in the West and the fifth leading cause of cancer-related death. The use of PSMA radioligands has represented an important advance both in its diagnosis, through PET molecular imaging, and in its treatment in advanced stages of the disease. This article reviews the contribution of PET studies with PSMA radioligands in initial staging, in tumor detection in biochemical recurrence (elevation of PSA) after treatment with curative intent, and in the more advanced stages of the disease (castration resistant PC or CRPC). The contribution of PSMA radioligand therapy (PSMA-RLT) in CRPC patients who progress to standard therapy is also analyzed.
Collapse
Affiliation(s)
- M Rodríguez-Fraile
- Servicio de Medicina Nuclear, Clínica Universidad de Navarra, Pamplona, Spain.
| | - P Tamayo Alonso
- Servicio de Medicina Nuclear, Hospital Universitario de Salamanca, Salamanca, Spain
| | - J J Rosales
- Servicio de Medicina Nuclear, Clínica Universidad de Navarra, Pamplona, Spain
| | | | | | | | | | - Marc Simó Perdigó
- Servicio de Medicina Nuclear Hospital Vall d'Hebron, Barcelona, Spain
| | - R C Delgado Bolton
- Servicio de Medicina Nuclear, Hospital San Pedro - Centro de Investigación Biomédica de La Rioja (CIBIR), La Rioja, Spain
| | - C Artigas Guix
- Servicio de Medicina Nuclear, Institut Jules Bordet, Université Libre de Bruxelles, Bruselas, Belgium
| | | |
Collapse
|
158
|
PARP Inhibitors and Radiometabolic Approaches in Metastatic Castration-Resistant Prostate Cancer: What’s Now, What’s New, and What’s Coming? Cancers (Basel) 2022; 14:cancers14040907. [PMID: 35205654 PMCID: PMC8869833 DOI: 10.3390/cancers14040907] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/03/2022] [Accepted: 02/08/2022] [Indexed: 12/04/2022] Open
Abstract
Simple Summary Prostate cancer still represents an important health problem in men, considering its high frequency. Over the last decade, novel treatment options have emerged, leading to notable clinical benefits. These recent scientific acquisitions are creating the basis to widen the treatment scenario of this tumor, evolving from targeting the androgen receptor axis or the traditional chemotherapy approach. Abstract In recent years, the advances in the knowledge on the molecular characteristics of prostate cancer is allowing to explore novel treatment scenarios. Furthermore, technological discoveries are widening diagnostic and treatment weapons at the clinician disposal. Among these, great relevance is being gained by PARP inhibitors and radiometabolic approaches. The result is that DNA repair genes need to be altered in a high percentage of patients with metastatic prostate cancer, making these patients optimal candidates for PARP inhibitors. These compounds have already been proved to be active in pretreated patients and are currently being investigated in other settings. Radiometabolic approaches combine specific prostate cancer cell ligands to radioactive particles, thus allowing to deliver cytotoxic radiations in cancer cells. Among these, radium-223 and lutetium-177 have shown promising activity in metastatic pretreated prostate cancer patients and further studies are ongoing to expand the applications of this therapeutic approach. In addition, nuclear medicine techniques also have an important diagnostic role in prostate cancer. Herein, we report the state of the art on the knowledge on PARP inhibitors and radiometabolic approaches in advanced prostate cancer and present ongoing clinical trials that will hopefully expand these two treatment fields.
Collapse
|
159
|
Berliner C, Kesch C, Fendler WP, Eiber M, Maurer T. [Prostate-specific membrane antigen positron emission tomography (PSMA PET) for urologists-when and which tracer?]. Urologe A 2022; 61:384-391. [PMID: 35138414 DOI: 10.1007/s00120-022-01766-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Prostate-specific membrane antigen positron emission tomography (PSMA PET) is increasingly replacing conventional imaging for staging of prostate cancer. A major challenge is its appropriate use and correct interpretation. OBJECTIVES How and when is PSMA PET used in patient care to optimally direct therapy? MATERIALS AND METHODS Systematic presentation and discussion of the current state of knowledge, guidelines and expert knowledge on PSMA PET with a summary of ongoing studies. CONCLUSION PSMA PET is the new standard for systemic staging of prostate cancer, enabling precision patient care with novel local, oligometastatic, and systemic treatment approaches.
Collapse
Affiliation(s)
- Christoph Berliner
- Klinik für Nuklearmedizin, Universitätsklinikum Essen, Hufelandstr. 55, 45147, Essen, Deutschland.
| | - Claudia Kesch
- Klinik für Urologie, Universitätsklinikum Essen, Essen, Deutschland
| | - Wolfgang P Fendler
- Klinik für Nuklearmedizin, Universitätsklinikum Essen, Hufelandstr. 55, 45147, Essen, Deutschland
| | - Matthias Eiber
- Klinik für Nuklearmedizin, Klinikum Rechts , der Isar, Technische Universität München, München, Deutschland
| | - Tobias Maurer
- Martini-Klinik Prostatakrebszentrum, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Deutschland.,Klinik für Urologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Deutschland
| |
Collapse
|
160
|
Farolfi A, Mei R, Ali S, Castellucci P. Theragnostics in prostate cancer. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF... 2022; 65:333-341. [PMID: 35133097 DOI: 10.23736/s1824-4785.21.03419-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Prostate-specific membrane antigen (PSMA) is a molecular target for both imaging diagnostics and therapeutics, i.e., a theragnostics target. There has been a growing body of evidence supporting PSMA theragnostics approaches in the management of prostate cancer (PCa) for tailored precision medicine. Tumor characterization through PSMA-ligand PET imaging is crucial for assessing the molecular signature and eligibility for PSMA radioligand therapy. Recent U.S. Food and Drug Administration (FDA) approval of two new drug applications for PSMA PET imaging contribute to reinforce PSMA as an oncologic blockbuster. Additionally, relevant progress in the PSMA treatment has been made in the last five years. [177Lu]Lu-PSMA-617 radioligand therapy for patients with progressive PSMA-avid metastatic castration-resistant PCa (mCRPC) significantly increased overall survival and radiographic progression-free survival, according to the results of an international, prospective, open label, multicenter, randomized, phase III study (VISION trial). The objective of this comprehensive review is to highlight the recent advances in PCa theragnostics, focusing on actual clinical applications and future perspectives.
Collapse
Affiliation(s)
- Andrea Farolfi
- Division of Nuclear Medicine, IRCCS University Hospital of Bologna, Bologna, Italy -
| | - Riccardo Mei
- Division of Nuclear Medicine, IRCCS University Hospital of Bologna, Bologna, Italy
| | - Sakaria Ali
- Department of Pediatrics, University College London Hospital, London, UK
| | - Paolo Castellucci
- Division of Nuclear Medicine, IRCCS University Hospital of Bologna, Bologna, Italy
| |
Collapse
|
161
|
Zhang-Yin J, Montravers F, Montagne S, Hennequin C, Renard-Penna R. Diagnosis of early biochemical recurrence after radical prostatectomy or radiation therapy in patients with prostate cancer: State of the art. Diagn Interv Imaging 2022; 103:191-199. [DOI: 10.1016/j.diii.2022.02.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/11/2022] [Accepted: 02/13/2022] [Indexed: 12/30/2022]
|
162
|
Chavoshi M, Mirshahvalad SA, Metser U, Veit-Haibach P. 68Ga-PSMA PET in prostate cancer: a systematic review and meta-analysis of the observer agreement. Eur J Nucl Med Mol Imaging 2022; 49:1021-1029. [PMID: 34767046 DOI: 10.1007/s00259-021-05616-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Accepted: 11/02/2021] [Indexed: 01/25/2023]
Abstract
PURPOSE The performance of 68 Ga-PSMA PET/CT-MR has been evaluated in prostate cancer (PCa), showing significant results. However, even a technically accurate imaging procedure requires a high interobserver agreement in its interpretation to implement in patients' management. This study aims to perform a systematic review and meta-analysis on the interobserver variability in 68 Ga-PSMA PET/CT-MR imaging in PCa patients. METHODS We conducted a systematic review and meta-analysis on the interobserver variability, including studies: (1) providing Kappa (K) as the inter-observer agreement test or the essential data to calculate it, (2) providing the K confidence interval or the essential crude data to calculate it, (3) measuring K statistic based on the appropriate use criteria for the inter-observer agreement. RESULTS Twelve studies, providing 1585 68 Ga-PSMA PET/CT-MR studies reviewed by 62 independent readers, were included. In general, the pooled inter-observer agreement was interpreted as substantial for all analyzed groups, including tumoral lesions in the prostate bed, lymphadenopathies, bone metastasis, and soft-tissue metastasis (all between 0.6 and 0.8). The regional lymphadenopathy group (0.74) obtained the highest agreement, while the lowest was for soft tissue metastasis (0.65). CONCLUSION This study showed a substantial interobserver agreement in the overall interpretation and detecting locoregional and distant involvement with 68 Ga-PSMA PET/CT-MR in PCa patients.
Collapse
Affiliation(s)
- Mohammadreza Chavoshi
- Department of Radiology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Ali Mirshahvalad
- Research Center for Nuclear Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- University Health Network, Toronto, Canada.
| | - Ur Metser
- University Health Network, Toronto, Canada
- Dept. Medical Imaging, University of Toronto, Toronto, Canada
| | - Patrick Veit-Haibach
- University Health Network, Toronto, Canada
- Dept. Medical Imaging, University of Toronto, Toronto, Canada
| |
Collapse
|
163
|
Guler OC, Onal C. In Regard to Harmon et al. Int J Radiat Oncol Biol Phys 2022; 112:572-573. [PMID: 34998536 DOI: 10.1016/j.ijrobp.2021.09.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 09/21/2021] [Indexed: 10/19/2022]
Affiliation(s)
- Ozan Cem Guler
- Başkent University Faculty of Medicine, Adana Dr Turgut Noyan Research and Treatment Center, Department of Radiation Oncology, Adana, Turkey
| | - Cem Onal
- Başkent University Faculty of Medicine, Adana Dr Turgut Noyan Research and Treatment Center, Department of Radiation Oncology, Adana, Turkey
| |
Collapse
|
164
|
Marhold M, Kramer G, Krainer M, Le Magnen C. The prostate cancer landscape in Europe: Current challenges, future opportunities. Cancer Lett 2022; 526:304-310. [PMID: 34863887 DOI: 10.1016/j.canlet.2021.11.033] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 11/24/2021] [Accepted: 11/30/2021] [Indexed: 01/03/2023]
Abstract
Prostate cancer (PCa) is the most common non-cutaneous cancer in men in Europe and is predicted to exhibit declining mortality in the European Union (EU) due to various recent improvements in treatment. The goal of this short review is to give insight into the European treatment landscape of PCa, while focusing on improvements in care.
Collapse
Affiliation(s)
- Maximilian Marhold
- Division of Oncology, Department for Medicine I, Medical University of Vienna, Vienna, Austria.
| | - Gero Kramer
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Michael Krainer
- Division of Oncology, Department for Medicine I, Medical University of Vienna, Vienna, Austria
| | - Clémentine Le Magnen
- Pathology, Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Switzerland; Department of Urology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
165
|
Surcel C, Kretschmer A, Mirvald C, Sinescu I, Heidegger I, Tsaur I. Molecular Mechanisms Related with Oligometastatic Prostate Cancer-Is It Just a Matter of Numbers? Cancers (Basel) 2022; 14:cancers14030766. [PMID: 35159033 PMCID: PMC8833728 DOI: 10.3390/cancers14030766] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 02/07/2023] Open
Abstract
During the last decade, the body of knowledge regarding the oligometastatic state has increased exponentially. Several molecular frameworks have been established, aiding our understanding of metastatic spread caused by genetically unstable cells that adapt to a tissue environment which is distant from the primary tumor. In the current narrative review, we provide an overview of the current treatment landscape of oligometastatic cancer, focusing on the current biomarkers used in the identification of true oligometastatic disease and highlighting the impact of molecular imaging on stage shift in different scenarios. Finally, we address current and future directions regarding the use of genetic and epigenetic targeting treatments in oligometastatic prostate cancer.
Collapse
Affiliation(s)
- Cristian Surcel
- Center of Urologic Surgery, Dialysis and Renal Transplantation, Fundeni Clinical Institute, “Carol Davila” University of Medicine and Pharmacy, 00238 Bucharest, Romania; (C.M.); (I.S.)
- Correspondence:
| | | | - Cristian Mirvald
- Center of Urologic Surgery, Dialysis and Renal Transplantation, Fundeni Clinical Institute, “Carol Davila” University of Medicine and Pharmacy, 00238 Bucharest, Romania; (C.M.); (I.S.)
| | - Ioanel Sinescu
- Center of Urologic Surgery, Dialysis and Renal Transplantation, Fundeni Clinical Institute, “Carol Davila” University of Medicine and Pharmacy, 00238 Bucharest, Romania; (C.M.); (I.S.)
| | - Isabel Heidegger
- Department of Urology, Medical University Innsbruck, 6020 Innsbruck, Austria;
| | - Igor Tsaur
- Department of Urology and Pediatric Urology, University Medical Center Mainz, 55131 Mainz, Germany;
| |
Collapse
|
166
|
Wang G, Hong H, Zang J, Liu Q, Jiang Y, Fan X, Zhu Z, Zhu L, Kung HF. Head-to-head comparison of [ 68 Ga]Ga-P16-093 and [ 68 Ga]Ga-PSMA-617 in dynamic PET/CT evaluation of the same group of recurrent prostate cancer patients. Eur J Nucl Med Mol Imaging 2022; 49:1052-1062. [PMID: 34557930 DOI: 10.1007/s00259-021-05539-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 08/23/2021] [Indexed: 12/14/2022]
Abstract
PURPOSE This study was prospectively designed to evaluate the early dynamic organ distribution and tumor detection capability of [68 Ga]Ga-P16-093, which was compared with [68 Ga]Ga-PSMA-617 in the same group of recurrent prostate cancer patients. METHODS Twenty patients with recurrent prostate cancer were enrolled. In 2 consecutive days, each patient underwent a 60-min dynamic PET/CT scan after intravenous administration of 148-185 MBq (4-5 mCi) [68 Ga]Ga-P16-093 and [68 Ga]Ga-PSMA-617, respectively. Following a low-dose CT scan, serial dynamic PET scans were performed from head to proximal thigh at 9 time points (30 s/bed at 4, 7, 10, 13, and 16 min; 1 min/bed at 20, 30, and 45 min; and 2 min/bed at 60 min). Standardized uptake values were measured for semi-quantitative comparison. RESULTS [68 Ga]Ga-P16-093 PET/CT revealed a significantly higher tumor uptake at 4 min (SUVmax 7.88 ± 5.26 vs. 6.01 ± 3.88, P < 0.001), less blood pool retention at 4 min (SUVmean 5.12 ± 1.16 vs. 6.14 ± 0.98, P < 0.001), and lower bladder accumulation at 60 min (SUVmean 31.33 ± 27.47 vs. 48.74 ± 34.01, P = 0.042) than [68 Ga]Ga-PSMA-617 scan. Significantly higher [68 Ga]Ga-P16-093 uptakes were also observed in the parotid gland, liver, spleen, and kidney. Besides, [68 Ga]Ga-P16-093 exhibited a better detectability of tumor than [68 Ga]Ga-PSMA-617 (366 vs. 321, P = 0.009). CONCLUSIONS [68 Ga]Ga-P16-093 showed advantages over [68 Ga]Ga-PSMA-617 with higher tumor uptakes, tumor-to-blood pool ratio and detection capability, less blood pool, and bladder accumulation in recurrent prostate cancer patients. TRIAL REGISTRATION [68 Ga]Ga-P16-093 and [68 Ga]Ga-PSMA-617 PET/CT Imaging in the Same Group of Prostate Cancer Patients (NCT04796467, Registered 12 March 2021, retrospectively registered) URL of registry: https://clinicaltrials.gov/ct2/show/NCT04796467.
Collapse
Affiliation(s)
- 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
| | - Haiyan Hong
- College of Chemistry, Key Laboratory of Radiopharmaceuticals, Ministry of Education, Beijing Normal University, Beijing, 100875, China
| | - 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
| | - Qingxing Liu
- 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
| | - Yuanyuan Jiang
- 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
| | - Xinrong Fan
- Department of Urology, 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
- College of Chemistry, Key Laboratory of Radiopharmaceuticals, Ministry of Education, Beijing Normal University, Beijing, 100875, China.
| | - Hank F Kung
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
167
|
In Reply to Guler and Onal. Int J Radiat Oncol Biol Phys 2022; 112:573-574. [PMID: 34998537 DOI: 10.1016/j.ijrobp.2021.09.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 09/21/2021] [Indexed: 11/22/2022]
|
168
|
Tsechelidis I, Vrachimis A. PSMA PET in Imaging Prostate Cancer. Front Oncol 2022; 12:831429. [PMID: 35155262 PMCID: PMC8832487 DOI: 10.3389/fonc.2022.831429] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/12/2022] [Indexed: 12/18/2022] Open
Abstract
After prostate malignancy diagnosis, precise determination of disease extent are fundamental steps for tailored made therapy. The earlier the diagnosis of the burden of the disease, the longer the survival in many cases. National and international guidelines are based on “classic” imaging technics combining radiological and nuclear medicine scans like CT, MRI and bone scintigraphy (BS). The most recent nuclear medicine development is the prostate specific membrane antigen (PSMA) PET and is emerging as the most promising tool of medical imaging, gaining ground every day. Nevertheless, the different onset among multiple studies fails to establish a worldwide admission and incorporation of this technique in guidelines and its position in workaday medical algorithms. It seems that the medical community agrees not to utilize PSMA PET for low-risk patients; intense debate and research is ongoing for its utility in intermediate risk patients. Contrariwise, in high-risk patients PSMA PET is confirmed outperforming CT and BS combined. Additionally, irrespectively to their castration status, patients with biochemical failure should be referred for PSMA PET. Even though PSMA PET reveals more extended disease than expected or exonerates equivalent lesions, thus impacting treatment optimization. Studies being in progress and future trials with clarify whether PSMA PET will be the new gold standard technic for specific groups of patients.
Collapse
Affiliation(s)
- Ioannis Tsechelidis
- Department of Nuclear Medicine, German Oncology Center, University Hospital of the European University, Limassol, Cyprus
- *Correspondence: Ioannis Tsechelidis,
| | - Alexis Vrachimis
- Department of Nuclear Medicine, German Oncology Center, University Hospital of the European University, Limassol, Cyprus
- Cancer Research and Innovation Center (CARIC), Limassol, Cyprus
| |
Collapse
|
169
|
Comparison of reprojected bone SPECT/CT and planar bone scintigraphy for the detection of bone metastases in breast and prostate cancer. Nucl Med Commun 2022; 43:510-517. [PMID: 35081092 DOI: 10.1097/mnm.0000000000001536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE The aim of this study was to compare reprojected bone SPECT/CT (RBS) against planar bone scintigraphy (BS) in the detection of bone metastases in breast and prostate cancer patients. METHODS Twenty-six breast and 105 prostate cancer patients with high risk for bone metastases underwent 99mTc-HMDP BS and whole-body SPECT/CT, 1.5-T whole-body diffusion-weighted MRI and 18F-NaF or 18F-PSMA-1007 PET/CT within two prospective clinical trials (NCT01339780 and NCT03537391). Consensus reading of all imaging modalities and follow-up data were used to define the reference standard diagnosis. The SPECT/CT data were reprojected into anterior and posterior views to produce RBS images. Both BS and RBS images were independently double read by two pairs of experienced nuclear medicine physicians. The findings were validated against the reference standard diagnosis and compared between BS and RBS on the patient, region and lesion levels. RESULTS All metastatic patients detected by BS were also detected by RBS. In addition, three metastatic patients were missed by BS but detected by RBS. The average patient-level sensitivity of two readers for metastases was 75% for BS and 87% for RBS, and the corresponding specificity was 79% for BS and 39% for RBS. The average region-level sensitivity of two readers was 64% for BS and 69% for RBS, and the corresponding specificity was 96% for BS and 87% for RBS. CONCLUSION Whole-body bone SPECT/CT can be reprojected into more familiar anterior and posterior planar images with excellent sensitivity for bone metastases, making additional acquisition of planar BS unnecessary.
Collapse
|
170
|
The Emerging Role of Next-Generation Imaging in Prostate Cancer. Curr Oncol Rep 2022; 24:33-42. [DOI: 10.1007/s11912-021-01156-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2021] [Indexed: 12/23/2022]
|
171
|
Management of patients with a persistently elevated PSA after radical prostatectomy: a narrative review. World J Urol 2022; 40:965-972. [PMID: 35024944 DOI: 10.1007/s00345-021-03923-y] [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: 11/30/2021] [Accepted: 12/28/2021] [Indexed: 10/19/2022] Open
Abstract
INTRODUCTION The management of the postoperative biological relapse of prostate cancer is most often based on salvage radiotherapy (SRT) with or without the addition of a variable duration of hormone therapy (HT). The indications for SRT +/- HT are established in the setting of a rising PSA level after a period where an undetectable PSA was achieved. However, in case of detectable PSA immediately after radical prostatectomy, the treatment options and prognosis are still unclear. MATERIALS AND METHODS We conducted a narrative review based on an analysis of the literature focusing on articles targeting the population of patients with postoperative persistently detectable PSA level. Case reports, original articles, clinical trials, and published reviews were studied for this purpose. CONCLUSION This article will describe current management of patients with detectable PSA immediately after radical prostatectomy, notably the contribution of modern imaging and new treatment options involving the combination of RT and HT.
Collapse
|
172
|
Jadvar H, Calais J, Fanti S, Feng F, Greene KL, Gulley JL, Hofman M, Koontz BF, Lin DW, Morris MJ, Rowe SP, Royce TJ, Salami S, Savir-Baruch B, Srinivas S, Hope TA. Appropriate Use Criteria for Prostate-Specific Membrane Antigen PET Imaging. J Nucl Med 2022; 63:59-68. [PMID: 34593595 PMCID: PMC8717184 DOI: 10.2967/jnumed.121.263262] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 09/21/2021] [Indexed: 11/16/2022] Open
Affiliation(s)
- Hossein Jadvar
- Department of Radiology, University of Southern California, Los Angeles, California
| | - Jeremie Calais
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California
| | | | - Felix Feng
- Department of Radiation Oncology, University of California, San Francisco, California
| | - Kirsten L. Greene
- Department of Urology, University of Virginia, Charlottesville, Virginia
| | | | - Michael Hofman
- Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia and Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria Australia
| | - Bridget F. Koontz
- Department of Radiation Oncology, Duke University, Durham, North Carolina
| | - Daniel W. Lin
- Department of Urology, University of Washington, Seattle, Washington
| | - Michael J. Morris
- Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Steve P. Rowe
- Department of Radiological Sciences, Johns Hopkins University, Baltimore, Maryland
| | - Trevor J. Royce
- Department of Radiation Oncology, University of North Carolina, Chapel Hill, North Carolina
| | - Simpa Salami
- Department of Urology, University of Michigan, Ann Arbor, Michigan
| | | | - Sandy Srinivas
- Department of Medicine (Oncology), Stanford University, California; and
| | - Thomas A. Hope
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, California
| |
Collapse
|
173
|
Razmaria AA, Schoder H, Morris MJ. Advances in Prostate Cancer Imaging. Urol Oncol 2022. [DOI: 10.1007/978-3-030-89891-5_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
174
|
Franquet E, Park H. Molecular imaging in oncology: Common PET/CT radiopharmaceuticals and applications. Eur J Radiol Open 2022; 9:100455. [DOI: 10.1016/j.ejro.2022.100455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 11/03/2022] [Accepted: 11/09/2022] [Indexed: 11/26/2022] Open
|
175
|
Solari EL, Gafita A, Schachoff S, Bogdanović B, Villagrán Asiares A, Amiel T, Hui W, Rauscher I, Visvikis D, Maurer T, Schwamborn K, Mustafa M, Weber W, Navab N, Eiber M, Hatt M, Nekolla SG. The added value of PSMA PET/MR radiomics for prostate cancer staging. Eur J Nucl Med Mol Imaging 2022; 49:527-538. [PMID: 34255130 PMCID: PMC8803696 DOI: 10.1007/s00259-021-05430-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/24/2021] [Indexed: 12/24/2022]
Abstract
PURPOSE To evaluate the performance of combined PET and multiparametric MRI (mpMRI) radiomics for the group-wise prediction of postsurgical Gleason scores (psGSs) in primary prostate cancer (PCa) patients. METHODS Patients with PCa, who underwent [68 Ga]Ga-PSMA-11 PET/MRI followed by radical prostatectomy, were included in this retrospective analysis (n = 101). Patients were grouped by psGS in three categories: ISUP grades 1-3, ISUP grade 4, and ISUP grade 5. mpMRI images included T1-weighted, T2-weighted, and apparent diffusion coefficient (ADC) map. Whole-prostate segmentations were performed on each modality, and image biomarker standardization initiative (IBSI)-compliant radiomic features were extracted. Nine support vector machine (SVM) models were trained: four single-modality radiomic models (PET, T1w, T2w, ADC); three PET + MRI double-modality models (PET + T1w, PET + T2w, PET + ADC), and two baseline models (one with patient data, one image-based) for comparison. A sixfold stratified cross-validation was performed, and balanced accuracies (bAcc) of the predictions of the best-performing models were reported and compared through Student's t-tests. The predictions of the best-performing model were compared against biopsy GS (bGS). RESULTS All radiomic models outperformed the baseline models. The best-performing (mean ± stdv [%]) single-modality model was the ADC model (76 ± 6%), although not significantly better (p > 0.05) than other single-modality models (T1w: 72 ± 3%, T2w: 73 ± 2%; PET: 75 ± 5%). The overall best-performing model combined PET + ADC radiomics (82 ± 5%). It significantly outperformed most other double-modality (PET + T1w: 74 ± 5%, p = 0.026; PET + T2w: 71 ± 4%, p = 0.003) and single-modality models (PET: p = 0.042; T1w: p = 0.002; T2w: p = 0.003), except the ADC-only model (p = 0.138). In this initial cohort, the PET + ADC model outperformed bGS overall (82.5% vs 72.4%) in the prediction of psGS. CONCLUSION All single- and double-modality models outperformed the baseline models, showing their potential in the prediction of GS, even with an unbalanced cohort. The best-performing model included PET + ADC radiomics, suggesting a complementary value of PSMA-PET and ADC radiomics.
Collapse
Affiliation(s)
- Esteban Lucas Solari
- School of Medicine, Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University Munich, Munich, Germany.
| | - Andrei Gafita
- School of Medicine, Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Sylvia Schachoff
- School of Medicine, Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Borjana Bogdanović
- School of Medicine, Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Alberto Villagrán Asiares
- School of Medicine, Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Thomas Amiel
- School of Medicine, Department of Urology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Wang Hui
- School of Medicine, Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Isabel Rauscher
- School of Medicine, Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | | | - Tobias Maurer
- Department of Urology and Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Kristina Schwamborn
- School of Medicine, Institute of Pathology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Mona Mustafa
- School of Medicine, Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Wolfgang Weber
- School of Medicine, Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Nassir Navab
- School of Computer Science, Computer Aided Medical Procedures and Augmented Reality, Technical University Munich, Munich, Germany
| | - Matthias Eiber
- School of Medicine, Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Mathieu Hatt
- INSERM, UMR 1101, LaTIM, Univ Brest, Brest, France
| | - Stephan G Nekolla
- School of Medicine, Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| |
Collapse
|
176
|
Jimbo M, Andrews JR, Ahmed ME, Dundar A, Karnes RJ, Bryce AH, Kendi AT, Kwon ED, Lowe VJ, Bold MS. Prognostic role of 11C-choline PET/CT scan in patients with metastatic castrate resistant prostate cancer undergoing primary docetaxel chemotherapy. Prostate 2022; 82:41-48. [PMID: 34633087 DOI: 10.1002/pros.24246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 07/30/2021] [Accepted: 09/07/2021] [Indexed: 11/08/2022]
Abstract
BACKGROUND We sought to assess the prognostic utility of 11C-choline positron emission tomography/computed tomography (PET/CT) in patients with metastatic castrate resistant prostate cancer (mCRPC) undergoing primary docetaxel chemotherapy. METHODS We performed a single institution retrospective analysis of 77 mCRPC patients who were treated with 6 cycles of docetaxel chemotherapy, and who also underwent 11C-choline PET/CT scans at baseline (before chemotherapy), mid-course (after 3 cycles), and posttherapy (after 6 cycles). We evaluated treatment response based on percent change in blood pool-corrected maximum standardized uptake value (SUVmax) of the target lesion on PET/CT, as well as percent change in serum prostate specific antigen (PSA). Logistic regression analysis was used to identify factors associated with complete treatment response. Progression free survival (PFS) analysis was performed using log-rank test and shown on Kaplan-Meier plot. RESULTS Percent change in blood pool-corrected SUVmax on mid-course scan was a significant predictor of complete response (odds ratio [OR]: 0.98, 95% confidence interval [CI]: 0.96-0.99, p = .0003), whereas percent change in PSA was not (OR: 0.99, 95% CI: 0.99-1.01, p = .6025). 57 of 77 patients (74%) achieved ≥20% reduction in blood pool-corrected SUVmax on mid-course; these patients were 3.6 times more likely to achieve complete response after full 6 cycles of docetaxel chemotherapy, compared to patients with <20% reduction in blood pool-corrected SUVmax (OR: 3.56, 95% CI: 1.04-16.52, p = .0420). Median PFS in the complete response group was 35.1 months (95% CI: 26.0-52.7 months), compared to 9.4 months (95% CI: 6.9-13.0 months) in the incomplete response group (p = .0005). CONCLUSIONS Our study showed that mid-course and posttherapy 11C-choline PET/CT evaluation for mCRPC patients undergoing primary docetaxel chemotherapy can predict full course treatment response and PFS, respectively. 11C-choline PET/CT imaging may provide valuable prognostic information to guide treatment choices for patients with mCRPC.
Collapse
Affiliation(s)
- Masaya Jimbo
- Department of Urology, Mayo Clinic, Rochester, Minnesota, USA
| | - Jack R Andrews
- Department of Urology, Mayo Clinic, Rochester, Minnesota, USA
| | - Mohamed E Ahmed
- Department of Urology, Mayo Clinic, Rochester, Minnesota, USA
| | - Ayca Dundar
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Alan H Bryce
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Mayo Clinic, Phoenix, Arizona, USA
| | - Ayse T Kendi
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Eugene D Kwon
- Department of Urology, Mayo Clinic, Rochester, Minnesota, USA
| | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Michael S Bold
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
177
|
Vogl UM, Beer TM, Davis ID, Shore ND, Sweeney CJ, Ost P, Attard G, Bossi A, de Bono J, Drake CG, Efstathiou E, Fanti S, Fizazi K, Halabi S, James N, Mottet N, Padhani AR, Roach M, Rubin M, Sartor O, Small E, Smith MR, Soule H, Sydes MR, Tombal B, Omlin A, Gillessen S. Lack of consensus identifies important areas for future clinical research: Advanced Prostate Cancer Consensus Conference (APCCC) 2019 findings. Eur J Cancer 2022; 160:24-60. [PMID: 34844839 DOI: 10.1016/j.ejca.2021.09.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 09/23/2021] [Indexed: 11/03/2022]
Abstract
BACKGROUND Innovations in treatments, imaging and molecular characterisation have improved outcomes for people with advanced prostate cancer; however, many aspects of clinical management are devoid of high-level evidence. At the Advanced Prostate Cancer Consensus Conference (APCCC) 2019, many of these topics were addressed, and consensus was not always reached. The results from clinical trials will most reliably plus the gaps. METHODS An invited panel of 57 experts voted on 123 multiple-choice questions on clinical management at APCCC 2019. No consensus was reached on 88 (71.5%) questions defined as <75% of panellists voting for the same answer option. We reviewed clinicaltrials.gov to identify relevant ongoing phase III trials in these areas of non-consensus. RESULTS A number of ongoing phase III trials were identified that are relevant to these non-consensus issues. However, many non-consensus issues appear not to be addressed by current clinical trials. Of note, no phase III but only phase II trials were identified, investigating side effects of hormonal treatments and their management. CONCLUSIONS Lack of consensus almost invariably indicates gaps in existing evidence. The high percentage of questions lacking consensus at APCCC 2019 highlights the complexity of advanced prostate cancer care and the need for robust, clinically relevant trials that can fill current gaps with high-level evidence. Our review of these areas of non-consensus and ongoing trials provides a useful summary, indicating areas in which future consensus may soon be reached. This review may facilitate academic investigators to identify and prioritise topics for future research.
Collapse
Affiliation(s)
- Ursula M Vogl
- Oncology Institute of Southern Switzerland, EOC, Bellinzona, Switzerland
| | - Tomasz M Beer
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Ian D Davis
- Monash University and Eastern Health, Victoria, Australia
| | - Neal D Shore
- Carolina Urologic Research Center, Myrtle Beach, SC, USA
| | - Christopher J Sweeney
- Dana-Farber Cancer Institute, Boston, MA, USA; Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Piet Ost
- Radiation Oncology, Ghent University Hospital, Ghent, Belgium
| | | | - Alberto Bossi
- Genito Urinary Oncology, Prostate Brachytherapy Unit, Goustave Roussy, Paris, France
| | - Johann de Bono
- The Institute of Cancer Research/Royal Marsden NHS Foundation Trust, Surrey, UK
| | - Charles G Drake
- Division of Haematology/Oncology, Columbia University Medical Center, New York, NY, USA
| | | | | | - Karim Fizazi
- Institut Gustave Roussy, University of Paris Sud, Villejuif, France
| | - Susan Halabi
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | | | | | - Anwar R Padhani
- Mount Vernon Cancer Centre and Institute of Cancer Research, London, UK
| | - Mack Roach
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Mark Rubin
- Bern Center for Precision Medicine, Bern, Switzerland; Department for Biomedical Research, University of Bern, Bern, Switzerland
| | | | - Eric Small
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Matthew R Smith
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Howard Soule
- Prostate Cancer Foundation, Santa Monica, CA, USA
| | - Matthew R Sydes
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK
| | | | - Aurelius Omlin
- Department of Medical Oncology and Haematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Silke Gillessen
- Oncology Institute of Southern Switzerland, EOC, Bellinzona, Switzerland; University of Bern, Bern, Switzerland; Università della Svizzera Italiana, Lugano, Switzerland; Division of Cancer Science, University of Manchester, Manchester, UK.
| |
Collapse
|
178
|
Daryanani A, Turkbey B. Recent Advancements in CT and MR Imaging of Prostate Cancer. Semin Nucl Med 2021; 52:365-373. [PMID: 34930627 PMCID: PMC9038642 DOI: 10.1053/j.semnuclmed.2021.11.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/19/2021] [Accepted: 11/23/2021] [Indexed: 11/11/2022]
Abstract
CT and MRI are both commonly used in prostate cancer (PCa) management, which includes a large spectrum from screening positive pre-diagnosis phase to metastatic disease. CT and MRI have continually evolved to meet the changing demands for PCa management. For CT, novel techniques such as dual energy CT and photon counting CT show promising results for tissue characterization and quantification. For MRI, the detection, staging, and management of prostate cancer has been significantly improved by the development of multiparametric, biparametric, and whole-body MRI techniques. Additionally, research on ultrasmall superparamagnetic particles of iron oxide contrast-enhanced MRI has revealed promising results for nodal staging of PCa. In this manuscript we aim to outline the current status and recent advancements of CT and MRI in PCa imaging.
Collapse
Affiliation(s)
- Asha Daryanani
- Molecular Imaging Branch, National Cancer Institute, NIH, Bethesda, MD
| | - Baris Turkbey
- Molecular Imaging Branch, National Cancer Institute, NIH, Bethesda, MD.
| |
Collapse
|
179
|
Harmon G, Chan D, Lee B, Miller C, Gorbonos A, Gupta G, Quek M, Woods M, Savir-Baruch B, Harkenrider MM, Solanki AA. Validating Modern NRG Oncology Pelvic Nodal and Groupe Francophone de Radiothérapie Urologique Prostate Bed Contouring Guidelines for Post-Prostatectomy Salvage Radiation: A Secondary Analysis of the LOCATE Trial. Int J Radiat Oncol Biol Phys 2021; 111:1195-1203. [PMID: 34015368 DOI: 10.1016/j.ijrobp.2021.05.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/23/2021] [Accepted: 05/01/2021] [Indexed: 12/14/2022]
Abstract
PURPOSE We used the patterns of recurrence on 18F-fluciclovine positron emission tomography (PET)-computed tomography (CT) in patients enrolled in the LOCATE trial after prostatectomy to evaluate how well the most recent NRG Oncology and Groupe Francophone de Radiothérapie Urologique (GFRU) contouring recommendations encompassed all sites of recurrence in the prostate fossa and pelvic nodes in comparison to former Radiation Therapy Oncology Group (RTOG) recommendations. METHODS AND MATERIALS Patients with biochemically recurrent prostate cancer after radical prostatectomy with a positive finding within the prostate fossa or pelvic nodes on 18F-fluciclovine PET/CTs were identified from the LOCATE patient population. Areas of gross disease were delineated. Prostate fossa contours were delineated using both the 2010 RTOG consensus guidelines and the recently published 2020 GFRU consensus guidelines. Pelvic nodes were contoured with both the 2009 RTOG consensus guidelines and the 2020 NRG consensus guidelines. The performance of the contouring guidelines was assessed by determining what proportion of gross recurrent lesions were encompassed completely or marginally. RESULTS Of the 213 patients within the LOCATE trial, 45 patients were eligible for analysis with positive 18F-fluciclovine PET findings. Of the 30 total prostate fossa recurrences, the 2010 RTOG contour covered 20 (67%) and missed or marginally covered 10 (33%). The 2020 GFRU contour covered 27 recurrences (90%), and missed or marginally covered 3 (10%). Of the 43 total nodal recurrences, the 2009 RTOG pelvic nodal contour covered 29 nodes (67%), and missed or marginally covered 14 (32%). The 2020 NRG pelvic nodal contour covered 43 nodes (100%), with no misses or marginal coverage. CONCLUSIONS This secondary analysis of the LOCATE trial exemplifies the improved coverage of the latest prostate fossa contouring recommendations from the GFRU. Similarly, it also validates the updated 2020 NRG pelvic nodal contouring guidelines by demonstrating improved coverage of recurrent disease in this patient population.
Collapse
Affiliation(s)
- Grant Harmon
- Department of Radiation Oncology, Stritch School of Medicine, Loyola University, Chicago, Illinois
| | - Dennis Chan
- Department of Radiation Oncology, Stritch School of Medicine, Loyola University, Chicago, Illinois
| | - Brian Lee
- Department of Radiation Oncology, Stritch School of Medicine, Loyola University, Chicago, Illinois
| | - Chelsea Miller
- Department of Radiation Oncology, Warren Alpert Medical School of Brown University, Providence, Rhode Island
| | - Alex Gorbonos
- Department of Urology, Stritch School of Medicine, Loyola University, Chicago, Illinois
| | - Gopal Gupta
- Department of Urology, Stritch School of Medicine, Loyola University, Chicago, Illinois
| | - Marcus Quek
- Department of Urology, Stritch School of Medicine, Loyola University, Chicago, Illinois
| | - Michael Woods
- Department of Urology, Stritch School of Medicine, Loyola University, Chicago, Illinois
| | - Bital Savir-Baruch
- Department of Radiology, Stritch School of Medicine, Loyola University, Chicago, Illinois
| | - Matthew M Harkenrider
- Department of Radiation Oncology, Stritch School of Medicine, Loyola University, Chicago, Illinois
| | - Abhishek A Solanki
- Department of Radiation Oncology, Stritch School of Medicine, Loyola University, Chicago, Illinois.
| |
Collapse
|
180
|
Rais-Bahrami S, Efstathiou JA, Turnbull CM, Camper SB, Kenwright A, Schuster DM, Scarsbrook AF. 18F-Fluciclovine PET/CT performance in biochemical recurrence of prostate cancer: a systematic review. Prostate Cancer Prostatic Dis 2021; 24:997-1006. [PMID: 34012062 PMCID: PMC8616758 DOI: 10.1038/s41391-021-00382-9] [Citation(s) in RCA: 9] [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: 01/28/2021] [Revised: 04/13/2021] [Accepted: 04/28/2021] [Indexed: 11/16/2022]
Abstract
BACKGROUND A systematic literature review of the performance of 18Fluorine-fluciclovine PET/CT for imaging of men with recurrent prostate cancer was performed. METHODS Scientific literature databases (MEDLINE, ScienceDirect and Cochrane Libraries) were searched systematically during Oct 2020 using PRISMA criteria. No limit was put on the date of publication. Prospective studies reporting a patient-level 18F-fluciclovine detection rate (DR) from ≥25 patients with recurrent prostate cancer were sought. Proceedings of relevant meetings held from 2018 through Oct 2020 were searched for abstracts meeting criteria. RESULTS Searches identified 321 unique articles. In total, nine articles (six papers and three conference abstracts), comprising a total of 850 patients met inclusion criteria. Most studies (n = 6) relied on ASTRO-Phoenix Criteria, EAU-ESTRO-SIOG, and/or ASTRO-AUA guidelines to identify patients with biochemical recurrence. Patients' PSA levels ranged from 0.02-301.7 ng/mL (median level per study, 0.34-4.10 ng/mL [n = 8]). Approximately 64% of patients had undergone prostatectomy, but three studies focused solely on post-prostatectomy patients. Adherence to imaging protocol guidelines was heterogeneous, with variance seen in administered activity, uptake and scan times. Overall patient-level DR varied between studies from 26% to 83%, with 78% of studies reporting a DR > 50%. DR was proportional to PSA, but even at PSA < 0.5 ng/mL DR of up to 53% were reported. Prostate/bed DR (n = 7) ranged from 18% to 78% and extra-prostatic rates (n = 6) from 8% to 72%. Pelvic node and bone lesion DR ranged from 8% to 47% and 0% to 26%, respectively (n = 5). 18F-Fluciclovine PET/CT was shown to impact patient management and outcomes. Two studies reported 59-63% of patients to have a management change post-scan. A further study showed significant increase in failure-free survival following 18F-fluciclovine-guided compared with conventional imaging-guided radiotherapy planning. CONCLUSIONS 18F-Fluciclovine PET/CT shows good performance in patients with recurrent prostate cancer leading to measurable clinical benefits. Careful adherence to recommended imaging protocols may help optimize DR.
Collapse
Affiliation(s)
- Soroush Rais-Bahrami
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL, USA
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL, USA
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jason A Efstathiou
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | - David M Schuster
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, USA
| | - Andrew F Scarsbrook
- Department of Radiology, Leeds Teaching Hospitals NHS Trust, Leeds, UK.
- Leeds Institute of Health Research, University of Leeds, Leeds, UK.
| |
Collapse
|
181
|
Oprea-Lager DE, Cysouw MC, Boellaard R, Deroose CM, de Geus-Oei LF, Lopci E, Bidaut L, Herrmann K, Fournier LS, Bäuerle T, deSouza NM, Lecouvet FE. Bone Metastases Are Measurable: The Role of Whole-Body MRI and Positron Emission Tomography. Front Oncol 2021; 11:772530. [PMID: 34869009 PMCID: PMC8640187 DOI: 10.3389/fonc.2021.772530] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/04/2021] [Indexed: 12/14/2022] Open
Abstract
Metastatic tumor deposits in bone marrow elicit differential bone responses that vary with the type of malignancy. This results in either sclerotic, lytic, or mixed bone lesions, which can change in morphology due to treatment effects and/or secondary bone remodeling. Hence, morphological imaging is regarded unsuitable for response assessment of bone metastases and in the current Response Evaluation Criteria In Solid Tumors 1.1 (RECIST1.1) guideline bone metastases are deemed unmeasurable. Nevertheless, the advent of functional and molecular imaging modalities such as whole-body magnetic resonance imaging (WB-MRI) and positron emission tomography (PET) has improved the ability for follow-up of bone metastases, regardless of their morphology. Both these modalities not only have improved sensitivity for visual detection of bone lesions, but also allow for objective measurements of bone lesion characteristics. WB-MRI provides a global assessment of skeletal metastases and for a one-step "all-organ" approach of metastatic disease. Novel MRI techniques include diffusion-weighted imaging (DWI) targeting highly cellular lesions, dynamic contrast-enhanced MRI (DCE-MRI) for quantitative assessment of bone lesion vascularization, and multiparametric MRI (mpMRI) combining anatomical and functional sequences. Recommendations for a homogenization of MRI image acquisitions and generalizable response criteria have been developed. For PET, many metabolic and molecular radiotracers are available, some targeting tumor characteristics not confined to cancer type (e.g. 18F-FDG) while other targeted radiotracers target specific molecular characteristics, such as prostate specific membrane antigen (PSMA) ligands for prostate cancer. Supporting data on quantitative PET analysis regarding repeatability, reproducibility, and harmonization of PET/CT system performance is available. Bone metastases detected on PET and MRI can be quantitatively assessed using validated methodologies, both on a whole-body and individual lesion basis. Both have the advantage of covering not only bone lesions but visceral and nodal lesions as well. Hybrid imaging, combining PET with MRI, may provide complementary parameters on the morphologic, functional, metabolic and molecular level of bone metastases in one examination. For clinical implementation of measuring bone metastases in response assessment using WB-MRI and PET, current RECIST1.1 guidelines need to be adapted. This review summarizes available data and insights into imaging of bone metastases using MRI and PET.
Collapse
Affiliation(s)
- Daniela E. Oprea-Lager
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Matthijs C.F. Cysouw
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Ronald Boellaard
- Department of Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Christophe M. Deroose
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
- Nuclear Medicine & Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Lioe-Fee de Geus-Oei
- Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
- Biomedical Photonic Imaging Group, University of Twente, Enschede, Netherlands
| | - Egesta Lopci
- Nuclear Medicine Unit, IRCCS – Humanitas Research Hospital, Milan, Italy
| | - Luc Bidaut
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- College of Science, University of Lincoln, Lincoln, United Kingdom
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen, and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | - Laure S. Fournier
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Paris Cardiovascular Research Center (PARCC), Institut National de la Santé et de la Recherche Médicale (INSERM), Radiology Department, Assistance Publique-Hôpitaux de Paris (AP-HP), Hopital europeen Georges Pompidou, Université de Paris, Paris, France
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
| | - Tobias Bäuerle
- Institute of Radiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Nandita M. deSouza
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- European Imaging Biomarkers Alliance (EIBALL), European Society of Radiology, Vienna, Austria
- Division of Radiotherapy and Imaging, The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Frederic E. Lecouvet
- Imaging Group, European Organisation of Research and Treatment in Cancer (EORTC), Brussels, Belgium
- Department of Radiology, Institut de Recherche Expérimentale et Clinique (IREC), Cliniques Universitaires Saint Luc, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
182
|
Osborne JR, Bander NH, Tagawa ST. Prostate-Specific Membrane Antigen Positron Emission Tomography and the New Algorithm for Patients With Prostate Cancer Prior to Prostatectomy. JAMA Oncol 2021; 7:1642-1643. [PMID: 34529003 DOI: 10.1001/jamaoncol.2021.3762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Joseph R Osborne
- Division of Molecular Imaging and Therapeutics, Department of Radiology, Weill Cornell Medicine, New York, New York.,Meyer Cancer Center, Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, New York
| | - Neil H Bander
- Meyer Cancer Center, Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, New York.,Department of Urology, Weill Cornell Medicine, New York, New York
| | - Scott T Tagawa
- Meyer Cancer Center, Weill Cornell Medicine, NewYork-Presbyterian Hospital, New York, New York.,Department of Urology, Weill Cornell Medicine, New York, New York.,Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, New York
| |
Collapse
|
183
|
Abstract
More than 40% of men with intermediate-risk or high-risk prostate cancer will experience a biochemical recurrence after radical prostatectomy. Clinical guidelines for the management of these patients largely focus on the use of salvage radiotherapy with or without systemic therapy. However, not all patients with biochemical recurrence will go on to develop metastases or die from their disease. The optimal pre-salvage therapy investigational workup for patients who experience biochemical recurrence should, therefore, include novel techniques such as PET imaging and genomic analysis of radical prostatectomy specimen tissue, as well as consideration of more traditional clinical variables such as PSA value, PSA kinetics, Gleason score and pathological stage of disease. In patients without metastatic disease, the only known curative intervention is salvage radiotherapy but, given the therapeutic burden of this treatment, importance must be placed on accurate timing of treatment, radiation dose, fractionation and field size. Systemic therapy also has a role in the salvage setting, both concurrently with radiotherapy and as salvage monotherapy.
Collapse
|
184
|
Latorzeff I, Ploussard G, Schick U, Benziane N, Pasquier D, Sargos P. [Salvage radiotherapy after prostatectomy - what do do in case of elevated post operative PSA or macroscopic relapse?]. Cancer Radiother 2021; 25:822-829. [PMID: 34702645 DOI: 10.1016/j.canrad.2021.08.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/12/2021] [Accepted: 08/13/2021] [Indexed: 02/03/2023]
Abstract
The management of the postoperative biological relapse of prostate cancer is most often based on salvage radiotherapy (RT) and a variable duration of hormone therapy (HT) in addition. The indications for RT±HT become more consensual for the adjuvant postoperative situation or in the event of a rising PSA level after a period where an undetectable PSA level was achieved. On the other hand, in the event of detectable PSA immediately postoperatively or in the event of a biological recurrence with macroscopic relapse in the prostate bed seen on conventional imaging assessment, the treatment options are still being evaluated. This article will describe these 2 situations and their current management but also will come through assessments with the contribution of modern imaging and new treatment options in terms of RT dose and RT±HT combination.
Collapse
Affiliation(s)
- I Latorzeff
- Département de radiothérapie-oncologie, Bât Atrium, clinique Pasteur, 1, rue de la Petite-Vitesse, 31300 Toulouse, France.
| | - G Ploussard
- Département d'urologie, clinique La Croix du Sud, Quint-Fonsegrives, France
| | - U Schick
- Département de radiothérapie-oncologie, CHU de Brest, Brest, France
| | - N Benziane
- Département de radiothérapie, institut Bergonié, 229 cours de l'Argonne, 33076 Bordeaux cedex, France
| | - D Pasquier
- Département universitaire de radiothérapie, centre O.-Lambret, 3, rue F.-Combemale, 59020 Lille, France
| | - P Sargos
- Département de radiothérapie, institut Bergonié, 229 cours de l'Argonne, 33076 Bordeaux cedex, France
| |
Collapse
|
185
|
Van Damme J, Tombal B, Collette L, Van Nieuwenhove S, Pasoglou V, Gérard T, Jamar F, Lhommel R, Lecouvet FE. Comparison of 68Ga-Prostate Specific Membrane Antigen (PSMA) Positron Emission Tomography Computed Tomography (PET-CT) and Whole-Body Magnetic Resonance Imaging (WB-MRI) with Diffusion Sequences (DWI) in the Staging of Advanced Prostate Cancer. Cancers (Basel) 2021; 13:cancers13215286. [PMID: 34771449 PMCID: PMC8582508 DOI: 10.3390/cancers13215286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/04/2021] [Accepted: 10/14/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Precise staging is key for the optimal management of advanced prostate cancer. PSMA PET-CT and WB-MRI outperform standard imaging technology for staging high-risk prostate cancer, but direct comparison between both modalities is lacking. The primary endpoint of our study was to compare the diagnostic accuracy of both techniques in the detection of lymph node, bone and visceral metastases against a best valuable comparator (BVC), defined as a consensus adjudication of all lesions on the basis of baseline and follow-up imaging, biological and clinical data and histopathologic confirmation when available. Knowing the diagnostic accuracy of both next generation imaging modalities might influence the diagnostic and therapeutic strategy in prostate cancer by tailoring therapy. However, the impact on treatment and patient outcome of an improved detection of metastases has not been determined yet. Abstract Background: Prostate specific membrane antigen (PSMA) positron emission tomography computed tomography (PET-CT) and whole-body magnetic resonance imaging (WB-MRI) outperform standard imaging technology for the detection of metastasis in prostate cancer (PCa). There are few direct comparisons between both modalities. This paper compares the diagnostic accuracy of PSMA PET-CT and WB-MRI for the detection of metastasis in PCa. One hundred thirty-four patients with newly diagnosed PCa (n = 81) or biochemical recurrence after curative treatment (n = 53) with high-risk features prospectively underwent PSMA PET-CT and WB-MRI. The diagnostic accuracy of both techniques for lymph node, skeletal and visceral metastases was compared against a best valuable comparator (BVC). Overall, no significant difference was detected between PSMA PET-CT and WB-MRI to identify metastatic patients when considering lymph nodes, skeletal and visceral metastases together (AUC = 0.96 (0.92–0.99) vs. 0.90 (0.85–0.95); p = 0.09). PSMA PET-CT, however, outperformed WB-MRI in the subgroup of patients with newly diagnosed PCa for the detection of lymph node metastases (AUC = 0.96 (0.92–0.99) vs. 0.86 (0.79–0.92); p = 0.0096). In conclusion, PSMA PET-CT outperforms WB-MRI for the detection of nodal metastases in primary staging of PCa.
Collapse
Affiliation(s)
- Julien Van Damme
- Department of Urology, Institut de Recherche Expérimentale et Clinique (IREC), Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, B-1200 Brussels, Belgium; (J.V.D.); (B.T.)
| | - Bertrand Tombal
- Department of Urology, Institut de Recherche Expérimentale et Clinique (IREC), Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, B-1200 Brussels, Belgium; (J.V.D.); (B.T.)
| | - Laurence Collette
- International Drug Development Institute (IDDI), B-1341 Louvain-la-Neuve, Belgium;
| | - Sandy Van Nieuwenhove
- Department of Radiology, Institut de Recherche Expérimentale et Clinique (IREC-IMAG), Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, B-1200 Brussels, Belgium; (S.V.N.); (V.P.)
| | - Vassiliki Pasoglou
- Department of Radiology, Institut de Recherche Expérimentale et Clinique (IREC-IMAG), Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, B-1200 Brussels, Belgium; (S.V.N.); (V.P.)
| | - Thomas Gérard
- Department of Nuclear Medicine, Institut de Recherche Expérimentale et Clinique (IREC-MIRO), Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, B-1200 Brussels, Belgium; (T.G.); (F.J.); (R.L.)
| | - François Jamar
- Department of Nuclear Medicine, Institut de Recherche Expérimentale et Clinique (IREC-MIRO), Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, B-1200 Brussels, Belgium; (T.G.); (F.J.); (R.L.)
| | - Renaud Lhommel
- Department of Nuclear Medicine, Institut de Recherche Expérimentale et Clinique (IREC-MIRO), Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, B-1200 Brussels, Belgium; (T.G.); (F.J.); (R.L.)
| | - Frédéric E. Lecouvet
- Department of Radiology, Institut de Recherche Expérimentale et Clinique (IREC-IMAG), Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, B-1200 Brussels, Belgium; (S.V.N.); (V.P.)
- Correspondence:
| |
Collapse
|
186
|
Miyahira AK, Soule HR. The History of Prostate-Specific Membrane Antigen as a Theranostic Target in Prostate Cancer: The Foundational Role of the Prostate Cancer Foundation. J Nucl Med 2021; 63:331-338. [PMID: 34675109 DOI: 10.2967/jnumed.121.262997] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/08/2021] [Indexed: 11/16/2022] Open
Abstract
Prostate-Specific Membrane Antigen (PSMA) is a credentialed imaging and therapy (theranostic) target for the detection and treatment of prostate cancer. PSMA-targeted positron emission tomography (PET) imaging and molecular radiotherapy (MRT) are promising evolving technologies that will improve the outcomes of prostate cancer patients. In anticipation of this new era in prostate cancer theranostics, this article will review the history of PSMA from discovery, through early and late stage clinical trials. Since 1993, the Prostate Cancer Foundation (PCF) has funded critical and foundational PSMA research that established this theranostic revolution. The history and role of PCF funding in this field will be discussed.
Collapse
|
187
|
Marcus C, Abiodun-Ojo OA, Jani AB, Schuster DM. Clinical utility of 18F-Fluciclovine PET/CT in recurrent prostate cancer with very low (≤0.3 ng/mL) prostate-specific antigen levels. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2021; 11:406-414. [PMID: 34754611 PMCID: PMC8569337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 09/22/2021] [Indexed: 06/13/2023]
Abstract
The purpose of this study was to evaluate 18F-fluciclovine PET/CT detection rates in the evaluation of biochemical recurrence in prostate cancer patients with very low (≤0.3 ng/mL) serum prostate-specific antigen (PSA) levels following definitive treatment. Prostate cancer patients with biochemical recurrence and very low serum PSA (≤0.3 ng/mL) who underwent clinical 18F-fluciclovine PET/CT were included in this single-institution retrospective study. PET/CT clinical reports at the time of interpretation were reviewed and categorized as positive or negative. In patients who had further evaluation with imaging and/or biopsy, the results were recorded to determine the true detection rate. Of the 64 eligible patients with very low serum PSA (median serum PSA of 0.17 ng/mL), 57.8% (37/64) scans were categorized as positive. Stratified by PSA levels, positivity rates were 43.8% (7/16), 60.0% (15/25) and 65.2% (15/23) for PSA<0.1 ng/mL, 0.1-<0.2 ng/mL and 0.2-≤0.3 ng/mL, respectively. The most common location of disease was the prostate bed (73%), followed by pelvic lymph nodes (22%) and distant disease (14%). In the small subset of patients who had further evaluation after a positive study (n=7), all had confirmed disease with a positive predictive value of 100%. In conclusion, among prostate cancer patients with biochemical recurrence, 18F-fluciclovine PET/CT is useful in patients with very low serum PSA of ≤0.3 ng/mL, with a 57.8% positivity rate, higher than previously reported. Though standard of truth could only be ascertained in 19% (7/37) of patients with a positive study, the positive predictive value was 100%.
Collapse
Affiliation(s)
- Charles Marcus
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology and Imaging Sciences, Emory University School of MedicineAtlanta, GA, USA
| | - Olayinka A Abiodun-Ojo
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology and Imaging Sciences, Emory University School of MedicineAtlanta, GA, USA
| | - Ashesh B Jani
- Winship Cancer Institute, Department of Radiation Oncology, Emory University School of MedicineAtlanta, GA, USA
| | - David M Schuster
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology and Imaging Sciences, Emory University School of MedicineAtlanta, GA, USA
| |
Collapse
|
188
|
Watanabe M, Kawai-Miyake K, Fushimi Y, Ishimori T, Nakajima A, Yoshimura M, Kikuchi M, Ohno K, Nakamoto Y. Application of a Flexible PET Scanner Combined with 3 T MRI Using Non-local Means Reconstruction: Qualitative and Quantitative Comparison with Whole-Body PET/CT. Mol Imaging Biol 2021; 24:167-176. [PMID: 34642900 DOI: 10.1007/s11307-021-01651-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/21/2021] [Accepted: 09/01/2021] [Indexed: 11/28/2022]
Abstract
PURPOSE Flexible positron emission tomography (fxPET) employing a non-local means reconstruction algorithm was designed to fit existing magnetic resonance imaging (MRI) systems. We aimed to compare the qualitative and quantitative performance of fxPET among fxPET with MR-based attenuation correction (MRAC), fxPET with CT-based attenuation correction (CTAC) using CT as a part of WB PET/CT, and whole-body (WB) PET/CT. PROCEDURES Sixteen patients with suspected head and neck cancer underwent 2-deoxy-2-[18F]fluoro-D-glucose WB PET/CT scans, followed by fxPET and 3 T MRI scans. Phantom data were compared among the three datasets. For registration accuracy, we measured the distance between the center of the tumor determined by fxPET and that in MRI. We compared image quality, detection rates, and quantitative values including maximal standardized uptake value (SUVmax), metabolic tumor volume (MTV), total lesion glycolysis (TLG), and tumor-to-muscle ratio (TMR) among the three datasets. RESULTS The phantom data in fxPET, except the percent contrast recoveries of 17-mm and 22-mm hot spheres, were inferior to those in WB PET/CT. The mean registration accuracy was 4.4 mm between fxPET using MRAC and MRI. The image quality was comparable between two fxPET datasets, but significantly inferior to WB PET/CT (p < 0.0001). In contrast, detection rates were comparable among the three datasets. SUVmax was significantly higher, and MTV and TLG were significantly lower in the two fxPET datasets compared with the WB PET/CT dataset (p < 0.005). There were no significant differences in SUVmax, MTV, and TLG between the two fxPET datasets or in TMR among the three datasets. All quantitative values had significantly positive correlations. CONCLUSIONS Compared with WB PET/CT, the phantom data and image quality were inferior in fxPET. However, the results of the detection rates and quantitative values suggested the clinical feasibility of fxPET.
Collapse
Affiliation(s)
- Masao Watanabe
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.,Department of Radiology, Kobe City Medical Center General Hospital, 2-1-1 Minatojima Minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Kanae Kawai-Miyake
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yasutaka Fushimi
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Takayoshi Ishimori
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Aya Nakajima
- Department of Radiation Oncology and Image-Applied Therapy, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Michio Yoshimura
- Department of Radiation Oncology and Image-Applied Therapy, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Masahiro Kikuchi
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kazuko Ohno
- Department of Radiological Technology, Kyoto College of Medical Science, 1-3 Imakita, Oyamahigashi-cho, Sonobe-cho, Nantan, Kyoto, 622-0041, Japan
| | - Yuji Nakamoto
- Department of Diagnostic Imaging and Nuclear Medicine, Graduate School of Medicine, Kyoto University, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.
| |
Collapse
|
189
|
Bratt O. Has the time come for routine use of PET/CT for guiding treatment of biochemical recurrence after radical prostatectomy? RE: JaniAB, et al. 18F-fluciclovine-PET/CT imaging versus conventional imaging alone to guide postprostatectomy salvage radiotherapy for prostate cancer (EMPIRE-1): a single centre, open-label, phase 2/3 randomised controlled trial. Lancet. 2021 May 22;55:1895-1904. Scand J Urol 2021; 55:420-421. [PMID: 34633894 DOI: 10.1080/21681805.2021.1942192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Ola Bratt
- Department of Urology, Institute of Clinical Science, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
190
|
Artigas C, Diamand R, Shagera QA, Plouznikoff N, Fokoue F, Otte FX, Gil T, Peltier A, Van Gestel D, Flamen P. Oligometastatic Disease Detection with 68Ga-PSMA-11 PET/CT in Hormone-Sensitive Prostate Cancer Patients (HSPC) with Biochemical Recurrence after Radical Prostatectomy: Predictive Factors and Clinical Impact. Cancers (Basel) 2021; 13:cancers13194982. [PMID: 34638466 PMCID: PMC8508549 DOI: 10.3390/cancers13194982] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 11/16/2022] Open
Abstract
Metastasis-directed therapy (MDT) in oligometastatic prostate cancer has the potential of delaying the start of androgen deprivation therapy (ADT) and disease progression. We aimed to analyze the efficacy of PSMA-PET/CT in detecting oligometastatic disease (OMD), to look for predictive factors of OMD, and to evaluate the impact of PSMA-PET/CT findings on clinical management. We retrospectively analyzed a homogeneous population of 196 hormone-sensitive prostate cancer patients (HSPC), considered potential candidates for MDT, with a PSMA-PET/CT performed at biochemical recurrence (BCR) after radical prostatectomy (RP). Multivariable logistic regression analysis was performed based on several clinico-pathological factors. Changes in clinical management before and after PSMA-PET/CT were analyzed. The OMD detection rate was 44% for a total positivity rate of 60%. PSMA-PET/CT positivity was independently related to PSA (OR (95% CI), p) (1.7 (1.3-2.3), p < 0.0001) and PSAdt (0.4 (0.2-0.8), p = 0.013), and OMD detection was independently related to PSA (1.6 (1.2-2.2), p = 0.001) and no previous salvage therapy (0.3 (0.1-0.9), p = 0.038). A treatment change was observed in 58% of patients, mostly to perform MDT after OMD detection (60% of changes). This study showed that PSMA-PET/CT is an excellent imaging technique to detect OMD early in HSPC patients with BCR after RP, changing therapeutic management mostly into MDT.
Collapse
Affiliation(s)
- Carlos Artigas
- Department of Nuclear Medicine, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (Q.A.S.); (F.F.); (P.F.)
- Correspondence: ; Tel.: +32-2-541-32-40
| | - Romain Diamand
- Department of Urology, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (R.D.); (A.P.)
| | - Qaid Ahmed Shagera
- Department of Nuclear Medicine, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (Q.A.S.); (F.F.); (P.F.)
| | - Nicolas Plouznikoff
- Department of Nuclear Medicine, Centre Hospitalier de l’Université de Montréal, Montréal, QC H2X 3E4, Canada;
| | - Fabrice Fokoue
- Department of Nuclear Medicine, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (Q.A.S.); (F.F.); (P.F.)
| | - François-Xavier Otte
- Department of Radiation Oncology, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (F.-X.O.); (D.V.G.)
| | - Thierry Gil
- Department of Medical Oncology, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium;
| | - Alexandre Peltier
- Department of Urology, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (R.D.); (A.P.)
| | - Dirk Van Gestel
- Department of Radiation Oncology, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (F.-X.O.); (D.V.G.)
| | - Patrick Flamen
- Department of Nuclear Medicine, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (Q.A.S.); (F.F.); (P.F.)
| |
Collapse
|
191
|
Stuschke M, Hadaschik B. [Radiation therapy in high-risk and very high-risk localized or locally advanced prostate cancer : Prostate-only versus prostate plus whole-pelvic?]. Strahlenther Onkol 2021; 197:939-942. [PMID: 34476530 PMCID: PMC8458169 DOI: 10.1007/s00066-021-01825-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/06/2021] [Indexed: 11/11/2022]
Affiliation(s)
- Martin Stuschke
- Klinik für Strahlentherapie, Universitätsklinikum Essen, Hufelandstr. 55, 45147, Essen, Deutschland.
| | - Boris Hadaschik
- Klinik für Urologie, Universitätsklinikum Essen, Hufelandstr. 55, 45147, Essen, Deutschland.
| |
Collapse
|
192
|
Sandhu S, Moore CM, Chiong E, Beltran H, Bristow RG, Williams SG. Prostate cancer. Lancet 2021; 398:1075-1090. [PMID: 34370973 DOI: 10.1016/s0140-6736(21)00950-8] [Citation(s) in RCA: 320] [Impact Index Per Article: 80.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 12/12/2022]
Abstract
The management of prostate cancer continues to evolve rapidly, with substantial advances being made in understanding the genomic landscape and biology underpinning both primary and metastatic prostate cancer. Similarly, the emergence of more sensitive imaging methods has improved diagnostic and staging accuracy and refined surveillance strategies. These advances have introduced personalised therapeutics to clinical practice, with treatments targeting genomic alterations in DNA repair pathways now clinically validated. An important shift in the therapeutic framework for metastatic disease has taken place, with metastatic-directed therapies being evaluated for oligometastatic disease, aggressive management of the primary lesion shown to benefit patients with low-volume metastatic disease, and with several novel androgen pathway inhibitors significantly improving survival when used as a first-line therapy for metastatic disease. Research into the molecular characterisation of localised, recurrent, and progressive disease will undoubtedly have an impact on clinical management. Similarly, emerging research into novel therapeutics, such as targeted radioisotopes and immunotherapy, holds much promise for improving the lives of patients with prostate cancer.
Collapse
Affiliation(s)
- Shahneen Sandhu
- Division of Cancer Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | | | - Edmund Chiong
- Department of Urology and Department of Surgery, National University of Singapore, Singapore
| | | | - Robert G Bristow
- Manchester Cancer Research Centre and University of Manchester, Manchester, UK
| | - Scott G Williams
- Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
193
|
le Guevelou J, Achard V, Mainta I, Zaidi H, Garibotto V, Latorzeff I, Sargos P, Ménard C, Zilli T. PET/CT-Based Salvage Radiotherapy for Recurrent Prostate Cancer After Radical Prostatectomy: Impact on Treatment Management and Future Directions. Front Oncol 2021; 11:742093. [PMID: 34532294 PMCID: PMC8438304 DOI: 10.3389/fonc.2021.742093] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 08/09/2021] [Indexed: 12/25/2022] Open
Abstract
Biochemical recurrence is a clinical situation experienced by 20 to 40% of prostate cancer patients treated with radical prostatectomy (RP). Prostate bed (PB) radiation therapy (RT) remains the mainstay salvage treatment, although it remains non-curative for up to 30% of patients developing further recurrence. Positron emission tomography with computed tomography (PET/CT) using prostate cancer-targeting radiotracers has emerged in the last decade as a new-generation imaging technique characterized by a better restaging accuracy compared to conventional imaging. By adapting targeting of recurrence sites and modulating treatment management, implementation in clinical practice of restaging PET/CT is challenging the established therapeutic standards born from randomized controlled trials. This article reviews the potential impact of restaging PET/CT on changes in the management of recurrent prostate cancer after RP. Based on PET/CT findings, it addresses potential adaptation of RT target volumes and doses, as well as use of androgen-deprivation therapy (ADT). However, the impact of such management changes on the oncological outcomes of PET/CT-based salvage RT strategies is as yet unknown.
Collapse
Affiliation(s)
- Jennifer le Guevelou
- Division of Radiation Oncology, Geneva University Hospital, Geneva, Switzerland.,Division of Radiation Oncology, Centre François Baclesse, Caen, France
| | - Vérane Achard
- Division of Radiation Oncology, Geneva University Hospital, Geneva, Switzerland.,Faculty of Medicine, Geneva University, Geneva, Switzerland
| | - Ismini Mainta
- Division of Nuclear Medicine and Molecular Imaging, Diagnostic Department, Geneva University Hospital, Geneva, Switzerland
| | - Habib Zaidi
- Faculty of Medicine, Geneva University, Geneva, Switzerland.,Division of Nuclear Medicine and Molecular Imaging, Diagnostic Department, Geneva University Hospital, Geneva, Switzerland.,Geneva Neuroscience Center, Geneva University, Geneva, Switzerland.,Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, Netherlands.,Department of Nuclear Medicine, University of Southern Denmark, Odense, Denmark
| | - Valentina Garibotto
- Faculty of Medicine, Geneva University, Geneva, Switzerland.,Division of Nuclear Medicine and Molecular Imaging, Diagnostic Department, Geneva University Hospital, Geneva, Switzerland.,Geneva Neuroscience Center, Geneva University, Geneva, Switzerland
| | - Igor Latorzeff
- Department of Radiation Oncology, Groupe Oncorad-Garonne, Clinique Pasteur, Toulouse, France
| | - Paul Sargos
- Department of Radiation Oncology, Institut Bergonié, Bordeaux, France
| | - Cynthia Ménard
- Department of Radiation Oncology, Centre Hospitalier de l'Université de Montréal (CHUM), Montréal, QC, Canada
| | - Thomas Zilli
- Division of Radiation Oncology, Geneva University Hospital, Geneva, Switzerland.,Faculty of Medicine, Geneva University, Geneva, Switzerland
| |
Collapse
|
194
|
Hope TA, Eiber M, Armstrong WR, Juarez R, Murthy V, Lawhn-Heath C, Behr SC, Zhang L, Barbato F, Ceci F, Farolfi A, Schwarzenböck SM, Unterrainer M, Zacho HD, Nguyen HG, Cooperberg MR, Carroll PR, Reiter RE, Holden S, Herrmann K, Zhu S, Fendler WP, Czernin J, Calais J. Diagnostic Accuracy of 68Ga-PSMA-11 PET for Pelvic Nodal Metastasis Detection Prior to Radical Prostatectomy and Pelvic Lymph Node Dissection: A Multicenter Prospective Phase 3 Imaging Trial. JAMA Oncol 2021; 7:1635-1642. [PMID: 34529005 DOI: 10.1001/jamaoncol.2021.3771] [Citation(s) in RCA: 208] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Importance The presence of pelvic nodal metastases at radical prostatectomy is associated with biochemical recurrence after prostatectomy. Objective To assess the accuracy of prostate-specific membrane antigen (PSMA) 68Ga-PSMA-11 positron emission tomographic (PET) imaging for the detection of pelvic nodal metastases compared with histopathology at time of radical prostatectomy and pelvic lymph node dissection. Design, Setting, and Participants This investigator-initiated prospective multicenter single-arm open-label phase 3 imaging trial of diagnostic efficacy enrolled 764 patients with intermediate- to high-risk prostate cancer considered for prostatectomy at University of California, San Francisco and University of California, Los Angeles from December 2015 to December 2019. Data analysis took place from October 2018 to July 2021. Interventions Imaging scan with 3 to 7 mCi of 68Ga-PSMA-11 PET. Main Outcomes and Measures The primary end point was the sensitivity and specificity for the detection pelvic lymph nodes compared with histopathology on a per-patient basis using nodal region correlation. Each scan was read centrally by 3 blinded independent central readers, and a majority rule was used for analysis. Results A total of 764 men (median [interquartile range] age, 69 [63-73] years) underwent 1 68Ga-PSMA-11 PET imaging scan for primary staging, and 277 of 764 (36%) subsequently underwent prostatectomy with lymph node dissection (efficacy analysis cohort). Based on pathology reports, 75 of 277 patients (27%) had pelvic nodal metastasis. Results of 68Ga-PSMA-11 PET were positive in 40 of 277 (14%), 2 of 277 (1%), and 7 of 277 (3%) of patients for pelvic nodal, extrapelvic nodal, and bone metastatic disease. Sensitivity, specificity, positive predictive value, and negative predictive value for pelvic nodal metastases were 0.40 (95% CI, 0.34-0.46), 0.95 (95% CI, 0.92-0.97), 0.75 (95% CI, 0.70-0.80), and 0.81 (95% CI, 0.76-0.85), respectively. Of the 764 patients, 487 (64%) did not undergo prostatectomy, of which 108 were lost to follow-up. Patients with follow-up instead underwent radiotherapy (262 of 379 [69%]), systemic therapy (82 of 379 [22%]), surveillance (16 of 379 [4%]), or other treatments (19 of 379 [5%]). Conclusions and Relevance This phase 3 diagnostic efficacy trial found that in men with intermediate- to high-risk prostate cancer who underwent radical prostatectomy and lymph node dissection, the sensitivity and specificity of 68Ga-PSMA-11 PET were 0.40 and 0.95, respectively. This academic collaboration is the largest known to date and formed the foundation of a New Drug Application for 68Ga-PSMA-11. Trial Registration ClinicalTrials.gov Identifiers: NCT03368547, NCT02611882, and NCT02919111.
Collapse
Affiliation(s)
- Thomas A Hope
- Department of Radiology and Biomedical Imaging, University of California, San Francisco.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco.,Department of Radiology and Biomedical Imaging, San Francisco VA Medical Center, San Francisco, California
| | - Matthias Eiber
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles.,Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Nuclear Medicine, Munich, Germany
| | - Wesley R Armstrong
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles
| | - Roxanna Juarez
- Department of Radiology and Biomedical Imaging, University of California, San Francisco
| | - Vishnu Murthy
- Department of Radiology and Biomedical Imaging, University of California, San Francisco
| | - Courtney Lawhn-Heath
- Department of Radiology and Biomedical Imaging, University of California, San Francisco
| | - Spencer C Behr
- Department of Radiology and Biomedical Imaging, University of California, San Francisco
| | - Li Zhang
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco.,Department of Radiology and Biomedical Imaging, San Francisco VA Medical Center, San Francisco, California
| | - Francesco Barbato
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Nuclear Medicine, Munich, Germany
| | - Francesco Ceci
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Andrea Farolfi
- Division of Nuclear Medicine, Azienda Ospedaliero-Universitaria Di Bologna, Bologna, Italy
| | | | - Marcus Unterrainer
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Helle D Zacho
- Department of Nuclear Medicine and Clinical Cancer Research Centre, Aalborg University Hospital, Aalborg, Denmark
| | - Hao G Nguyen
- Department of Urology, University of California, San Francisco
| | | | - Peter R Carroll
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco.,Department of Urology, University of California, San Francisco
| | - Robert E Reiter
- Institute of Urologic Oncology, University of California, Los Angeles.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles
| | - Stuart Holden
- Institute of Urologic Oncology, University of California, Los Angeles.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles
| | - Ken Herrmann
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles.,Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | - Shaojun Zhu
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles
| | - Wolfgang P Fendler
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles.,Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | - Johannes Czernin
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles.,Institute of Urologic Oncology, University of California, Los Angeles.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles
| | - Jeremie Calais
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles.,Institute of Urologic Oncology, University of California, Los Angeles.,Jonsson Comprehensive Cancer Center, University of California, Los Angeles
| |
Collapse
|
195
|
Margel D, Bernstine H, Groshar D, Ber Y, Nezrit O, Segal N, Yakimov M, Baniel J, Domachevsky L. Diagnostic Performance of 68Ga Prostate-specific Membrane Antigen PET/MRI Compared with Multiparametric MRI for Detecting Clinically Significant Prostate Cancer. Radiology 2021; 301:379-386. [PMID: 34463555 DOI: 10.1148/radiol.2021204093] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Background Gallium 68 (68Ga) prostate-specific membrane antigen (PSMA) PET/MRI may improve detection of clinically significant prostate cancer (CSPC). Purpose To compare the sensitivity and specificity of 68Ga-PSMA PET/MRI with multiparametric MRI for detecting CSPC. Materials and Methods Men with prostate specific antigen levels of 2.5-20 ng/mL prospectively underwent 68Ga-PSMA PET/MRI, including multiparametric MRI sequences, between June 2019 and March 2020. Imaging was evaluated independently by two radiologists by using the Prostate Imaging Reporting and Data System (PI-RADS) version 2.1. Sensitivity and specificity for CSPC (International Society of Urological Pathology grade group ≥ 2) were compared for 68Ga-PSMA PET/MRI and multiparametric MRI by using the McNemar test. Decision curve analysis compared the net benefit of each imaging strategy. Results Ninety-nine men (median age, 67 years; interquartile range, 62-71 years) were included; 79% (78 of 99) underwent biopsy. CSPC was detected in 32% (25 of 78). For CSPC, specificity was higher for 68Ga-PSMA PET/MRI than multiparametric MRI (76% [95% CI: 62, 86] vs 49% [95% CI: 35, 63], respectively; P < .001). Sensitivity was similar (88% [95% CI: 69, 98] vs 92% [95% CI: 74, 99], respectively; P > .99). For PI-RADS 3 lesions, specificity was also higher for 68Ga-PSMA PET/MRI than for multiparametric MRI: 86% (95% CI: 73, 95) versus 59% (95% CI: 43, 74), respectively (P = .002). Decision curve analysis showed that biopsies targeted to PSMA uptake increased the net benefit of multiparametric MRI only among PI-RADS 3 lesions. The net benefit of targeted biopsy for a PI-RADS 3 lesion with PSMA uptake was higher across all threshold probabilities over 8%. The net benefit of targeted biopsy was similar for PI-RADS 4 and 5 lesions, regardless of PSMA uptake. Conclusions Gallium 68 prostate-specific membrane antigen PET/MRI improved specificity for clinically significant prostate cancer compared with multiparametric MRI, particularly in Prostate Imaging Reporting and Data System grade 3 lesions. © RSNA, 2021 Online supplemental material is available for this article. See also the editorial by Williams and Estes in this issue.
Collapse
Affiliation(s)
- David Margel
- From the Division of Urology (D.M., Y.B., O.N., N.S., J.B.), Department of Nuclear Medicine (H.B., D.G.), and Department of Pathology (M.Y.), Rabin Medical Center, 39 Jabotinski Rd, Petah-Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel (D.M., H.B., D.G., J.B., L.D.); Department of Nuclear Medicine, Assuta Medical Center, Tel Aviv, Israel (H.B., D.G.); and Department of Nuclear Medicine, Sheba Medical Center, Tel Hashomer, Israel (L.D.)
| | - Hanna Bernstine
- From the Division of Urology (D.M., Y.B., O.N., N.S., J.B.), Department of Nuclear Medicine (H.B., D.G.), and Department of Pathology (M.Y.), Rabin Medical Center, 39 Jabotinski Rd, Petah-Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel (D.M., H.B., D.G., J.B., L.D.); Department of Nuclear Medicine, Assuta Medical Center, Tel Aviv, Israel (H.B., D.G.); and Department of Nuclear Medicine, Sheba Medical Center, Tel Hashomer, Israel (L.D.)
| | - David Groshar
- From the Division of Urology (D.M., Y.B., O.N., N.S., J.B.), Department of Nuclear Medicine (H.B., D.G.), and Department of Pathology (M.Y.), Rabin Medical Center, 39 Jabotinski Rd, Petah-Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel (D.M., H.B., D.G., J.B., L.D.); Department of Nuclear Medicine, Assuta Medical Center, Tel Aviv, Israel (H.B., D.G.); and Department of Nuclear Medicine, Sheba Medical Center, Tel Hashomer, Israel (L.D.)
| | - Yaara Ber
- From the Division of Urology (D.M., Y.B., O.N., N.S., J.B.), Department of Nuclear Medicine (H.B., D.G.), and Department of Pathology (M.Y.), Rabin Medical Center, 39 Jabotinski Rd, Petah-Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel (D.M., H.B., D.G., J.B., L.D.); Department of Nuclear Medicine, Assuta Medical Center, Tel Aviv, Israel (H.B., D.G.); and Department of Nuclear Medicine, Sheba Medical Center, Tel Hashomer, Israel (L.D.)
| | - Orian Nezrit
- From the Division of Urology (D.M., Y.B., O.N., N.S., J.B.), Department of Nuclear Medicine (H.B., D.G.), and Department of Pathology (M.Y.), Rabin Medical Center, 39 Jabotinski Rd, Petah-Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel (D.M., H.B., D.G., J.B., L.D.); Department of Nuclear Medicine, Assuta Medical Center, Tel Aviv, Israel (H.B., D.G.); and Department of Nuclear Medicine, Sheba Medical Center, Tel Hashomer, Israel (L.D.)
| | - Niv Segal
- From the Division of Urology (D.M., Y.B., O.N., N.S., J.B.), Department of Nuclear Medicine (H.B., D.G.), and Department of Pathology (M.Y.), Rabin Medical Center, 39 Jabotinski Rd, Petah-Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel (D.M., H.B., D.G., J.B., L.D.); Department of Nuclear Medicine, Assuta Medical Center, Tel Aviv, Israel (H.B., D.G.); and Department of Nuclear Medicine, Sheba Medical Center, Tel Hashomer, Israel (L.D.)
| | - Maxim Yakimov
- From the Division of Urology (D.M., Y.B., O.N., N.S., J.B.), Department of Nuclear Medicine (H.B., D.G.), and Department of Pathology (M.Y.), Rabin Medical Center, 39 Jabotinski Rd, Petah-Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel (D.M., H.B., D.G., J.B., L.D.); Department of Nuclear Medicine, Assuta Medical Center, Tel Aviv, Israel (H.B., D.G.); and Department of Nuclear Medicine, Sheba Medical Center, Tel Hashomer, Israel (L.D.)
| | - Jack Baniel
- From the Division of Urology (D.M., Y.B., O.N., N.S., J.B.), Department of Nuclear Medicine (H.B., D.G.), and Department of Pathology (M.Y.), Rabin Medical Center, 39 Jabotinski Rd, Petah-Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel (D.M., H.B., D.G., J.B., L.D.); Department of Nuclear Medicine, Assuta Medical Center, Tel Aviv, Israel (H.B., D.G.); and Department of Nuclear Medicine, Sheba Medical Center, Tel Hashomer, Israel (L.D.)
| | - Liran Domachevsky
- From the Division of Urology (D.M., Y.B., O.N., N.S., J.B.), Department of Nuclear Medicine (H.B., D.G.), and Department of Pathology (M.Y.), Rabin Medical Center, 39 Jabotinski Rd, Petah-Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel (D.M., H.B., D.G., J.B., L.D.); Department of Nuclear Medicine, Assuta Medical Center, Tel Aviv, Israel (H.B., D.G.); and Department of Nuclear Medicine, Sheba Medical Center, Tel Hashomer, Israel (L.D.)
| |
Collapse
|
196
|
Brenneman RJ, Baumann BC, Gay HA, Michalski JM. Regarding the Use of PSMA PET-CT Versus Conventional Imaging for Assessing the Value of Prophylactic Whole-Pelvis Radiation for High-Risk Prostate Cancer. J Clin Oncol 2021; 39:2847-2848. [PMID: 34086488 DOI: 10.1200/jco.21.00634] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Randall J Brenneman
- Randall J. Brenneman, MD, PhD, Brian C. Baumann, MD, Hiram A. Gay, MD, and Jeff M. Michalski, MD, MBA, Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO
| | - Brian C Baumann
- Randall J. Brenneman, MD, PhD, Brian C. Baumann, MD, Hiram A. Gay, MD, and Jeff M. Michalski, MD, MBA, Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO
| | - Hiram A Gay
- Randall J. Brenneman, MD, PhD, Brian C. Baumann, MD, Hiram A. Gay, MD, and Jeff M. Michalski, MD, MBA, Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO
| | - Jeff M Michalski
- Randall J. Brenneman, MD, PhD, Brian C. Baumann, MD, Hiram A. Gay, MD, and Jeff M. Michalski, MD, MBA, Department of Radiation Oncology, Washington University School of Medicine, St Louis, MO
| |
Collapse
|
197
|
Galgano SJ, McDonald AM, Rais-Bahrami S, Porter KK, Choudhary G, Burgan C, Bhambhvani P, Nix JW, Morgan DE, Li Y, Thomas JV, McConathy J. Utility of 18F-Fluciclovine PET/MRI for Staging Newly Diagnosed High-Risk Prostate Cancer and Evaluating Response to Initial Androgen Deprivation Therapy: A Prospective Single-Arm Pilot Study. AJR Am J Roentgenol 2021; 217:720-729. [PMID: 33052718 PMCID: PMC9170127 DOI: 10.2214/ajr.20.24509] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND. Despite advances in prostate cancer treatment, rates of biochemical recurrence remain high, relating to lack of detection of small-volume metastatic disease using conventional imaging for initial staging. OBJECTIVE. The purpose of this study was to assess the potential use of 18F-fluciclovine PET/MRI for initial staging of high-risk prostate cancer and evaluating response to androgen deprivation therapy (ADT). METHODS. This prospective clinical trial enrolled 14 men with newly diagnosed high-risk prostate cancer and negative or equivocal conventional staging imaging for metastatic disease between January 2018 and February 2019. All patients underwent pretreatment 18F-fluciclovine PET/MRI including multiparametric prostate MRI; 12 underwent 18F-fluciclovine PET/MRI after surgery or between ADT and radiotherapy. Confidence in identification of the primary intraprostatic lesion and nodal metastases was independently rated on a 0-3 Likert scale by three readers with nuclear medicine experience for 18F-fluciclovine PET/MRI and three readers with abdominal imaging experience for MRI alone. Findings scored as 2 or 3 by at least two readers of a given modality were considered positive. A single reader measured SUVmean, SUVmax, and volume of the MRI-defined intraprostatic lesion and SUVmax of suspicious lymph nodes on PET before and after initiation of ADT. Changes in SUV were analyzed using nonparametric Wilcox-on signed-rank tests. RESULTS. The biopsy-proven lesion in the prostate gland was accurately identified in all 14 patients on both MRI and 18F-fluciclovine PET/MRI. Suspected nodal metastases were detected in three patients on MRI and seven patients on 18F-fluciclovine PET/MRI. After ADT, all patients showed decreased activity within the intraprostatic lesion and/or all suspicious lymph nodes. The primary lesion SUVmean was 4.5 ± 1.1 (range, 2.7-6.5) before treatment and 2.4 ± 1.1 (range, 0.0-3.6) after initiation of ADT (p = .008). For suspicious lymph nodes, the pretreatment SUVmax was 5.5 ± 3.7 (range, 2.8-12.7) and the post-treatment SUVmax was 2.8 ± 1.4 (range, 1.4-5.5) (p = .03). CONCLUSION.18F-labeled fluciclovine PET/MRI shows potential utility in initial staging of high-risk prostate cancer and in evaluating response to ADT. CLINICAL IMPACT. Given the FDA approval and widespread availability of 18F-fluciclovine, the findings could have an impact in the immediate future in guiding initial management of patients with prostate cancer. TRIAL REGISTRATION. ClinicalTrials.gov NCT03264456.
Collapse
Affiliation(s)
- Samuel J Galgano
- Department of Radiology, University of Alabama at Birmingham, 619 19th St S, JT N325, Birmingham, AL 35249
- O'Neal Comprehensive Cancer Center at UAB, University of Alabama at Birmingham, Birmingham, AL
| | - Andrew M McDonald
- O'Neal Comprehensive Cancer Center at UAB, University of Alabama at Birmingham, Birmingham, AL
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham, Birmingham, AL
| | - Soroush Rais-Bahrami
- Department of Radiology, University of Alabama at Birmingham, 619 19 St S, JT N325, Birmingham, AL 35249
- O'Neal Comprehensive Cancer Center at UAB, University of Alabama at Birmingham, Birmingham, AL
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL
| | - Kristin K Porter
- Department of Radiology, University of Alabama at Birmingham, 619 19 St S, JT N325, Birmingham, AL 35249
| | - Gagandeep Choudhary
- Department of Radiology, University of Alabama at Birmingham, 619 19 St S, JT N325, Birmingham, AL 35249
| | - Constantine Burgan
- Department of Radiology, University of Alabama at Birmingham, 619 19 St S, JT N325, Birmingham, AL 35249
| | - Pradeep Bhambhvani
- Department of Radiology, University of Alabama at Birmingham, 619 19 St S, JT N325, Birmingham, AL 35249
| | - Jeffrey W Nix
- O'Neal Comprehensive Cancer Center at UAB, University of Alabama at Birmingham, Birmingham, AL
- Department of Urology, University of Alabama at Birmingham, Birmingham, AL
| | - Desiree E Morgan
- Department of Radiology, University of Alabama at Birmingham, 619 19 St S, JT N325, Birmingham, AL 35249
- O'Neal Comprehensive Cancer Center at UAB, University of Alabama at Birmingham, Birmingham, AL
| | - Yufeng Li
- Department of Radiology, University of Alabama at Birmingham, 619 19 St S, JT N325, Birmingham, AL 35249
- O'Neal Comprehensive Cancer Center at UAB, University of Alabama at Birmingham, Birmingham, AL
- Division of Preventive Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - John V Thomas
- Department of Radiology, University of Alabama at Birmingham, 619 19 St S, JT N325, Birmingham, AL 35249
| | - Jonathan McConathy
- Department of Radiology, University of Alabama at Birmingham, 619 19 St S, JT N325, Birmingham, AL 35249
- O'Neal Comprehensive Cancer Center at UAB, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
198
|
de Kouchkovsky I, Aggarwal R, Hope TA. Prostate-specific membrane antigen (PSMA)-based imaging in localized and advanced prostate cancer: a narrative review. Transl Androl Urol 2021; 10:3130-3143. [PMID: 34430416 PMCID: PMC8350242 DOI: 10.21037/tau-20-1047] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/30/2020] [Indexed: 12/13/2022] Open
Abstract
Combined molecular and morphologic imaging modalities have emerged in recent years as an alternative to conventional imaging in prostate cancer (PC). In particular, novel prostate-specific membrane antigen (PSMA) radiotracers have demonstrated increased sensitivity and specificity for the initial staging of men with clinically localized PC, as well as for PC detection in the setting of biochemical recurrence (BCR). Molecular imaging is increasingly used to guide treatment decisions in these patients—though its impact on survival has yet to be established. Improved PC detection in men with BCR has also helped to identify a subset of patients with oligometastatic disease. The optimal management of oligometastatic PC and the role of metastasis-directed therapies (MDT) are the subjects of ongoing studies. In comparison to clinically localized or biochemically recurrent PC, the role of molecular imaging in men with advanced disease is less established. In metastatic castration-resistant PC (mCRPC), PSMA-based imaging has primarily been investigated as a companion diagnostic tool to predict and monitor response to PSMA-targeted radioligand therapy (RLT). More recent efforts have focused on using molecular imaging to monitor treatment response to conventional chemohormonal therapies. However, despite promising early results, several barriers remain to the widespread use of PSMA-based imaging in metastatic PC: temporary flares in PSMA uptake have been described in a subset of patients after initiation of therapy, and the underlying mechanism and clinical implications of this phenomenon are still poorly understood. Furthermore, whereas PSMA is invariably expressed in hormone-sensitive PC, loss of PSMA expression is increasingly recognized in a subset of mCRPC patients with aggressive disease. Although this may limit the use of PSMA-based imaging as a standalone modality in advanced PC, loss of PSMA uptake may also provide non-invasive and clinically relevant molecular insight on patients’ underlying tumor biology.
Collapse
Affiliation(s)
- Ivan de Kouchkovsky
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Rahul Aggarwal
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Thomas A Hope
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA.,Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
199
|
Highlighting recent treatment advances in metastatic prostate cancer: expanding the treatment arsenal. Curr Opin Oncol 2021; 33:252-256. [PMID: 33818543 DOI: 10.1097/cco.0000000000000723] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE OF REVIEW Present highlights from recent research examining the treatment of advanced prostate cancer. RECENT FINDINGS Although debate remains about the optimal sequencing of docetaxel and novel androgen directed therapies in addition to androgen deprivation therapy (ADT) in the treatment of men with new metastatic prostate cancer, the novel LHRH antagonist relugolix seems poised to become an appealing option in a choice of initial ADT. Novel radioisotopes, genomically selected therapies, and immune therapy combinations show progress in opening up new treatment options for men with castration-resistant prostate cancer. SUMMARY Although no clear consensus has emerged, evolving data continue to refine the selection of systemic therapies in treatment naïve metastatic prostate cancer. With potentially less cardiotoxic androgen deprivation therapies, novel radioisotopes, targeted pharmaceuticals, and immune therapy combinations, progress appears to be on the horizon in improving outcomes for men with advanced prostate cancer.
Collapse
|
200
|
Abstract
The role of PET imaging with 11C-choline and 18F-fluciclovine in evaluating patients with prostate cancer (PCa) has become more important over the years and has been incorporated into the NCCN guidelines. A new generation of PET radiotracers targeting the prostate-specific membrane antigen (PSMA) is widely used outside the United States to evaluate patients with primary PCa and PCa recurrence. PET imaging influences treatment planning and demonstrates a significantly higher disease detection rate than conventional imaging such as computed tomography and MR imaging. Early data indicate that using PET radiotracers such as 18F-fluciclovine and PSMA improves patient outcomes. 68-Ga-PSMA-11 and 18F-DCFPyL-PET/CT were recently approved by the US Food & Drug Administration (FDA) for clinical use. Other PSMA radiotracers, including fluorinated variants, will likely gain FDA approval in the not-too-distant future.
Collapse
|