1
|
Chen G, Li Y, Geng S, Lv L, Wang Y, Li X, Chen S, Shi B. Evaluating the Heterogeneity of Advanced Prostate Cancer by 18F-DCFPyL and 18F-FDG PET/CT in a Prospective Cohort. Prostate 2025; 85:749-757. [PMID: 40045414 DOI: 10.1002/pros.24881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/20/2025] [Accepted: 02/18/2025] [Indexed: 04/30/2025]
Abstract
PURPOSE 18F-DCFPyL (targeted PSMA) and 18F-FDG dual-tracer PET/CT combination with next-generation sequencing was applied in a prospective cohort of men with prostate cancer to identify the clinical and genetic characteristics with heterogeneous PET/CT imaging features. METHODS 104 men with documented prostate cancer underwent 18F-DCFPyL and 18F-FDG PET/CT, of which 83 underwent next-generation sequencing for detecting variation of AR, TP53, RB1, PTEN, etc. Lesions were classified into DCFPyL+FDG± lesions and DCFPyL-FDG+ lesions and analyzed for heterogeneous distribution. We divided the patients with positive lesions into DCFPyL+FDG± group and DCFPyL-FDG+ group, then compared the differences in clinical features and genetic mutations between the two groups with CRPC. RESULTS Overall, 92 men had positive lesions detected. By comparing lesion distribution with the DCFPyL+FDG ± , DCFPyL-FDG+ disease had higher proportions of visceral metastases (4.1% vs. 1.0%, p = 0.002). DCFPyL-FDG+ was more frequently found in CRPC cohorts, and in the CRPC cohort, patients with DCFPyL-FDG+ lesions often had worse PSA response. Exploratory analysis showed that TP53 and/or RB1 mutations might be a risk factor for DCFPyL-FDG+ disease (OR = 10.625, 95% CI 3.492-32.332, p < 0.001). CONCLUSION Patients with DCFPyL-FDG+ lesions were more likely to have visceral metastases detected, be found in castration-resistant cohorts, have TP53 and/or RB1 mutations detected, and have poor therapeutic response compared to patients with DCFPyL+FDG± lesions. Therefore, dual-tracer (18F-DCFPyL and 18F-FDG) PET/CT is recommended for patients with low PSMA expression incompatible with the true burden of the disease and those with TP53 and/or RB1 mutations to better evaluate the disease burden, tumor heterogeneity, and prognosis.
Collapse
Affiliation(s)
- GuangHao Chen
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - YueKai Li
- Department of Nuclear Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - ShangZhen Geng
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - LinChen Lv
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Yong Wang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Xin Li
- Department of Nuclear Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - ShouZhen Chen
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - BenKang Shi
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
2
|
Belge Bilgin G, Lucien-Matteoni F, Chaudhuri AA, Orme JJ, Childs DS, Muniz M, Li GG, Chauhan PS, Lee S, Gupta S, Thorpe MP, Johnson DR, Johnson GB, Kendi AT, Sartor O. Current and future directions in theranostics for neuroendocrine prostate cancer. Cancer Treat Rev 2025; 136:102941. [PMID: 40239461 DOI: 10.1016/j.ctrv.2025.102941] [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: 02/05/2025] [Revised: 04/05/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
Neuroendocrine prostate cancer (NEPC) is rare at the time of initial diagnosis but much more common in patients treated with the combination of androgen deprivation therapy (ADT) and androgen receptor pathway inhibitors (ARPI) such as abiraterone and enzalutamide. NEPC is typically characterized by the loss of prostate-specific membrane antigen (PSMA) expression while exhibiting variable neuroendocrine markers. Recent advancements in nuclear medicine have provided a promising avenue for the development of molecular imaging techniques and targeted therapies tailored to NEPC. This review examines the current and future role of theranostics in the diagnosis and management of NEPC and explores potential future directions in this rapidly evolving field.
Collapse
Affiliation(s)
| | | | | | - Jacob J Orme
- Department of Oncology, Mayo Clinic Rochester, MN, USA
| | | | - Miguel Muniz
- Department of Oncology, Mayo Clinic Rochester, MN, USA
| | | | | | - SeungBaek Lee
- Department of Radiology, Mayo Clinic Rochester, MN, USA
| | - Sounak Gupta
- Department of Laboratory Medicine and Pathology, Mayo Clinic Rochester, MN, USA
| | | | | | - Geoffrey B Johnson
- Department of Radiology, Mayo Clinic Rochester, MN, USA; Department of Immunology, Mayo Clinic Rochester, MN, USA
| | | | - Oliver Sartor
- Department of Radiology, Mayo Clinic Rochester, MN, USA; Department of Urology, Mayo Clinic Rochester, MN, USA; Department of Oncology, Mayo Clinic Rochester, MN, USA
| |
Collapse
|
3
|
Telli T, Lopes L, Karpinski M, Pabst KM, Grünwald V, Shi K, Hadaschik B, Kesch C, Umutlu L, Herrmann K, Seifert R, Fendler WP. Prognostic value of [ 18F]FDG- and PSMA-PET in patients evaluated for [ 177Lu]Lu-PSMA therapy of mCRPC. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07198-y. [PMID: 40113645 DOI: 10.1007/s00259-025-07198-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 03/05/2025] [Indexed: 03/22/2025]
Abstract
PURPOSE To improve [177Lu]Lu-Prostate-specific membrane antigen therapy (LuPSMA) selection, this study investigates the prognostic value of PSMA and 2-[18F]fluoro-2-deoxy-D-glucose ([18F]FDG)-PET in metastatic castration-resistant prostate cancer (mCRPC) patients considered for LuPSMA therapy. METHODS We conducted a retrospective analysis in 152 mCRPC patients referred for LuPSMA therapy who underwent PSMA and [18F]FDG-PET/CT. Of these, 104 patients (68.4%) underwent LuPSMA therapy, while 48 (31.6%) received other standard of care (SOC). PET/CT analyses included visual assessment and semiquantitative measurements. Clinical and laboratory parameters were recorded. Overall survival (OS) and PSA response (decline > 50%) were primary and secondary endpoints, respectively. RESULTS Baseline [18F]FDG-derived total tumor volume was the only independent predictor of overall survival both in patients subsequently treated with LuPSMA (HR 1.28 [95%CI 1.02-1.61]; p = 0.03) or in those under other SOC (HR 1.61 [95%CI 1.02-2.56]; p = 0.04), respectively. In other SOC patients, additional independent predictors of OS were total lesion PSMA uptake (PSMA-TL; HR 1.14 [95%CI 1.03-1.26]; p = 0.01), [18F]FDG mean SUV (HR 20.88 [95%CI 1.2-364.74]; p = 0.04), and [18F]FDG total lesion glycolysis (HR 1.61 [95%CI 1.02-2.56]; p = 0.04). In LuPSMA patients, PSMA-PET SUVmean was a significant independent predictor of PSA decline ≥ 50% (OR 2.97 [95%CI 1.27-8.16]; p = 0.02). CONCLUSION PSMA-PET and [18F]FDG-PET provide imaging biomarkers of outcome in candidates for LuPSMA. FDG-PET total tumor volume was an independent predictor of overall survival in candidates for LuPSMA therapy, irrespective of subsequent treatment decision. PSMA-PET SUVmean was associated with biochemical response to LuPSMA. Dual tracer imaging should further be assessed in prospective trials for mCRPC treatment guidance.
Collapse
Affiliation(s)
- Tugce Telli
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany.
- German Cancer Consortium (DKTK), Partner Site, Partnership Between DKFZ and University Hospital Essenaq , Essen, Germany.
- West German Cancer (WTZ), Essen, Germany.
| | - Leonor Lopes
- Department of Nuclear Medicine, University Hospital Bern, Bern, Switzerland
| | - Madeleine Karpinski
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site, Partnership Between DKFZ and University Hospital Essenaq , Essen, Germany
- West German Cancer (WTZ), Essen, Germany
| | - Kim M Pabst
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site, Partnership Between DKFZ and University Hospital Essenaq , Essen, Germany
- West German Cancer (WTZ), Essen, Germany
| | - Viktor Grünwald
- German Cancer Consortium (DKTK), Partner Site, Partnership Between DKFZ and University Hospital Essenaq , Essen, Germany
- West German Cancer (WTZ), Essen, Germany
- Department of Urology, University Hospital Essen, Essen, Germany
| | - Kuangyu Shi
- Department of Nuclear Medicine, University Hospital Bern, Bern, Switzerland
| | - Boris Hadaschik
- German Cancer Consortium (DKTK), Partner Site, Partnership Between DKFZ and University Hospital Essenaq , Essen, Germany
- West German Cancer (WTZ), Essen, Germany
- Department of Urology, University Hospital Essen, Essen, Germany
| | - Claudia Kesch
- German Cancer Consortium (DKTK), Partner Site, Partnership Between DKFZ and University Hospital Essenaq , Essen, Germany
- West German Cancer (WTZ), Essen, Germany
- Department of Urology, University Hospital Essen, Essen, Germany
| | - Lale Umutlu
- German Cancer Consortium (DKTK), Partner Site, Partnership Between DKFZ and University Hospital Essenaq , Essen, Germany
- Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Essen, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site, Partnership Between DKFZ and University Hospital Essenaq , Essen, Germany
- West German Cancer (WTZ), Essen, Germany
| | - Robert Seifert
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site, Partnership Between DKFZ and University Hospital Essenaq , Essen, Germany
- West German Cancer (WTZ), Essen, Germany
- Department of Nuclear Medicine, University Hospital Bern, Bern, Switzerland
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site, Partnership Between DKFZ and University Hospital Essenaq , Essen, Germany
- West German Cancer (WTZ), Essen, Germany
| |
Collapse
|
4
|
Bakht MK, Beltran H. Biological determinants of PSMA expression, regulation and heterogeneity in prostate cancer. Nat Rev Urol 2025; 22:26-45. [PMID: 38977769 PMCID: PMC11841200 DOI: 10.1038/s41585-024-00900-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/21/2024] [Indexed: 07/10/2024]
Abstract
Prostate-specific membrane antigen (PSMA) is an important cell-surface imaging biomarker and therapeutic target in prostate cancer. The PSMA-targeted theranostic 177Lu-PSMA-617 was approved in 2022 for men with PSMA-PET-positive metastatic castration-resistant prostate cancer. However, not all patients respond to PSMA-radioligand therapy, in part owing to the heterogeneity of PSMA expression in the tumour. The PSMA regulatory network is composed of a PSMA transcription complex, an upstream enhancer that loops to the FOLH1 (PSMA) gene promoter, intergenic enhancers and differentially methylated regions. Our understanding of the PSMA regulatory network and the mechanisms underlying PSMA suppression is evolving. Clinically, molecular imaging provides a unique window into PSMA dynamics that occur on therapy and with disease progression, although challenges arise owing to the limited resolution of PET. PSMA regulation and heterogeneity - including intertumoural and inter-patient heterogeneity, temporal changes, lineage dynamics and the tumour microenvironment - affect PSMA theranostics. PSMA response and resistance to radioligand therapy are mediated by a number of potential mechanisms, and complementary biomarkers beyond PSMA are under development. Understanding the biological determinants of cell surface target regulation and heterogeneity can inform precision medicine approaches to PSMA theranostics as well as other emerging therapies.
Collapse
Affiliation(s)
- Martin K Bakht
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
5
|
Ismuha RR, Ritawidya R, Daruwati I, Muchtaridi M. Future Prospect of Low-Molecular-Weight Prostate-Specific Membrane Antigen Radioisotopes Labeled as Theranostic Agents for Metastatic Castration-Resistant Prostate Cancer. Molecules 2024; 29:6062. [PMID: 39770150 PMCID: PMC11679579 DOI: 10.3390/molecules29246062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Prostate cancer ranks as the fourth most common cancer among men, with approximately 1.47 million new cases reported annually. The emergence of prostate-specific membrane antigen (PSMA) as a critical biomarker has revolutionized the diagnosis and treatment of prostate cancer. Recent advancements in low-molecular-weight PSMA inhibitors, with their diverse chemical structures and binding properties, have opened new avenues for research and therapeutic applications in prostate cancer management. These novel agents exhibit enhanced tumor targeting and specificity due to their small size, facilitating rapid uptake and localization at the target site while minimizing the retention in non-target tissues. The primary aim of this study is to evaluate the potential of low-molecular-weight PSMA inhibitors labeled with radioisotopes as theranostic agents for prostate cancer. This includes assessing their efficacy in targeted imaging and therapy and understanding their pharmacokinetic properties and mechanisms of action. This study is a literature review focusing on in vitro and clinical research data. The in vitro studies utilize PSMA-targeted radioligands labeled with radioisotopes to assess their binding affinity, specificity, and internalization in prostate cancer cell lines. Additionally, the clinical studies evaluate the safety, effectiveness, and biodistribution of radiolabeled PSMA ligands in patients with advanced prostate cancer. The findings indicate promising outcomes regarding the safety and efficacy of PSMA-targeted radiopharmaceuticals in clinical settings. The specific accumulation of these agents in prostate tumor lesions suggests their potential for various applications, including imaging and therapy. This research underscores the promise of radiopharmaceuticals targeting PSMA in advancing the diagnosis and treatment of prostate cancer. These agents improve diagnostic accuracy and patients' outcomes by enhancing imaging capabilities and enabling personalized treatment strategies.
Collapse
Affiliation(s)
- Ratu Ralna Ismuha
- Department of Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia;
- Department of Pharmacy, Dharmais Cancer Hospital—National Cancer Center, Jakarta 11420, Indonesia
| | - Rien Ritawidya
- Center for Research on Radioisotope Technology, Radiopharmaceuticals, and Biodosimetry, National Research and Innovation Agency (BRIN), South Tangerang 15314, Indonesia; (R.R.); (I.D.)
- Research Collaboration Centre for Radiopharmaceuticals Theranostic, National Research and Innovation Agency (BRIN), Sumedang 45363, Indonesia
| | - Isti Daruwati
- Center for Research on Radioisotope Technology, Radiopharmaceuticals, and Biodosimetry, National Research and Innovation Agency (BRIN), South Tangerang 15314, Indonesia; (R.R.); (I.D.)
- Research Collaboration Centre for Radiopharmaceuticals Theranostic, National Research and Innovation Agency (BRIN), Sumedang 45363, Indonesia
| | - Muchtaridi Muchtaridi
- Department of Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia;
- Research Collaboration Centre for Radiopharmaceuticals Theranostic, National Research and Innovation Agency (BRIN), Sumedang 45363, Indonesia
| |
Collapse
|
6
|
Michalski K, Kosmala A, Hartrampf PE, Heinrich M, Serfling SE, Schlötelburg W, Buck AK, Meining A, Werner RA, Weich A. [ 18F]FDG and [ 68Ga]Ga-FAPI-04-Directed Imaging for Outcome Prediction in Patients with High-Grade Neuroendocrine Neoplasms. J Nucl Med 2024; 65:1899-1903. [PMID: 39477500 PMCID: PMC11619591 DOI: 10.2967/jnumed.124.268288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/30/2024] [Indexed: 12/08/2024] Open
Abstract
We aimed to quantitatively investigate the prognostic value of PET-based biomarkers on [18F]FDG and [68Ga]Ga-fibroblast activation protein inhibitor (FAPI)-04 PET/CT in patients with highly aggressive neuroendocrine neoplasms (NENs) and to compare the visually assessed differences in uptake on both examinations with progression-free survival (PFS). Methods: In this single-center retrospective analysis, 20 patients with high-grade NENs had undergone [18F]FDG and [68Ga]Ga-FAPI-04 PET. Both PET scans were visually compared, and the presence of [18F]FDG-positive, [68Ga]Ga-FAPI-04-negative (FDG+/FAPI-) lesions was noted. In addition, we assessed maximum, peak, and mean SUV; tumor volume (TV); and total lesion uptake (TLU = TV × SUVmean) for both radiotracers using a 40% lesion-based threshold. The results of quantitative and visual analysis were correlated with PFS using log-rank analysis or univariate Cox regression. PFS was defined radiographically using RECIST 1.1., clinically using signs of disease progression, or as death. Results: Most primary tumors were located in the gastrointestinal tract (13/20 patients, 65%) or were cancer of unknown primary (5/20 patients, 25%). FDG+/FAPI- lesions were found in 9 of 20 patients (45%). Patients with FDG+/FAPI- lesions had a significantly decreased PFS of 4 mo, compared with 9 mo for patients without FDG+/FAPI- metastases (P = 0.0063 [log-rank test]; hazard ratio [HR], 5.637; 95% CI 1.619-26.16; P = 0.0110 [univariate Cox regression]). On univariate analysis, a significant correlation was also found between PFS and TV for both radiotracers ([18F]FDG: mean TV, 258 ± 588 cm3; HR, 1.024 [per 10 cm3]; 95% CI, 1.007-1.046; P = 0.0204) ([68Ga]Ga-FAPI-04: mean TV, 130 ± 192 cm3; HR, 1.032 [per 10 cm3]; 95% CI, 1.001-1.062; P = 0.0277) and TLU on [18F]FDG PET (mean TLU, 1,931 ± 4,248 cm3; HR, 1.004 [per 10 cm3]; 95% CI, 1.001-1.007; P = 0.0135). Conclusion: The presence of discordant FDG+/FAPI- lesions is associated with a significantly shorter PFS, which might indicate more aggressive disease prone to early progression. Dual-tracer PET/CT of patients with highly aggressive NENs could help guide treatment decisions or identify high-risk lesions for additional local therapeutic approaches.
Collapse
Affiliation(s)
- Kerstin Michalski
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany;
| | - Aleksander Kosmala
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Philipp E Hartrampf
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
- NET-Zentrum Würzburg, European Neuroendocrine Tumor Society Center of Excellence, University Hospital Würzburg, Würzburg, Germany
| | - Marieke Heinrich
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | | | - Wiebke Schlötelburg
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Alexander Meining
- NET-Zentrum Würzburg, European Neuroendocrine Tumor Society Center of Excellence, University Hospital Würzburg, Würzburg, Germany
- Department of Internal Medicine II, Gastroenterology, University Hospital Würzburg, Würzburg, Germany
| | - Rudolf A Werner
- NET-Zentrum Würzburg, European Neuroendocrine Tumor Society Center of Excellence, University Hospital Würzburg, Würzburg, Germany
- Department of Nuclear Medicine, Clinic for Radiology and Nuclear Medicine, University Hospital, Goethe University Frankfurt, Frankfurt, Germany; and
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Alexander Weich
- NET-Zentrum Würzburg, European Neuroendocrine Tumor Society Center of Excellence, University Hospital Würzburg, Würzburg, Germany
- Department of Internal Medicine II, Gastroenterology, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
7
|
Zeng T, Xie Y, Chai K, Sang H. The Application of Prostate Specific Membrane Antigen in the Diagnosis and Treatment of Prostate Cancer: Status and Challenge. Onco Targets Ther 2024; 17:991-1015. [PMID: 39564453 PMCID: PMC11573878 DOI: 10.2147/ott.s485869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 10/22/2024] [Indexed: 11/21/2024] Open
Abstract
In recent years, the incidence of prostate cancer has been increasing globally. Early stage of the disease can obtain a better clinical prognosis from surgery and endocrine therapy. The progression of advanced stage varies significantly between individuals, with some patients developing metastatic castration-resistant prostate cancer after standardized treatment. Therefore, staging of prostate cancer by accurate imaging is particularly important for the clinical management of patients. Simultaneously, the development of targeted therapy is also urgent for the treatment of advanced prostate cancer. Prostate specific membrane antigen as a prostate specific target has been widely used in the diagnosis and treatment of prostate cancer. This review summarizes the latest research progress of targeted prostate specific membrane antigen in the diagnosis and treatment of prostate cancer in detail, analyzes their value and challenges.
Collapse
Affiliation(s)
- Tongwei Zeng
- Department of Urology, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, 730900, People's Republic of China
| | - Yongqiang Xie
- Department of Urology, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, 730900, People's Republic of China
| | - Keqiang Chai
- Department of Urology, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, 730900, People's Republic of China
| | - Hui Sang
- Department of Urology, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, 730900, People's Republic of China
| |
Collapse
|
8
|
Demirci RA, Ghodsi A, Gulati R, Behnia S, Nelson PS, Cheng HH, Yezefski TA, Haffner MC, Hawley JE, Montgomery RB, Yu EY, Schweizer MT, Chen DL, Iravani A. PET-Based TheraP Eligibility and Outcomes of VISION-Eligible Patients with Metastatic Castration-Resistant Prostate Cancer Who Received 177Lu-PSMA-617: Importance of 18F-FDG-Avid Discordant Findings. J Nucl Med 2024:jnumed.124.268167. [PMID: 39542703 DOI: 10.2967/jnumed.124.268167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 10/15/2024] [Indexed: 11/17/2024] Open
Abstract
The VISION and TheraP trials introduced different PET-based criteria for patient selection for treatment with 177Lu-PSMA-617 (LuPSMA). TheraP used a higher prostate-specific membrane antigen (PSMA) uptake threshold than VISION and required 18F-FDG PET to exclude patients with discordant findings. Although the screen-failed patients had shorter overall survival (OS) than those treated with LuPSMA, it remains unclear whether their outcomes might have been modified if they had been exposed to LuPSMA. In this study, we evaluated associations between the TheraP eligibility criteria and subgroups and the treatment outcomes of patients who were deemed suitable and treated on the basis of VISION criteria. Methods: Consecutive patients who were treated with LuPSMA and who underwent pretreatment PSMA and 18F-FDG PET were classified as TheraP-eligible (TheraP-E) and TheraP-ineligible (TheraP-I), the latter of which were subclassified as low PSMA or discordant. Odds ratios for an at least 50% decline in prostate-specific antigen (PSA50) were computed using logistic regression, and hazard ratios (HRs) for PSA progression-free survival (PSA-PFS) and OS were computed using Cox regressions. Multivariable analyses were adjusted for baseline imaging and clinical parameters. Results: Of 75 patients, 31 (41%) were deemed TheraP-I; of those, 24 were subclassified as having discordant disease. TheraP-I patients had a lower PSA50 rate than that of TheraP-E patients (28% vs. 67%; odds ratio, 0.19; 95% CI, 0.06-0.52; P = 0.002) and a higher risk of PSA progression (HR, 2.0; 95% CI, 1.2-3.3; P = 0.007). OS in the TheraP-I group was numerically shorter than in the TheraP-E group, but the comparison was only marginally significant (10.4 mo vs. not reached; HR, 1.9; 95% CI, 1.0-3.7; P = 0.054). TheraP-I patients with low PSMA had no significantly different risk of death (P = 0.9) from that of TheraP-E patients, but those with discordant findings had higher risk of death (HR, 2.3; 95% CI, 1.1-4.6; P = 0.02). Discordant disease remained prognostic for OS after adjusting for baseline imaging and clinical parameters (HR, 3.0; 95% CI, 1.3-6.8; P = 0.01). Conclusion: In VISION-eligible patients who were treated with LuPSMA, TheraP-I patients with discordant findings had lower PSA50, PSA-PFS, and OS. Our study suggests that the shorter OS of TheraP-I patients is mainly driven by the presence of discordant disease.
Collapse
Affiliation(s)
| | - Alireza Ghodsi
- Department of Radiology, University of Washington, Seattle, Washington
| | - Roman Gulati
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Sanaz Behnia
- Department of Radiology, University of Washington, Seattle, Washington
| | - Peter S Nelson
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, Washington
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington; and
| | - Heather H Cheng
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, Washington
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Todd A Yezefski
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, Washington
| | - Michael C Haffner
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington; and
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Jessica E Hawley
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, Washington
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Robert B Montgomery
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, Washington
| | - Evan Y Yu
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, Washington
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Michael T Schweizer
- Division of Hematology and Oncology, Department of Medicine, University of Washington, Seattle, Washington
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Delphine L Chen
- Department of Radiology, University of Washington, Seattle, Washington
| | - Amir Iravani
- Department of Radiology, University of Washington, Seattle, Washington;
| |
Collapse
|
9
|
Ni X, Wei Y, Li X, Pan J, Fang B, Zhang T, Lu Y, Ye D, Zhu Y. From biology to the clinic - exploring liver metastasis in prostate cancer. Nat Rev Urol 2024; 21:593-614. [PMID: 38671281 DOI: 10.1038/s41585-024-00875-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2024] [Indexed: 04/28/2024]
Abstract
Liver metastases from prostate cancer are associated with an aggressive disease course and poor prognosis. Results from autopsy studies indicate a liver metastasis prevalence of up to 25% in patients with advanced prostate cancer. Population data estimate that ~3-10% of patients with metastatic castration-resistant prostate cancer harbour liver metastases at the baseline, rising to 20-30% in post-treatment cohorts, suggesting that selective pressure imposed by novel therapies might promote metastatic spread to the liver. Liver metastases are associated with more aggressive tumour biology than lung metastases. Molecular profiling of liver lesions showed an enrichment of low androgen receptor, neuroendocrine phenotypes and high genomic instability. Despite advancements in molecular imaging modalities such as prostate-specific membrane antigen PET-CT, and liquid biopsy markers such as circulating tumour DNA, early detection of liver metastases from prostate cancer remains challenging, as both approaches are hampered by false positive and false negative results, impeding the accurate identification of early liver lesions. Current therapeutic strategies showed limited efficacy in this patient population. Emerging targeted radionuclide therapies, metastasis-directed therapy, and novel systemic agents have shown preliminary activity against liver metastases, but require further validation. Treatment with various novel prostate cancer therapies might lead to an increase in the prevalence of liver metastasis, underscoring the urgent need for coordinated efforts across preclinical and clinical researchers to improve characterization, monitoring, and management of liver metastases from prostate cancer. Elucidating molecular drivers of liver tropism and interactions with the liver microenvironment might ultimately help to identify actionable targets to enhance survival in this high-risk patient group.
Collapse
Affiliation(s)
- Xudong Ni
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Yu Wei
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Xiaomeng Li
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Jian Pan
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Bangwei Fang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Tingwei Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Ying Lu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Shanghai Medical College of Fudan University, Shanghai, China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
| | - Yao Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Genitourinary Cancer Institute, Shanghai, China.
| |
Collapse
|
10
|
Flippot R, Telli T, Velev M, Fléchon A, De Vries-Brilland M, Turpin L, Bergman A, Turco F, Mahammedi H, Fendler WP, Giraudet AL, Josset Q, Montravers F, Vogel W, Gillessen S, Berardi Vilei S, Herrmann K, Kryza D, Paone G, Hadaschik B, Merlin C, Dufour PA, Bernard-Tessier A, Naoun N, Patrikidou A, Garcia C, Foulon S, Pagès A, Fizazi K. Activity of Lutetium-177 Prostate-specific Membrane Antigen and Determinants of Outcomes in Patients with Metastatic Castration-resistant Prostate Cancer Previously Treated with Cabazitaxel: The PACAP Study. Eur Urol Oncol 2024; 7:1132-1140. [PMID: 38664139 DOI: 10.1016/j.euo.2024.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/26/2024] [Accepted: 03/26/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND Both cabazitaxel and lutetium-177 prostate-specific membrane antigen (Lu-PSMA) improve survival in metastatic castration-resistant prostate cancer (mCRPC) after an androgen receptor pathway inhibitor and docetaxel, but there are limited data regarding Lu-PSMA activity after cabazitaxel. OBJECTIVE To assess the activity of Lu-PSMA and determinants of outcomes after cabazitaxel in mCRPC. DESIGN, SETTING, AND PARTICIPANTS A retrospective analysis was conducted of consecutive mCRPC patients from eight European centers treated with Lu-PSMA after cabazitaxel. INTERVENTION Lu-PSMA every 6-8 wk at a dose of 6-7.6 GBq. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The primary endpoint was radiographic progression-free survival (rPFS). The secondary endpoints included time to prostate-specific antigen (PSA) progression (TTPSA), overall survival (OS), PSA decline, objective response rate (ORR), clinical benefit, and safety. RESULTS AND LIMITATIONS Of 126 patients, 68% had International Society of Urological Pathology (ISUP) grade 4-5 disease, 21% had visceral metastases, and 7% had lymph node disease only. DNA damage repair (DDR) alterations were detected in 11/50 (22%) patients with available testing. Patients received a median number of 3 Lu-PSMA cycles (interquartile range 2-4). With a median follow-up of 12.0 mo, the median rPFS was 4.4 mo (95% confidence interval [CI] 3.2-5.4), TTPSA 3.5 mo (95% CI 3.0-4.6), and OS 8.9 mo (95% CI 6.5-12.7). The ORR was 35%, and 55 patients (44%) experienced a PSA decline of ≥50%. The time to castration resistance of <12 mo was associated with shorter rPFS (p = 0.01). A similar trend was observed for ISUP grade 4-5 (p = 0.08), and baseline positron-emission tomography parameters including PSMA mean standardized uptake value (SUV) and maximum SUV (respectively, p = 0.06 and 0.05). The duration of previous cabazitaxel or DDR status did not impact outcomes. Patients experiencing a PSA decline of ≥ 50% on therapy demonstrated longer rPFS, TTPSA, and OS (all p < 0.0001). Limitations include retrospective data collection and investigator-based rPFS assessment. CONCLUSIONS Lu-PSMA demonstrated a substantial PSA decline but limited rPFS after cabazitaxel in a real-life setting. Adverse baseline characteristics, baseline positron-emission tomography parameters, and quality of PSA response may help identify patients less likely to benefit from Lu-PSMA. PATIENT SUMMARY Lutetium-177 prostate-specific membrane antigen (Lu-PSMA) improved outcomes in patients with castration-resistant prostate cancer, but there are limited data about its activity after cabazitaxel, a chemotherapy that is also the standard of care in this setting. We conducted a study across eight European centers and showed substantial responses on Lu-PSMA after cabazitaxel, although activity was short lived in a heavily pretreated population. Our findings prompt for real-life evaluation of Lu-PSMA in earlier settings to define the best therapeutic sequence.
Collapse
Affiliation(s)
- Ronan Flippot
- Department of Cancer Medicine, Gustave Roussy, Paris Saclay University, Villejuif, France.
| | - Tugce Telli
- Department of Nuclear Medicine, University of Duisburg-Essen, Essen, Germany; German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | - Maud Velev
- Department of Cancer Medicine, Gustave Roussy, Paris Saclay University, Villejuif, France
| | - Aude Fléchon
- Department of Medical Oncology, Centre Leon Berard, Lyon, France
| | | | - Léa Turpin
- Department of Nuclear Medicine, Tenon University Hospital, Paris, France
| | - Andries Bergman
- Division of Medical Oncology, the Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Fabio Turco
- Istituto Oncologico della Svizzera Italiana, EOC, Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland; Department of Oncology, at Division of Medical Oncology, San Luigi Gonzaga Hospital, University of Turin, Orbassano, Turin, Italy
| | - Hakim Mahammedi
- Department of Medical Oncology, Centre Jean Perrin, Clermont-Ferrand, France
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University of Duisburg-Essen, Essen, Germany; German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | | | - Quentin Josset
- Department of Medical Oncology, Institut de Cancérologie de l'Ouest, Angers, France
| | | | - Wouter Vogel
- Division of Medical Oncology, the Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Silke Gillessen
- Istituto Oncologico della Svizzera Italiana, EOC, Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Simona Berardi Vilei
- Istituto Oncologico della Svizzera Italiana, EOC, Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen, Essen, Germany; German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | - David Kryza
- Department of Nuclear Medicine, Centre Leon Bérard, Lyon, France
| | - Gaetano Paone
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland; Clinic of Nuclear Medicine and Molecular Imaging, Imaging Institute of Southern Switzerland, EOC, Bellinzona, Switzerland
| | - Boris Hadaschik
- German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany; Department of Urology, University of Duisburg-Essen, Essen, Germany
| | - Charles Merlin
- Department of Nuclear Medicine, Centre Jean Perrin, Clermont-Ferrand, France
| | - Pierre-Alban Dufour
- Department of Nuclear Medicine, Institut de Cancérologie de l'Ouest, Angers, France
| | - Alice Bernard-Tessier
- Department of Cancer Medicine, Gustave Roussy, Paris Saclay University, Villejuif, France
| | - Natacha Naoun
- Department of Cancer Medicine, Gustave Roussy, Paris Saclay University, Villejuif, France
| | - Anna Patrikidou
- Department of Cancer Medicine, Gustave Roussy, Paris Saclay University, Villejuif, France
| | - Camilo Garcia
- Department of Nuclear Medicine, Gustave Roussy, Paris Saclay University, Villejuif, France
| | - Stéphanie Foulon
- Department of Biostatistics and Epidemiology, INSERM UMR 1018 "Oncostat", Gustave Roussy, Paris Saclay University, Villejuif, France
| | - Arnaud Pagès
- Department of Biostatistics and Epidemiology, INSERM UMR 1018 "Oncostat", Gustave Roussy, Paris Saclay University, Villejuif, France
| | - Karim Fizazi
- Department of Cancer Medicine, Gustave Roussy, Paris Saclay University, Villejuif, France
| |
Collapse
|
11
|
代 洪, 黄 淑, 田 甜, 侯 乃, 曾 浩, 魏 强, 黄 蕤. [Clinical Value of Dual Tracer PET Imaging With 68Ga-PSMA and 18F-FDG in Patients With Metastatic Prostate Cancer]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:1063-1070. [PMID: 39507973 PMCID: PMC11536228 DOI: 10.12182/20240960201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Indexed: 11/08/2024]
Abstract
Objective In this study, we retrospectively analyzed the imaging characteristics of dual-tracer 68Ga-prostate specific membrane antigen (PSMA) and 18F-flurodeoxyglucose (FDG) positron emission tomography (PET)/computed tomography (CT) in metastatic prostate cancer (mPCa) patients. We analyzed the uptake modes of the dual tracers, explored clinical pathological parameters affecting the 18F-FDG uptake in the lesions, and evaluated their prognostic implications for prostate specific antigen progression-free survival (PSA-PFS). Methods A total of 41 mPCa patients who underwent dual-tracer PET/CT (68Ga-PSMA and 18F-FDG) scans between September 2021 and January 2024 were retrospectively enrolled. One patient had negative uptake of both PSMA and FDG. According to the uptake patterns of the 2 tracers, the other patients, 40 in total, were categorized in 2 groups, including group A consisting of 33 cases who showed PSMA and FDG dual and those who showed FDG only avidity, and group B consisting of 7 cases who showed PSMA avidity only. Comparative analyses of clinical pathological characteristics between group A and group B were conducted. The relationship between various parameters and PSA-PFS was analyzed by the Kaplan-Meier method. Results A total of 26 patients (63.4%) were diagnosed with metastatic castration-resistant prostate cancer (mCRPC), and 38 cases (92.7%) had a Gleason score of 8-9. Bone metastasis, the predominant type of distant metastasis, occurred in 36 cases (87.8%). The skeletal and distant lymph node metastases mostly showed a dual positive uptake pattern for both PSMA and FDG (85.7% [24/28] and 81.8% [9/11]). 37.5% (3/8) of the metastases to organs showed FDG only positive uptake pattern. The serum levels of prostate specific antigen (PSA) in group A were significantly higher than those in group B (P=0.013). A total of 13 patients of special pathological classification (intraductal carcinoma and neuroendocrine differentiation) were all found to be in group A. Among the 41 cases, 16 were lost to follow-up. Of the 25 patients who completed follow-up, 9 patients, with a median PSA value of 104 ng/mL, experienced PSA progression, while the 16 other patients, with a median PSA of 0.34 ng/mL, did not incur any PSA progression. There was significant difference in the median PSA between patients showing PSA progression and those who did not show PSA progression (P<0.001). Kaplan-Meier survival analysis revealed that the median PSA-PFS of patients of specific pathological classifications was 7 months, which was shorter than the 16 months of the patients with typical prostate cancer, with the difference between the two groups being statistically meaningful (P=0.043). The median PSA-PFS for group A was 30 months. With more than half of the patients in the group not experiencing any PSA progression, group B did not reach the median PSA-PFS (P=0.645). Conclusion Dual-tracer PET/CT imaging with 68Ga-PSMA and 18F-FDG commonly exhibits avidity for both tracers in mPCa. Serum PSA level is a reliable biomarker for predicting FDG-positive lesions. mPCa presented with intraductal carcinoma and neuroendocrine differentiation tends to exhibit FDG avidity and is more susceptible to PSA progression.
Collapse
Affiliation(s)
- 洪媛 代
- 四川大学华西医院 核医学科 (成都 610041)Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 淑辉 黄
- 四川大学华西医院 核医学科 (成都 610041)Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 甜 田
- 四川大学华西医院 核医学科 (成都 610041)Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 乃峰 侯
- 四川大学华西医院 核医学科 (成都 610041)Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 浩 曾
- 四川大学华西医院 核医学科 (成都 610041)Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 强 魏
- 四川大学华西医院 核医学科 (成都 610041)Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 蕤 黄
- 四川大学华西医院 核医学科 (成都 610041)Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
12
|
Kuo PH, Morris MJ, Hesterman J, Kendi AT, Rahbar K, Wei XX, Fang B, Adra N, Garje R, Michalski JM, Chi K, de Bono J, Fizazi K, Krause B, Sartor O, Tagawa ST, Ghebremariam S, Brackman M, Wong CC, Catafau AM, Benson T, Armstrong AJ, Herrmann K, Atzen S. Quantitative 68Ga-PSMA-11 PET and Clinical Outcomes in Metastatic Castration-resistant Prostate Cancer Following 177Lu-PSMA-617 (VISION Trial). Radiology 2024; 312:e233460. [PMID: 39162634 PMCID: PMC11366674 DOI: 10.1148/radiol.233460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/22/2024] [Accepted: 05/14/2024] [Indexed: 08/21/2024]
Abstract
Background Lutetium 177 [177Lu]Lu-PSMA-617 (177Lu-PSMA-617) is a prostate-specific membrane antigen (PSMA)-targeted radioligand therapy for metastatic castration-resistant prostate cancer (mCRPC). Quantitative PSMA PET/CT analysis could provide information on 177Lu-PSMA-617 treatment benefits. Purpose To explore the association between quantitative baseline gallium 68 [68Ga]Ga-PSMA-11 (68Ga-PSMA-11) PET/CT parameters and treatment response and outcomes in the VISION trial. Materials and Methods This was an exploratory secondary analysis of the VISION trial. Eligible participants were randomized (June 2018 to October 2019) in a 2:1 ratio to 177Lu-PSMA-617 therapy (7.4 GBq every 6 weeks for up to six cycles) plus standard of care (SOC) or to SOC only. Baseline 68Ga-PSMA-11 PET parameters, including the mean and maximum standardized uptake value (SUVmean and SUVmax), PSMA-positive tumor volume, and tumor load, were extracted from five anatomic regions and the whole body. Associations of quantitative PET parameters with radiographic progression-free survival (rPFS), overall survival (OS), objective response rate, and prostate-specific antigen response were investigated using univariable and multivariable analyses (with treatment as the only other covariate). Outcomes were assessed in subgroups based on SUVmean quartiles. Results Quantitative PET parameters were well balanced between study arms for the 826 participants included. The median whole-body tumor SUVmean was 7.6 (IQR, 5.8-9.9). Whole-body tumor SUVmean was the best predictor of 177Lu-PSMA-617 efficacy, with a hazard ratio (HR) range of 0.86-1.43 for all outcomes (all P < .001). A 1-unit whole-body tumor SUVmean increase was associated with a 12% and 10% decrease in risk of an rPFS event and death, respectively. 177Lu-PSMA-617 plus SOC prolonged rPFS and OS in all SUVmean quartiles versus SOC only, with no identifiable optimum among participants receiving 177Lu-PSMA-617. Higher baseline PSMA-positive tumor volume and tumor load were associated with worse rPFS (HR range, 1.44-1.53 [P < .05] and 1.02-1.03 [P < .001], respectively) and OS (HR range, 1.36-2.12 [P < .006] and 1.04 [P < .001], respectively). Conclusion Baseline 68Ga-PSMA-11 PET/CT whole-body tumor SUVmean was the best predictor of 177Lu-PSMA-617 efficacy in participants in the VISION trial. Improvements in rPFS and OS with 177Lu-PSMA-617 plus SOC were greater among participants with higher whole-body tumor SUVmean, with evidence for benefit at all SUVmean levels. ClinicalTrials.gov identifier: NCT03511664 Published under a CC BY 4.0 license. Supplemental material is available for this article.
Collapse
Affiliation(s)
| | | | - Jacob Hesterman
- From the University of Arizona, Tucson, Ariz (P.H.K.); Memorial
Sloan-Kettering Cancer Center, New York, NY (M.J.M.); Invicro, Needham, Mass
(J.H.); Mayo Clinic, Rochester, Minn (A.T.K., O.S.); Department of Nuclear
Medicine, University Hospital Münster, Münster, Germany (K.R.);
West German Cancer Center, Münster and Essen, Germany (K.R.); Dana-Farber
Cancer Institute, Boston, Mass (X.X.W.); Astera Cancer Care, East Brunswick, NJ
(B.F.); Indiana University Simon Comprehensive Cancer Center, Indianapolis, Ind
(N.A.); Miami Cancer Institute, Baptist Health South Florida, Miami, Fla (R.G.);
Washington University, St. Louis, Mo (J.M.M.); British Columbia Cancer Agency,
Vancouver, British Columbia, Canada (K.C.); The Institute of Cancer Research and
Royal Marsden Hospital, London, United Kingdom (J.d.B.); Gustave Roussy
Institute, University of Paris-Saclay, Villejuif, France (K.F.); Rostock
University Medical Center, Rostock, Germany (B.K.); Weill Cornell Medicine, New
York, NY (S.T.T.); Novartis Pharmaceuticals, East Hanover, NJ (S.G.); Novartis
Pharmaceuticals, Indianapolis, Ind (M.B.); Novartis Pharmaceuticals, Cambridge,
Mass (C.C.W.); Novartis Pharmaceuticals, Geneva, Switzerland (A.M.C.); Novartis
Pharmaceuticals, St. George, Utah (T.B.); Duke Cancer Institute Center for
Prostate and Urologic Cancers, Duke University, Durham, NC (A.J.A.); and
University Hospital Essen and German Cancer Consortium, Hufelandstr. 55, 45147
Essen, Germany (K.H.)
| | - A. Tuba Kendi
- From the University of Arizona, Tucson, Ariz (P.H.K.); Memorial
Sloan-Kettering Cancer Center, New York, NY (M.J.M.); Invicro, Needham, Mass
(J.H.); Mayo Clinic, Rochester, Minn (A.T.K., O.S.); Department of Nuclear
Medicine, University Hospital Münster, Münster, Germany (K.R.);
West German Cancer Center, Münster and Essen, Germany (K.R.); Dana-Farber
Cancer Institute, Boston, Mass (X.X.W.); Astera Cancer Care, East Brunswick, NJ
(B.F.); Indiana University Simon Comprehensive Cancer Center, Indianapolis, Ind
(N.A.); Miami Cancer Institute, Baptist Health South Florida, Miami, Fla (R.G.);
Washington University, St. Louis, Mo (J.M.M.); British Columbia Cancer Agency,
Vancouver, British Columbia, Canada (K.C.); The Institute of Cancer Research and
Royal Marsden Hospital, London, United Kingdom (J.d.B.); Gustave Roussy
Institute, University of Paris-Saclay, Villejuif, France (K.F.); Rostock
University Medical Center, Rostock, Germany (B.K.); Weill Cornell Medicine, New
York, NY (S.T.T.); Novartis Pharmaceuticals, East Hanover, NJ (S.G.); Novartis
Pharmaceuticals, Indianapolis, Ind (M.B.); Novartis Pharmaceuticals, Cambridge,
Mass (C.C.W.); Novartis Pharmaceuticals, Geneva, Switzerland (A.M.C.); Novartis
Pharmaceuticals, St. George, Utah (T.B.); Duke Cancer Institute Center for
Prostate and Urologic Cancers, Duke University, Durham, NC (A.J.A.); and
University Hospital Essen and German Cancer Consortium, Hufelandstr. 55, 45147
Essen, Germany (K.H.)
| | - Kambiz Rahbar
- From the University of Arizona, Tucson, Ariz (P.H.K.); Memorial
Sloan-Kettering Cancer Center, New York, NY (M.J.M.); Invicro, Needham, Mass
(J.H.); Mayo Clinic, Rochester, Minn (A.T.K., O.S.); Department of Nuclear
Medicine, University Hospital Münster, Münster, Germany (K.R.);
West German Cancer Center, Münster and Essen, Germany (K.R.); Dana-Farber
Cancer Institute, Boston, Mass (X.X.W.); Astera Cancer Care, East Brunswick, NJ
(B.F.); Indiana University Simon Comprehensive Cancer Center, Indianapolis, Ind
(N.A.); Miami Cancer Institute, Baptist Health South Florida, Miami, Fla (R.G.);
Washington University, St. Louis, Mo (J.M.M.); British Columbia Cancer Agency,
Vancouver, British Columbia, Canada (K.C.); The Institute of Cancer Research and
Royal Marsden Hospital, London, United Kingdom (J.d.B.); Gustave Roussy
Institute, University of Paris-Saclay, Villejuif, France (K.F.); Rostock
University Medical Center, Rostock, Germany (B.K.); Weill Cornell Medicine, New
York, NY (S.T.T.); Novartis Pharmaceuticals, East Hanover, NJ (S.G.); Novartis
Pharmaceuticals, Indianapolis, Ind (M.B.); Novartis Pharmaceuticals, Cambridge,
Mass (C.C.W.); Novartis Pharmaceuticals, Geneva, Switzerland (A.M.C.); Novartis
Pharmaceuticals, St. George, Utah (T.B.); Duke Cancer Institute Center for
Prostate and Urologic Cancers, Duke University, Durham, NC (A.J.A.); and
University Hospital Essen and German Cancer Consortium, Hufelandstr. 55, 45147
Essen, Germany (K.H.)
| | - Xiao X. Wei
- From the University of Arizona, Tucson, Ariz (P.H.K.); Memorial
Sloan-Kettering Cancer Center, New York, NY (M.J.M.); Invicro, Needham, Mass
(J.H.); Mayo Clinic, Rochester, Minn (A.T.K., O.S.); Department of Nuclear
Medicine, University Hospital Münster, Münster, Germany (K.R.);
West German Cancer Center, Münster and Essen, Germany (K.R.); Dana-Farber
Cancer Institute, Boston, Mass (X.X.W.); Astera Cancer Care, East Brunswick, NJ
(B.F.); Indiana University Simon Comprehensive Cancer Center, Indianapolis, Ind
(N.A.); Miami Cancer Institute, Baptist Health South Florida, Miami, Fla (R.G.);
Washington University, St. Louis, Mo (J.M.M.); British Columbia Cancer Agency,
Vancouver, British Columbia, Canada (K.C.); The Institute of Cancer Research and
Royal Marsden Hospital, London, United Kingdom (J.d.B.); Gustave Roussy
Institute, University of Paris-Saclay, Villejuif, France (K.F.); Rostock
University Medical Center, Rostock, Germany (B.K.); Weill Cornell Medicine, New
York, NY (S.T.T.); Novartis Pharmaceuticals, East Hanover, NJ (S.G.); Novartis
Pharmaceuticals, Indianapolis, Ind (M.B.); Novartis Pharmaceuticals, Cambridge,
Mass (C.C.W.); Novartis Pharmaceuticals, Geneva, Switzerland (A.M.C.); Novartis
Pharmaceuticals, St. George, Utah (T.B.); Duke Cancer Institute Center for
Prostate and Urologic Cancers, Duke University, Durham, NC (A.J.A.); and
University Hospital Essen and German Cancer Consortium, Hufelandstr. 55, 45147
Essen, Germany (K.H.)
| | - Bruno Fang
- From the University of Arizona, Tucson, Ariz (P.H.K.); Memorial
Sloan-Kettering Cancer Center, New York, NY (M.J.M.); Invicro, Needham, Mass
(J.H.); Mayo Clinic, Rochester, Minn (A.T.K., O.S.); Department of Nuclear
Medicine, University Hospital Münster, Münster, Germany (K.R.);
West German Cancer Center, Münster and Essen, Germany (K.R.); Dana-Farber
Cancer Institute, Boston, Mass (X.X.W.); Astera Cancer Care, East Brunswick, NJ
(B.F.); Indiana University Simon Comprehensive Cancer Center, Indianapolis, Ind
(N.A.); Miami Cancer Institute, Baptist Health South Florida, Miami, Fla (R.G.);
Washington University, St. Louis, Mo (J.M.M.); British Columbia Cancer Agency,
Vancouver, British Columbia, Canada (K.C.); The Institute of Cancer Research and
Royal Marsden Hospital, London, United Kingdom (J.d.B.); Gustave Roussy
Institute, University of Paris-Saclay, Villejuif, France (K.F.); Rostock
University Medical Center, Rostock, Germany (B.K.); Weill Cornell Medicine, New
York, NY (S.T.T.); Novartis Pharmaceuticals, East Hanover, NJ (S.G.); Novartis
Pharmaceuticals, Indianapolis, Ind (M.B.); Novartis Pharmaceuticals, Cambridge,
Mass (C.C.W.); Novartis Pharmaceuticals, Geneva, Switzerland (A.M.C.); Novartis
Pharmaceuticals, St. George, Utah (T.B.); Duke Cancer Institute Center for
Prostate and Urologic Cancers, Duke University, Durham, NC (A.J.A.); and
University Hospital Essen and German Cancer Consortium, Hufelandstr. 55, 45147
Essen, Germany (K.H.)
| | - Nabil Adra
- From the University of Arizona, Tucson, Ariz (P.H.K.); Memorial
Sloan-Kettering Cancer Center, New York, NY (M.J.M.); Invicro, Needham, Mass
(J.H.); Mayo Clinic, Rochester, Minn (A.T.K., O.S.); Department of Nuclear
Medicine, University Hospital Münster, Münster, Germany (K.R.);
West German Cancer Center, Münster and Essen, Germany (K.R.); Dana-Farber
Cancer Institute, Boston, Mass (X.X.W.); Astera Cancer Care, East Brunswick, NJ
(B.F.); Indiana University Simon Comprehensive Cancer Center, Indianapolis, Ind
(N.A.); Miami Cancer Institute, Baptist Health South Florida, Miami, Fla (R.G.);
Washington University, St. Louis, Mo (J.M.M.); British Columbia Cancer Agency,
Vancouver, British Columbia, Canada (K.C.); The Institute of Cancer Research and
Royal Marsden Hospital, London, United Kingdom (J.d.B.); Gustave Roussy
Institute, University of Paris-Saclay, Villejuif, France (K.F.); Rostock
University Medical Center, Rostock, Germany (B.K.); Weill Cornell Medicine, New
York, NY (S.T.T.); Novartis Pharmaceuticals, East Hanover, NJ (S.G.); Novartis
Pharmaceuticals, Indianapolis, Ind (M.B.); Novartis Pharmaceuticals, Cambridge,
Mass (C.C.W.); Novartis Pharmaceuticals, Geneva, Switzerland (A.M.C.); Novartis
Pharmaceuticals, St. George, Utah (T.B.); Duke Cancer Institute Center for
Prostate and Urologic Cancers, Duke University, Durham, NC (A.J.A.); and
University Hospital Essen and German Cancer Consortium, Hufelandstr. 55, 45147
Essen, Germany (K.H.)
| | - Rohan Garje
- From the University of Arizona, Tucson, Ariz (P.H.K.); Memorial
Sloan-Kettering Cancer Center, New York, NY (M.J.M.); Invicro, Needham, Mass
(J.H.); Mayo Clinic, Rochester, Minn (A.T.K., O.S.); Department of Nuclear
Medicine, University Hospital Münster, Münster, Germany (K.R.);
West German Cancer Center, Münster and Essen, Germany (K.R.); Dana-Farber
Cancer Institute, Boston, Mass (X.X.W.); Astera Cancer Care, East Brunswick, NJ
(B.F.); Indiana University Simon Comprehensive Cancer Center, Indianapolis, Ind
(N.A.); Miami Cancer Institute, Baptist Health South Florida, Miami, Fla (R.G.);
Washington University, St. Louis, Mo (J.M.M.); British Columbia Cancer Agency,
Vancouver, British Columbia, Canada (K.C.); The Institute of Cancer Research and
Royal Marsden Hospital, London, United Kingdom (J.d.B.); Gustave Roussy
Institute, University of Paris-Saclay, Villejuif, France (K.F.); Rostock
University Medical Center, Rostock, Germany (B.K.); Weill Cornell Medicine, New
York, NY (S.T.T.); Novartis Pharmaceuticals, East Hanover, NJ (S.G.); Novartis
Pharmaceuticals, Indianapolis, Ind (M.B.); Novartis Pharmaceuticals, Cambridge,
Mass (C.C.W.); Novartis Pharmaceuticals, Geneva, Switzerland (A.M.C.); Novartis
Pharmaceuticals, St. George, Utah (T.B.); Duke Cancer Institute Center for
Prostate and Urologic Cancers, Duke University, Durham, NC (A.J.A.); and
University Hospital Essen and German Cancer Consortium, Hufelandstr. 55, 45147
Essen, Germany (K.H.)
| | - Jeff M. Michalski
- From the University of Arizona, Tucson, Ariz (P.H.K.); Memorial
Sloan-Kettering Cancer Center, New York, NY (M.J.M.); Invicro, Needham, Mass
(J.H.); Mayo Clinic, Rochester, Minn (A.T.K., O.S.); Department of Nuclear
Medicine, University Hospital Münster, Münster, Germany (K.R.);
West German Cancer Center, Münster and Essen, Germany (K.R.); Dana-Farber
Cancer Institute, Boston, Mass (X.X.W.); Astera Cancer Care, East Brunswick, NJ
(B.F.); Indiana University Simon Comprehensive Cancer Center, Indianapolis, Ind
(N.A.); Miami Cancer Institute, Baptist Health South Florida, Miami, Fla (R.G.);
Washington University, St. Louis, Mo (J.M.M.); British Columbia Cancer Agency,
Vancouver, British Columbia, Canada (K.C.); The Institute of Cancer Research and
Royal Marsden Hospital, London, United Kingdom (J.d.B.); Gustave Roussy
Institute, University of Paris-Saclay, Villejuif, France (K.F.); Rostock
University Medical Center, Rostock, Germany (B.K.); Weill Cornell Medicine, New
York, NY (S.T.T.); Novartis Pharmaceuticals, East Hanover, NJ (S.G.); Novartis
Pharmaceuticals, Indianapolis, Ind (M.B.); Novartis Pharmaceuticals, Cambridge,
Mass (C.C.W.); Novartis Pharmaceuticals, Geneva, Switzerland (A.M.C.); Novartis
Pharmaceuticals, St. George, Utah (T.B.); Duke Cancer Institute Center for
Prostate and Urologic Cancers, Duke University, Durham, NC (A.J.A.); and
University Hospital Essen and German Cancer Consortium, Hufelandstr. 55, 45147
Essen, Germany (K.H.)
| | - Kim Chi
- From the University of Arizona, Tucson, Ariz (P.H.K.); Memorial
Sloan-Kettering Cancer Center, New York, NY (M.J.M.); Invicro, Needham, Mass
(J.H.); Mayo Clinic, Rochester, Minn (A.T.K., O.S.); Department of Nuclear
Medicine, University Hospital Münster, Münster, Germany (K.R.);
West German Cancer Center, Münster and Essen, Germany (K.R.); Dana-Farber
Cancer Institute, Boston, Mass (X.X.W.); Astera Cancer Care, East Brunswick, NJ
(B.F.); Indiana University Simon Comprehensive Cancer Center, Indianapolis, Ind
(N.A.); Miami Cancer Institute, Baptist Health South Florida, Miami, Fla (R.G.);
Washington University, St. Louis, Mo (J.M.M.); British Columbia Cancer Agency,
Vancouver, British Columbia, Canada (K.C.); The Institute of Cancer Research and
Royal Marsden Hospital, London, United Kingdom (J.d.B.); Gustave Roussy
Institute, University of Paris-Saclay, Villejuif, France (K.F.); Rostock
University Medical Center, Rostock, Germany (B.K.); Weill Cornell Medicine, New
York, NY (S.T.T.); Novartis Pharmaceuticals, East Hanover, NJ (S.G.); Novartis
Pharmaceuticals, Indianapolis, Ind (M.B.); Novartis Pharmaceuticals, Cambridge,
Mass (C.C.W.); Novartis Pharmaceuticals, Geneva, Switzerland (A.M.C.); Novartis
Pharmaceuticals, St. George, Utah (T.B.); Duke Cancer Institute Center for
Prostate and Urologic Cancers, Duke University, Durham, NC (A.J.A.); and
University Hospital Essen and German Cancer Consortium, Hufelandstr. 55, 45147
Essen, Germany (K.H.)
| | - Johann de Bono
- From the University of Arizona, Tucson, Ariz (P.H.K.); Memorial
Sloan-Kettering Cancer Center, New York, NY (M.J.M.); Invicro, Needham, Mass
(J.H.); Mayo Clinic, Rochester, Minn (A.T.K., O.S.); Department of Nuclear
Medicine, University Hospital Münster, Münster, Germany (K.R.);
West German Cancer Center, Münster and Essen, Germany (K.R.); Dana-Farber
Cancer Institute, Boston, Mass (X.X.W.); Astera Cancer Care, East Brunswick, NJ
(B.F.); Indiana University Simon Comprehensive Cancer Center, Indianapolis, Ind
(N.A.); Miami Cancer Institute, Baptist Health South Florida, Miami, Fla (R.G.);
Washington University, St. Louis, Mo (J.M.M.); British Columbia Cancer Agency,
Vancouver, British Columbia, Canada (K.C.); The Institute of Cancer Research and
Royal Marsden Hospital, London, United Kingdom (J.d.B.); Gustave Roussy
Institute, University of Paris-Saclay, Villejuif, France (K.F.); Rostock
University Medical Center, Rostock, Germany (B.K.); Weill Cornell Medicine, New
York, NY (S.T.T.); Novartis Pharmaceuticals, East Hanover, NJ (S.G.); Novartis
Pharmaceuticals, Indianapolis, Ind (M.B.); Novartis Pharmaceuticals, Cambridge,
Mass (C.C.W.); Novartis Pharmaceuticals, Geneva, Switzerland (A.M.C.); Novartis
Pharmaceuticals, St. George, Utah (T.B.); Duke Cancer Institute Center for
Prostate and Urologic Cancers, Duke University, Durham, NC (A.J.A.); and
University Hospital Essen and German Cancer Consortium, Hufelandstr. 55, 45147
Essen, Germany (K.H.)
| | - Karim Fizazi
- From the University of Arizona, Tucson, Ariz (P.H.K.); Memorial
Sloan-Kettering Cancer Center, New York, NY (M.J.M.); Invicro, Needham, Mass
(J.H.); Mayo Clinic, Rochester, Minn (A.T.K., O.S.); Department of Nuclear
Medicine, University Hospital Münster, Münster, Germany (K.R.);
West German Cancer Center, Münster and Essen, Germany (K.R.); Dana-Farber
Cancer Institute, Boston, Mass (X.X.W.); Astera Cancer Care, East Brunswick, NJ
(B.F.); Indiana University Simon Comprehensive Cancer Center, Indianapolis, Ind
(N.A.); Miami Cancer Institute, Baptist Health South Florida, Miami, Fla (R.G.);
Washington University, St. Louis, Mo (J.M.M.); British Columbia Cancer Agency,
Vancouver, British Columbia, Canada (K.C.); The Institute of Cancer Research and
Royal Marsden Hospital, London, United Kingdom (J.d.B.); Gustave Roussy
Institute, University of Paris-Saclay, Villejuif, France (K.F.); Rostock
University Medical Center, Rostock, Germany (B.K.); Weill Cornell Medicine, New
York, NY (S.T.T.); Novartis Pharmaceuticals, East Hanover, NJ (S.G.); Novartis
Pharmaceuticals, Indianapolis, Ind (M.B.); Novartis Pharmaceuticals, Cambridge,
Mass (C.C.W.); Novartis Pharmaceuticals, Geneva, Switzerland (A.M.C.); Novartis
Pharmaceuticals, St. George, Utah (T.B.); Duke Cancer Institute Center for
Prostate and Urologic Cancers, Duke University, Durham, NC (A.J.A.); and
University Hospital Essen and German Cancer Consortium, Hufelandstr. 55, 45147
Essen, Germany (K.H.)
| | - Bernd Krause
- From the University of Arizona, Tucson, Ariz (P.H.K.); Memorial
Sloan-Kettering Cancer Center, New York, NY (M.J.M.); Invicro, Needham, Mass
(J.H.); Mayo Clinic, Rochester, Minn (A.T.K., O.S.); Department of Nuclear
Medicine, University Hospital Münster, Münster, Germany (K.R.);
West German Cancer Center, Münster and Essen, Germany (K.R.); Dana-Farber
Cancer Institute, Boston, Mass (X.X.W.); Astera Cancer Care, East Brunswick, NJ
(B.F.); Indiana University Simon Comprehensive Cancer Center, Indianapolis, Ind
(N.A.); Miami Cancer Institute, Baptist Health South Florida, Miami, Fla (R.G.);
Washington University, St. Louis, Mo (J.M.M.); British Columbia Cancer Agency,
Vancouver, British Columbia, Canada (K.C.); The Institute of Cancer Research and
Royal Marsden Hospital, London, United Kingdom (J.d.B.); Gustave Roussy
Institute, University of Paris-Saclay, Villejuif, France (K.F.); Rostock
University Medical Center, Rostock, Germany (B.K.); Weill Cornell Medicine, New
York, NY (S.T.T.); Novartis Pharmaceuticals, East Hanover, NJ (S.G.); Novartis
Pharmaceuticals, Indianapolis, Ind (M.B.); Novartis Pharmaceuticals, Cambridge,
Mass (C.C.W.); Novartis Pharmaceuticals, Geneva, Switzerland (A.M.C.); Novartis
Pharmaceuticals, St. George, Utah (T.B.); Duke Cancer Institute Center for
Prostate and Urologic Cancers, Duke University, Durham, NC (A.J.A.); and
University Hospital Essen and German Cancer Consortium, Hufelandstr. 55, 45147
Essen, Germany (K.H.)
| | - Oliver Sartor
- From the University of Arizona, Tucson, Ariz (P.H.K.); Memorial
Sloan-Kettering Cancer Center, New York, NY (M.J.M.); Invicro, Needham, Mass
(J.H.); Mayo Clinic, Rochester, Minn (A.T.K., O.S.); Department of Nuclear
Medicine, University Hospital Münster, Münster, Germany (K.R.);
West German Cancer Center, Münster and Essen, Germany (K.R.); Dana-Farber
Cancer Institute, Boston, Mass (X.X.W.); Astera Cancer Care, East Brunswick, NJ
(B.F.); Indiana University Simon Comprehensive Cancer Center, Indianapolis, Ind
(N.A.); Miami Cancer Institute, Baptist Health South Florida, Miami, Fla (R.G.);
Washington University, St. Louis, Mo (J.M.M.); British Columbia Cancer Agency,
Vancouver, British Columbia, Canada (K.C.); The Institute of Cancer Research and
Royal Marsden Hospital, London, United Kingdom (J.d.B.); Gustave Roussy
Institute, University of Paris-Saclay, Villejuif, France (K.F.); Rostock
University Medical Center, Rostock, Germany (B.K.); Weill Cornell Medicine, New
York, NY (S.T.T.); Novartis Pharmaceuticals, East Hanover, NJ (S.G.); Novartis
Pharmaceuticals, Indianapolis, Ind (M.B.); Novartis Pharmaceuticals, Cambridge,
Mass (C.C.W.); Novartis Pharmaceuticals, Geneva, Switzerland (A.M.C.); Novartis
Pharmaceuticals, St. George, Utah (T.B.); Duke Cancer Institute Center for
Prostate and Urologic Cancers, Duke University, Durham, NC (A.J.A.); and
University Hospital Essen and German Cancer Consortium, Hufelandstr. 55, 45147
Essen, Germany (K.H.)
| | - Scott T. Tagawa
- From the University of Arizona, Tucson, Ariz (P.H.K.); Memorial
Sloan-Kettering Cancer Center, New York, NY (M.J.M.); Invicro, Needham, Mass
(J.H.); Mayo Clinic, Rochester, Minn (A.T.K., O.S.); Department of Nuclear
Medicine, University Hospital Münster, Münster, Germany (K.R.);
West German Cancer Center, Münster and Essen, Germany (K.R.); Dana-Farber
Cancer Institute, Boston, Mass (X.X.W.); Astera Cancer Care, East Brunswick, NJ
(B.F.); Indiana University Simon Comprehensive Cancer Center, Indianapolis, Ind
(N.A.); Miami Cancer Institute, Baptist Health South Florida, Miami, Fla (R.G.);
Washington University, St. Louis, Mo (J.M.M.); British Columbia Cancer Agency,
Vancouver, British Columbia, Canada (K.C.); The Institute of Cancer Research and
Royal Marsden Hospital, London, United Kingdom (J.d.B.); Gustave Roussy
Institute, University of Paris-Saclay, Villejuif, France (K.F.); Rostock
University Medical Center, Rostock, Germany (B.K.); Weill Cornell Medicine, New
York, NY (S.T.T.); Novartis Pharmaceuticals, East Hanover, NJ (S.G.); Novartis
Pharmaceuticals, Indianapolis, Ind (M.B.); Novartis Pharmaceuticals, Cambridge,
Mass (C.C.W.); Novartis Pharmaceuticals, Geneva, Switzerland (A.M.C.); Novartis
Pharmaceuticals, St. George, Utah (T.B.); Duke Cancer Institute Center for
Prostate and Urologic Cancers, Duke University, Durham, NC (A.J.A.); and
University Hospital Essen and German Cancer Consortium, Hufelandstr. 55, 45147
Essen, Germany (K.H.)
| | - Samson Ghebremariam
- From the University of Arizona, Tucson, Ariz (P.H.K.); Memorial
Sloan-Kettering Cancer Center, New York, NY (M.J.M.); Invicro, Needham, Mass
(J.H.); Mayo Clinic, Rochester, Minn (A.T.K., O.S.); Department of Nuclear
Medicine, University Hospital Münster, Münster, Germany (K.R.);
West German Cancer Center, Münster and Essen, Germany (K.R.); Dana-Farber
Cancer Institute, Boston, Mass (X.X.W.); Astera Cancer Care, East Brunswick, NJ
(B.F.); Indiana University Simon Comprehensive Cancer Center, Indianapolis, Ind
(N.A.); Miami Cancer Institute, Baptist Health South Florida, Miami, Fla (R.G.);
Washington University, St. Louis, Mo (J.M.M.); British Columbia Cancer Agency,
Vancouver, British Columbia, Canada (K.C.); The Institute of Cancer Research and
Royal Marsden Hospital, London, United Kingdom (J.d.B.); Gustave Roussy
Institute, University of Paris-Saclay, Villejuif, France (K.F.); Rostock
University Medical Center, Rostock, Germany (B.K.); Weill Cornell Medicine, New
York, NY (S.T.T.); Novartis Pharmaceuticals, East Hanover, NJ (S.G.); Novartis
Pharmaceuticals, Indianapolis, Ind (M.B.); Novartis Pharmaceuticals, Cambridge,
Mass (C.C.W.); Novartis Pharmaceuticals, Geneva, Switzerland (A.M.C.); Novartis
Pharmaceuticals, St. George, Utah (T.B.); Duke Cancer Institute Center for
Prostate and Urologic Cancers, Duke University, Durham, NC (A.J.A.); and
University Hospital Essen and German Cancer Consortium, Hufelandstr. 55, 45147
Essen, Germany (K.H.)
| | - Marcia Brackman
- From the University of Arizona, Tucson, Ariz (P.H.K.); Memorial
Sloan-Kettering Cancer Center, New York, NY (M.J.M.); Invicro, Needham, Mass
(J.H.); Mayo Clinic, Rochester, Minn (A.T.K., O.S.); Department of Nuclear
Medicine, University Hospital Münster, Münster, Germany (K.R.);
West German Cancer Center, Münster and Essen, Germany (K.R.); Dana-Farber
Cancer Institute, Boston, Mass (X.X.W.); Astera Cancer Care, East Brunswick, NJ
(B.F.); Indiana University Simon Comprehensive Cancer Center, Indianapolis, Ind
(N.A.); Miami Cancer Institute, Baptist Health South Florida, Miami, Fla (R.G.);
Washington University, St. Louis, Mo (J.M.M.); British Columbia Cancer Agency,
Vancouver, British Columbia, Canada (K.C.); The Institute of Cancer Research and
Royal Marsden Hospital, London, United Kingdom (J.d.B.); Gustave Roussy
Institute, University of Paris-Saclay, Villejuif, France (K.F.); Rostock
University Medical Center, Rostock, Germany (B.K.); Weill Cornell Medicine, New
York, NY (S.T.T.); Novartis Pharmaceuticals, East Hanover, NJ (S.G.); Novartis
Pharmaceuticals, Indianapolis, Ind (M.B.); Novartis Pharmaceuticals, Cambridge,
Mass (C.C.W.); Novartis Pharmaceuticals, Geneva, Switzerland (A.M.C.); Novartis
Pharmaceuticals, St. George, Utah (T.B.); Duke Cancer Institute Center for
Prostate and Urologic Cancers, Duke University, Durham, NC (A.J.A.); and
University Hospital Essen and German Cancer Consortium, Hufelandstr. 55, 45147
Essen, Germany (K.H.)
| | - Connie C. Wong
- From the University of Arizona, Tucson, Ariz (P.H.K.); Memorial
Sloan-Kettering Cancer Center, New York, NY (M.J.M.); Invicro, Needham, Mass
(J.H.); Mayo Clinic, Rochester, Minn (A.T.K., O.S.); Department of Nuclear
Medicine, University Hospital Münster, Münster, Germany (K.R.);
West German Cancer Center, Münster and Essen, Germany (K.R.); Dana-Farber
Cancer Institute, Boston, Mass (X.X.W.); Astera Cancer Care, East Brunswick, NJ
(B.F.); Indiana University Simon Comprehensive Cancer Center, Indianapolis, Ind
(N.A.); Miami Cancer Institute, Baptist Health South Florida, Miami, Fla (R.G.);
Washington University, St. Louis, Mo (J.M.M.); British Columbia Cancer Agency,
Vancouver, British Columbia, Canada (K.C.); The Institute of Cancer Research and
Royal Marsden Hospital, London, United Kingdom (J.d.B.); Gustave Roussy
Institute, University of Paris-Saclay, Villejuif, France (K.F.); Rostock
University Medical Center, Rostock, Germany (B.K.); Weill Cornell Medicine, New
York, NY (S.T.T.); Novartis Pharmaceuticals, East Hanover, NJ (S.G.); Novartis
Pharmaceuticals, Indianapolis, Ind (M.B.); Novartis Pharmaceuticals, Cambridge,
Mass (C.C.W.); Novartis Pharmaceuticals, Geneva, Switzerland (A.M.C.); Novartis
Pharmaceuticals, St. George, Utah (T.B.); Duke Cancer Institute Center for
Prostate and Urologic Cancers, Duke University, Durham, NC (A.J.A.); and
University Hospital Essen and German Cancer Consortium, Hufelandstr. 55, 45147
Essen, Germany (K.H.)
| | - Ana M. Catafau
- From the University of Arizona, Tucson, Ariz (P.H.K.); Memorial
Sloan-Kettering Cancer Center, New York, NY (M.J.M.); Invicro, Needham, Mass
(J.H.); Mayo Clinic, Rochester, Minn (A.T.K., O.S.); Department of Nuclear
Medicine, University Hospital Münster, Münster, Germany (K.R.);
West German Cancer Center, Münster and Essen, Germany (K.R.); Dana-Farber
Cancer Institute, Boston, Mass (X.X.W.); Astera Cancer Care, East Brunswick, NJ
(B.F.); Indiana University Simon Comprehensive Cancer Center, Indianapolis, Ind
(N.A.); Miami Cancer Institute, Baptist Health South Florida, Miami, Fla (R.G.);
Washington University, St. Louis, Mo (J.M.M.); British Columbia Cancer Agency,
Vancouver, British Columbia, Canada (K.C.); The Institute of Cancer Research and
Royal Marsden Hospital, London, United Kingdom (J.d.B.); Gustave Roussy
Institute, University of Paris-Saclay, Villejuif, France (K.F.); Rostock
University Medical Center, Rostock, Germany (B.K.); Weill Cornell Medicine, New
York, NY (S.T.T.); Novartis Pharmaceuticals, East Hanover, NJ (S.G.); Novartis
Pharmaceuticals, Indianapolis, Ind (M.B.); Novartis Pharmaceuticals, Cambridge,
Mass (C.C.W.); Novartis Pharmaceuticals, Geneva, Switzerland (A.M.C.); Novartis
Pharmaceuticals, St. George, Utah (T.B.); Duke Cancer Institute Center for
Prostate and Urologic Cancers, Duke University, Durham, NC (A.J.A.); and
University Hospital Essen and German Cancer Consortium, Hufelandstr. 55, 45147
Essen, Germany (K.H.)
| | - Taylor Benson
- From the University of Arizona, Tucson, Ariz (P.H.K.); Memorial
Sloan-Kettering Cancer Center, New York, NY (M.J.M.); Invicro, Needham, Mass
(J.H.); Mayo Clinic, Rochester, Minn (A.T.K., O.S.); Department of Nuclear
Medicine, University Hospital Münster, Münster, Germany (K.R.);
West German Cancer Center, Münster and Essen, Germany (K.R.); Dana-Farber
Cancer Institute, Boston, Mass (X.X.W.); Astera Cancer Care, East Brunswick, NJ
(B.F.); Indiana University Simon Comprehensive Cancer Center, Indianapolis, Ind
(N.A.); Miami Cancer Institute, Baptist Health South Florida, Miami, Fla (R.G.);
Washington University, St. Louis, Mo (J.M.M.); British Columbia Cancer Agency,
Vancouver, British Columbia, Canada (K.C.); The Institute of Cancer Research and
Royal Marsden Hospital, London, United Kingdom (J.d.B.); Gustave Roussy
Institute, University of Paris-Saclay, Villejuif, France (K.F.); Rostock
University Medical Center, Rostock, Germany (B.K.); Weill Cornell Medicine, New
York, NY (S.T.T.); Novartis Pharmaceuticals, East Hanover, NJ (S.G.); Novartis
Pharmaceuticals, Indianapolis, Ind (M.B.); Novartis Pharmaceuticals, Cambridge,
Mass (C.C.W.); Novartis Pharmaceuticals, Geneva, Switzerland (A.M.C.); Novartis
Pharmaceuticals, St. George, Utah (T.B.); Duke Cancer Institute Center for
Prostate and Urologic Cancers, Duke University, Durham, NC (A.J.A.); and
University Hospital Essen and German Cancer Consortium, Hufelandstr. 55, 45147
Essen, Germany (K.H.)
| | | | | | - Sarah Atzen
- From the University of Arizona, Tucson, Ariz (P.H.K.); Memorial
Sloan-Kettering Cancer Center, New York, NY (M.J.M.); Invicro, Needham, Mass
(J.H.); Mayo Clinic, Rochester, Minn (A.T.K., O.S.); Department of Nuclear
Medicine, University Hospital Münster, Münster, Germany (K.R.);
West German Cancer Center, Münster and Essen, Germany (K.R.); Dana-Farber
Cancer Institute, Boston, Mass (X.X.W.); Astera Cancer Care, East Brunswick, NJ
(B.F.); Indiana University Simon Comprehensive Cancer Center, Indianapolis, Ind
(N.A.); Miami Cancer Institute, Baptist Health South Florida, Miami, Fla (R.G.);
Washington University, St. Louis, Mo (J.M.M.); British Columbia Cancer Agency,
Vancouver, British Columbia, Canada (K.C.); The Institute of Cancer Research and
Royal Marsden Hospital, London, United Kingdom (J.d.B.); Gustave Roussy
Institute, University of Paris-Saclay, Villejuif, France (K.F.); Rostock
University Medical Center, Rostock, Germany (B.K.); Weill Cornell Medicine, New
York, NY (S.T.T.); Novartis Pharmaceuticals, East Hanover, NJ (S.G.); Novartis
Pharmaceuticals, Indianapolis, Ind (M.B.); Novartis Pharmaceuticals, Cambridge,
Mass (C.C.W.); Novartis Pharmaceuticals, Geneva, Switzerland (A.M.C.); Novartis
Pharmaceuticals, St. George, Utah (T.B.); Duke Cancer Institute Center for
Prostate and Urologic Cancers, Duke University, Durham, NC (A.J.A.); and
University Hospital Essen and German Cancer Consortium, Hufelandstr. 55, 45147
Essen, Germany (K.H.)
| |
Collapse
|
13
|
Green A, Temsah P, Goldfarb L, Sanfolippo K, Knoche E, Muzaffar R, Osman MM. Evaluating appropriateness of 18F-fluciclovine PET/CT relative to standard of care imaging guidelines and the impact of ADT on positivity: a prospective study in 62 Veterans Administration patients at a single institution. Nucl Med Commun 2024; 45:526-535. [PMID: 38517329 DOI: 10.1097/mnm.0000000000001836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
BACKGROUND According to the National Comprehensive Cancer Network Guidelines, 18F-fluciclovine PET/CT is considered appropriate after negative standard of care (SOC) imaging. OBJECTIVE To prospectively compare 18F-fluciclovine to SOC imaging, investigate whether it should be done when SOC imaging is (+), and evaluate its detection rate in patients receiving androgen deprivation therapy. METHODS We recruited 57 prostate cancer patients with biochemical recurrence with 18F-fluciclovine PET/CT and SOC imaging within 30 days. Prostate-specific antigen (PSA) level, Gleason score (GS), history of radical prostatectomy (RP), radiation therapy (RT) or hormone therapy (HT) were reviewed. RESULTS The 57 patients had a median PSA of 2.6 and average GS of 7.4; 27 (47.4%) had RP, 28 (49.1%) had RT, 1 (1.75%) had HT and 1 (1.75%) observation only. 18F-fluciclovine identified disease recurrence in 45/57 patients (78.9%), including oligometastasis in 18/45 (40%). SOC imaging identified recurrent disease in 12/57 patients (21.1%) while 18F-fluciclvoine identified additional sites of disease in 11/12 (91.7%). The (+) 18F-fluciclovine studies had a median PSA 2.6 ng/ml compared to 6.0 ng/ml in the (+) SOC studies. CONCLUSION 18F-fluciclovine was superior to SOC imaging for lesion detection, identification of oligometastasis and identification of additional sites of disease.
Collapse
Affiliation(s)
- Aileen Green
- Department of Radiology, Saint Louis VA Medical Center, and
| | - Peter Temsah
- Division of Nuclear Medicine, Department of Radiology, Saint Louis University and
| | | | - Kristen Sanfolippo
- Department of Internal Medicine, Saint Louis VA Medical Center, Saint Louis, Missouri, USA
| | - Eric Knoche
- Department of Internal Medicine, Saint Louis VA Medical Center, Saint Louis, Missouri, USA
| | - Razi Muzaffar
- Division of Nuclear Medicine, Department of Radiology, Saint Louis University and
| | - Medhat M Osman
- Department of Radiology, Saint Louis VA Medical Center, and
- Division of Nuclear Medicine, Department of Radiology, Saint Louis University and
| |
Collapse
|
14
|
Bhattacharya S, Stillahn A, Smith K, Muders M, Datta K, Dutta S. Understanding the molecular regulators of neuroendocrine prostate cancer. Adv Cancer Res 2024; 161:403-429. [PMID: 39032955 DOI: 10.1016/bs.acr.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Worldwide, prostate cancer (PCa) remains a leading cause of death in men. Histologically, the majority of PCa cases are classified as adenocarcinomas, which are mainly composed of androgen receptor-positive luminal cells. PCa is initially driven by the androgen receptor axis, where androgen-mediated activation of the receptor is one of the primary culprits for disease progression. Therefore, in advanced stage PCa, patients are generally treated with androgen deprivation therapies alone or in combination with androgen receptor pathway inhibitors. However, after an initial decrease, the cancer recurs for majority patients. At this stage, cancer is known as castration-resistant prostate cancer (CRPC). Majority of CRPC tumors still depend on androgen receptor axis for its progression to metastasis. However, in around 20-30% of cases, CRPC progresses via an androgen receptor-independent pathway and is often presented as neuroendocrine cancer (NE). This NE phenotype is highly aggressive with poor overall survival as compared to CRPC adenocarcinoma. NE cancers are resistant to standard taxane chemotherapies, which are often used to treat metastatic disease. Pathologically and morphologically, NE cancers are highly diverse and often co-exist with adenocarcinoma. Due to the lack of proper biomarkers, it is often difficult to make an early diagnosis of this lethal disease. Moreover, increased tumor heterogeneity and admixtures of adeno and NE subtypes in the same tumor make early detection of NE tumors very difficult. With the advancement of our knowledge and sequencing technology, we are now able to better understand the molecular mediators of this transformation pathway. This current study will give an update on how various molecular regulators are involved in these lineage transformation processes and what challenges we are still facing to detect and treat this cancer.
Collapse
Affiliation(s)
- Sreyashi Bhattacharya
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, United States; Department of Biochemistry and Molecular Biology, Massy Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Avery Stillahn
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, United States
| | - Kaitlin Smith
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, United States
| | | | - Kaustubh Datta
- Department of Biochemistry and Molecular Biology, Massy Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Samikshan Dutta
- Department of Biochemistry and Molecular Biology, Massy Cancer Center, Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
15
|
Chen DC, Huang S, Buteau JP, Kashyap R, Hofman MS. Clinical Positron Emission Tomography/Computed Tomography: Quarter-Century Transformation of Prostate Cancer Molecular Imaging. PET Clin 2024; 19:261-279. [PMID: 38199918 DOI: 10.1016/j.cpet.2023.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Although positron emission tomography/computed tomography (PET/CT) underwent rapid growth during the last quarter-century, becoming a new standard-of-care for imaging most cancer types, CT and bone scan remained the gold standard for patients with prostate cancer. This occurred as 2-fluorine-18-fluoro-2-deoxy-d-glucose was perceived to have a limited role owing to low sensitivity in many patients. A resurgence of interest occurred with the use of fluorine-18-sodium-fluoride PET/CT as a replacement for bone scintigraphy, and then choline, fluciclovine, and dihydrotestosterone (DHT) PET/CT as prostate "specific" radiotracers. The last decade, however, has seen a true revolution with the meteoric rise of prostate-specific membrane antigen PET/CT.
Collapse
Affiliation(s)
- David C Chen
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Siyu Huang
- Department of Surgery, The University of Melbourne
| | - James P Buteau
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Raghava Kashyap
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Michael S Hofman
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
16
|
Moran S, Cheng HH, Weg E, Kim EH, Chen DL, Iravani A, Ippolito JE. Prostate-specific membrane antigen-positron emission tomography (PSMA-PET) of prostate cancer: current and emerging applications. Abdom Radiol (NY) 2024; 49:1288-1305. [PMID: 38386156 DOI: 10.1007/s00261-024-04188-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 01/03/2024] [Accepted: 01/07/2024] [Indexed: 02/23/2024]
Abstract
Prostate-specific membrane antigen-positron emission tomography (PSMA-PET) is transforming the management of patients with prostate cancer. In appropriately selected patients, PSMA-PET offers superior sensitivity and specificity compared to conventional imaging (e.g., computed tomography and bone scintigraphy) as well as choline and fluciclovine PET, with the added benefit of consolidating bone and soft tissue evaluation into a single study. Despite being a newly available imaging tool, PSMA-PET has established indications, interpretation guidelines, and reporting criteria, which will be reviewed. The prostate cancer care team, from imaging specialists to those delivering treatment, should have knowledge of physiologic PSMA radiotracer uptake, patterns of disease spread, and the strengths and limitations of PSMA-PET. In this review, current and emerging applications of PSMA-PET, including appropriateness use criteria as well as image interpretation and pitfalls, will be provided with an emphasis on clinical implications.
Collapse
Affiliation(s)
- Shamus Moran
- Department of Radiology, University of Washington School of Medicine, Seattle, WA, USA
| | - Heather H Cheng
- Division of Oncology, Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Emily Weg
- Department of Radiation Oncology, University of Washington School of Medicine, Seattle, WA, USA
| | - Eric H Kim
- Division of Urologic Surgery, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Delphine L Chen
- Division of Nuclear Medicine, Department of Radiology, University of Washington School of Medicine, Seattle, WA, USA
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Amir Iravani
- Division of Nuclear Medicine, Department of Radiology, University of Washington School of Medicine, Seattle, WA, USA
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Joseph E Ippolito
- Mallinckrodt Institute of Radiology, Washington University School of Medicine in St. Louis, 4559 Scott Ave., Mail Stop Code: 8131, St. Louis, MO, 63110, USA.
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
17
|
Giunta EF, Brighi N, Gurioli G, Matteucci F, Paganelli G, De Giorgi U. 177Lu-PSMA therapy in metastatic prostate cancer: An updated review of prognostic and predictive biomarkers. Cancer Treat Rev 2024; 125:102699. [PMID: 38422894 DOI: 10.1016/j.ctrv.2024.102699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/14/2024] [Accepted: 02/16/2024] [Indexed: 03/02/2024]
Abstract
177Lu-PSMA has been approved for the treatment of PSMA-positive metastatic castration-resistant (mCRPC) patients who progressed to androgen receptor pathway inhibitors (ARPIs) and taxane-based chemotherapy. However, a higher proportion of patients do not respond to this type of radioligand therapy (RLT). To date, there is a lack of validated prognostic and predictive biomarkers for 177Lu-PSMA therapy in prostate cancer. Several studies have investigated the prognostic and predictive role of clinical and molecular factors and also the metabolic features of PET imaging. In this review, we aim to take stock of the current scenario, focusing on new emerging data from retrospective/prospective series and clinical trials. Given the high costs and the possibility of primary resistance, it seems essential to identify clinical and molecular characteristics that could allow clinicians to choose the right patient to treat with 177Lu-PSMA. Biomarker-based clinical trials are urgently needed in this field.
Collapse
Affiliation(s)
- Emilio Francesco Giunta
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy.
| | - Nicole Brighi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giorgia Gurioli
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Federica Matteucci
- Nuclear Medicine Operative Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giovanni Paganelli
- Nuclear Medicine Operative Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Ugo De Giorgi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| |
Collapse
|
18
|
Bauckneht M, Ciccarese C, Laudicella R, Mosillo C, D'Amico F, Anghelone A, Strusi A, Beccia V, Bracarda S, Fornarini G, Tortora G, Iacovelli R. Theranostics revolution in prostate cancer: Basics, clinical applications, open issues and future perspectives. Cancer Treat Rev 2024; 124:102698. [PMID: 38359590 DOI: 10.1016/j.ctrv.2024.102698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/06/2024] [Accepted: 02/09/2024] [Indexed: 02/17/2024]
Abstract
In the last years, theranostics has expanded the therapeutic options available for prostate cancer patients. In this review, we explore this dynamic field and its potential to revolutionize precision medicine for prostate cancer. We delve into the foundational principles, clinical applications, and emerging opportunities, emphasizing the potential synergy between radioligand therapy and other systemic treatments. Additionally, we address the ongoing challenges, including optimizing patient selection, assessing treatment responses, and determining the role of theranostics within the broader landscape of prostate cancer treatment.
Collapse
Affiliation(s)
- Matteo Bauckneht
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Genova, Italy; Department of Health Sciences (DISSAL), University of Genova, Genova, Italy.
| | - Chiara Ciccarese
- Medical Oncology, Fondazione Policlinico A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Riccardo Laudicella
- Nuclear Medicine Unit, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, 98124 Messina, Italy
| | - Claudia Mosillo
- Oncologia Medica e Traslazionale, Azienda Ospedaliera Santa Maria di Terni, Terni, Italy
| | - Francesca D'Amico
- Department of Health Sciences (DISSAL), University of Genova, Genova, Italy
| | - Annunziato Anghelone
- Medical Oncology, Fondazione Policlinico A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Alessandro Strusi
- Medical Oncology, Fondazione Policlinico A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Viria Beccia
- Medical Oncology, Fondazione Policlinico A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy
| | - Sergio Bracarda
- Oncologia Medica e Traslazionale, Azienda Ospedaliera Santa Maria di Terni, Terni, Italy
| | - Giuseppe Fornarini
- Medical Oncology 1, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Giampaolo Tortora
- Medical Oncology, Fondazione Policlinico A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; Università Cattolica del Sacro Cuore, Rome, Italy
| | - Roberto Iacovelli
- Medical Oncology, Fondazione Policlinico A. Gemelli IRCCS, Largo Agostino Gemelli 8, 00168 Rome, Italy; Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
19
|
Ergül N, Çermik TF, Alçın G, Arslan E, Erol Fenercioğlu Ö, Beyhan E, Şahin R, Baloğlu MC, Baykal Koca S, Türkay R, Yücetaş U. Contribution of 68 Ga-DOTA-FAPI-04 PET/CT to Prostate Cancer Imaging : Complementary Role in PSMA-Negative Cases. Clin Nucl Med 2024; 49:e105-e110. [PMID: 38271254 DOI: 10.1097/rlu.0000000000005064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Abstract
PURPOSE Prostate-specific membrane antigen (PSMA)-targeted PET/CT is a well-established imaging method in prostate cancer (PC) for both staging and restaging, and also for theranostic applications. An alternative imaging method is crucial for 15% PSMA-negative cases. We aimed to investigate the contribution of 68 Ga-DOTA-FAPI-04 PET/CT to PC imaging. PATIENTS AND METHODS Thirty-six patients diagnosed with PC were included. Patients underwent both 68 Ga-PSMA PET/CT and 68 Ga-DOTA-FAPI-04 PET/CT imaging within 1 week. In staging group, primary tumor uptake values were compared, and also correlations were done with histopathological findings, MRI findings, and total PSA levels. In biochemical recurrence group, the uptake values in prostatic region and metastases were evaluated to define the local recurrence or metastatic disease. RESULTS In staging group, PSMA PET showed increased uptake in the primary lesion area in 14/27 (52%) patients, whereas 20/27 (74%) patients were positive in FAPI-04 PET. FAPI-04 positivity was found to be quite high, such as 54%, in PSMA-negative patients. A significant difference was observed between ISUP grade 1-3 patients and ISUP grade 4-5 patients in FAPI-04 PET ( P = 0.03). Local recurrence was detected in 3 patients, pelvic lymph node metastasis in 1 patient, and sacrum metastasis in 1 patient in biochemical recurrence group, and all of the lesions had more intense uptake in PSMA PET than FAPI-04 PET. CONCLUSIONS FAPI PET imaging seems to have a potential to contribute PSMA PET imaging with FAPI positivity in more than half of PSMA-negative cases. Also, FAPI-targeted radionuclide therapy may be a promising method in patients resistant to PSMA-targeted therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Mehmnet Can Baloğlu
- Pathology, Istanbul Training and Research Hospital, University of Health Sciences
| | - Sevim Baykal Koca
- Pathology, Istanbul Training and Research Hospital, University of Health Sciences
| | - Rüştü Türkay
- Clinic of Radiology, Haseki Training and Research Hospital, University of Health Sciences
| | - Uğur Yücetaş
- Clinic of Urology, Istanbul Training and Research Hospital, University of Health Sciences, Istanbul, Türkiye
| |
Collapse
|
20
|
Al Saffar H, Chen DC, Delgado C, Ingvar J, Hofman MS, Lawrentschuk N, Perera M, Murphy DG, Eapen R. The Current Landscape of Prostate-Specific Membrane Antigen (PSMA) Imaging Biomarkers for Aggressive Prostate Cancer. Cancers (Basel) 2024; 16:939. [PMID: 38473301 PMCID: PMC10931387 DOI: 10.3390/cancers16050939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
The review examines the vital role of prostate-specific membrane antigen (PSMA) positron emission tomography/computed tomography (PET/CT) in the diagnosis, staging, and treatment of prostate cancer (PCa). It focuses on the superior diagnostic abilities of PSMA PET/CT for identifying both nodal and distant PCa, and its potential as a prognostic indicator for biochemical recurrence and overall survival. Additionally, we focused on the variability of PSMA's expression and its impact on personalised treatment, particularly the use of [177Lu] Lu-PSMA-617 radioligand therapy. This review emphasises the essential role of PSMA PET/CT in enhancing treatment approaches, improving patient outcomes, and reducing unnecessary interventions, positioning it as a key element in personalised PCa management.
Collapse
Affiliation(s)
- Haidar Al Saffar
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia; (D.C.C.); (J.I.); (N.L.); (M.P.); (D.G.M.); (R.E.)
| | - David C. Chen
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia; (D.C.C.); (J.I.); (N.L.); (M.P.); (D.G.M.); (R.E.)
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia;
- Department of Surgery, Austin Health, Heidelberg, VIC 3084, Australia
| | - Carlos Delgado
- School of Medicine and Health Sciences, Tecnologico de Monterrey, Monterrey 64849, Mexico;
| | - Jacob Ingvar
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia; (D.C.C.); (J.I.); (N.L.); (M.P.); (D.G.M.); (R.E.)
| | - Michael S. Hofman
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia;
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Nathan Lawrentschuk
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia; (D.C.C.); (J.I.); (N.L.); (M.P.); (D.G.M.); (R.E.)
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3052, Australia
- Department of Surgery (Urology), Royal Melbourne Hospital, Melbourne, VIC 3052, Australia
- EJ Whitten Prostate Cancer Research Centre, Epworth Hospital, Richmond, VIC 3121, Australia
| | - Marlon Perera
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia; (D.C.C.); (J.I.); (N.L.); (M.P.); (D.G.M.); (R.E.)
- Department of Surgery, Austin Health, Heidelberg, VIC 3084, Australia
| | - Declan G. Murphy
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia; (D.C.C.); (J.I.); (N.L.); (M.P.); (D.G.M.); (R.E.)
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia;
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Renu Eapen
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia; (D.C.C.); (J.I.); (N.L.); (M.P.); (D.G.M.); (R.E.)
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia;
- Department of Surgery, Austin Health, Heidelberg, VIC 3084, Australia
| |
Collapse
|
21
|
Michalski K, Kosmala A, Werner RA, Serfling SE, Seitz AK, Lapa C, Buck AK, Hartrampf PE. Comparison of PET/CT-based eligibility according to VISION and TheraP trial criteria in end-stage prostate cancer patients undergoing radioligand therapy. Ann Nucl Med 2024; 38:87-95. [PMID: 37891376 PMCID: PMC10822822 DOI: 10.1007/s12149-023-01874-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023]
Abstract
BACKGROUND Two randomized clinical trials demonstrated the efficacy of prostate-specific membrane antigen (PSMA) radioligand therapy (PSMA RLT) in metastatic castration-resistant prostate cancer (mCRPC). While the VISION trial used criteria within PSMA PET/CT for inclusion, the TheraP trial used dual tracer imaging including FDG PET/CT. Therefore, we investigated whether the application of the VISION criteria leads to a benefit in overall survival (OS) or progression-free survival (PFS) for men with mCRPC after PSMA RLT. METHODS Thirty-five men with mCRPC who had received PSMA RLT as a last-line option and who had undergone pretherapeutic imaging with FDG and [68Ga]Ga-PSMA I&T or [18F]PSMA-1007 were studied. Therapeutic eligibility was retrospectively evaluated using the VISION and TheraP study criteria. RESULTS 26 of 35 (74%) treated patients fulfilled the VISION criteria (= VISION+) and only 17 of 35 (49%) fulfilled the TheraP criteria (= TheraP+). Significantly reduced OS and PFS after PSMA RLT was observed in patients rated VISION- compared to VISION+ (OS: VISION-: 3 vs. VISION+: 12 months, hazard ratio (HR) 3.1, 95% confidence interval (CI) 1.0-9.1, p < 0.01; PFS: VISION-: 1 vs. VISION+: 5 months, HR 2.7, 95% CI 1.0-7.8, p < 0.01). For patients rated TheraP-, no significant difference in OS but in PFS was observed compared to TheraP+ patients (OS: TheraP-: 5.5 vs. TheraP+: 11 months, HR 1.6, 95% CI 0.8-3.3, p = 0.2; PFS: TheraP-: 1 vs. TheraP+: 6 months, HR 2.2, 95% CI 1.0-4.5, p < 0.01). CONCLUSION Retrospective application of the inclusion criteria of the VISION study leads to a benefit in OS and PFS after PSMA RL, whereas TheraP criteria appear to be too strict in patients with end-stage prostate cancer. Thus, performing PSMA PET/CT including a contrast-enhanced CT as proposed in the VISION trial might be sufficient for treatment eligibility of end-stage prostate cancer patients.
Collapse
Affiliation(s)
- Kerstin Michalski
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Straße 6, 97080, Würzburg, Germany.
| | - Aleksander Kosmala
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Straße 6, 97080, Würzburg, Germany
| | - Rudolf A Werner
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Straße 6, 97080, Würzburg, Germany
| | - Sebastian E Serfling
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Straße 6, 97080, Würzburg, Germany
| | - Anna K Seitz
- Department of Urology and Paediatric Urology, University Hospital Würzburg, Oberdürrbacher Straße 6, 97080, Würzburg, Germany
| | - Constantin Lapa
- Nuclear Medicine, Medical Faculty, University of Augsburg, Stenglinstr. 2, 86156, Augsburg, Germany
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Straße 6, 97080, Würzburg, Germany
| | - Philipp E Hartrampf
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Straße 6, 97080, Würzburg, Germany
| |
Collapse
|
22
|
Weiner AB, Agrawal R, Valle LF, Sonni I, Kishan AU, Rettig MB, Raman SS, Calais J, Boutros PC, Reiter RE. Impact of PSMA PET on Prostate Cancer Management. Curr Treat Options Oncol 2024; 25:191-205. [PMID: 38270802 PMCID: PMC11034977 DOI: 10.1007/s11864-024-01181-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2024] [Indexed: 01/26/2024]
Abstract
OPINION STATEMENT PSMA-PET has been a practice-changing imaging biomarker for the management of men with PCa. Research suggests improved accuracy over conventional imaging and other PET radiotracers in many contexts. With multiple approved PSMA-targeting radiotracers, PSMA PET will become even more available in clinical practice. Its increased use requires an understanding of the prospective data available and caution when extrapolating from prior trial data that utilized other imaging modalities. Future trials leveraging PSMA PET for treatment optimization and management decision-making will ultimately drive its clinical utility.
Collapse
Affiliation(s)
- Adam B Weiner
- Department of Urology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA.
- Institute for Precision Health, University of California-Los Angeles, Los Angeles, CA, USA.
| | - Raag Agrawal
- Institute for Precision Health, University of California-Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, CA, USA
| | - Luca F Valle
- Department of Radiation Oncology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Ida Sonni
- Department of Radiological Sciences, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- Department of Clinical and Experimental Medicine, University Magna Graecia, Catanzaro, Italy
| | - Amar U Kishan
- Department of Radiation Oncology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
| | - Matthew B Rettig
- Department of Urology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
| | - Steven S Raman
- Department of Radiological Sciences, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
| | - Jeremie Calais
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
| | - Paul C Boutros
- Department of Urology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- Institute for Precision Health, University of California-Los Angeles, Los Angeles, CA, USA
- Department of Human Genetics, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, CA, USA
| | - Robert E Reiter
- Department of Urology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
23
|
Klyuzhin IS, Chaussé G, Bloise I, Harsini S, Ferres JL, Uribe C, Rahmim A. PSMA-Hornet: Fully-automated, multi-target segmentation of healthy organs in PSMA PET/CT images. Med Phys 2024; 51:1203-1216. [PMID: 37544015 DOI: 10.1002/mp.16658] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 03/28/2023] [Accepted: 05/26/2023] [Indexed: 08/08/2023] Open
Abstract
BACKGROUND Prostate-specific membrane antigen (PSMA) PET imaging represents a valuable source of information reflecting disease stage, response rate, and treatment optimization options, particularly with PSMA radioligand therapy. Quantification of radiopharmaceutical uptake in healthy organs from PSMA images has the potential to minimize toxicity by extrapolation of the radiation dose delivery towards personalization of therapy. However, segmentation and quantification of uptake in organs requires labor-intensive organ delineations that are often not feasible in the clinic nor scalable for large clinical trials. PURPOSE In this work we develop and test the PSMA Healthy organ segmentation network (PSMA-Hornet), a fully-automated deep neural net for simultaneous segmentation of 14 healthy organs representing the normal biodistribution of [18 F]DCFPyL on PET/CT images. We also propose a modified U-net architecture, a self-supervised pre-training method for PET/CT images, a multi-target Dice loss, and multi-target batch balancing to effectively train PSMA-Hornet and similar networks. METHODS The study used manually-segmented [18 F]DCFPyL PET/CT images from 100 subjects, and 526 similar images without segmentations. The unsegmented images were used for self-supervised model pretraining. For supervised training, Monte-Carlo cross-validation was used to evaluate the network performance, with 85 subjects in each trial reserved for model training, 5 for validation, and 10 for testing. Image segmentation and quantification metrics were evaluated on the test folds with respect to manual segmentations by a nuclear medicine physician, and compared to inter-rater agreement. The model's segmentation performance was also evaluated on a separate set of 19 images with high tumor load. RESULTS With our best model, the lowest mean Dice coefficient on the test set was 0.826 for the sublingual gland, and the highest was 0.964 for liver. The highest mean error in tracer uptake quantification was 13.9% in the sublingual gland. Self-supervised pretraining improved training convergence, train-to-test generalization, and segmentation quality. In addition, we found that a multi-target network produced significantly higher segmentation accuracy than single-organ networks. CONCLUSIONS The developed network can be used to automatically obtain high-quality organ segmentations for PSMA image analysis tasks. It can be used to reproducibly extract imaging data, and holds promise for clinical applications such as personalized radiation dosimetry and improved radioligand therapy.
Collapse
Affiliation(s)
- Ivan S Klyuzhin
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- AI for Health, Microsoft, Redmond, Washington, USA
- Deparment of Radiology, University of British Columbia, Vancouver, BC, Canada
| | - Guillaume Chaussé
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Ingrid Bloise
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Sara Harsini
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | | | - Carlos Uribe
- Deparment of Radiology, University of British Columbia, Vancouver, BC, Canada
- Department of Functional Imaging, BC Cancer, Vancouver, BC, Canada
| | - Arman Rahmim
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Deparment of Radiology, University of British Columbia, Vancouver, BC, Canada
- Deparment of Physics and Astronomy, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
24
|
Pan J, Zhang T, Chen S, Bu T, Zhao J, Ni X, Shi B, Gan H, Wei Y, Wang Q, Wang B, Wu J, Song S, Wang F, Liu C, Ye D, Zhu Y. Nomogram to predict the presence of PSMA-negative but FDG-positive lesion in castration-resistant prostate cancer: a multicenter cohort study. Ther Adv Med Oncol 2024; 16:17588359231220506. [PMID: 38188464 PMCID: PMC10771757 DOI: 10.1177/17588359231220506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 11/21/2023] [Indexed: 01/09/2024] Open
Abstract
Background PSMA-negative but FDG-positive (PSMA-/FDG+) lesion in dual-tracer (68Ga-PSMA and 18F-FDG) positron emission tomography/computed tomography (PET/CT) is associated with an unfavorable response to Lutetium-177 (177Lu)-PSMA-617. This study sought to develop both radiomics and clinical models for the precise prediction of the presence of PSMA-/FDG+ lesions in patients with castration-resistant prostate cancer (CPRC). Methods A cohort of 298 patients who underwent dual-tracer PET/CT with a less than 5-day interval was included. The evaluation of the prognostic performance of the radiomics model drew upon the survival data derived from 40 patients with CRPC treated with 177Lu-PSMA-617 in an external cohort. Two endpoints were evaluated: (a) prostate-specific antigen (PSA) response rate, defined as a reduction exceeding 50% from baseline and (b) overall survival (OS), measured from the initiation of 177Lu-PSMA-617 to death from any cause. Results PSMA-/FDG+ lesions were identified in 56 (18.8%) CRPC patients. Both radiomics (area under the curve [AUC], 0.83) and clinical models (AUC, 0.78) demonstrated robust performance in PSMA-/FDG+ lesion prediction. Decision curve analysis revealed that the radiomics model yielded a net benefit over the 'screen all' strategy at a threshold probability of ⩾4%. At a 5% probability threshold, the radiomics model facilitated a 21% reduction in 18F-FDG PET/CT scans while only missing 2% of PSMA-/FDG+ cases. Patients with a low estimated score exhibited significantly prolonged OS (hazard ratio = 0.49, p = 0.029) and a higher PSA response rate (75% versus 35%, p = 0.011) compared to those with a high estimated score. Conclusion This study successfully developed two models with accurate estimations of the risk associated with PSMA-/FDG+ lesions in CRPC patients. These models held potential utility in aiding the selection of candidates for 177Lu-PSMA-617 treatment and guiding 68Ga-PSMA PET/CT-directed radiotherapy.
Collapse
Affiliation(s)
- Jian Pan
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Tingwei Zhang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shouzhen Chen
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong Province, China
| | - Ting Bu
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
- Department of Nuclear Medicine, The Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, China
| | - Jinou Zhao
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xudong Ni
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Benkang Shi
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Shandong Province, China
| | - Hualei Gan
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yu Wei
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qifeng Wang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Beihe Wang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Junlong Wu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shaoli Song
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Feng Wang
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Chang Liu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yao Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Genitourinary Cancer Institute, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
25
|
Sutherland DEK, Azad AA, Murphy DG, Eapen RS, Kostos L, Hofman MS. Role of FDG PET/CT in Management of Patients with Prostate Cancer. Semin Nucl Med 2024; 54:4-13. [PMID: 37400321 DOI: 10.1053/j.semnuclmed.2023.06.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 07/05/2023]
Abstract
Prostate cancer is the second most common cancer in men worldwide. [18F]FDG PET/CT imaging, a well-known and effective technique for detecting malignancies, has not been considered a useful tool for prostate cancer imaging by many because of its perceived low [18F]FDG uptake. Incidentally detected focal [18F]FDG uptake in the prostate is not uncommon, and typically benign. Imaging features that would increase concern for an underlying prostatic carcinoma, include focal uptake in the periphery near the gland margin without calcifications. [18F]FDG PET/CT imaging provides little value in the initial staging of prostate cancer, particularly in the era of prostate specific membrane antigen (PSMA) radiotracer. In cases of biochemical recurrence, the value of [18F]FDG PET/CT increases notably when Grade group 4 or 5 and elevated PSA levels are present. Active research is underway for theranostic approaches to prostate cancer, including [177Lu]Lu-PSMA therapy. Dual tracer staging using FDG and PSMA imaging significantly enhances the accuracy of disease site assessment. Specifically, the addition of [18F]FDG PET/CT imaging allows for the evaluation of discordant disease (PSMA negative/FDG positive). The maximal benefit from [177Lu]Lu-PSMA therapy relies on significant PSMA accumulation across all disease sites, and the identification of discordant disease suggests that these patients may derive less benefit from the treatment. The genuine value of [18F]FDG PET/CT imaging lies in advanced prostate cancer, PSMA-negative disease, as a prognostic biomarker, and the realm of new targeted theranostic agents.
Collapse
Affiliation(s)
- Duncan E K Sutherland
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Arun A Azad
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia; Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Declan G Murphy
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia; Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Renu S Eapen
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia; Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Louise Kostos
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia; Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Michael S Hofman
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
26
|
Abstract
Prostate cancer (PC) is a significant health concern worldwide, with high incidence and mortality rates. Early and accurate detection and localization of recurrent disease at biochemical recurrence (BCR) is critical for guiding subsequent therapeutic decisions and improving patient outcomes. At BCR, conventional imaging consisting of CT, MRI, and bone scintigraphy are recommended by US and European guidelines, however, these modalities all bear certain limitations in detecting metastatic disease, particularly in low-volume relapse at low prostate-specific antigen (PSA) levels. Molecular imaging with PET/CT or PET/MRI using prostate-specific membrane antigen (PSMA) targeting radiopharmaceuticals has revolutionized imaging of PC. Particularly at BCR PC, PSMA PET has shown better diagnostic performance compared to conventional imaging in detecting local relapse and metastases, even at very low PSA levels. The most recent version of the National Comprehensive Cancer Network (NCCN) guideline has included PSMA-targeted PET/CT or PET/MRI for the localization of BCR PC. There are several different PSMA-targeting radiopharmaceuticals labeled with different radioisotopes, each with slightly different characteristics, but overall similar high sensitivity and specificity for PC. PSMA-targeted PET has the potential to significantly impact patient care by guiding personalized treatment decisions and thus improving outcomes in BCR PC patients.
Collapse
Affiliation(s)
- Heying Duan
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, Stanford University, Stanford, CA
| | - Andrei Iagaru
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, Stanford University, Stanford, CA.
| |
Collapse
|
27
|
Yu XY, Zhu YQ, Liu X, Tian R, Chen JJ, Liu GQ, Yang DY, Zhang XP, Li B, Zhao HJ, Li X. Case report: 177Lu DOTA-TATE: a new scheme for the treatment of prostate neuroendocrine cancer. Front Oncol 2023; 13:1289272. [PMID: 38152366 PMCID: PMC10752594 DOI: 10.3389/fonc.2023.1289272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/27/2023] [Indexed: 12/29/2023] Open
Abstract
Background Most instances of small cell carcinoma originate from the lungs, while the gastrointestinal tract serves as a secondary site. Only a minuscule proportion of cases manifest within the urogenital system. Prostate small cell carcinoma (SCCP) represents an exceedingly uncommon pathological subtype within the realm of prostate cancer, displaying significant rarity in clinical settings. This scarcity has resulted in a paucity of adequate foundational and clinical research for SCCP treatment. While investigations have unveiled a certain therapeutic efficacy of radiotherapy and chemotherapy for SCCP, clinical practice has revealed suboptimal treatment outcomes. We hereby present a case report detailing the utilization of 177Lu-DOTA-TATE in the treatment of SCCP, aiming to investigate the therapeutic efficacy of 177Lu-DOTA-TATE for SCCP. Case presentation A male patient in his 80s presented with elevated prostate-specific antigen (PSA) levels and underwent a biopsy that revealed prostate adenocarcinoma. The patient received CAB (bicalutamide + goserelin) therapy. One year later, disease progression was detected, and a second biopsy confirmed the presence of prostate small cell carcinoma. Following the diagnosis of prostate small cell carcinoma, the patient underwent two cycles of 177Lu-DOTA-TATE treatment. Subsequent to the treatment, the original lesions showed shrinkage, metastatic lesions disappeared, and there was significant improvement, approaching complete remission. Conclusion SCCP exhibits a high degree of malignancy and aggressive invasiveness, currently lacking effective therapeutic modalities. The treatment course of this patient serves as compelling evidence for the efficacy of 177Lu-DOTA-TATE in managing SCCP, thereby opening new avenues for future SCCP treatments.
Collapse
Affiliation(s)
- Xin-yuan Yu
- Department of Urology Surgery, The First Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Yu-qin Zhu
- Department of Intensive Care, Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Xin Liu
- Department of Urology Surgery, The First Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Rong Tian
- Department of Urology Surgery, The First Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Jun-jie Chen
- Department of Urology Surgery, The First Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Guo-qing Liu
- Department of Urology Surgery, The First Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Dong-yu Yang
- Department of Urology Surgery, The First Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Xue-ping Zhang
- Department of Urology Surgery, The First Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Bao Li
- Department of Urology Surgery, The First Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Hong-jun Zhao
- Department of Urology Surgery, The First Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| | - Xiao Li
- Department of Urology Surgery, The First Affiliated Hospital of Weifang Medical University, Weifang, Shandong, China
| |
Collapse
|
28
|
Gaal S, Huang K, Rogasch JMM, Jochens HV, De Santis M, Erber B, Amthauer H. Prognostic Value of the De Ritis Ratio for Overall Survival in Patients with Metastatic Castration-Resistant Prostate Cancer Undergoing [ 177Lu]Lu-PSMA-617 Radioligand Therapy. Cancers (Basel) 2023; 15:4907. [PMID: 37894274 PMCID: PMC10605155 DOI: 10.3390/cancers15204907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/28/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
The De Ritis ratio (=aspartate transaminase/alanine transaminase) has shown prognostic value in different cancer types. This is the first such analysis in prostate cancer patients undergoing radioligand therapy (RLT) with [177Lu]Lu-PSMA-617. This retrospective monocentric analysis included 91 patients with a median of 3 RLT cycles (range 1-6) and median cumulative activity of 17.3 GBq. Univariable Cox regression regarding overall survival (OS) included age, different types of previous treatment, metastatic patterns and different laboratory parameters before RLT. Based on multivariable Cox regression, a prognostic score was derived. Seventy-two patients (79%) died (median follow-up in survivors: 19.8 months). A higher number of previous chemotherapy lines, the presence of liver metastases, brain metastases, a higher tumor load on PSMA-PET, a higher prostate-specific antigen (PSA) level, lower red blood cell count, lower hemoglobin, higher neutrophil-lymphocyte ratio and higher De Ritis ratio were associated with shorter OS (each p < 0.05). In multivariable Cox, a higher number of chemotherapy lines (range, 0-2; p = 0.036), brain metastases (p < 0.001), higher PSA (p = 0.004) and higher De Ritis ratio before RLT (hazard ratio, 1.27 per unit increase; p = 0.023) remained significant. This prognostic score separated five groups with a significantly different median OS ranging from 4.9 to 28.1 months (log-rank test, p < 0.001). If validated independently, the De Ritis ratio could enhance multifactorial models for OS after RLT.
Collapse
Affiliation(s)
- Sebastian Gaal
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Kai Huang
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Praxen für Diagnostische und Therapeutische Nuklearmedizin, Düppelstr. 30, 12163 Berlin, Germany
| | - Julian M M Rogasch
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
- Berlin Institute of Health, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Hans V Jochens
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Maria De Santis
- Department of Urology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
- Department of Urology, Medical University of Vienna, 1090 Vienna, Austria
| | - Barbara Erber
- Department of Urology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Holger Amthauer
- Department of Nuclear Medicine, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| |
Collapse
|
29
|
Laarhuis BI, Janssen MJR, Simons M, van Kalmthout LWM, van der Doelen MJ, Peters SMB, Westdorp H, van Oort IM, Litjens G, Gotthardt M, Nagarajah J, Mehra N, Privé BM. Tumoral Ki67 and PSMA Expression in Fresh Pre-PSMA-RLT Biopsies and Its Relation With PSMA-PET Imaging and Outcomes of PSMA-RLT in Patients With mCRPC. Clin Genitourin Cancer 2023; 21:e352-e361. [PMID: 37164814 DOI: 10.1016/j.clgc.2023.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 04/13/2023] [Indexed: 05/12/2023]
Abstract
INTRODUCTION Prostate specific membrane antigen (PSMA) directed radioligand therapy (RLT) is a novel therapy for metastatic castration-resistant prostate cancer (mCRPC) patients. However, it is still poorly understood why approximately 40% of the patients does not respond to PSMA-RLT. The aims of this study were to evaluate the pretreatment PSMA expression on immunohistochemistry (IHC) and PSMA uptake on PET/CT imaging in mCRPC patients who underwent PSMA-RLT. We correlated these parameters and a cell proliferation marker (Ki67) to the therapeutic efficacy of PSMA-RLT. PATIENTS AND METHODS In this retrospective study, mCRPC patients who underwent PSMA-RLT were analyzed. Patients biopsies were scored for immunohistochemical Ki67 expression, PSMA staining intensity and percentage of cells with PSMA expression. Moreover, the PSMA tracer uptake of the tumor lesion(s) and healthy organs on PET/CT imaging was assessed. The primary outcome was to evaluate the association between histological PSMA protein expression of tumor in pre-PSMA-RLT biopsies and the PSMA uptake on PSMA PET/CT imaging of the biopsied lesion. Secondary outcomes were to assess the relationship between PSMA expression and Ki67 on IHC and the progression free survival (PFS) and overall survival (OS) following PSMA-RLT. RESULTS In total, 22 mCRPC patients were included in this study. Nineteen (86%) patients showed a high and homogenous PSMA expression of >80% on IHC. Three (14%) patients had low PSMA expression on IHC. Although there was limited PSMA uptake on PET/CT imaging, these 3 patients had lower PSMA uptake on PET/CT imaging compared to the patients with high PSMA expression on IHC. Yet, no correlation was found between PSMA uptake on PET/CT imaging and PSMA expression on IHC (SUVmax: R2 = 0.046 and SUVavg: R2 = 0.036). The 3 patients had a shorter PFS compared to the patients with high PSMA expression on IHC (HR: 4.76, 95% CI: 1.14-19.99; P = .033). Patients with low Ki67 expression had a longer PFS and OS compared to patients with a high Ki67 expression (HR: 0.40, 95% CI: 0.15-1.06; P = .013) CONCLUSION: The PSMA uptake on PSMA-PET/CT generally followed the PSMA expression on IHC. However, heterogeneity may be missed on PSMA-PET/CT. Immunohistochemical PSMA and Ki67 expression in fresh tumor biopsies, may contribute to predict treatment efficacy of PSMA-RLT in mCRPC patients. This needs to be further explored in prospective cohorts.
Collapse
Affiliation(s)
- Babette I Laarhuis
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marcel J R Janssen
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Michiel Simons
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Maarten J van der Doelen
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Urology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Steffie M B Peters
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Harm Westdorp
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Inge M van Oort
- Department of Urology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Geert Litjens
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martin Gotthardt
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - James Nagarajah
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Niven Mehra
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Bastiaan M Privé
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
30
|
Volpe F, Nappi C, Piscopo L, Zampella E, Mainolfi CG, Ponsiglione A, Imbriaco M, Cuocolo A, Klain M. Emerging Role of Nuclear Medicine in Prostate Cancer: Current State and Future Perspectives. Cancers (Basel) 2023; 15:4746. [PMID: 37835440 PMCID: PMC10571937 DOI: 10.3390/cancers15194746] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
Prostate cancer is the most frequent epithelial neoplasia after skin cancer in men starting from 50 years and prostate-specific antigen (PSA) dosage can be used as an early screening tool. Prostate cancer imaging includes several radiological modalities, ranging from ultrasonography, computed tomography (CT), and magnetic resonance to nuclear medicine hybrid techniques such as single-photon emission computed tomography (SPECT)/CT and positron emission tomography (PET)/CT. Innovation in radiopharmaceutical compounds has introduced specific tracers with diagnostic and therapeutic indications, opening the horizons to targeted and very effective clinical care for patients with prostate cancer. The aim of the present review is to illustrate the current knowledge and future perspectives of nuclear medicine, including stand-alone diagnostic techniques and theragnostic approaches, in the clinical management of patients with prostate cancer from initial staging to advanced disease.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Michele Klain
- Department of Advanced Biomedical Sciences, University of Naples Federico II, 80138 Naples, Italy; (F.V.); (C.N.); (L.P.); (E.Z.); (C.G.M.); (A.P.); (M.I.); (A.C.)
| |
Collapse
|
31
|
Abouzayed A, Seitova K, Lundmark F, Bodenko V, Oroujeni M, Tolmachev V, Rosenström U, Orlova A. 177Lu-labeled PSMA targeting therapeutic with optimized linker for treatment of disseminated prostate cancer; evaluation of biodistribution and dosimetry. Front Oncol 2023; 13:1221103. [PMID: 37829345 PMCID: PMC10565663 DOI: 10.3389/fonc.2023.1221103] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 09/07/2023] [Indexed: 10/14/2023] Open
Abstract
Introduction Prostate specific membrane antigen (PSMA), highly expressed in metastatic castration-resistant prostate cancer (mCRPC), is an established therapeutic target. Theranostic PSMA-targeting agents are widely used in patient management and has shown improved outcomes for mCRPC patients. Earlier, we optimized a urea-based probe for radionuclide visualization of PSMA-expression in vivo using computer modeling. With the purpose to develop a targeting agent equally suitable for radionuclide imaging and therapy, the agent containing DOTA chelator was designed (BQ7876). The aim of the study was to test the hypothesis that 177Lu-labeled BQ7876 possesses target binding and biodistribution properties potentially enabling its use for radiotherapy. Methods BQ7876 was synthesized and labeled with Lu-177. Specificity and affinity of [177Lu]Lu-BQ7876 to PSMA-expressing PC3-pip cells was evaluated and its processing after binding to cells was studied. Animal studies in mice were performed to assess its biodistribution in vivo, target specificity and dosimetry. [177Lu]Lu-PSMA-617 was simultaneously evaluated for comparison. Results BQ7876 was labeled with Lu-177 with radiochemical yield >99%. Its binding to PSMA was specific in vitro and in vivo when tested in antigen saturation conditions as well as in PSMA-negative PC-3 tumors. The binding of [177Lu]Lu-BQ7876 to living cells was characterized by rapid association, while the dissociation included a rapid and a slow phase with affinities KD1 = 3.8 nM and KD2 = 25 nM. The half-maximal inhibitory concentration for natLu-BQ7876 was 59 nM that is equal to 61 nM for natLu-PSMA-617. Cellular processing of [177Lu]Lu-BQ7876 was accompanied by slow internalization. [177Lu]Lu-BQ7876 was cleared from blood and normal tissues rapidly. Initial elevated uptake in kidneys decreased rapidly, and by 3 h post injection, the renal uptake (13 ± 3%ID/g) did not differ significantly from tumor uptake (9 ± 3%ID/g). Tumor uptake was stable between 1 and 3 h followed by a slow decline. The highest absorbed dose was in kidneys, followed by organs and tissues in abdomen. Discussion Biodistribution studies in mice demonstrated that targeting properties of [177Lu]Lu-BQ7876 are not inferior to properties of [177Lu]Lu-PSMA-617, but do not offer any decisive advantages.
Collapse
Affiliation(s)
- Ayman Abouzayed
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Kamila Seitova
- Scientific and Research Laboratory of Chemical and Pharmaceutical Research, Siberian State Medical University, Tomsk, Russia
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk, Russia
| | - Fanny Lundmark
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Vitalina Bodenko
- Scientific and Research Laboratory of Chemical and Pharmaceutical Research, Siberian State Medical University, Tomsk, Russia
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk, Russia
| | - Maryam Oroujeni
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Affibody AB, Solna, Sweden
| | - Vladimir Tolmachev
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, Tomsk, Russia
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Ulrika Rosenström
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
- Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
32
|
Sathekge M, Bruchertseifer F, Vorster M, Lawal IO, Mokoala K, Reed J, Maseremule L, Ndlovu H, Hlongwa K, Maes A, Morgenstern A, Van de Wiele C. 225Ac-PSMA-617 radioligand therapy of de novo metastatic hormone-sensitive prostate carcinoma (mHSPC): preliminary clinical findings. Eur J Nucl Med Mol Imaging 2023; 50:2210-2218. [PMID: 36864360 PMCID: PMC10199874 DOI: 10.1007/s00259-023-06165-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 02/19/2023] [Indexed: 03/04/2023]
Abstract
PURPOSE 225Ac-PSMA-617 has demonstrated good anti-tumor effect as a treatment option for metastatic castration-resistant prostate cancer (mCRPC) patients. No study has previously assessed treatment outcome and survival following 225Ac-PSMA-617 treatment of de novo metastatic hormone-sensitive prostate carcinoma (mHSPC) patients. Based on the potential side effects that are known and explained to the patients by the oncologist, some of the patients refused the standard treatment and are seeking alternative therapies. Thus, we report our preliminary findings in a retrospective series of 21 mHSPC patients that refused standard treatment options and were treated with 225Ac-PSMA-617. METHODS We retrospectively reviewed patients with histologically confirmed de novo treatment-naïve bone ± visceral mHSPC that were treated with 225Ac-PSMA-617 radioligand therapy (RLT). Inclusion criteria included an Eastern Cooperative Oncology Group (ECOG) performance status of 0 to 2, treatment-naive bone ± visceral mHSPC, and patients refusal for ADT ± docetaxel, abiraterone acetate, or enzalutamide. We evaluated the response to treatment using prostate-specific antigen (PSA) response and the progression-free survival (PFS) and overall survival (OS) as well as the toxicities. RESULTS Twenty-one mHSPC patients were included in this preliminary work. Following treatment, twenty patients (95%) had any decline in PSA and eighteen patients (86%) presented with a PSA decline of ≥ 50% including 4 patients in whom PSA became undetectable. A lower percentage decrease in PSA following treatment was associated with increased mortality and shorter progression-free survival. Overall, administration of 225Ac-PSMA-617 was well tolerated. The commonest toxicity seen was grade I/II dry mouth observed in 94% of patients. CONCLUSIONS Given these favorable results, randomized prospective multicenter trials assessing the clinical value of 225Ac-PSMA-617 as a therapeutic agent for mHSPC administered either as monotherapy or administered concomitant with ADT are of interest.
Collapse
Affiliation(s)
- Mike Sathekge
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria, 0001, South Africa.
- Nuclear Medicine Research Infrastructure (NuMeRI), Pretoria, South Africa.
| | | | - Mariza Vorster
- Department of Nuclear Medicine, University of Kwa-Zulu Natal & Inkosi Albert Luthuli Central Academic Hospital, Durban, South Africa
| | - Ismaheel O Lawal
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria, 0001, South Africa
- Nuclear Medicine Research Infrastructure (NuMeRI), Pretoria, South Africa
| | - Kgomotso Mokoala
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria, 0001, South Africa
- Nuclear Medicine Research Infrastructure (NuMeRI), Pretoria, South Africa
| | - Janet Reed
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria, 0001, South Africa
- Nuclear Medicine Research Infrastructure (NuMeRI), Pretoria, South Africa
| | - Letjie Maseremule
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria, 0001, South Africa
- Nuclear Medicine Research Infrastructure (NuMeRI), Pretoria, South Africa
| | - Honest Ndlovu
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria, 0001, South Africa
- Nuclear Medicine Research Infrastructure (NuMeRI), Pretoria, South Africa
| | - Khanyi Hlongwa
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria, 0001, South Africa
- Nuclear Medicine Research Infrastructure (NuMeRI), Pretoria, South Africa
| | - Alex Maes
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria, 0001, South Africa
- Katholieke University Leuven, Kortrijk, Belgium
| | - Alfred Morgenstern
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria, 0001, South Africa
- European Commission, Joint Research Centre, Karlsruhe, Germany
| | - Christophe Van de Wiele
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Pretoria, 0001, South Africa
- Ghent University, Ghent, Belgium
| |
Collapse
|
33
|
Murce E, Beekman S, Spaan E, Handula M, Stuurman D, de Ridder C, Seimbille Y. Preclinical Evaluation of a PSMA-Targeting Homodimer with an Optimized Linker for Imaging of Prostate Cancer. Molecules 2023; 28:molecules28104022. [PMID: 37241763 DOI: 10.3390/molecules28104022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/08/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Prostate-specific membrane antigen (PSMA) targeting radiopharmaceuticals have been successfully used for diagnosis and therapy of prostate cancer. Optimization of the available agents is desirable to improve tumor uptake and reduce side effects to non-target organs. This can be achieved, for instance, via linker modifications or multimerization approaches. In this study, we evaluated a small library of PSMA-targeting derivatives with modified linker residues, and selected the best candidate based on its binding affinity to PSMA. The lead compound was coupled to a chelator for radiolabeling, and subject to dimerization. The resulting molecules, 22 and 30, were highly PSMA specific (IC50 = 1.0-1.6 nM) and stable when radiolabeled with indium-111 (>90% stable in PBS and mouse serum up to 24 h). Moreover, [111In]In-30 presented a high uptake in PSMA expressing LS174T cells, with 92.6% internalization compared to 34.1% for PSMA-617. Biodistribution studies in LS174T mice xenograft models showed that [111In]In-30 had a higher tumor and kidney uptake compared to [111In]In-PSMA-617, but increasing T/K and T/M ratios at 24 h p.i. Tumors could be clearly visualized at 1 h p.i. by SPECT/CT after administration of [111In]In-22 and [111In]In-PSMA-617, while [111In]In-30 showed a clear signal at later time-points (e.g., 24 h p.i.).
Collapse
Affiliation(s)
- Erika Murce
- Department of Radiology and Nuclear Medicine, University Medical Center Rotterdam, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Savanne Beekman
- Department of Radiology and Nuclear Medicine, University Medical Center Rotterdam, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Evelien Spaan
- Department of Radiology and Nuclear Medicine, University Medical Center Rotterdam, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Maryana Handula
- Department of Radiology and Nuclear Medicine, University Medical Center Rotterdam, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Debra Stuurman
- Department of Radiology and Nuclear Medicine, University Medical Center Rotterdam, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Corrina de Ridder
- Department of Radiology and Nuclear Medicine, University Medical Center Rotterdam, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
| | - Yann Seimbille
- Department of Radiology and Nuclear Medicine, University Medical Center Rotterdam, Erasmus MC, 3015 GD Rotterdam, The Netherlands
- Erasmus MC Cancer Institute, 3015 GD Rotterdam, The Netherlands
- Life Sciences Division, TRIUMF, Vancouver, BC V6T 2A3, Canada
| |
Collapse
|
34
|
Bakht MK, Yamada Y, Ku SY, Venkadakrishnan VB, Korsen JA, Kalidindi TM, Mizuno K, Ahn SH, Seo JH, Garcia MM, Khani F, Elemento O, Long HW, Chaglassian A, Pillarsetty N, Lewis JS, Freedman M, Belanger AP, Nguyen QD, Beltran H. Landscape of prostate-specific membrane antigen heterogeneity and regulation in AR-positive and AR-negative metastatic prostate cancer. NATURE CANCER 2023; 4:699-715. [PMID: 37038004 PMCID: PMC10867901 DOI: 10.1038/s43018-023-00539-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 03/06/2023] [Indexed: 04/12/2023]
Abstract
Tumor expression of prostate-specific membrane antigen (PSMA) is lost in 15-20% of men with castration-resistant prostate cancer (CRPC), yet the underlying mechanisms remain poorly defined. In androgen receptor (AR)-positive CRPC, we observed lower PSMA expression in liver lesions versus other sites, suggesting a role of the microenvironment in modulating PSMA. PSMA suppression was associated with promoter histone 3 lysine 27 methylation and higher levels of neutral amino acid transporters, correlating with 18F-fluciclovine uptake on positron emission tomography imaging. While PSMA is regulated by AR, we identified a subset of AR-negative CRPC with high PSMA. HOXB13 and AR co-occupancy at the PSMA enhancer and knockout models point to HOXB13 as an upstream regulator of PSMA in AR-positive and AR-negative prostate cancer. These data demonstrate how PSMA expression is differentially regulated across metastatic lesions and in the context of the AR, which may inform selection for PSMA-targeted therapies and development of complementary biomarkers.
Collapse
Affiliation(s)
- Martin K Bakht
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Yasutaka Yamada
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Sheng-Yu Ku
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | - Joshua A Korsen
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Teja M Kalidindi
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kei Mizuno
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Shin Hye Ahn
- Harvard Medical School, Boston, MA, USA
- Molecular Cancer Imaging Facility, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ji-Heui Seo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Maria Mica Garcia
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Francesca Khani
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical Center, New York Presbyterian Hospital, New York, NY, USA
| | - Olivier Elemento
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Henry W Long
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Matthew Freedman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Anthony P Belanger
- Harvard Medical School, Boston, MA, USA
- Molecular Cancer Imaging Facility, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Quang-De Nguyen
- Harvard Medical School, Boston, MA, USA
- Lurie Family Imaging Center, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
35
|
Seifert R, Emmett L, Rowe SP, Herrmann K, Hadaschik B, Calais J, Giesel FL, Reiter R, Maurer T, Heck M, Gafita A, Morris MJ, Fanti S, Weber WA, Hope TA, Hofman MS, Fendler WP, Eiber M. Second Version of the Prostate Cancer Molecular Imaging Standardized Evaluation Framework Including Response Evaluation for Clinical Trials (PROMISE V2). Eur Urol 2023; 83:405-412. [PMID: 36935345 DOI: 10.1016/j.eururo.2023.02.002] [Citation(s) in RCA: 116] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/18/2022] [Accepted: 02/01/2023] [Indexed: 03/19/2023]
Abstract
CONTEXT Prostate-specific membrane antigen (PSMA) targeting positron emission tomography (PET) is emerging to become a reference imaging tool for the staging and restaging of patients with prostate cancer for both clinical routine and trials. The prostate cancer molecular imaging standardized evaluation (PROMISE) criteria have been proposed as a framework for whole-body staging (molecular imaging TNM staging, denoted miTNM staging) to describe the prostate cancer disease extent on PSMA-PET. OBJECTIVE To create a comprehensive and integrated framework for PSMA-PET image interpretation and reporting. EVIDENCE ACQUISITION We propose the PROMISE V2 framework, which integrates an updated miTNM system, improved assessment of local disease, and a slightly modified PSMA-expression score for clinical routine. We have added a response monitoring framework defining qualitative and quantitative parameters to be recorded for a longitudinal assessment in clinical trials. EVIDENCE SYNTHESIS We provide a comprehensive literature review on the current use of the PROMISE framework in clinical research and prospective trials. PROMISE variables demonstrate a clear association with survival. PSMA expression assessed by the PSMA-expression score was used in several trials, and a low PSMA-expression score is a negative prognosticator of overall survival after 177Lu-PSMA radioligand therapy. The proposed imaging parameters recorded for response assessment in clinical trials can be utilized to determine response according to PSMA-PET progression (PPP) or Response Evaluation Criteria in PSMA-PET/Computed Tomography (RECIP) frameworks, but also future response criteria. CONCLUSIONS PROMISE V2 offers standardized reporting of disease extent for clinical routine and research. Parameters recorded within clinical trials facilitate objective response assessment. PATIENT SUMMARY Prostate-specific membrane antigen (PSMA) targeting positron emission tomography (PET) has become a standard imaging examination for prostate cancer. We propose a comprehensive framework for the analysis and reporting of PSMA-PET findings that will improve the communication between imaging experts and uro-oncologists.
Collapse
Affiliation(s)
- Robert Seifert
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany.
| | - Louise Emmett
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, NSW, Australia
| | - Steven P Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins, University School of Medicine, Baltimore, MD, USA; The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany; Ahmanson Translational Theranostics, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, University of California Los Angeles, CA, USA
| | - Boris Hadaschik
- Department of Urology, University Hospital Essen, Essen, Germany
| | - Jeremie Calais
- Ahmanson Translational Theranostics, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, University of California Los Angeles, CA, USA
| | - Frederik L Giesel
- Department of Nuclear Medicine, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Robert Reiter
- Department of Urology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Tobias Maurer
- Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Matthias Heck
- Department of Urology, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany; Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| | - Andrei Gafita
- Ahmanson Translational Theranostics, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, University of California Los Angeles, CA, USA
| | - Michael J Morris
- Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Wolfgang A Weber
- Department of Nuclear Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Thomas A Hope
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Michael S Hofman
- Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Wolfgang Peter Fendler
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany; PET Committee of the German Society of Nuclear Medicine, Göttingen, Germany
| | - Matthias Eiber
- Bavarian Cancer Research Center (BZKF), Erlangen, Germany; Department of Nuclear Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| |
Collapse
|
36
|
Telli T, Tuncel M, Karabulut E, Aksoy S, Erman M, Akdogan B, Caglar M. Prognostic factors of overall and prostate-specific antigen-progression-free survival in metastatic castration-resistant prostate cancer patients treated with 177 Lu-PSMA-617. A single-center prospective observational study. Prostate 2023; 83:792-800. [PMID: 36919876 DOI: 10.1002/pros.24518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 02/22/2023] [Indexed: 03/16/2023]
Abstract
BACKGROUND Metastatic castration-resistant prostate cancer (mCRPC) is characterized by heterogeneity among patients as well as therapy responses due to diverse genetic, epigenetic differences, and resistance mechanisms. At this stage of the disease, therapy modalities should be individualized in light of the patients' clinical state, symptoms, and genetic characteristics. In this prospective study, we aimed to evaluate the outcome of patients with mCRPC treated with 177 Lutetium labeled PSMA-617 therapy (PSMA-RLT), as well as baseline and therapy-related parameters associated with survival. METHODS This prospective study included 52 patients who received two to six cycles of PSMA-RLT. Primary endpoints were overall survival (OS) and prostate-specific antigen (PSA)-progression-free survival (PFS). 18 F-Fluorodeoxyglucose (FDG) and 68 Ga-PSMA (PSMA) Positron Emission Tomography/Computer Tomography (PET/CT) scans were performed for a comprehensive assessment of tumor burden and heterogeneity. Biochemical, imaging, clinical, and therapy-related parameters were analyzed with the Kaplan-Meier, log-rank, and Cox regression analyses to predict OS and PFS. RESULTS Median OS and PSA-PFS were 17.7 (95% confidence interval [CI]: 15.2-20.2) and 6.6 months (95% CI: 4.5-8.8), respectively. Primary resistance to PSMA-RLT (hazard ratio [HR]: 12.57, 95% CI: 2.4-65.2, p: 0.003), <30% PSA response rate after first cycle of PSMA-RLT (HR: 1.016, 95% CI: 1.006-1.03, p: 0.003), FDG > PSMA disease (HR: 4.9, 95% CI: 1.19-20.62, p: 0.03), PSA doubling time (PSA DT) of ≤2.4 months (HR: 15.7, 95% CI: 3.7-66.4, p: <0.0001), and low hemoglobin levels (HR: 0.59, 95% CI: 0.41-0.83, p: 0.003) were correlated with poor OS in the multivariate analysis. Bone scintigraphy > PSMA disease (HR: 5.6; 95% CI: 1.8-17, p: 0.002) and high C-reactive protein (HR: 1.4, 95% CI: 1.1-1.7, p: 0.001) were significant predictive biomarkers for PFS in the multivariate analysis. CONCLUSION PSA response rate and pattern to PSMA-RLT are the most important predictors of survival in patients receiving PSMA-RLT. Being a strong predictive biomarker, combined FDG and PSMA PET can be helpful for the decision of PSMA-RLT eligibility.
Collapse
Affiliation(s)
- Tugce Telli
- Department of Nuclear Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Murat Tuncel
- Department of Nuclear Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Erdem Karabulut
- Department of Biostatistics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Sercan Aksoy
- Department of Medical Oncology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Mustafa Erman
- Department of Medical Oncology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Bulent Akdogan
- Department of Urology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Meltem Caglar
- Department of Nuclear Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
37
|
Jadvar H, Colletti PM. Clinical Trials of Prostate-Specific Membrane Antigen Radiopharmaceutical Therapy. J Nucl Med Technol 2023; 51:16-21. [PMID: 36599704 DOI: 10.2967/jnmt.122.264928] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 12/06/2022] [Accepted: 12/06/2022] [Indexed: 01/06/2023] Open
Abstract
Prostate-specific membrane antigen (PSMA) theranostics has been a momentous triumph for nuclear medicine. The recent approvals of PSMA-targeted imaging agents (68Ga-PSMA-11, 18F-DCFPyL) and radiopharmaceutical therapy (177Lu-PSMA-617) have paved the way for theranostics as a viable care strategy for men with metastatic castration-resistant prostate cancer. The imaging clinical trials OSPREY, CONDOR, and those conducted at the University of California (Los Angeles and San Francisco), as well as the randomized phase 3 therapy trial VISION, have been the fruitful beginnings for PSMA theranostics. There are currently several ongoing clinical trials to expand the reach of PSMA theranostics to the earlier phases of prostate cancer and to optimize its utility in combination therapeutic regimens. We provide a brief narrative review of the many PSMA-directed radiopharmaceutical therapy clinical trials with the β-emitter 177Lu-PSMA-617 and the α-emitter 225Ac-PSMA-617 in prostate cancer.
Collapse
Affiliation(s)
- Hossein Jadvar
- Division of Nuclear Medicine, Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Patrick M Colletti
- Division of Nuclear Medicine, Department of Radiology, Keck School of Medicine, University of Southern California, Los Angeles, California
| |
Collapse
|
38
|
Collins K, Cheng L. Reprint of: morphologic spectrum of treatment-related changes in prostate tissue and prostate cancer: an updated review. Hum Pathol 2023; 133:92-101. [PMID: 36898948 DOI: 10.1016/j.humpath.2023.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/05/2022] [Indexed: 03/11/2023]
Abstract
A wide range of treatment options are available to patients with prostate cancer. Some treatments are standard (currently used) while some are emerging therapies. Androgen deprivation therapy is typically reserved for localized or metastatic prostate cancer not amenable to surgery. Radiation therapy may be offered to individuals for local therapy with curative intent in low- or intermediate-risk disease that may have a high probability of progression on active surveillance or where surgery is not suitable. Focal therapy/ablation treatment is an alternative approach for those who prefer to avoid radical prostatectomy for localized disease of low- or intermediate-risk or as salvage therapy after failed radiation therapy. Chemotherapy and immunotherapy remain under investigation and are currently used for androgen-independent disease or hormone-refractory prostate cancer; however, a better understanding of therapeutic efficacy is needed. Histopathologic changes observed in benign and malignant prostate tissue induced by hormonal therapies and radiation therapy are well described, whereas treatment-related effects secondary to novel therapies continue to be documented although their clinical significance is not absolutely clear. An informed and accurate evaluation of post-treatment prostate specimens requires pathologists with diagnostic acumen and knowledge relating to the histopathologic spectrum associated with each treatment option. In situations when clinical history is lacking, but morphologic features are suggestive of prior treatment, pathologists are encouraged to consult clinical colleagues regarding prior treatment history including details of when treatment was initiated and duration of therapy. This review aims to provide a concise update of current and emerging therapies for prostate cancer, histologic alterations and recommendations on Gleason grading.
Collapse
Affiliation(s)
- Katrina Collins
- Department of Pathology, Indiana University, Indianapolis, IN 46202, USA.
| | - Liang Cheng
- Department of Pathology, Indiana University, Indianapolis, IN 46202, USA
| |
Collapse
|
39
|
Gravestock P, Somani BK, Tokas T, Rai BP. A Review of Modern Imaging Landscape for Prostate Cancer: A Comprehensive Clinical Guide. J Clin Med 2023; 12:jcm12031186. [PMID: 36769834 PMCID: PMC9918161 DOI: 10.3390/jcm12031186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 01/29/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
The development of prostate cancer imaging is rapidly evolving, with many changes to the way patients are diagnosed, staged, and monitored for recurrence following treatment. New developments, including the potential role of imaging in screening and the combined diagnostic and therapeutic applications in the field of theranostics, are underway. In this paper, we aim to outline the current landscape in prostate cancer imaging and look to the future at the potential modalities and applications to come.
Collapse
Affiliation(s)
- Paul Gravestock
- Department of Urology, Freeman Hospital, Newcastle upon Tyne NE7 7DN, UK
| | - Bhaskar Kumar Somani
- Department of Urology, University Hospital Southampton NHS Trust, Southampton SO16 6YD, UK
| | - Theodoros Tokas
- Department of Urology and Andrology, General Hospital Hall in Tirol, 6060 Hall in Tirol, Austria
- Training and Research in Urological Surgery and Technology (T.R.U.S.T.)-Group, 6060 Hall in Tirol, Austria
| | - Bhavan Prasad Rai
- Department of Urology, Freeman Hospital, Newcastle upon Tyne NE7 7DN, UK
- Correspondence:
| |
Collapse
|
40
|
Parihar AS, Hofman MS, Iravani A. 177Lu-Prostate-specific Membrane Antigen Radioligand Therapy in Patients with Metastatic Castration-resistant Prostate Cancer. Radiology 2023; 306:e220859. [PMID: 36125377 DOI: 10.1148/radiol.220859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
A 76-year-old man with metastatic castration-resistant prostate carcinoma progressing with antiandrogen and taxane therapy was treated with lutetium 177 prostate-specific membrane antigen (PSMA)-617 and showed marked biochemical and imaging response, with improvement in clinical status and osseous pain. A summary of nuclear medicine theranostics with emphasis on PSMA targeting agents is presented.
Collapse
Affiliation(s)
- Ashwin Singh Parihar
- From the Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4525 Scott Ave, Suite 3433, MIR East Building, St Louis, MO 63110 (A.S.P., A.I.); and Department of Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia (M.S.H.)
| | - Michael S Hofman
- From the Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4525 Scott Ave, Suite 3433, MIR East Building, St Louis, MO 63110 (A.S.P., A.I.); and Department of Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia (M.S.H.)
| | - Amir Iravani
- From the Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4525 Scott Ave, Suite 3433, MIR East Building, St Louis, MO 63110 (A.S.P., A.I.); and Department of Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia (M.S.H.)
| |
Collapse
|
41
|
Grubmüller B, Huebner NA, Rasul S, Clauser P, Pötsch N, Grubmüller KH, Hacker M, Hartenbach S, Shariat SF, Hartenbach M, Baltzer P. Dual-Tracer PET-MRI-Derived Imaging Biomarkers for Prediction of Clinically Significant Prostate Cancer. Curr Oncol 2023; 30:1683-1691. [PMID: 36826090 PMCID: PMC9954891 DOI: 10.3390/curroncol30020129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/11/2023] [Accepted: 01/24/2023] [Indexed: 02/03/2023] Open
Abstract
PURPOSE To investigate if imaging biomarkers derived from 3-Tesla dual-tracer [(18)F]fluoromethylcholine (FMC) and [68Ga]Ga-PSMAHBED-CC conjugate 11 (PSMA)-positron emission tomography can adequately predict clinically significant prostate cancer (csPC). METHODS We assessed 77 biopsy-proven PC patients who underwent 3T dual-tracer PET/mpMRI followed by radical prostatectomy (RP) between 2014 and 2017. We performed a retrospective lesion-based analysis of all cancer foci and compared it to whole-mount histopathology of the RP specimen. The primary aim was to investigate the pretherapeutic role of the imaging biomarkers FMC- and PSMA-maximum standardized uptake values (SUVmax) for the prediction of csPC and to compare it to the mpMRI-methods and PI-RADS score. RESULTS Overall, we identified 104 cancer foci, 69 were clinically significant (66.3%) and 35 were clinically insignificant (33.7%). We found that the combined FMC+PSMA SUVmax were the only significant parameters (p < 0.001 and p = 0.049) for the prediction of csPC. ROC analysis showed an AUC for the prediction of csPC of 0.695 for PI-RADS scoring (95% CI 0.591 to 0.786), 0.792 for FMC SUVmax (95% CI 0.696 to 0.869), 0.852 for FMC+PSMA SUVmax (95% CI 0.764 to 0.917), and 0.852 for the multivariable CHAID model (95% CI 0.763 to 0.916). Comparing the AUCs, we found that FMC+PSMA SUVmax and the multivariable model were significantly more accurate for the prediction of csPC compared to PI-RADS scoring (p = 0.0123, p = 0.0253, respectively). CONCLUSIONS Combined FMC+PSMA SUVmax seems to be a reliable parameter for the prediction of csPC and might overcome the limitations of PI-RADS scoring. Further prospective studies are necessary to confirm these promising preliminary results.
Collapse
Affiliation(s)
- Bernhard Grubmüller
- Department of Urology, Medical University of Vienna, 1090 Vienna, Austria
- Department of Urology and Andrology, University Hospital Krems, 3500 Krems, Austria
- Karl Landsteiner University of Health Sciences, 3500 Krems, Austria
- Working Group of Diagnostic Imaging in Urology, Austrian Society of Urology, 1090 Vienna, Austria
| | - Nicolai A. Huebner
- Department of Urology, Medical University of Vienna, 1090 Vienna, Austria
- Working Group of Diagnostic Imaging in Urology, Austrian Society of Urology, 1090 Vienna, Austria
| | - Sazan Rasul
- Department of Biomedical Imaging and Image Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Paola Clauser
- Department of Biomedical Imaging and Image Guided Therapy, Division of General and Pediatric Radiology, Medical University of Vienna, 1090 Vienna, Austria
| | - Nina Pötsch
- Department of Biomedical Imaging and Image Guided Therapy, Division of General and Pediatric Radiology, Medical University of Vienna, 1090 Vienna, Austria
| | - Karl Hermann Grubmüller
- Department of Urology and Andrology, University Hospital Krems, 3500 Krems, Austria
- Karl Landsteiner University of Health Sciences, 3500 Krems, Austria
| | - Marcus Hacker
- Department of Biomedical Imaging and Image Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | | | - Shahrokh F. Shariat
- Department of Urology, Medical University of Vienna, 1090 Vienna, Austria
- Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
- Department of Urology, Weill Medical College of Cornell University, New York, NY 10021, USA
- Department of Urology, University of Texas Southwestern, Dallas, TX 75390, USA
- Department of Urology, Second Faculty of Medicine, Charles University, 116 36 Prague, Czech Republic
- Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman 19328, Jordan
- Karl Landsteiner Institute of Urology and Andrology, 1010 Vienna, Austria
| | - Markus Hartenbach
- Department of Biomedical Imaging and Image Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Pascal Baltzer
- Department of Biomedical Imaging and Image Guided Therapy, Division of General and Pediatric Radiology, Medical University of Vienna, 1090 Vienna, Austria
- Correspondence:
| |
Collapse
|
42
|
Roberts MJ, Maurer T, Perera M, Eiber M, Hope TA, Ost P, Siva S, Hofman MS, Murphy DG, Emmett L, Fendler WP. Using PSMA imaging for prognostication in localized and advanced prostate cancer. Nat Rev Urol 2023; 20:23-47. [PMID: 36473945 DOI: 10.1038/s41585-022-00670-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2022] [Indexed: 12/12/2022]
Abstract
The use of prostate-specific membrane antigen (PSMA)-directed applications in modern prostate cancer management has evolved rapidly over the past few years, helping to establish new treatment pathways and provide further insights into prostate cancer biology. However, the prognostic implications of PSMA-PET have not been studied systematically, owing to rapid clinical implementation without long follow-up periods to determine intermediate-term and long-term oncological outcomes. Currently available data suggest that traditional prognostic factors and survival outcomes are associated with high PSMA expression (both according to immunohistochemistry and PET uptake) in men with localized and biochemically recurrent disease. Treatment with curative intent (primary and/or salvage) often fails when PSMA-positive metastases are present; however, the sensitivity of PSMA-PET in detecting all metastases is poor. Low PSMA-PET uptake in recurrent disease is a favourable prognostic factor; however, it can be associated with poor prognosis in conjunction with high 18F-fluorodeoxyglucose uptake in metastatic castration-resistant prostate cancer. Clinical trials embedding PSMA-PET for guiding management with reliable oncological outcomes are needed to support ongoing clinical use.
Collapse
Affiliation(s)
- Matthew J Roberts
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia.
- University of Queensland Centre for Clinical Research, Faculty of Medicine, Brisbane, Queensland, Australia.
- Department of Urology, Redcliffe Hospital, Brisbane, Queensland, Australia.
| | - Tobias Maurer
- Martini-Klinik Prostate Cancer Center, Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Marlon Perera
- Department of Surgery, Austin Health, Heidelberg, Victoria, Australia
| | - Matthias Eiber
- Department of Nuclear Medicine, Technical University of Munich, Munich, Germany
| | - Thomas A Hope
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Piet Ost
- Department of Radiation Oncology, Iridium Network, GZA Ziekenhuizen, Antwerp, Belgium
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Shankar Siva
- Peter MacCallum Cancer Centre, Radiation Oncology, Parkville, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, Melbourne University, Parkville, Victoria, Australia
| | - Michael S Hofman
- Sir Peter MacCallum Department of Oncology, Melbourne University, Parkville, Victoria, Australia
- Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Declan G Murphy
- Sir Peter MacCallum Department of Oncology, Melbourne University, Parkville, Victoria, Australia
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Louise Emmett
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, New South Wales, Australia
- Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University of Duisburg-Essen, Essen, Germany
- PET Committee of the German Society of Nuclear Medicine, Goettingen, Germany
| |
Collapse
|
43
|
Chandran E, Figg WD, Madan R. Lutetium-177-PSMA-617: A Vision of the Future. Cancer Biol Ther 2022; 23:186-190. [PMID: 35220877 PMCID: PMC8890398 DOI: 10.1080/15384047.2022.2037985] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 01/24/2022] [Accepted: 01/27/2022] [Indexed: 12/16/2022] Open
Abstract
In the last decade, many life-prolonging therapeutic options have emerged for metastatic castration-resistant prostate cancer (mCRPC). The recent VISION trial is the first to demonstrate a survival benefit of Lutetium-177[177Lu]Lu-PSMA-617 in post-chemotherapy mCRPC. This journal club reviews the VISION trial in the context of the earlier TheraP trial of [177Lu]Lu-PSMA-617 in mCRPC post docetaxel and androgen pathway inhibition, to provide direction for the real-world application of [177Lu]Lu-PSMA-617. Treatment in the control groups differed significantly between both trials and may have influenced outcomes: TheraP mandated cabazitaxel whereas VISION's design could not allow it. In both trials, [177Lu]Lu-PSMA-617 had a good safety profile, with common adverse events being fatigue, nausea, dry mouth, marrow suppression and diarrhea. Given its efficacy and favorable safety even in heavily pre-treated patients, [177Lu]Lu-PSMA-617 provides hope to mCRPC patients and may be applied to earlier disease stages in future investigations.
Collapse
Affiliation(s)
- Elias Chandran
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - William D. Figg
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ravi Madan
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
44
|
AlSadi R, Bouhali O, Dewji S, Djekidel M. 177Lu-PSMA Therapy for Metastatic Castration-Resistant Prostate Cancer: A Mini-Review of State-of-the-Art. Oncologist 2022; 27:e957-e966. [PMID: 36288537 DOI: 10.1093/oncolo/oyac216] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 09/16/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Prostate specific membrane antigen (PSMA) ligand labeled with Lutetium-177 (177Lu) is a promising therapeutic option for metastatic castration-resistant prostate cancer (mCRPC). Several prospective and retrospective studies as well as clinical trials are completed or underway. This has ultimately led to the approval of this therapy by the US Food and Drug Administration (FDA) on March 23 2022. Our work aims to present a mini-review of the most recent research performed and the potential future directions of 177Lu-PSMA-radioligand therapy (RLT) for mCRPC patients. MAIN BODY For patients with mCRPCwho have met the eligibility criteria for 177Lu-PSMA RLT, numerous studies and trials are either ongoing or have been completed. The studies included in this review have reported overall biochemical response, defined as a prostate-specific antigen (PSA) decline of at least 50%, in at least 44% of patients with mCRPC. The median ranges of overall survival (OS) and radiographic progression-free survival (rPFS) were reported within 10.7-56 and 3.6-16 months, respectively. With data from several retrospective and prospective studies published, the safety of 177Lu-PSMA RLT in mCRPC has been confirmed and demonstrated by its low toxicity profile. Various studies have published pharmacokinetic/pharmacodynamic models to better understand the absorption, distribution, metabolism, and excretion of the RLT in this patient population. Findings have been published for 177Lu-PSMA RLT alone and in combination with other agents. We summarize their findings in our review. CONCLUSIONS The efficacy of 177Lu-PSMA RLT for patients with mCRPC has been proven thus far with promising results: PSA response, OS and rPFS when used alone or in combination with other treatment options, relative to the standard treatment options alone. The low toxicity profile noted also proves the safety of 177Lu-PSMA RLT in these patients.
Collapse
Affiliation(s)
- Rahaf AlSadi
- Science Department, Texas A&M University at Qatar, Doha, Qatar
| | - Othmane Bouhali
- Science Department, Texas A&M University at Qatar, Doha, Qatar.,Qatar Computing Research Institute, Hamad Bin Khalifa University, Doha, Qatar
| | - Shaheen Dewji
- Department of Nuclear & Radiological Engineering and Medical Physics, Georgia Institute of Technology, North Avenue Atlanta, GA, USA
| | | |
Collapse
|
45
|
Pan J, Zhao J, Ni X, Gan H, Wei Y, Wu J, Zhang T, Wang Q, Freedland SJ, Wang B, Song S, Ye D, Liu C, Zhu Y. The prevalence and prognosis of next-generation therapeutic targets in metastatic castration-resistant prostate cancer. Mol Oncol 2022; 16:4011-4022. [PMID: 36209367 PMCID: PMC9718110 DOI: 10.1002/1878-0261.13320] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 09/18/2022] [Accepted: 10/07/2022] [Indexed: 12/24/2022] Open
Abstract
The success of the PROfound, IPATential150, and TheraP trials promoted the transition from sequential treatment to therapeutic targets (TTs)-guided precision treatment in metastatic castration-resistant prostate cancer (mCRPC). The objective of this study was to evaluate the prevalence and prognostic value of TTs from these three trials. All included Chinese mCRPC patients underwent circulating tumor DNA (ctDNA) sequencing, PTEN status assessment, and dual-tracer [68 Ga-prostate-specific membrane antigen (PSMA) and 18 F-fluorodexyglucose (FDG)] positron emission tomography/computed tomography (PET/CT). Previous treatment with cabazitaxel, Lu-PSMA or olaparib was unallowed. Patients with known significant sarcomatoid or spindle cell or neuroendocrine small cell components were also excluded. TTs were defined as positive as follows: (a) high PSMA and no PSMA-/FDG+ disease on dual-tracer PET/CT scans; (b) defects in homologous recombination repair (HRR) genes in ctDNA; and (c) loss of PTEN immunohistochemistry staining in tumor tissue. The prevalence and prognostic value on progression-free survival (PFS) of TTs were evaluated. A total of 106 consecutive mCRPC patients were included. The prevalence of positive PET/CT, HRR defect, and PTEN loss was 30%, 29% and 16%, respectively. Sixty-three patients had at least one TT. Metastatic volume (odds ratio = 5.0; P = 0.017) was the only independent factor of positive TT in multivariate analysis. Seventy-four patients received abiraterone after TT screening. Patients with positive PET/CT (P = 0.011) and HRR defect (P = 0.002) had a significantly shorter PFS after receiving abiraterone than patients with negative TTs. However, PTEN status was unrelated to PFS, which may be due to a less number of patients with PTEN loss (P = 0.952). Overall, patients with any positive TTs had a significantly shorter PFS after abiraterone than patients with negative TTs (P = 0.009). Nearly 60% of Chinese patients with mCRPC who had a poor prognosis on abiraterone were candidates for precision treatments based on the specific criteria of TTs.
Collapse
Affiliation(s)
- Jian Pan
- Department of UrologyFudan University Shanghai Cancer CenterChina,Shanghai Genitourinary Cancer InstituteChina,Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Jinou Zhao
- Department of UrologyFudan University Shanghai Cancer CenterChina,Shanghai Genitourinary Cancer InstituteChina,Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Xudong Ni
- Department of UrologyFudan University Shanghai Cancer CenterChina,Shanghai Genitourinary Cancer InstituteChina,Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Hualei Gan
- Shanghai Genitourinary Cancer InstituteChina,Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Yu Wei
- Department of UrologyFudan University Shanghai Cancer CenterChina,Shanghai Genitourinary Cancer InstituteChina,Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Junlong Wu
- Department of UrologyFudan University Shanghai Cancer CenterChina,Shanghai Genitourinary Cancer InstituteChina,Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Tingwei Zhang
- Department of UrologyFudan University Shanghai Cancer CenterChina,Shanghai Genitourinary Cancer InstituteChina,Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Qifeng Wang
- Shanghai Genitourinary Cancer InstituteChina,Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Stephen J. Freedland
- Department of Nuclear MedicineFudan University Shanghai Cancer CenterChina,Department of Surgery, Division of Urology and Samuel Oschin Comprehensive Cancer InstituteCedars‐Sinai Medical CenterLos AngelesCAUSA,Urology Section, Department of SurgeryVeterans Affairs Medical CenterDurhamNCUSA
| | - Beihe Wang
- Department of UrologyFudan University Shanghai Cancer CenterChina,Shanghai Genitourinary Cancer InstituteChina,Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Shaoli Song
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina,Department of PathologyFudan University Shanghai Cancer CenterChina
| | - Dingwei Ye
- Department of UrologyFudan University Shanghai Cancer CenterChina,Shanghai Genitourinary Cancer InstituteChina,Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Chang Liu
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina,Department of PathologyFudan University Shanghai Cancer CenterChina
| | - Yao Zhu
- Department of UrologyFudan University Shanghai Cancer CenterChina,Shanghai Genitourinary Cancer InstituteChina,Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| |
Collapse
|
46
|
Tumor-Derived Extracellular Vesicles Predict Clinical Outcomes in Oligometastatic Prostate Cancer and Suppress Antitumor Immunity. Int J Radiat Oncol Biol Phys 2022; 114:725-737. [PMID: 35671867 PMCID: PMC9869345 DOI: 10.1016/j.ijrobp.2022.05.037] [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: 03/28/2022] [Revised: 05/17/2022] [Accepted: 05/23/2022] [Indexed: 01/26/2023]
Abstract
PURPOSE SABR has demonstrated clinical benefit in oligometastatic prostate cancer. However, the risk of developing new distant metastatic lesions remains high, and only a minority of patients experience durable progression-free response. Therefore, there is a critical need to identify which patients will benefit from SABR alone versus combination SABR and systemic agents. Herein we provide, to our knowledge, the first proof-of-concept of circulating prostate cancer-specific extracellular vesicles (PCEVs) as a noninvasive predictor of outcomes in oligometastatic castration-resistant prostate cancer (omCRPC) treated with SABR. METHODS AND MATERIALS We analyzed the levels and kinetics of PCEVs in the peripheral blood of 79 patients with omCRPC at baseline and days 1, 7, and 14 after SABR using nanoscale flow cytometry and compared with baseline values from cohorts with localized and widely metastatic prostate cancer. The association of omCRPC PCEV levels with oncological outcomes was determined with Cox regression models. RESULTS Levels of PCEVs were highest in mCRPC followed by omCRPC and were lowest in localized prostate cancer. High PCEV levels at baseline predicted a shorter median time to distant recurrence (3.5 vs 6.6 months; P = .0087). After SABR, PCEV levels peaked on day 7, and median overall survival was significantly longer in patients with elevated PCEV levels (32.7 vs 27.6 months; P = .003). This suggests that pretreatment PCEV levels reflect tumor burden, whereas early changes in PCEV levels after treatment predict response to SABR. In contrast, radiomic features of 11C-choline positron emission tomography and computed tomography before and after SABR were not predictive of clinical outcomes. Interestingly, PCEV levels and peripheral tumor-reactive CD8 T cells (TTR; CD8+ CD11ahigh) were correlated. CONCLUSIONS This original study demonstrates that circulating PCEVs can serve as prognostic and predictive markers to SABR to identify patients with "true" omCRPC. In addition, it provides novel insights into the global crosstalk, mediated by PCEVs, between tumors and immune cells that leads to systemic suppression of immunity against CRPC. This work lays the foundation for future studies to investigate the underpinnings of metastatic progression and provide new therapeutic targets (eg, PCEVs) to improve SABR efficacy and clinical outcomes in treatment-resistant CRPC.
Collapse
|
47
|
Buteau JP, Martin AJ, Emmett L, Iravani A, Sandhu S, Joshua AM, Francis RJ, Zhang AY, Scott AM, Lee ST, Azad AA, McJannett MM, Stockler MR, Williams SG, Davis ID, Hofman MS, Akhurst T, Alipour R, Azad AA, Banks P, Beaulieu A, Buteau JP, Chua W, Davis ID, Dhiantravan N, Emmett L, Ford K, Hofman MS, Francis RJ, Gedye C, Goh JC, Guminski A, Hamid A, Haskali MB, Hicks RJ, Hsiao E, Iravani A, Joshua AM, Kirkwood ID, Kong G, Kwan EM, Langford A, Lawrence N, Lee ST, Lewin J, Lin P, Martin AJ, McDonald W, McJannett MM, Moodie K, Murphy DG, Ng S, Pattison DA, Pokorski I, Ramdave S, Ravi Kumar AS, Redfern AD, Rutherford NK, Saghebi J, Sandhu S, Scott AM, Spain L, Stockler MR, Subramaniam S, Tan TH, Thang SP, Tran B, Wallace R, Weickhardt A, Williams SG, Yip S, Zhang AY. PSMA and FDG-PET as predictive and prognostic biomarkers in patients given [177Lu]Lu-PSMA-617 versus cabazitaxel for metastatic castration-resistant prostate cancer (TheraP): a biomarker analysis from a randomised, open-label, phase 2 trial. Lancet Oncol 2022; 23:1389-1397. [DOI: 10.1016/s1470-2045(22)00605-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 11/05/2022]
|
48
|
A Treatment Paradigm Shift: Targeted Radionuclide Therapies for Metastatic Castrate Resistant Prostate Cancer. Cancers (Basel) 2022; 14:cancers14174276. [PMID: 36077820 PMCID: PMC9454920 DOI: 10.3390/cancers14174276] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Metastatic prostate cancer has traditionally been treated with a combination of hormonal and chemotherapy regimens. With the recent FDA approval of targeted radionuclide therapeutics, there is now a new class of therapy that is routinely available to patients and clinicians. This review explores the most commonly studied therapeutic radiopharmaceuticals and their appropriate use and contraindications. Additionally, we detail how these therapeutic radiopharmaceuticals can fit into the common medical oncology practice and future directions of this field of medicine. Abstract The recent approval of 177Lu PSMA-617 (Pluvicto®) by the United States Food and Drug Administration (FDA) is the culmination of decades of work in advancing the field of targeted radionuclide therapy for metastatic prostate cancer. 177Lu PSMA-617, along with the bone specific radiotherapeutic agent, 223RaCl2 (Xofigo®), are now commonly used in routine clinical care as a tertiary line of therapy for men with metastatic castrate resistant prostate cancer and for osseus metastatic disease respectively. While these radiopharmaceuticals are changing how metastatic prostate cancer is classified and treated, there is relatively little guidance to the practitioner and patient as to how best utilize these therapies, especially in conjunction with other more well-established regimens including hormonal, immunologic, and chemotherapeutic agents. This review article will go into detail about the mechanism and effectiveness of these radiopharmaceuticals and less well-known classes of targeted radionuclide radiopharmaceuticals including alpha emitting prostate specific membrane antigen (PSMA)-, gastrin-releasing peptide receptor (GRPR)-, and somatostatin targeted radionuclide therapeutics. Additionally, a thorough discussion of the clinical approach of these agents is included and required futures studies.
Collapse
|
49
|
Clinical Applications of PSMA PET Examination in Patients with Prostate Cancer. Cancers (Basel) 2022; 14:cancers14153768. [PMID: 35954432 PMCID: PMC9367427 DOI: 10.3390/cancers14153768] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/21/2022] [Accepted: 07/31/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary The prostate specific membrane antigens, abbreviated as PSMAs, are type II membrane proteins that are highly ex-pressed on the surface of malignant prostate tissue in prostate cancer (PCa), particularly in aggressive, andro-gen-deprived, metastatic, and hormone-refractory PCa. Today, radionuclides that bind to these PSMA peptides are widely available for diagnostic and therapeutic purposes to specifically image and target prostate tumor cells at molec-ular level. In this descriptive review, we aimed to emphasize the usefulness of PSMA positron emission tomography (PET) examination in the management of patients with various stages of PCa. In addition, we outlined the main pitfalls and limitations of this scan to avoid misinterpretation of the results and to improve the decision making process in rela-tion to the patient’s further treatment. We concluded that PSMA PET examination in primary PCa patients has an es-sential role in the high-risk group. It is the new imaging standard in patients with in biochemical recurrence PCa and plays an important role in treatment decision. Furthermore, PSMA PET scan is a gold standard for the evaluation of PSMA targeted therapies in patients having progress of the disease. Future prospective studies, particularly on the im-pact of PSMA PET on therapy stratification, may further strengthen the role of PSMA in the treatment of PCa patients. Abstract With the progressive aging of the population in industrially developed countries, as well as advances in diagnostic and biopsy techniques and improvements in patient awareness, the incidence of prostate cancer (PCa) is continuously increasing worldwide. Therefore, PCa is currently considered as the second leading cause of tumor-related death. Early detection of the tumor and its metastasis is essential, as the rate of disease recurrence is high and occurs in 27% to 53% of all patients who underwent curative therapy with radical prostatectomy or local radiotherapy. In this regard, the prostate specific membrane antigens, abbreviated as PSMAs, are type II membrane proteins that are highly expressed on the surface of malignant prostate tissue in PCa, particularly in aggressive, androgen-deprived, metastatic, and hormone-refractory PCa, and they are inversely associated with the androgen level. Up to 95% of adenocarcinomas of the prostate express PSMA receptors on their surface. Today, radionuclides that bind to these PSMA peptides are widely accepted for diagnostic and therapeutic purposes to specifically image and target prostate tumor cells at the molecular level, a process referred to as targeted theranostics. Numerous studies have demonstrated that the integration of these peptides into diagnostic and therapeutic procedures plays a critical role in the primary staging and treatment decisions of especially high-risk PCa, expands therapeutic options for patients with advanced stage of prostate tumor, and prolongs patients’ survival rate. In this review article, we intend to briefly spotlight the latest clinical utilization of the PSMA-targeted radioligand PET imaging modality in patients with different stages of PCa. Furthermore, limitations and pitfalls of this diagnostic technique are presented.
Collapse
|
50
|
Mizuno K, Beltran H. Future directions for precision oncology in prostate cancer. Prostate 2022; 82 Suppl 1:S86-S96. [PMID: 35657153 PMCID: PMC9942493 DOI: 10.1002/pros.24354] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 03/28/2022] [Indexed: 11/06/2022]
Abstract
Clinical genomic testing is becoming routine in prostate cancer, as biomarker-driven therapies such as poly-ADP ribose polymerase (PARP) inhibitors and anti-PD1 immunotherapy are now approved for select men with castration-resistant prostate cancer harboring alterations in DNA repair genes. Challenges for precision medicine in prostate cancer include an overall low prevalence of actionable genomic alterations and a still limited understanding of the impact of tumor heterogeneity and co-occurring alterations on treatment response and outcomes across diverse patient populations. Expanded tissue-based technologies such as whole-genome sequencing, transcriptome analysis, epigenetic analysis, and single-cell RNA sequencing have not yet entered the clinical realm and could potentially improve upon our understanding of how molecular features of tumors, intratumoral heterogeneity, and the tumor microenvironment impact therapy response and resistance. Blood-based technologies including cell-free DNA, circulating tumor cells (CTCs), and extracellular vesicles (EVs) are less invasive molecular profiling resources that could also help capture intraindividual tumor heterogeneity and track dynamic changes that occur in the context of specific therapies. Furthermore, molecular imaging is an important biomarker tool within the framework of prostate cancer precision medicine with a capability to detect heterogeneity across metastases and potential therapeutic targets less invasively. Here, we review recent technological advances that may help promote the future implementation and value of precision oncology testing for patients with advanced prostate cancer.
Collapse
Affiliation(s)
- Kei Mizuno
- Department of Medical Oncology, Dana Farber Cancer Institute
| | - Himisha Beltran
- Department of Medical Oncology, Dana Farber Cancer Institute
| |
Collapse
|