1
|
Voter AF, Amindarolzarbi A, Shen CJ, Wang J, Kang H, Sharma R, Solnes LB, Pomper MG, Bishop JA, Rowe SP, Kiess AP. A pilot study of PSMA-targeted F-18-DCFPyL PET imaging of patients with adenoid cystic carcinoma. Sci Rep 2025; 15:17104. [PMID: 40379702 DOI: 10.1038/s41598-025-01515-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Accepted: 05/06/2025] [Indexed: 05/19/2025] Open
Abstract
Adenoid cystic carcinoma (ACC) is a rare malignancy of the salivary glands with poor long-term outcomes and with a need for improved imaging and therapeutic options. Prostate-specific membrane antigen (PSMA) expression has been observed in ACC and preliminary studies have demonstrated PET imaging using 68Ga-functionalized PSMA agents for positron emission tomography (PET). We aimed to assess the extent of PSMA expression in a collection of archival ACC samples and demonstrate the feasibility of using 18F-DCFPyL for PSMA PET imaging of ACC. PSMA expression levels were assessed in 77 ACC and 11 unaffected parotid gland control samples by immunohistochemistry and quantified by mean H-score. Three patients with metastatic ACC, who had previously undergone local resection, were imaged with18F-DCFPyL PSMA PET in a prospective, pilot trial setting. Demographic, oncologic, and treatment history from the PET patient cohort were acquired at the time of imaging. Maximum standardized uptake values (SUVmax) were obtained from representative presumptive metastatic lesions on the 18F-DCFPyL scans by manual placement of regions-of-interest. PSMA expression was detected in 52% of archival ACC samples, compared to 18% in the unaffected salivary glands. Moderate or high levels of PSMA expression (H-score > 5) were seen in 37% of samples. Radiotracer avid disease was identified on PET imaging of all three patients with tumor SUVmax values of 4.1, 4.0, and 2.0, corresponding to tumor-to-liver ratios of 0.7, 1.0, and 0.4 respectively. We find that PSMA is expressed in a majority of histologic samples from patients with ACC. We also demonstrated the feasibility of 18F-DCFPyL PSMA-targeted PET imaging in the assessment of ACC. Overall, the tumor uptake on 18F-DCFPyL PET was modest compared to lesional uptake seen in prostate cancer. Given the potential role of PSMA-targeted agents in the management of ACC, broadening access to PSMA PET through F-18-labeled PSMA PET agents is important. Clinical trials investigating the use of PSMA-targeted radioligand therapies for ACC are underway.
Collapse
Affiliation(s)
- Andrew F Voter
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alireza Amindarolzarbi
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Colette J Shen
- Department of Radiation Oncology, University of North Carolina, Chapel Hill, NC, USA
| | - Jarey Wang
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hyunseok Kang
- Department of Oncology, University of California San Franscisco, San Fransico, CA, USA
| | | | - Lilja B Solnes
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Martin G Pomper
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Justin A Bishop
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Steven P Rowe
- Department of Radiology, University of North Carolina, Chapel Hill, NC, USA
| | - Ana P Kiess
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
2
|
Hoyek NE, Shi X, Jenkins J, Chen W. Role of PSMA PET/CT in imaging and management of prostate cancer. Curr Opin Oncol 2025; 37:233-239. [PMID: 40065665 DOI: 10.1097/cco.0000000000001131] [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: 04/05/2025]
Abstract
PURPOSE OF REVIEW In the era of precision medicine, the introduction of FDA-approved prostate-specific membrane antigen (PSMA) targeting tracers has revolutionized prostate cancer imaging. These tracers enable functional positron emission tomography (PET) imaging, allowing for precise identification of the location and extent of prostate cancer spread. This review serves as a practical guide for multidisciplinary teams caring for prostate cancer patients, outlining the current approved uses of PET imaging with PSMA tracers and exploring its future applications. RECENT FINDINGS PSMA PET/CT has become a reliable modality for initial staging in patients with intermediate-to-high risk prostate cancer, restaging in cases of biochemical recurrence and further clarifying disease status among patients with conventional imaging based nonmetastatic castrate resistant prostate cancer and metastatic prostate cancer. Additionally, it has promising roles in selecting patients for radioligand therapy, monitoring treatment response, and guiding therapeutic decision-making. SUMMARY PSMA PET/CT is currently a crucial imaging tool used at key stages of prostate cancer management, with ongoing research exploring its potential for additional clinical applications.
Collapse
Affiliation(s)
| | - Xiaolei Shi
- Division of Hematology & Oncology, Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jason Jenkins
- Department of Diagnostic Radiology and Nuclear Medicine
| | - Wengen Chen
- Department of Diagnostic Radiology and Nuclear Medicine
| |
Collapse
|
3
|
Wang Y, Yang Y, Li J, Cheng D, Xu H, Huang J. Dynamic FDG PET/CT imaging: quantitative assessment, advantages and application in the diagnosis of malignant solid tumors. Front Oncol 2025; 15:1539911. [PMID: 40297815 PMCID: PMC12034529 DOI: 10.3389/fonc.2025.1539911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Dynamic imaging has obtained remarkable achievements among a variety of malignant tumors due to the development of multiple simplified scanning protocols and the emergence of whole-body PET/CT scanners, which promote wider application of dynamic PET/CT. In this paper, we mainly review the acquisition protocols of dynamic imaging, related kinetic parameters, advantages and the application of dynamic PET/CT imaging in malignant tumors, including lung cancer, hepatocellular carcinoma, breast cancer, pancreatic carcinoma, prostate neoplasm, and cancer of head and neck. Dynamic PET/CT imaging is increasingly being applied the diagnosis, staging, efficacy monitoring, and prognosis evaluation of malignant tumors. Although standardized uptake value is the most frequently employed semi-quantitative assessment index for static imaging, it is susceptible to several factors, thus cannot be used to evaluate the tracer kinetic information of the lesion. Dynamic PET/CT imaging can be used to achieve continuous assessment of the metabolic activity of a lesion over a certain time frame through quantitative measurement of kinetic parameters, such as the net uptake rate constant. Compared with conventional static imaging, dynamic scanning can be used for the early estimation of minute metabolic changes in tumors. Besides, dynamic scanning can directly and effectively reflect tracer uptake. Nevertheless, the intricacy of parameter analysis and the lengthy scanning time related to dynamic scanning limits its clinical application. Dynamic imaging has obtained remarkable achievements among a variety of malignant tumors due to the development of multiple simplified scanning protocols and the emergence of whole-body PET/CT scanners, which promote wider application of dynamic PET/CT. In this paper, we mainly review the acquisition protocols of dynamic imaging, related kinetic parameters, advantages and the application of dynamic PET/CT imaging in malignant tumors, including lung cancer, hepatocellular carcinoma, breast cancer, pancreatic carcinoma, prostate neoplasm, and cancer of head and neck.
Collapse
Affiliation(s)
- Yiling Wang
- Nuclear Medicine Department, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Yuanshan Yang
- Nuclear Medicine Department, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Jinxin Li
- Nuclear Medicine Department, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Dezhou Cheng
- Nuclear Medicine Department, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Hai Xu
- Division of Endocrinology and Rheumatology, HuangPi People’s Hospital, The Third Affiliated Hospital of Jianghan University, Wuhan, China
| | - Jinbai Huang
- Nuclear Medicine Department, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| |
Collapse
|
4
|
Fernández Calvo O, Muñoz Iglesias J, Abou Jokh Casas E, Molina-Díaz A, Anido Herranz U, Casas Nebra J, García-Bernardo L, Martínez-Breijo S, Lázaro-Quintela M, Muñiz-García G, Vázquez-Estevez S. Recommendations from the Galician Oncological Society and the Galician Society of Nuclear Medicine for the use of 177Lu-PSMA-617 radioligand-therapy in prostate cancer. Clin Transl Oncol 2025; 27:1383-1397. [PMID: 39266875 PMCID: PMC12000182 DOI: 10.1007/s12094-024-03662-7] [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: 05/02/2024] [Accepted: 08/07/2024] [Indexed: 09/14/2024]
Abstract
Theragnostic is a type of precision medicine that uses molecules linked to radioactive isotopes for the diagnosis and treatment of diseases. In recent years, it has gained significant importance to treat neuroendocrine tumors and is currently being used in prostate cancer. Various radiopharmaceuticals have emerged for diagnosing and detecting lesions showing prostate-specific membrane antigen (PSMA) positivity on the Positron emission tomography/computed tomography scan, being the most widely used labeled with [68Ga] and [18F]. Its use as therapy in prostate cancer (PC) has been assessed in the VISION, TheraP, and PSMAfore clinical trials conducted with the radioligand [177Lu]Lu-PSMA-617, demonstrating significant antitumor activity. The aim of this article is to present practical recommendations, based on current available scientific evidence and on a multidisciplinary consensus, for the diagnosis and treatment with [177Lu]Lu-PSMA-617 in patients with PC.
Collapse
Affiliation(s)
- Ovidio Fernández Calvo
- Department of Medical Oncology, Complexo Hospitalario Universitario de Ourense, Ourense, Spain.
| | - José Muñoz Iglesias
- Department of Nuclear Medicine (SERGAS), University Hospital of Vigo, Meixoeiro Hospital, Vigo, Spain
| | | | - Aura Molina-Díaz
- Department of Medical Oncology, Complexo Hospitalario Universitario de A Coruña, A Coruña, Spain
| | - Urbano Anido Herranz
- Department of Medical Oncology, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Javier Casas Nebra
- Uro-Oncology Unit, Complexo Hospitalario Universitario de A Coruña, A Coruña, Spain
| | - Lucía García-Bernardo
- Department of Nuclear Medicine, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Sara Martínez-Breijo
- Department of Urology, Complexo Hospitalario Universitario de A Coruña, A Coruña, Spain
| | - Martín Lázaro-Quintela
- Department of Medical Oncology, University Hospital of Vigo, Meixoeiro Hospital, Vigo, Spain
| | - Gloria Muñiz-García
- Department of Nuclear Medicine, Complexo Hospitalario Universitario de Ourense, Ourense, Spain
| | - Sergio Vázquez-Estevez
- Department of Medical Oncology, Hospital Universitario Lucus Augusti de Lugo, Lugo, Spain
| |
Collapse
|
5
|
Nassour AJ, Jain A, Khanani H, Hui N, Thompson NJ, Sorensen B, Baskaranathan S, Bergersen P, Chalasani V, Dean T, Dias M, Wines M, Symons J, Tarlinton L, Woo H. Impact of Uptake Period on 18F-DCFPyL-PSMA PET/CT Maximum Standardised Uptake Value. Cancers (Basel) 2025; 17:960. [PMID: 40149296 PMCID: PMC11940267 DOI: 10.3390/cancers17060960] [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: 01/27/2025] [Revised: 03/04/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND The maximum standardised uptake value (SUVmax) can potentially be affected by the uptake period during PSMA PET imaging. The optimal image acquisition period for 2-(3-{1-carboxy-5-[(6-18F-fluoro-pyridine-3-carbonyl)-amino]-pentyl}-ureido)-pentanedioic acid (18F-DCFPyL)PSMA PET/CT is yet to be established. This study aims to evaluate the effect of the uptake period on the SUVmax in diagnosing localised, clinically significant prostate cancer using 18F-DCFPyL-PSMA PET/CT. METHODS Sixty biopsy-naive men with one or more PI-RADS 4 or 5 lesions of at least 10 mm on multiparametric MRI (mpMRI) were enrolled to undergo 18F-DCFPyL-PSMA PET/CT. SUVmax was prospectively measured following an uptake period of 60, 90 and 120 min post injection of 18F-DCFPyL-PSMA radiotracer. Concordance with biopsy results or final histopathology was recorded. RESULTS Mean absolute differences in SUVmax at 60 vs. 90, 60 vs. 120, and 90 vs. 120 min uptake periods were 3.23 (SD 4.76), 4.53 (SD 7.33), and 3.24 (SD 4.56), respectively. This represents a statistically significant systematic increase in SUVmax (p-value < 0.001) with increasing uptake period. The interval between the uptake period of 60 vs. 120 min represented the largest SUVmax change of 29.98%. CONCLUSIONS The SUVmax is a dynamic variable significantly affected by uptake period. Our study supports image acquisition at 120 min following injection of 18F-DCFPyL radiotracer. Further studies are needed to determine if this acquisition period can be applied to other Fluorine-18 based PSMA radiotracers.
Collapse
Affiliation(s)
- Anthony-Joe Nassour
- Department of Urology, Sydney Adventist Hospital, Wahroonga, NSW 2076, Australia
| | - Anika Jain
- Department of Urology, Sydney Adventist Hospital, Wahroonga, NSW 2076, Australia
| | - Hadia Khanani
- Department of Urology, Sydney Adventist Hospital, Wahroonga, NSW 2076, Australia
| | - Nicholas Hui
- Department of Urology, Sydney Adventist Hospital, Wahroonga, NSW 2076, Australia
| | - Nadine J. Thompson
- SAN Nuclear Medicine and Radiology, Sydney Adventist Hospital, Wahroonga, NSW 2076, Australia
| | - Brian Sorensen
- SAN Nuclear Medicine and Radiology, Sydney Adventist Hospital, Wahroonga, NSW 2076, Australia
| | - Sris Baskaranathan
- Department of Urology, Sydney Adventist Hospital, Wahroonga, NSW 2076, Australia
- SAN Prostate Centre of Excellence, Sydney Adventist Hospital, Wahroonga, NSW 2076, Australia
| | - Philip Bergersen
- Department of Urology, Sydney Adventist Hospital, Wahroonga, NSW 2076, Australia
- SAN Prostate Centre of Excellence, Sydney Adventist Hospital, Wahroonga, NSW 2076, Australia
| | - Venu Chalasani
- Department of Urology, Sydney Adventist Hospital, Wahroonga, NSW 2076, Australia
- SAN Prostate Centre of Excellence, Sydney Adventist Hospital, Wahroonga, NSW 2076, Australia
| | - Thomas Dean
- Department of Urology, Sydney Adventist Hospital, Wahroonga, NSW 2076, Australia
- SAN Prostate Centre of Excellence, Sydney Adventist Hospital, Wahroonga, NSW 2076, Australia
| | - Max Dias
- Department of Urology, Sydney Adventist Hospital, Wahroonga, NSW 2076, Australia
- SAN Prostate Centre of Excellence, Sydney Adventist Hospital, Wahroonga, NSW 2076, Australia
| | - Michael Wines
- Department of Urology, Sydney Adventist Hospital, Wahroonga, NSW 2076, Australia
- SAN Prostate Centre of Excellence, Sydney Adventist Hospital, Wahroonga, NSW 2076, Australia
| | - James Symons
- Department of Urology, Sydney Adventist Hospital, Wahroonga, NSW 2076, Australia
- SAN Prostate Centre of Excellence, Sydney Adventist Hospital, Wahroonga, NSW 2076, Australia
| | | | - Henry Woo
- Department of Urology, Blacktown and Mount Druitt Hospital, Blacktown, NSW 2148, Australia
- Blacktown Mount Druitt Clinical School, Western Sydney University, Blacktown, NSW 2148, Australia
| |
Collapse
|
6
|
Karimzadeh A, Lehnert W, Koehler D, Shenas F, Kisters A, Apostolova I, Klutmann S, Adam G, Sauer M. Overview of selected completed prospective studies on PSMA-targeted radioligand therapy with [177Lu]Lu-PSMA-617 in metastatic castration-resistant prostate cancer. ROFO-FORTSCHR RONTG 2025. [PMID: 39842443 DOI: 10.1055/a-2514-4523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
Theranostics in nuclear oncology combines diagnostic and therapeutic procedures using radiotracers to target tumor cells. Prostate-specific membrane antigen (PSMA) is a key target in metastatic prostate cancer, and the radioligand [177Lu]Lu-PSMA-617, which binds to PSMA, has shown promising results in treating metastatic castration-resistant prostate cancer (mCRPC), leading to its approval by the European Medicines Agency in 2022.In this narrative review, the current evidence of [177Lu]Lu-PSMA-617 in mCRPC was discussed in the context of selected studies and the joint EANM/SNMMI guidelines for Lutetium-177-labeled PSMA-targeted radioligand therapy.The use of [177Lu]Lu-PSMA-617 for post-chemotherapy mCRPC is supported by substantial evidence from the phase II TheraP and the phase III VISION trials, demonstrating its safety and efficacy. The theranostic approach identifies patients likely to benefit from [177Lu]Lu-PSMA-617, which is effective only in tumors with sufficient PSMA expression, as detected by PSMA-ligand PET/CT, which is also used for response assessment.The success of [177Lu]Lu-PSMA-617 in post-chemotherapy mCRPC patients has led to further ongoing studies evaluating its use earlier in the treatment sequence, prior to chemotherapy. To ensure beneficial treatment outcome, adequate patient selection and evaluation of imaging-based response through PSMA-ligand PET/CT is necessary. · Indications for [177Lu]Lu-PSMA-617 are based on the TheraP and VISION clinical trials.. · Adequate patient selection using PSMA-ligand PET/CT is essential for beneficial outcomes.. · Response evaluation is based on imaging, PSA levels, and the patient's clinical condition.. · Karimzadeh A, Lehnert W, Koehler D et al. Overview of selected completed prospective studies on PSMA-targeted radioligand therapy with [177Lu]Lu-PSMA-617 in metastatic castration-resistant prostate cancer. Rofo 2025; DOI 10.1055/a-2514-4523.
Collapse
Affiliation(s)
- Amir Karimzadeh
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Wencke Lehnert
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Daniel Koehler
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Farzad Shenas
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anna Kisters
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ivayla Apostolova
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Susanne Klutmann
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gerhard Adam
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Markus Sauer
- Department of Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
7
|
Heilinger J, Roth KS, Weis H, Fink A, Weindler J, Dietlein F, Krapf P, Schomäcker K, Neumaier B, Dietlein M, Drzezga A, Kobe C. Do you know your PSMA-tracer? Variability in the biodistribution of different PSMA ligands and its potential impact on defining PSMA-positivity prior to PSMA-targeted therapy. EJNMMI Res 2025; 15:4. [PMID: 39792324 PMCID: PMC11723865 DOI: 10.1186/s13550-024-01190-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 12/15/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND In clinical practice, several radiopharmaceuticals are used for PSMA-PET imaging, each with distinct biodistribution patterns. This may impact treatment decisions and outcomes, as eligibility for PSMA-directed radioligand therapy is usually assessed by comparing tumoral uptake to normal liver uptake as a reference. In this study, we aimed to compare tracer uptake intraindividually in various reference regions including liver, parotid gland and spleen as well as the respective tumor-to-background ratios (TBR) of different 18F-labeled PSMA ligands to today's standard radiopharmaceutical 68Ga-PSMA-11 in a series of patients with biochemical recurrence of prostate cancer who underwent a dual PSMA-PET examination as part of an individualized diagnostic approach. RESULTS Differences in background activity among different PSMA-PET tracers lead to variations in tumor-to-background ratios (TBR). In [18F]F-DCFPyL-PET, TBR with the liver as the reference organ (TBRliver) was comparable to [68Ga]Ga-PSMA-11-PET, while [18F]F-PSMA-1007-PET and [18F]F-JK-PSMA-7-PET showed significantly lower values. Using the parotid gland as the reference (TBRparotidgland), [18F]F-DCFPyL-PET exhibited significantly higher values, whereas [18F]F-PSMA-1007-PET and [18F]F-JK-PSMA-7-PET were comparable. For the spleen (TBRspleen), [18F]F-JK-PSMA-7-PET was comparable, but [18F]F-DCFPyL-PET and [18F]F-PSMA-1007-PET showed significantly higher and lower values, respectively. An additional Bland-Altman analyses revealed low bias for [18F]F-DCFPyL-PET in TBRparotidgland, whereas significant differences in TBRliver and TBRspleen for the other tracers resulted in higher bias. CONCLUSION Different PSMA-PET tracers exhibit distinct biodistribution patterns, leading to variations in tumor-to-background ratios (TBR) in reference organs such as the liver, parotid gland, and spleen. Patient selection for PSMA-directed radioligand therapy is currently based on a semiquantitative approach using the liver as a reference region in [68Ga]Ga-PSMA-11-PET. Thus, the use of alternative [18F]-labeled tracers may result in under- or overestimation of a patient's suitability for therapy. This highlights the importance of a comprehensive understanding of the differences in tracer-specific uptake behavior for accurate decisions regarding PSMA-expression levels. However, as the patient cohort in this study is at earlier disease stages, the generalizability of these findings to later-stage patients remains unclear and requires further investigation.
Collapse
Affiliation(s)
- Jan Heilinger
- Department of Nuclear Medicine, University Hospital of Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Katrin Sabine Roth
- Department of Nuclear Medicine, University Hospital of Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Henning Weis
- Department of Nuclear Medicine, University Hospital of Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Antonis Fink
- Department of Nuclear Medicine, University Hospital of Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Jasmin Weindler
- Department of Nuclear Medicine, University Hospital of Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Felix Dietlein
- Department of Nuclear Medicine, University Hospital of Cologne, Kerpener Straße 62, 50937, Cologne, Germany
- Computational Health Informatics Program, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Philipp Krapf
- Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428, Jülich, Germany
| | - Klaus Schomäcker
- Department of Nuclear Medicine, University Hospital of Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Bernd Neumaier
- Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428, Jülich, Germany
- Institute of Radiochemistry and Experimental Molecular Imaging, University Hospital of Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Markus Dietlein
- Department of Nuclear Medicine, University Hospital of Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Alexander Drzezga
- Department of Nuclear Medicine, University Hospital of Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Carsten Kobe
- Department of Nuclear Medicine, University Hospital of Cologne, Kerpener Straße 62, 50937, Cologne, Germany.
| |
Collapse
|
8
|
Huang S, Ong S, McKenzie D, Mirabelli A, Chen DC, Chengodu T, Murphy DG, Hofman MS, Lawrentschuk N, Perera M. Comparison of 18F-based PSMA radiotracers with [ 68Ga]Ga-PSMA-11 in PET/CT imaging of prostate cancer-a systematic review and meta-analysis. Prostate Cancer Prostatic Dis 2024; 27:654-664. [PMID: 38017295 PMCID: PMC11543591 DOI: 10.1038/s41391-023-00755-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/12/2023] [Accepted: 11/03/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND Prostate-specific membrane antigen (PSMA) positron emission tomography (PET)/computed tomography (CT) has become an increasingly established imaging modality in the staging of prostate cancer (PCa). Numerous PSMA-based tracers are currently available, however, there is a lack of consensus on the optimal radiotracer(s) for PSMA PET/CT. This study aims to investigate whether Fluorine-18 (18F)-labelled PSMA PET/CT is significantly different from Gallium-68 (68Ga) in primary diagnosis and/or secondary staging of prostate cancer following biochemical recurrence. METHODS A critical review of MEDLINE, EMBASE, PubMed and Web of Science databases was performed in May 2023 according to the Preferred Reporting Items for Systematic Review and Meta-analysis (PRISMA) statement. Studies that directly compared 18F-based PSMA radiotracers and [68Ga]Ga-PSMA-11 in terms of the normal organ SUV or the lesion SUV or the detection rate were assessed. Quality was assessed using the Quality Assessment of Diagnostic Accuracy Studies-2 (QUADAS-2). RESULTS Twenty-four studies were analysed. [18F]DCFPyL and [18F]PSMA-1007 were the two most commonly studied 18F based PSMA tracers. [18F]JK-PSMA-7, [18F]rhPSMA-7, [18F]AlF-PSMA-11 were the new tracers evaluated in a limited number of studies. Overall, [18F]DCFPyL was observed to have a similar lesion detection rate to [68Ga]Ga-PSMA-11 with no increase in false positive rates. [18F]PSMA-1007 was found to have a greater local lesion detection rate because of its predominant hepatobiliary excretory route. However, [68Ga]Ga-PSMA-11 was observed to have a similar local lesion detection rate in studies that administer patients with furosemide prior to the scan. In addition, [18F]PSMA-1007 was found to have a significant number of benign bone uptakes. CONCLUSIONS [18F]DCFPyL was observed to be similar to [68Ga]Ga-PSMA-11. [18F]PSMA-1007 was observed to be less preferrable to [68Ga]Ga-PSMA-11 due to its high benign bone uptakes. Overall, there was not enough evidence in differentiating the radiotracers based on their clinical impacts.
Collapse
Affiliation(s)
- Siyu Huang
- Department of Surgery, University of Melbourne, Parkville, VIC, Australia.
| | - Sean Ong
- Department of Surgery, University of Melbourne, Parkville, VIC, Australia
- EJ Whitten Prostate Cancer Research Centre, Epworth HealthCare, Melbourne, VIC, Australia
| | - Dean McKenzie
- Research Development & Governance Unit, Epworth HealthCare, Melbourne, VIC, Australia
- Department of Health Science and Biostatistics, Swinburne University of Technology, Melbourne, VIC, Australia
| | - Adam Mirabelli
- Department of Surgery, University of Melbourne, Parkville, VIC, Australia
| | - David C Chen
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Young Urology Researchers Organisation (YURO), Melbourne, VIC, Australia
| | - Thilakavathi Chengodu
- EJ Whitten Prostate Cancer Research Centre, Epworth HealthCare, Melbourne, VIC, Australia
| | - Declan G Murphy
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Michael S Hofman
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Nathan Lawrentschuk
- Department of Surgery, University of Melbourne, Parkville, VIC, Australia
- EJ Whitten Prostate Cancer Research Centre, Epworth HealthCare, Melbourne, VIC, Australia
- Department of Urology, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Marlon Perera
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Department of Surgery, Austin Health, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
9
|
Penny R, Fongenie B, Davis P, Sykes J. Normal-organ distribution of PSMA-targeting PET radiopharmaceutical 18F-flotufolastat: a post hoc analysis of the LIGHTHOUSE and SPOTLIGHT studies. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2024; 14:337-344. [PMID: 39583908 PMCID: PMC11578810 DOI: 10.62347/incg3525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 07/28/2024] [Indexed: 11/26/2024]
Abstract
BACKGROUND High-affinity radiohybrid PSMA-targeting radiopharmaceutical 18F-flotufolastat (18F-rhPSMA-7.3) is newly approved for diagnostic imaging of prostate cancer. Here, we conduct a post hoc analysis of two phase 3 studies to quantify 18F-flotufolastat uptake in a range of normal organs. METHODS All 718 evaluable 18F-flotufolastat scans from LIGHTHOUSE and SPOTLIGHT were re-evaluated. Additionally, patients' medical records were reviewed and any patients with high tumor burden (PSA>20 ng/mL), altered biodistribution (e.g., chronic kidney disease), major anatomical changes to normal organs (e.g., nephrectomy), or any other history of cancer were excluded. A medical physicist defined volumes of interest over specific organs for evaluation of SUVmean and SUVpeak per PERCIST 1.0 criteria. Normally distributed data are reported as mean (SD) and non-normally distributed data as median (IQR). The co-efficient of variation (CoV; calculated as SD/mean for normally distributed data and IQR/median for non-normally distributed data) was used to quantify variability of SUV metrics. RESULTS In total, scans from 546 patients (244 primary, 302 recurrent) were eligible for this analysis. All organs were considered to be normally distributed except for the bladder and spleen. In the liver, the mean SUVmean was 6.7 (SD 1.7), CoV 26%, while the bladder median SUVmean was 10.6 (IQR 11.9), CoV 112%. The mean SUVpeak in the liver was 8.2 (SD 2.1), CoV 26% and median SUVpeak in the bladder was 16.0 (IQR 18.5), CoV 116%. CONCLUSIONS Physiological 18F-flotufolastat uptake in normal organs was broadly consistent with other renally-cleared radiopharmaceuticals, which may have clinically significant implications when considering patient selection for radioligand therapy. Additionally, the bladder median SUVpeak for 18F-flotufolastat was lower than that previously reported for 68Ga-PSMA-11 and 18F-DCFPyL.
Collapse
Affiliation(s)
- Ross Penny
- Blue Earth Diagnostics Ltd.The Oxford Science Park, Magdalen Centre, Robert Robinson Avenue, Oxford, OX4 4GA, UK
| | - Benjamin Fongenie
- Blue Earth Therapeutics Ltd.The Oxford Science Park, Magdalen Centre, Robert Robinson Avenue, Oxford, OX4 4GA, UK
| | - Phillip Davis
- Blue Earth Diagnostics Inc.No. 259 Prospect Plains Road, Building H, Suite 100, Monroe Township, New Jersey 08831, USA
| | - James Sykes
- Blue Earth Diagnostics Ltd.The Oxford Science Park, Magdalen Centre, Robert Robinson Avenue, Oxford, OX4 4GA, UK
| |
Collapse
|
10
|
Osman MM, Iravani A, Mitchell C, Hicks RJ, Perry E, Hofman MS. 18F-DCFPyL PSMA PET/CT Tracheobronchial Uptake in Patients with Prostate Cancer: Incidence and Etiology. J Nucl Med 2024; 65:1383-1386. [PMID: 39089815 PMCID: PMC11372262 DOI: 10.2967/jnumed.124.267772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/25/2024] [Indexed: 08/04/2024] Open
Abstract
We evaluated the incidence and potential etiology of tracheobronchial uptake in patients being evaluated by 18F-DCFPyL PET/CT for prostate cancer (PCa). Methods: The study included a consecutive 100 PCa patients referred for 18F-DCFPyL PET/CT. The PET/CT scans were retrospectively reviewed. The presence or absence of physiologic tracheobronchial uptake on PET/CT was recorded. To further evaluate tracheal prostate-specific membrane antigen (PSMA) expression, immunohistochemistry was performed on tracheal samples taken from 2 men who had surgical resection of lung cancer. Results: Tracheal uptake was present in 31 of 100 patients (31%). When tracheal uptake was present, the SUVmax was significantly higher in the left main bronchus (mean, 2.7) than in the right (mean, 2.3) (P < 0.001). Histopathologic testing of tracheobronchial samples showed PSMA expression in bronchial submucosal glands. Conclusion: In PCa patients undergoing 18F-DCFPyL PET/CT, tracheobronchial uptake occurred in 31% of patients. This is attributed to normal physiologic PSMA expression in bronchial submucosal glands.
Collapse
Affiliation(s)
- Medhat M Osman
- Department of Radiology, Saint Louis University Hospital, St. Louis, Missouri;
| | - Amir Iravani
- Department of Radiology, Fred Hutchinson Cancer Center, University of Washington, Seattle, Washington
| | - Catherine Mitchell
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Rodney J Hicks
- Melbourne Theranostic Innovation Centre, Melbourne, Victoria, Australia
| | - Elisa Perry
- Department of Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; and
| | - Michael S Hofman
- Department of Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; and
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
11
|
Gelikman DG, Mena E, Lindenberg L, Azar WS, Rathi N, Yilmaz EC, Harmon SA, Schuppe K, Hsueh J, Huth H, Wood BJ, Gurram S, Choyke PL, Pinto PA, Turkbey B. Reducing False-Positives Due to Urinary Stagnation in the Prostatic Urethra on 18 F-DCFPyL PSMA PET/CT With MRI. Clin Nucl Med 2024; 49:630-636. [PMID: 38651785 PMCID: PMC11150104 DOI: 10.1097/rlu.0000000000005220] [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] [Indexed: 04/25/2024]
Abstract
PURPOSE Prostate-specific membrane antigen (PSMA)-targeting PET radiotracers reveal physiologic uptake in the urinary system, potentially misrepresenting activity in the prostatic urethra as an intraprostatic lesion. This study examined the correlation between midline 18 F-DCFPyL activity in the prostate and hyperintensity on T2-weighted (T2W) MRI as an indication of retained urine in the prostatic urethra. PATIENTS AND METHODS Eighty-five patients who underwent both 18 F-DCFPyL PSMA PET/CT and prostate MRI between July 2017 and September 2023 were retrospectively analyzed for midline radiotracer activity and retained urine on postvoid T2W MRIs. Fisher's exact tests and unpaired t tests were used to compare residual urine presence and prostatic urethra measurements between patients with and without midline radiotracer activity. The influence of anatomical factors including prostate volume and urethral curvature on urinary stagnation was also explored. RESULTS Midline activity on PSMA PET imaging was seen in 14 patients included in the case group, whereas the remaining 71 with no midline activity constituted the control group. A total of 71.4% (10/14) and 29.6% (21/71) of patients in the case and control groups had urethral hyperintensity on T2W MRI, respectively ( P < 0.01). Patients in the case group had significantly larger mean urethral dimensions, larger prostate volumes, and higher incidence of severe urethral curvature compared with the controls. CONCLUSIONS Stagnated urine within the prostatic urethra is a potential confounding factor on PSMA PET scans. Integrating PET imaging with T2W MRI can mitigate false-positive calls, especially as PSMA PET/CT continues to gain traction in diagnosing localized prostate cancer.
Collapse
Affiliation(s)
- David G. Gelikman
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Esther Mena
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Liza Lindenberg
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - William S. Azar
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nityam Rathi
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Enis C. Yilmaz
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Stephanie A. Harmon
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kyle Schuppe
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jessica Hsueh
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Hannah Huth
- Center for Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Department of Radiology, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Bradford J. Wood
- Center for Interventional Oncology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Department of Radiology, Clinical Center, National Institutes of Health, Bethesda, MD, USA
| | - Sandeep Gurram
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Peter L. Choyke
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Peter A. Pinto
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Baris Turkbey
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
12
|
Yadav S, Kim ST, Tuchayi AM, Jiang F, Morley A, Saelee R, Wang Y, Juarez R, Lawnh-Heath C, Koshkin VS, Hope TA. Comparison of 18F-DCFPyL and 68Ga-PSMA-11 for 177Lu-PSMA-617 therapy patient selection. Front Oncol 2024; 14:1382582. [PMID: 38993644 PMCID: PMC11238039 DOI: 10.3389/fonc.2024.1382582] [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: 02/08/2024] [Accepted: 05/30/2024] [Indexed: 07/13/2024] Open
Abstract
Purpose 68Ga-PSMA-11 is recommended for the selection of patients for treatment in the package insert for 177Lu-PSMA-617. We aimed to compare imaging properties and post-treatment outcomes from radioligand therapy (RLT) of patients selected with 68Ga-PSMA-11 and 18F-DCFPyL. Methods We retrospectively evaluated 80 patients undergoing PSMA RLT, who had pretreatment imaging using either 68Ga-PSMA-11 or 18F-DCFPyL. For both groups, we compared the biodistribution and lesion uptake and the PSA response to treatment. Results Both agents had comparable biodistribution. Patients initially imaged with 18F-DCFPyL had a higher PSA response (66% vs. 42%), and more patients had a PSA50 response (72% vs. 43%) compared to patients imaged with 68Ga-PSMA-11. Conclusion 18F-DCFPyL and 68Ga-PSMA-11 had comparable biodistribution and lesion uptake. Patients imaged with 18F-DCFPyL demonstrated clinical benefit to PSMA RLT comparable to those imaged with 68Ga-PSMA-11, and either agent can be used for screening patients.
Collapse
Affiliation(s)
- Surekha Yadav
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| | - Sarasa T. Kim
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| | - Abuzar Moradi Tuchayi
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| | - Fei Jiang
- Department of Epidemiology & Biostatistics, University of California, San Francisco, San Francisco, CA, United States
| | - Amanda Morley
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| | - Rachelle Saelee
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| | - Yingbing Wang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| | - Roxanna Juarez
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| | - Courtney Lawnh-Heath
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
| | - Vadim S. Koshkin
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA, United States
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, United States
| | - Thomas A. Hope
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, United States
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, United States
- Department of Radiology, San Francisco Veterans Affairs (VA) Medical Center, San Francisco, CA, United States
| |
Collapse
|
13
|
Lucaroni L, Oehler S, Georgiev T, Müller M, Bocci M, De Luca R, Favalli N, Neri D, Cazzamalli S, Prati L. DNA-encoded chemical libraries enable the discovery of potent PSMA-ligands with substantially reduced affinity towards the GCPIII anti-target. Chem Sci 2024; 15:6789-6799. [PMID: 38725500 PMCID: PMC11077555 DOI: 10.1039/d3sc06668a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 03/17/2024] [Indexed: 05/12/2024] Open
Abstract
Prostate-specific membrane antigen (PSMA) is a tumor-associated protein that has been successfully targeted with small organic ligands and monoclonal antibodies. Pluvicto™ is a PSMA-targeted radioligand therapeutic (RLT) recently approved by the FDA for the treatment of metastatic castration-resistant prostate cancer (2022 FDA marketing authorization). Although a large Phase III clinical trial (VISION trial) demonstrated clinical benefits in patients treated with Pluvicto™, the therapeutic window of the drug is narrowed by its undesired accumulation in healthy organs. Glutamate carboxypeptidase III (GCPIII), an enzyme sharing 70% identity with PSMA, may be responsible for the off-target accumulation of PSMA-RLTs in salivary glands and kidneys. In this work, we designed and synthesized affinity and selectivity maturation DNA-encoded chemical libraries (ASM-DELs) comprising 18'284'658 compounds that were screened in parallel against PSMA and GCPIII with the aim to identify potent and selective PSMA ligands for tumor-targeting applications. Compound A70-B104 was isolated as the most potent and selective ligand (KD of 900 pM for PSMA, KD of 40 nM for GCPIII). 177Lu-A70-B104-DOTA, a radiolabeled derivative of compound A70-B104, presented selective accumulation in PSMA-positive cancer lesions (i.e., 7.4% ID g-1, 2 hour time point) after systemic administration in tumor-bearing mice. The results of autoradiography experiments showed that 177Lu-A70-B104-DOTA selectively binds to PSMA-positive cancer tissues, while negligible binding on human salivary glands was observed.
Collapse
Affiliation(s)
- Laura Lucaroni
- Philochem AG, R&D Department CH-8112 Otelfingen Switzerland +41 43 544 88 19
| | - Sebastian Oehler
- Philochem AG, R&D Department CH-8112 Otelfingen Switzerland +41 43 544 88 19
| | - Tony Georgiev
- Philochem AG, R&D Department CH-8112 Otelfingen Switzerland +41 43 544 88 19
| | - Marco Müller
- Philochem AG, R&D Department CH-8112 Otelfingen Switzerland +41 43 544 88 19
| | - Matilde Bocci
- Philochem AG, R&D Department CH-8112 Otelfingen Switzerland +41 43 544 88 19
| | - Roberto De Luca
- Philochem AG, R&D Department CH-8112 Otelfingen Switzerland +41 43 544 88 19
| | - Nicholas Favalli
- Philochem AG, R&D Department CH-8112 Otelfingen Switzerland +41 43 544 88 19
| | - Dario Neri
- Philochem AG, R&D Department CH-8112 Otelfingen Switzerland +41 43 544 88 19
- Swiss Federal Institute of Technology, Department of Chemistry and Applied Biosciences CH-8093 Zurich Switzerland
- Philogen S.p.A. 53100 Siena Italy +39 0577 178 16 59
| | - Samuele Cazzamalli
- Philochem AG, R&D Department CH-8112 Otelfingen Switzerland +41 43 544 88 19
| | - Luca Prati
- Philogen S.p.A. 53100 Siena Italy +39 0577 178 16 59
| |
Collapse
|
14
|
Wang H, Zhu H, Li G, Dai J, Huang H, Jia Q. Effect of 18F-DCFPyL PET on changes in management of patients with prostate cancer: a systematic review and meta-analysis. Front Med (Lausanne) 2024; 11:1355236. [PMID: 38725467 PMCID: PMC11079165 DOI: 10.3389/fmed.2024.1355236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/15/2024] [Indexed: 05/12/2024] Open
Abstract
Purpose Prostate-specific membrane antigen (PSMA)-targeted imaging has gained increasing interest in its application in prostate cancer lesion detection. Compared with 68Galium (68Ga), 18Fluoride (18F)-labeled imaging agent has easier syntheses, lower price, and a longer half-time. 2-(3-{1-Carboxy-5-[(6-[18F]fluoro-pyridine-3-carbonyl)-amino]-pentyl}-ureido)-pentanedioic acid positron emission tomography (18F-DCFPyL PET) has been recently approved by the U.S. Food and Drug Administration. Several studies have proven its superiority to conventional imaging techniques in detecting prostate cancer lesions. However, the impact of 18F-DCFPyL PET on the management of patients with prostate cancer is not well established. Thus, we performed a systematic review and meta-analysis of available data to evaluate the impact of 18F-DCFPyL PET on the management of patients with prostate cancer. Methods The PubMed, Embase, Scopus, and Cochrane databases were searched up to April 2024. Studies that reported the proportion of changes in management after 18F-DCFPyL PET was performed in patients with prostate cancer were included. The Grading of Recommendations Assessment, Development, and Evaluation system was used for the quality evaluation of the included studies. The proportion of changes in management was pooled using a random effects model. Meta-regression analyses were performed to assess the potential correlation between the PET positivity and management changes. Results Fourteen studies (3,078 patients with prostate cancer) were included in our review and analysis. The pooled percentage of management changes was 43.5% (95% confidence interval [CI]: 33-54%). In patients with biochemical recurrent and for primary staging, the pooled percentage was 50% (95% CI: 39-60%) and 22% (95% CI: 15-29%), respectively. In the meta-regression analyses, PET positivity was detected as a significant predictor of management change (p = 0.0023). Conclusion 18F-DCFPyL PET significantly affects the management of patients with prostate cancer. Higher PET positivity rate significantly correlated with a higher proportion of management changes in patients with prostate cancer. However, more studies are still needed to confirm the important role of 18F-DCFPyL PET in the management of prostate cancer. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/#myprospero, CRD42022339178.
Collapse
Affiliation(s)
- Hui Wang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - HongMei Zhu
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - GuanNan Li
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - JiaoNa Dai
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - HeXiao Huang
- Department of Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Qiong Jia
- Department of Pediatrics, Peking University Third Hospital, Beijing, China
| |
Collapse
|
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
|
Jiang Z, Guo J, Hu L, Yang S, Meng B, Tang Q. Diagnostic performance of 18F‑DCFPyL PET vs. 68Ga‑PSMA PET/CT in patients with suspected prostate cancer: A systemic review and meta‑analysis. Oncol Lett 2024; 27:188. [PMID: 38486944 PMCID: PMC10938285 DOI: 10.3892/ol.2024.14321] [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: 10/26/2023] [Accepted: 02/13/2024] [Indexed: 03/17/2024] Open
Abstract
In this systematic review and meta-analysis, the diagnostic performance of 68Ga-prostate-specific membrane antigen (PSMA) positron emission tomography (PET)/CT was compared with that of 18F-DCFPyL PET for patients with suspected prostate cancer (PCa). Up to September 2023, the PubMed, Embase and Web of Science databases were thoroughly searched for relevant papers. Studies examining the diagnostic performance of 18F-DCFPyL PET and 68Ga-PSMA PET/CT in patients with suspected PCa were included in the present review. The Quality Assessment of Diagnostic Performance Studies-2 tool was used to rate the diagnostic performance of each study. The diagnostic performance of 18F-DCFPyL PET and 68Ga-PSMA PET/CT for primary PCa was examined by 13 studies included, comprising 1,178 patients. The pooled sensitivity and specificity of 18F-DCFPyL PET were 0.92 (95% CI, 0.85-0.96) and 0.59 (95% CI, 0.08-0.96), respectively. For 68Ga-PSMA PET/CT, the pooled sensitivity and specificity were 0.96 (95% CI, 0.88-0.99) and 0.71 (95% CI, 0.57-0.82), respectively. 18F-DCFPyL PET and 68Ga-PSMA PET/CT both had an area under the receiver operating characteristic curve of 0.92 (95% CI, 0.89-0.94). In addition, the Fagan nomogram revealed that the post-test probabilities for 18F-DCFPyL PET and 68Ga-PSMA PET/CT could rise to 69 and 77% when the pre-test probability was set at 50%. In conclusion, a comparable diagnostic performance for patients with suspected PCa was determined for 18F-DCFPyL PET and 68Ga-PSMA PET/CT. However, it is crucial to keep in mind that the findings of the present meta-analysis come from investigations with modest sample sizes. Therefore, more extensive research is required to obtain more solid data.
Collapse
Affiliation(s)
- Zhibing Jiang
- Department of Clinical Medicine, Medical College, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Jinjing Guo
- Department of Clinical Medicine, Medical College, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Liang Hu
- Department of Clinical Medicine, Medical College, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Siyu Yang
- Department of Clinical Medicine, Medical College, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Bin Meng
- Department of Clinical Medicine, Medical College, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| | - Qun Tang
- Department of Clinical Medicine, Medical College, Hunan University of Chinese Medicine, Changsha, Hunan 410208, P.R. China
| |
Collapse
|
18
|
Pruis IJ, van Doormaal PJ, Balvers RK, van den Bent MJ, Harteveld AA, de Jong LC, Konijnenberg MW, Segbers M, Valkema R, Verburg FA, Smits M, Veldhuijzen van Zanten SEM. Potential of PSMA-targeting radioligand therapy for malignant primary and secondary brain tumours using super-selective intra-arterial administration: a single centre, open label, non-randomised prospective imaging study. EBioMedicine 2024; 102:105068. [PMID: 38518652 PMCID: PMC10981001 DOI: 10.1016/j.ebiom.2024.105068] [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: 08/24/2023] [Revised: 02/27/2024] [Accepted: 03/05/2024] [Indexed: 03/24/2024] Open
Abstract
BACKGROUND The aim of this study was to provide quantitative evidence for the potential of PSMA-targeting radioligand therapy (RLT) as treatment approach for malignant brain tumours, and to explore whether tumour uptake could be enhanced by super-selective intra-arterial (ssIA)-administration. METHODS Ten patients (n = 5 high-grade glioma, n = 5 brain metastasis) received 1.5 MBq/kg [68Ga]Ga-PSMA-11 intravenously and, within 7 days, intra-arterially (i.e., selectively in tumour-feeding arteries), followed twice by PET-MRI at 90, 165 and 240 min post-injection. Patient safety was monitored for each procedure. Standardised uptake values (SUVs) were obtained for tumour, healthy-brain, salivary glands and liver. Tumour-to-salivary-gland (T/SG) and tumour-to-liver (T/L) uptake-ratios were calculated. FINDINGS No adverse events requiring study termination occurred. All patients showed uptake of [68Ga]Ga-PSMA-11 at the tumour site. Uptake was a median 15-fold higher following ssIA-administration (SUVmax median: 142.8, IQR: 102.8-245.9) compared to IV-administration (10.5, IQR:7.5-13.0). According to the bootstrap analysis, mean SUVmax after ssIA (168.8, 95% CI: 110.6-227.0) was well beyond the 95% confidence-interval of IV administration (10.5, 95% CI: 8.4-12.7). Uptake in healthy-brain was negligible, independent of administration route (SUVmean <0.1-0.1). Off-target uptake was comparable, resulting in more favourable T/SG- and T/L-ratios of 8.4 (IQR: 4.4-11.5) and 26.5 (IQR: 14.0-46.4) following ssIA, versus 0.5 (IQR: 0.4-0.7) and 1.8 (IQR: 1.0-2.7) for IV-administration. INTERPRETATION ssIA-administration is safe and leads to a median fifteen-fold higher radioligand uptake at the tumour site, therewith qualifying more patients for treatment and enhancing the potential of therapy. These results open new avenues for the development of effective RLT-based treatment strategies for patients with brain tumours. FUNDING Semmy Foundation.
Collapse
Affiliation(s)
- Ilanah J Pruis
- Department of Radiology and Nuclear Medicine, Erasmus MC, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands; Brain Tumour Centre, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Pieter Jan van Doormaal
- Department of Radiology and Nuclear Medicine, Erasmus MC, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Rutger K Balvers
- Brain Tumour Centre, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands; Department of Neurosurgery, Erasmus MC, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Martin J van den Bent
- Department of Neurology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Anita A Harteveld
- Department of Radiology and Nuclear Medicine, Erasmus MC, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Linda C de Jong
- Department of Radiology and Nuclear Medicine, Erasmus MC, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Mark W Konijnenberg
- Department of Radiology and Nuclear Medicine, Erasmus MC, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Marcel Segbers
- Department of Radiology and Nuclear Medicine, Erasmus MC, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Roelf Valkema
- Department of Radiology and Nuclear Medicine, Erasmus MC, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Frederik A Verburg
- Department of Radiology and Nuclear Medicine, Erasmus MC, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Marion Smits
- Department of Radiology and Nuclear Medicine, Erasmus MC, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands; Brain Tumour Centre, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands; Medical Delta, Delft, Huismansingel 4, 2629 JH, Delft, the Netherlands
| | - Sophie E M Veldhuijzen van Zanten
- Department of Radiology and Nuclear Medicine, Erasmus MC, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands; Brain Tumour Centre, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands.
| |
Collapse
|
19
|
Jewell K, Hofman MS, Ong JSL, Levy S. Emerging Theranostics for Prostate Cancer and a Model of Prostate-specific Membrane Antigen Therapy. Radiology 2024; 311:e231703. [PMID: 38563674 DOI: 10.1148/radiol.231703] [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: 04/04/2024]
Abstract
There is increasing demand worldwide to develop diagnostic and therapeutic (theranostic) markers for prostate cancer. One target of interest is prostate-specific membrane antigen (PSMA), a protein which is overexpressed in prostate cancer cells. Over the past decade, a growing body of literature has demonstrated that radiolabeled ligands that target PSMA show favorable clinical response and survival outcomes in patients with advanced prostate cancer. This focused review provides background to the development of PSMA as a target, an overview of key studies informing our current approach to radioligand-based imaging and therapy for prostate cancer, and a model for real-world implementation of PSMA theranostics based on an Australian experience.
Collapse
Affiliation(s)
- Kerry Jewell
- From the Department of Molecular Imaging and Therapeutic Nuclear Medicine and Department of Oncology, Prostate Theranostics and Imaging Centre of Excellence (ProsTIC); Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia (K.J., M.S.H., S.L.); University of Melbourne, Melbourne, Australia (M.S.H.); and Department of Nuclear Medicine, Fiona Stanley Hospital, Murdoch, Australia (J.S.L.O.)
| | - Michael S Hofman
- From the Department of Molecular Imaging and Therapeutic Nuclear Medicine and Department of Oncology, Prostate Theranostics and Imaging Centre of Excellence (ProsTIC); Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia (K.J., M.S.H., S.L.); University of Melbourne, Melbourne, Australia (M.S.H.); and Department of Nuclear Medicine, Fiona Stanley Hospital, Murdoch, Australia (J.S.L.O.)
| | - Jeremy S L Ong
- From the Department of Molecular Imaging and Therapeutic Nuclear Medicine and Department of Oncology, Prostate Theranostics and Imaging Centre of Excellence (ProsTIC); Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia (K.J., M.S.H., S.L.); University of Melbourne, Melbourne, Australia (M.S.H.); and Department of Nuclear Medicine, Fiona Stanley Hospital, Murdoch, Australia (J.S.L.O.)
| | - Sidney Levy
- From the Department of Molecular Imaging and Therapeutic Nuclear Medicine and Department of Oncology, Prostate Theranostics and Imaging Centre of Excellence (ProsTIC); Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC 3000, Australia (K.J., M.S.H., S.L.); University of Melbourne, Melbourne, Australia (M.S.H.); and Department of Nuclear Medicine, Fiona Stanley Hospital, Murdoch, Australia (J.S.L.O.)
| |
Collapse
|
20
|
Ali I, Rezk M, Hamouda D, Talaat O, Omar Y, Abdel Tawab M, Nasr I. Clinical value of 18F-PSMA-1007 PET/MRI in primary staging of patients with intermediate- to high-risk prostate cancer. Br J Radiol 2024; 97:622-631. [PMID: 38265254 PMCID: PMC11027301 DOI: 10.1093/bjr/tqae021] [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: 11/14/2023] [Revised: 01/09/2024] [Accepted: 01/18/2024] [Indexed: 01/25/2024] Open
Abstract
OBJECTIVE To assess the utility of 18F-PSMA-1007 PET/MRI in initial staging of intermediate- to high-risk prostate cancer (HRPCa). METHODS A total of 46 patients with pathologically verified intermediate and/or HRPCa who underwent 18F-PSMA-1007 PET/MRI with dedicated pelvic high-resolution multiparametric MRI (mpMRI) were included. RESULTS PET/MRI showed 100% sensitivity (SN), specificity (SP), positive predictive value (PPV), negative predictive value (NPV), and accuracy in detecting seminal vesicle (SV) and rectal invasion, versus 87.5%, 100%, 100% 93.8%, 95.7% and 50%, 100%,100%, 95.5%, and 95.7% for mpMRI respectively. However, PET/MRI had poor SN (40% and 0%) but high SP (94.4% and 100%) in detection of UB and neurovascular bundle (NV) invasion compared to 100% SN and SP for mpMRI. PET/MRI demonstrated stronger TNM staging agreement with the gold standard than mpMRI-WBMRI. It demonstrated concordance with T, N, and M stages in 40, 41, and 36 patients (k 0.84, 0.60, and 0.68, respectively) versus 29, 33, and 31 patients (k 0.54, 0.22, and 0.50) with accurate over all staging of 38/46 patients versus 30/46 patients (K 0.52 versus 0.22). CONCLUSION 18F-PSMA-1007 PET/MRI is a promising imaging modality with high diagnostic accuracy in staging intermediate- and HRPCa; it improves local tumour evaluation and provides precise TNM staging. ADVANCES IN KNOWLEDGE 18F-PSMA-1007 PET/MRI could have high diagnostic accuracy as shown in the current study for staging HRPCa patients that is crucial for treatment selection. We think that our study will contribute to the body of knowledge and improve the literature surrounding the clinical uses of integrated 18F-PSMA-1007 PET/MRI.
Collapse
Affiliation(s)
- Ismail Ali
- Radiology Department, Faculty of Human Medicine, Zagazig University, Zagazig, Faculty of medicine street, Zagazig, Sharkia, 44519, Egypt
| | - Mahmoud Rezk
- Radiology Department, National cancer Institute, Cairo University, Cairo, 11796, Egypt
| | - Dalia Hamouda
- Medical Oncology Department, Faculty of Human Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Omnia Talaat
- Radiation Oncology Department, National Cancer Institute, Cairo University, Cairo, 11796, Egypt
| | - Yehia Omar
- Director of PET/MRI unit, Misr Radiology Cente, Cairo, 11766, Egypt
| | - Mohamed Abdel Tawab
- Radiology Department, Faculty of Human Medicine, Alazhar University, Cairo, 11651, Egypt
| | - Ibrahim Nasr
- Clinical Oncology and Nuclear Medicine Department, Faculty of Human Medicine, Zagazig University, Zagazig, 44519, Egypt
| |
Collapse
|
21
|
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
|
22
|
Philbrook P, Casano KR, Lee DJ. Novel Biodistribution of PSMA Radiotracer in the Uvula of Patients Undergoing PSMA PET/CT. Clin Nucl Med 2024; 49:e80-e81. [PMID: 38049963 DOI: 10.1097/rlu.0000000000004993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/06/2023]
Abstract
ABSTRACT Prostate-specific membrane antigen (PSMA) PET/CT is an imaging technique that detects primary and metastatic prostate cancer and evaluates treatment effectiveness. The radioligands for PSMA PET/CT are known to have physiological off-target uptake in various tissues. These include the well-known off-target major and minor salivary glands. We report that, in addition to this location, radioligand uptake can be seen in the uvula, which we suggest is from salivary tissue in this location. PSMA uptake in the uvula is not reported in the literature and is a rare, but normal location for tracer biodistribution in some patients.
Collapse
Affiliation(s)
- Phaethon Philbrook
- From the School of Medicine, Louisiana State University Health Sciences Center New Orleans, New Orleans
| | - Kelsey R Casano
- Department of Radiology, Ochsner Clinic Foundation, Jefferson, LA
| | - Daniel J Lee
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
23
|
Maekawa S, Takata R, Obara W. Molecular Mechanisms of Prostate Cancer Development in the Precision Medicine Era: A Comprehensive Review. Cancers (Basel) 2024; 16:523. [PMID: 38339274 PMCID: PMC10854717 DOI: 10.3390/cancers16030523] [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: 11/24/2023] [Revised: 01/21/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
The progression of prostate cancer (PCa) relies on the activation of the androgen receptor (AR) by androgens. Despite efforts to block this pathway through androgen deprivation therapy, resistance can occur through several mechanisms, including the abnormal activation of AR, resulting in castration-resistant PCa following the introduction of treatment. Mutations, amplifications, and splicing variants in AR-related genes have garnered attention in this regard. Furthermore, recent large-scale next-generation sequencing analysis has revealed the critical roles of AR and AR-related genes, as well as the DNA repair, PI3K, and cell cycle pathways, in the onset and progression of PCa. Moreover, research on epigenomics and microRNA has increasingly become popular; however, it has not translated into the development of effective therapeutic strategies. Additionally, treatments targeting homologous recombination repair mutations and the PI3K/Akt pathway have been developed and are increasingly accessible, and multiple clinical trials have investigated the efficacy of immune checkpoint inhibitors. In this comprehensive review, we outline the status of PCa research in genomics and briefly explore potential future developments in the field of epigenetic modifications and microRNAs.
Collapse
Affiliation(s)
- Shigekatsu Maekawa
- Department of Urology, Iwate Medical University, Iwate 028-3694, Japan; (R.T.); (W.O.)
| | | | | |
Collapse
|
24
|
Fuscaldi LL, Sobral DV, Durante ACR, Mendonça FF, Miranda ACC, Salgueiro C, de Castiglia SG, Yamaga LYI, da Cunha ML, Malavolta L, de Barboza MF, Mejia J. Radiochemical and biological assessments of a PSMA-I&S cold kit for fast and inexpensive 99mTc-labeling for SPECT imaging and radioguided surgery in prostate cancer. Front Chem 2023; 11:1271176. [PMID: 37901160 PMCID: PMC10602725 DOI: 10.3389/fchem.2023.1271176] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/29/2023] [Indexed: 10/31/2023] Open
Abstract
The expression of prostate-specific membrane antigen (PSMA) is upregulated in prostate cancer (PCa) cells and PSMA-ligands have been radiolabeled and used as radiopharmaceuticals for targeted radionuclide therapy (TRT), single photon emission computed tomography (SPECT) or positron emission tomography (PET) molecular imaging, and radioguided surgery in PCa patients. Herein, we aimed at radiolabeling the PSMA-I&S cold kit with 99mTc, resulting in a radiopharmaceutical with high radiochemical yield (RCY) and stability for SPECT imaging and radioguided surgery in PCa malignancies. Various pre-clinical assays were conducted to evaluate the [99mTc]Tc-PSMA-I&S obtained by the cold kit. These assays included assessments of RCY, radiochemical stability in saline, lipophilicity, serum protein binding (SPB), affinity for LNCaP-PCa cells (binding and internalization studies), and ex vivo biodistribution profile in naive and LNCaP-PCa-bearing mice. The radiopharmaceutical was obtained with good RCY (92.05% ± 2.20%) and remained stable for 6 h. The lipophilicity was determined to be -2.41 ± 0.06, while the SPB was ∼97%. The binding percentages to LNCaP cells were 9.41% ± 0.57% (1 h) and 10.45% ± 0.45% (4 h), with 63.12 ± 0.93 (1 h) and 65.72% ± 1.28% (4 h) of the bound material being internalized. Blocking assays, employing an excess of unlabeled PSMA-I&S, resulted in a reduction in the binding percentage by 2.6 times. The ex vivo biodistribution profile confirmed high accumulation of [99mTc]Tc-PSMA-I&S in the tumor and the tumor-to-contralateral muscle ratio was ∼6.5. In conclusion, [99mTc]Tc-PSMA-I&S was successfully obtained by radiolabeling the cold kit using freshly eluted [99mTc]NaTcO4, exhibiting good RCY and radiochemical stability. The preclinical assays demonstrated that the radiopharmaceutical shows favorable characteristics for SPECT imaging and radioguided surgery in PCa patients.
Collapse
Affiliation(s)
- Leonardo Lima Fuscaldi
- Hospital Israelita Albert Einstein, Sao Paulo, Brazil
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, Sao Paulo, Brazil
| | - Danielle Vieira Sobral
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, Sao Paulo, Brazil
| | | | - Fernanda Ferreira Mendonça
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, Sao Paulo, Brazil
| | | | - Carla Salgueiro
- Departamento de Química, Universidad Kennedy, Buenos Aires, Argentina
| | | | | | | | - Luciana Malavolta
- Department of Physiological Sciences, Santa Casa de Sao Paulo School of Medical Sciences, Sao Paulo, Brazil
| | | | - Jorge Mejia
- Hospital Israelita Albert Einstein, Sao Paulo, Brazil
| |
Collapse
|
25
|
van Lith SAM, Pruis IJ, Tolboom N, Snijders TJ, Henssen D, Ter Laan M, Te Dorsthorst M, Leenders WPJ, Gotthardt M, Nagarajah J, Robe PA, De Witt Hamer P, Hendrikse H, Oprea-Lager DE, Yaqub M, Boellaard R, Wesseling P, Balvers RK, Verburg FA, Harteveld AA, Smits M, van den Bent M, van Zanten SEMV, van de Giessen E. PET Imaging and Protein Expression of Prostate-Specific Membrane Antigen in Glioblastoma: A Multicenter Inventory Study. J Nucl Med 2023; 64:1526-1531. [PMID: 37652540 DOI: 10.2967/jnumed.123.265738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/31/2023] [Indexed: 09/02/2023] Open
Abstract
Upregulation of prostate-specific membrane antigen (PSMA) in neovasculature has been described in glioblastoma multiforme (GBM), whereas vasculature in nonaffected brain shows hardly any expression of PSMA. It is unclear whether PSMA-targeting tracer uptake on PET is based on PSMA-specific binding to neovasculature or aspecific uptake in tumor. Here, we quantified uptake of various PSMA-targeting tracers in GBM and correlated this with PSMA expression in tumor biopsy samples from the same patients. Methods: Fourteen patients diagnosed with de novo (n = 8) or recurrent (n = 6) GBM underwent a preoperative PET scan after injection of 1.5 MBq/kg [68Ga]Ga-PSMA-11 (n = 7), 200 MBq of [18F]DCFpyl (n = 3), or 200 MBq of [18F]PSMA-1007 (n = 4). Uptake in tumor and tumor-to-background ratios, with contralateral nonaffected brain as background, were determined. In a subset of patients, PSMA expression levels from different regions in the tumor tissue samples (n = 40), determined using immunohistochemistry (n = 35) or RNA sequencing (n = 13), were correlated with tracer uptake on PET. Results: Moderate to high (SUVmax, 1.3-20.0) heterogeneous uptake was found in all tumors irrespective of the tracer type used. Uptake in nonaffected brain was low, resulting in high tumor-to-background ratios (6.1-359.0) calculated by dividing SUVmax of tumor by SUVmax of background. Immunohistochemistry showed variable PSMA expression on endothelial cells of tumor microvasculature, as well as on dispersed individual cells (of unknown origin), and granular staining of the neuropil. No correlation was found between in vivo uptake and PSMA expression levels (for immunohistochemistry, r = -0.173, P = 0.320; for RNA, r = -0.033, P = 0.915). Conclusion: Our results indicate the potential use of various PSMA-targeting tracers in GBM. However, we found no correlation between PSMA expression levels on immunohistochemistry and uptake intensity on PET. Whether this may be explained by methodologic reasons, such as the inability to measure functionally active PSMA with immunohistochemistry, tracer pharmacokinetics, or the contribution of a disturbed blood-brain barrier to tracer retention, should still be investigated.
Collapse
Affiliation(s)
- Sanne A M van Lith
- Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ilanah J Pruis
- Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Nelleke Tolboom
- Radiology and Nuclear Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Tom J Snijders
- Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Dylan Henssen
- Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mark Ter Laan
- Neurosurgery, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - William P J Leenders
- Biochemistry, Radboud University Medical Center, Nijmegen, The Netherlands
- Predica Diagnostics, Nijmegen, The Netherlands
| | - Martin Gotthardt
- Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - James Nagarajah
- Medical Imaging, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Pierre A Robe
- Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Harry Hendrikse
- Radiology and Nuclear Medicine, Amsterdam UMC, VUmc, Amsterdam, The Netherlands
| | | | - Maqsood Yaqub
- Radiology and Nuclear Medicine, Amsterdam UMC, VUmc, Amsterdam, The Netherlands
| | - Ronald Boellaard
- Radiology and Nuclear Medicine, Amsterdam UMC, VUmc, Amsterdam, The Netherlands
| | - Pieter Wesseling
- Pathology, Amsterdam UMC, VUmc, Amsterdam, The Netherlands
- Pathology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | | | | | - Anita A Harteveld
- Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Marion Smits
- Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
- Medical Delta, Delft, The Netherlands; and
| | - Martin van den Bent
- Brain Tumor Center at Erasmus MC Cancer Institute, Erasmus MC, Rotterdam, The Netherlands
| | | | | |
Collapse
|
26
|
Heilinger J, Weindler J, Roth KS, Krapf P, Schomäcker K, Dietlein M, Drzezga A, Kobe C. Threshold for defining PSMA-positivity prior to 177Lu-PSMA therapy: a comparison of [ 68Ga]Ga-PSMA-11 and [ 18F]F-DCFPyL in metastatic prostate cancer. EJNMMI Res 2023; 13:83. [PMID: 37731097 PMCID: PMC10511392 DOI: 10.1186/s13550-023-01033-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/13/2023] [Indexed: 09/22/2023] Open
Abstract
BACKGROUND In 2022, the American Food and Drug Administration and the European Medicines Agency approved [177Lu]Lu-PSMA-617 (PLUVICTO™, Novartis AG, Basel, Switzerland) for radionuclide therapy with prostate-specific membrane antigen (PSMA) ligands in metastatic prostate cancer. Theranostics require appropriate patients to be identified by positron emission tomography (PET) prior to radionuclide therapy, usually employing [68Ga]Ga-PSMA-11. Alternatively, several 18F-labelled PSMA-PET tracers are available and may increasingly replace 68Ga-labelled compounds, with respect to their image quality, availability and other practical advantages. However, alternative tracers may differ in uptake behaviour, and their comparability with regard to patient selection for [177Lu]Lu-PSMA therapy has not yet been established. Here, we analysed whether tumour-to-background ratios determined by PET using the 18F-labelled PSMA-specific radiopharmaceutical [18F]F-DCFPyL were comparable to those determined by PET using [68Ga]Ga-PSMA-11. RESULTS No differences could be observed between [68Ga]Ga-PSMA-11-PET and [18F]F-DCFPyL-PET regarding tumour-to-liver ratios or tumour-to-mediastinum ratios (e. g. tumour-to-liver ratios using maximum SUV of the tumour lesion for ultra-high definition reconstructed PET images with a median of 2.5 (0.6-9.0) on [68Ga]Ga-PSMA-11-PET vs. 2,0 (0.6-11.4) on [18F]F-DCFPyL-PET). However, significant differences were observed in terms of contrast-to-noise ratios, thereby demonstrating the better image quality obtained with [18F]F-DCFPyL-PET. CONCLUSIONS Our data showed that [18F]F-DCFPyl-PET and [68Ga]Ga-PSMA-11-PET provide comparable tumour-to-liver and tumour-to-mediastinum ratios. Therefore, a tumour uptake of [18F]F-DCFPyL above the liver background, like using [68Ga]Ga-PSMA-11, can be considered as equally suitable for defining PSMA-positivity by a semiquantitative assessment based on the liver background, e. g. prior to radioligand therapy with 177Lu-labelled PSMA ligands. In addition, our data suggest a tending advantage of [18F]F-DCFPyL in terms of lesion detectability.
Collapse
Affiliation(s)
- Jan Heilinger
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Jasmin Weindler
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Katrin Sabine Roth
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Philipp Krapf
- Institute of Neuroscience and Medicine, Nuclear Chemistry (INM-5), Forschungszentrum Jülich GmbH, Wilhelm-Johnen-Straße, 52428, Jülich, Germany
| | - Klaus Schomäcker
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Markus Dietlein
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Alexander Drzezga
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Straße 62, 50937, Cologne, Germany
| | - Carsten Kobe
- Department of Nuclear Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener Straße 62, 50937, Cologne, Germany.
| |
Collapse
|
27
|
Hope TA, Antonarakis ES, Bodei L, Calais J, Iravani A, Jacene H, Koo PJ, Morgans AK, Osborne JR, Tagawa ST, Taplin ME, Sartor O, Morris MJ. SNMMI Consensus Statement on Patient Selection and Appropriate Use of 177Lu-PSMA-617 Radionuclide Therapy. J Nucl Med 2023; 64:1417-1423. [PMID: 37290800 DOI: 10.2967/jnumed.123.265952] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 04/27/2023] [Indexed: 06/10/2023] Open
Affiliation(s)
- Thomas A Hope
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California;
| | | | - Lisa Bodei
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jeremie Calais
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Amir Iravani
- Department of Radiology, University of Washington, Seattle, Washington
| | - Heather Jacene
- Department of Radiology, Brigham and Women's Hospital, and Department of Imaging, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Phillip J Koo
- Banner M.D. Anderson Cancer Center, Phoenix, Arizona
| | - Alicia K Morgans
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Joseph R Osborne
- Molecular Imaging and Therapeutics, Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Scott T Tagawa
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Mary-Ellen Taplin
- Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | | | - Michael J Morris
- Genitourinary Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
28
|
Hotta M, Gafita A, Murthy V, Benz MR, Sonni I, Burger IA, Eiber M, Emmett L, Farolfi A, Fendler WP, Weber MM, Hofman MS, Hope TA, Kratochwil C, Czernin J, Calais J. PSMA PET Tumor-to-Salivary Gland Ratio to Predict Response to [ 177Lu]PSMA Radioligand Therapy: An International Multicenter Retrospective Study. J Nucl Med 2023; 64:1024-1029. [PMID: 36997329 PMCID: PMC11937727 DOI: 10.2967/jnumed.122.265242] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 02/10/2023] [Accepted: 02/10/2023] [Indexed: 04/01/2023] Open
Abstract
Prostate-specific membrane antigen (PSMA)-targeted radioligand therapy can improve the outcome of patients with advanced metastatic castration-resistant prostate cancer, but patients do not respond uniformly. We hypothesized that using the salivary glands as a reference organ can enable selective patient stratification. We aimed to establish a PSMA PET tumor-to-salivary gland ratio (PSG score) to predict outcomes after [177Lu]PSMA. Methods: In total, 237 men with metastatic castration-resistant prostate cancer treated with [177Lu]PSMA were included. A quantitative PSG (qPSG) score (SUVmean ratio of whole-body tumor to parotid glands) was semiautomatically calculated on baseline [68Ga]PSMA-11 PET images. Patients were divided into 3 groups: high (qPSG > 1.5), intermediate (qPSG = 0.5-1.5), and low (qPSG < 0.5) scores. Ten readers interpreted the 3-dimensional maximum-intensity-projection baseline [68Ga]PSMA-11 PET images and classified patients into 3 groups based on visual PSG (vPSG) score: high (most of the lesions showed higher uptake than the parotid glands) intermediate (neither low nor high), and low (most of the lesions showed lower uptake than the parotid glands). Outcome data included a more than 50% prostate-specific antigen decline, prostate-specific antigen (PSA) progression-free survival, and overall survival (OS). Results: Of the 237 patients, the numbers in the high, intermediate, and low groups were 56 (23.6%), 163 (68.8%), and 18 (7.6%), respectively, for qPSG score and 106 (44.7%), 96 (40.5%), and 35 (14.8%), respectively, for vPSG score. The interreader reproducibility of the vPSG score was substantial (Fleiss weighted κ, 0.68). The more than 50% prostate-specific antigen decline was better in patients with a higher PSG score (high vs. intermediate vs. low, 69.6% vs. 38.7% vs. 16.7%, respectively, for qPSG [P < 0.001] and 63.2% vs 33.3% vs 16.1%, respectively, for vPSG [P < 0.001]). The median PSA progression-free survival of the high, intermediate, and low groups by qPSG score was 7.2, 4.0, and 1.9 mo (P < 0.001), respectively, by qPSG score and 6.7, 3.8, and 1.9 mo (P < 0.001), respectively, by vPSG score. The median OS of the high, intermediate, and low groups was 15.0, 11.2, and 13.9 mo (P = 0.017), respectively, by qPSG score and 14.3, 9.6, and 12.9 mo (P = 0.018), respectively, by vPSG score. Conclusion: The PSG score was prognostic for PSA response and OS after [177Lu]PSMA. The visual PSG score assessed on 3-dimensional maximum-intensity-projection PET images yielded substantial reproducibility and comparable prognostic value to the quantitative score.
Collapse
Affiliation(s)
- Masatoshi Hotta
- Ahmanson Translational Theranostics Division, UCLA, Los Angeles, California;
| | - Andrei Gafita
- Ahmanson Translational Theranostics Division, UCLA, Los Angeles, California
| | - Vishnu Murthy
- Ahmanson Translational Theranostics Division, UCLA, Los Angeles, California
| | - Matthias R Benz
- Ahmanson Translational Theranostics Division, UCLA, Los Angeles, California
| | - Ida Sonni
- Ahmanson Translational Theranostics Division, UCLA, Los Angeles, California
| | - Irene A Burger
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Matthias Eiber
- Department of Nuclear Medicine, Technical University Munich, Munich, Germany
| | - Louise Emmett
- Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital, Sydney, New South Wales, Australia
| | - Andrea Farolfi
- Ahmanson Translational Theranostics Division, UCLA, Los Angeles, California
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium-University Hospital Essen, Essen, Germany
| | - Manuel M Weber
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium-University Hospital Essen, Essen, Germany
| | - Michael S Hofman
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Molecular Imaging Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Cancer Centre, and Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Thomas A Hope
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California; and
| | - Clemens Kratochwil
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Johannes Czernin
- Ahmanson Translational Theranostics Division, UCLA, Los Angeles, California
| | - Jeremie Calais
- Ahmanson Translational Theranostics Division, UCLA, Los Angeles, California
| |
Collapse
|
29
|
Wang YF, Lo CY, Chen LY, Chang CW, Huang YT, Huang YY, Huang YH. Comparing the Detection Performance Between Multiparametric Magnetic Resonance Imaging and Prostate-Specific Membrane Antigen PET/CT in Patients With Localized Prostate Cancer: A Systematic Review and Meta-analysis. Clin Nucl Med 2023; 48:e321-e331. [PMID: 37145456 DOI: 10.1097/rlu.0000000000004646] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
PURPOSE Multiparametric MRI (mpMRI) has been promoted as an auxiliary diagnostic tool for prostate biopsy. However, prostate-specific membrane antigen (PSMA) including 68 Ga-PSMA-11, 18 F-DCFPyL, and 18 F-PSMA-1007 applied PET/CT imaging was an emerging diagnostic tool in prostate cancer patients for staging or posttreatment follow-up, even early detecting. Many studies have used PSMA PET for comparison with mpMRI to test the diagnostic ability for early prostate cancer. Unfortunately, these studies have shown conflicting results. This meta-analysis aimed to compare the differences in diagnostic performance between PSMA PET and mpMRI for detecting and T staging localized prostatic tumors. METHODS This meta-analysis involved a systematic literature search of PubMed/MEDLINE and Cochrane Library databases. The pooling sensitivity and specificity of PSMA and mpMRI verified by pathological analysis were calculated and used to compare the differences between the 2 imaging tools. RESULTS Overall, 39 studies were included (3630 patients in total) from 2016 to 2022 in the current meta-analysis and found that the pooling sensitivity values for localized prostatic tumors and T staging T3a and T3b of PSMA PET were 0.84 (95% confidence interval [CI], 0.83-0.86), 0.61 (95% CI, 0.39-0.79), and 0.62 (95% CI, 0.46-0.76), respectively, whereas those of mpMRI were found to be 0.84 (95% 0.78-0.89), 0.67 (95% CI, 0.52-0.80), and 0.60 (95% CI, 0.45-0.73), respectively, without significant differences ( P > 0.05). However, in a subgroup analysis of radiotracer, the pooling sensitivity of 18 F-DCFPyL PET was higher than mpMRI (relative risk, 1.10; 95% CI, 1.03-1.17; P < 0.01). CONCLUSIONS This meta-analysis found that whereas 18 F-DCFPyL PET was superior to mpMRI at detecting localized prostatic tumors, the detection performance of PSMA PET for localized prostatic tumors and T staging was comparable to that of mpMRI.
Collapse
|
30
|
Gutiérrez Cardo AL, Vallejo Casas JA, García Garzón JR, Tirado Hospital JL, Medina López R, Freire Macías JM, Rodríguez Fernández A. 18F-DCFPyL PET/CT guidelines. Rev Esp Med Nucl Imagen Mol 2023; 42:203-208. [PMID: 36878314 DOI: 10.1016/j.remnie.2023.02.009] [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: 01/18/2023] [Revised: 01/29/2023] [Accepted: 01/31/2023] [Indexed: 03/07/2023]
Abstract
The objective of this guide is to provide to nuclear medicine physicians a tool based on scientific evidence and prepared by consensus of experts, to perform the 18F-DCFPyL PET/CT procedure with safely and efficiently for patients with prostate cancer who present PSMA overexpression. For them, some recommendations will be established for 18F-DCFPyL PET/CT examination: reconstruction parameters, presentation of the images and their interpretation. The possible false positives of the procedure will be analysed, how to interpret them and how to avoid them. Finally, all exploration should lead to the preparation of a report that answers the clinician's question. For this, it is recommended to prepare a structured report that includes the PROMISE criteria as well as the classification of the findings according to PSMA-RADS parameters.
Collapse
Affiliation(s)
| | - J A Vallejo Casas
- Servicio de Medicina Nuclear, Hospital Universitario Reina Sofía, Córdoba, Spain
| | | | - J L Tirado Hospital
- Servicio de Medicina Nuclear, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - R Medina López
- Unidad de Nefrourología, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - J M Freire Macías
- Servicio de Medicina Nuclear, Hospital Universitario Puerta del Mar, Cádiz, Spain
| | - A Rodríguez Fernández
- Servicio de Medicina Nuclear, Hospital Universitario Virgen de las Nieves, Granada, Spain.
| |
Collapse
|
31
|
Garje R, Hope TA, Rumble RB, Parikh RA. Systemic Therapy Update on 177Lutetium-PSMA-617 for Metastatic Castration-Resistant Prostate Cancer: ASCO Guideline Rapid Recommendation Q and A. JCO Oncol Pract 2023; 19:132-135. [PMID: 36693228 DOI: 10.1200/op.22.00753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Affiliation(s)
- Rohan Garje
- Miami Cancer Institute, Baptist Health South Florida, Miami, FL
| | - Thomas A Hope
- University of California, San Francisco, San Francisco, CA
| | | | | | | |
Collapse
|
32
|
Gutiérrez Cardo A, Vallejo Casas J, García Garzón J, Tirado Hospital J, Medina López R, Freire Macías J, Rodríguez Fernández A. Guía del procedimiento de la PET/TC con 18F-DCFPyL. Rev Esp Med Nucl Imagen Mol 2023. [DOI: 10.1016/j.remn.2023.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
|
33
|
Haj-Mirzaian A, Mahmood U, Heidari P. Targeted Molecular Imaging as a Biomarker in Urologic Oncology. Urol Clin North Am 2023; 50:115-131. [PMID: 36424076 PMCID: PMC10133841 DOI: 10.1016/j.ucl.2022.09.011] [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] [Indexed: 11/23/2022]
Abstract
Urologic malignancies constitute a large portion of annually diagnosed cancers. Timely diagnosis, accurate staging, and assessment of tumor heterogeneity are essential to devising the best treatment strategy for individual patients. The high sensitivity of molecular imaging allows for early and sensitive detection of lesions that were not readily detectable using conventional imaging techniques. Moreover, molecular imaging enables the interrogation of molecular processes used in targeted cancer therapies and predicts cancer response to treatment. Here we review the current advancements in molecular imaging of urologic cancers, including prostatic, vesical, renal testicular, and ureteral cancers.
Collapse
Affiliation(s)
- Arvin Haj-Mirzaian
- Department of Radiology, Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, 55 Fruit St, Wht 427, Boston, MA 02114, USA; Center for Precision Imaging, Athinoula A Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 55 Fruit St, Wht 427, Boston, MA 02114, USA
| | - Umar Mahmood
- Department of Radiology, Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, 55 Fruit St, Wht 427, Boston, MA 02114, USA; Center for Precision Imaging, Athinoula A Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 55 Fruit St, Wht 427, Boston, MA 02114, USA.
| | - Pedram Heidari
- Department of Radiology, Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, 55 Fruit St, Wht 427, Boston, MA 02114, USA; Center for Precision Imaging, Athinoula A Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 55 Fruit St, Wht 427, Boston, MA 02114, USA
| |
Collapse
|
34
|
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
|
35
|
Prostate-Specific Membrane Antigen Targeted Pet/CT Imaging in Patients with Colon, Gastric and Pancreatic Cancer. Cancers (Basel) 2022; 14:cancers14246209. [PMID: 36551695 PMCID: PMC9777210 DOI: 10.3390/cancers14246209] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/08/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Current imaging modalities frequently misjudge disease stage in colorectal, gastric and pancreatic cancer. As treatment decisions are dependent on disease stage, incorrect staging has serious consequences. Previous preclinical research and case reports indicate that prostate-specific membrane antigen (PSMA)-targeted PET/CT imaging might provide a solution to some of these challenges. This prospective clinical study aims to assess the feasibility of [18F]DCFPyL PET/CT imaging to target and visualize primary colon, gastric and pancreatic cancer. In this prospective clinical trial, patients with colon, gastric and pancreatic cancer were included and underwent both [18F]DCFPyL and [18F]FDG PET/CT scans prior to surgical resection or (for gastric cancer) neoadjuvant therapy. Semiquantitative analysis of immunohistochemical PSMA staining was performed on the surgical resection specimens, and the results were correlated to imaging parameters. The results of this study demonstrate detection of the primary tumor by [18F]DCFPyL PET/CT in 7 out of 10 patients with colon, gastric and pancreatic cancer, with a mean tumor-to-blood pool ratio (TBR) of 3.3 and mean SUVmax of 3.6. However, due to the high surrounding uptake, visual distinction of these tumors was difficult, and the SUVmax and TBR on [18F]FDG PET/CT were significantly higher than on [18F]DCFPyL PET/CT. In addition, no correlation between PSMA expression in the resection specimen and SUVmax on [18F]DCFPyL PET/CT was found. In conclusion, the detection of several gastrointestinal cancers using [18F]DCFPyL PET/CT is feasible. However, low tumor expression and high uptake physiologically in organs/background hamper the clear distinction of the tumor. As a result, [18F]FDG PET/CT was superior in detecting colon, gastric and pancreatic cancers.
Collapse
|
36
|
Reverse Liver Spleen Uptake on [ 68 Ga]Ga-PSMA-11 PET/CT. Clin Nucl Med 2022; 47:1063-1065. [PMID: 35619198 DOI: 10.1097/rlu.0000000000004285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
ABSTRACT An 80-year-old man underwent [ 68 Ga]Ga-PSMA-11 PET/CT for staging of high-risk prostate cancer. Homogeneously increased liver uptake, more than 3-fold the splenic uptake, was seen. There was no hepatic lesion evident on CT. A higher liver to splenic uptake is more typical of some 18 F-labeled PSMA PET/CT but unusual in 68 Ga-labeled PSMA PET/CT scan. Further evaluation revealed a history of impaired renal function, bilateral renal atrophy, relatively decreased renal uptake of [ 68 Ga]Ga-PSMA-11, and prominent bowel activity. We concluded that impaired renal function and subsequent poor excretion resulted in increased hepatic excretion, hence the unusual increased homogeneous hepatic uptake.
Collapse
|
37
|
Preclinical comparative study of [ 18F]AlF-PSMA-11 and [ 18F]PSMA-1007 in varying PSMA expressing tumors. Sci Rep 2022; 12:15744. [PMID: 36130980 PMCID: PMC9492661 DOI: 10.1038/s41598-022-20060-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
A wide variety of 18F-labeled PSMA-targeting PET radiotracers have been developed, including [18F]AlF-PSMA-11. As there is only limited data on the comparison with other 18F-labeled PSMA PET tracers, a comparative preclinical study between [18F]AlF-PSMA-11 and [18F]PSMA-1007 was conducted. Mice with varying PSMA expressing tumors (C4-2, 22Rv1 and PC-3, each n = 5) underwent two PET/CT scans with both [18F]AlF-PSMA-11 and [18F]PSMA-1007. Ten additional mice bearing C4-2 xenografts were subjected to ex vivo biodistribution with either [18F]AlF-PSMA-11 (n = 5) or [18F]PSMA-1007 (n = 5). Absolute SUVmean and SUVmax values were significantly higher for [18F]PSMA-1007 scans in both C4-2 tumors (p < 0.01) and 22Rv1 tumors (p < 0.01). In C4-2 xenograft bearing mice, the tumor-to-organ ratios did not significantly differ between [18F]AlF-PSMA-11 and [18F]PSMA-1007 for liver, muscle, blood and salivary glands (p > 0.05). However, in 22Rv1 xenograft bearing mice, all tumor-to-organ ratios were higher for [18F]AlF-PSMA-11 (p < 0.01). In healthy organs, [18F]PSMA-1007 uptake was higher in the liver, gallbladder, small intestines and glands. Biodistribution data confirmed the increased uptake in the heart, small intestines and liver with [18F]PSMA-1007. Absolute tumor uptake was higher with [18F]PSMA-1007 in all tumors. Tumor-to-organ ratios did not differ significantly in high PSMA expressing tumors, but were higher for [18F]AlF-PSMA-11 in low PSMA expressing tumors. Furthermore, [18F]PSMA-1007 showed higher uptake in healthy organs.
Collapse
|
38
|
Donswijk ML, Wondergem M, de Wit-van der Veen L, Bruin NM, van Leeuwen PJ, van der Poel HG, Stokkel MPM, Vogel WV. Effects of furosemide and tracer selection on urinary activity and peri-bladder artefacts in PSMA PET/CT: a single-centre retrospective study. EJNMMI Res 2022; 12:42. [PMID: 35895129 PMCID: PMC9329505 DOI: 10.1186/s13550-022-00913-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/09/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND High urinary activity in urinary bladder and ureters may hamper interpretation of prostate cancer and regional nodal metastases in prostate-specific membrane antigen (PSMA) PET/CT. The goal of this study was to assess effects of furosemide and choice of tracer on urinary activity in the bladder and ureters, as well as on occurrence of peri-bladder artefacts in PET/CT. METHODS Four cohorts with a total of 202 men staged with PSMA PET/CT for prostate cancer received either 68Ga-PSMA-11 as tracer, with (cohort G+) or without 10mg intravenous furosemide (G-) concurrent with tracer, or 18F-DCFPyL with (F+) or without furosemide (F-). SUVmax of bladder and ureters, presence, type, and severity of peri-bladder artefacts were compared between cohorts. The influence of furosemide and choice of tracer was determined while taking differences in biodistribution time into account. RESULTS Median SUVmax bladder was 43,5; 14,8; 61,7 and 22,8 in cohorts G-, G+, F- and F+, respectively, resulting in significant overall (p < 0.001) and between cohort differences (p adjusted < 0.001 to 0.003) except between G- and F+. Median SUVmax ureter was 6.4; 4.5; 8.1 and 6.0 in cohorts G-, G+, F- and F+, respectively, resulting in significant overall (p < 0.001) and between cohort differences for G+ : F- and F- : F+ (p < 0.001, respectively, 0.019). Significant effects of furosemide and choice of tracer on SUVmax bladder (p < 0.001 resp. p = 0.001) and of furosemide on SUVmax ureter (p < 0.001) were found, whereas differences in biodistribution time had not impacted these results significantly. Peri-bladder artefacts were present in 42/202 (21%) patients and were significantly more frequent in the F- cohort, respectively, less frequent in the G+ cohort (p = 0.001 resp. p < 0.001). Peri-bladder artefacts had a direct positive correlation with SUVmax bladder (p = 0.033). CONCLUSIONS Increased urinary activity and higher incidence of peri-bladder artefacts were found in 18F-DCFPyL compared to 68Ga-PSMA-11 PET/CT. Effective reduction of urinary activity may be reached through forced diuresis using 10mg intravenous furosemide, which is especially advantageous in 18F-DCFPyL PET/CT.
Collapse
Affiliation(s)
- Maarten L Donswijk
- Department of Nuclear Medicine, Antoni van Leeuwenhoek Nederlands Kanker Instituut, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands.
| | - Maurits Wondergem
- Department of Nuclear Medicine, Antoni van Leeuwenhoek Nederlands Kanker Instituut, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands
| | - Linda de Wit-van der Veen
- Department of Nuclear Medicine, Antoni van Leeuwenhoek Nederlands Kanker Instituut, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands
| | - Natascha M Bruin
- Department of Nuclear Medicine, Antoni van Leeuwenhoek Nederlands Kanker Instituut, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands.,Department of Radiation Oncology, Antoni van Leeuwenhoek Nederlands Kanker Instituut, Amsterdam, The Netherlands
| | - Pim J van Leeuwen
- Department of Urology, Antoni van Leeuwenhoek Nederlands Kanker Instituut, Prostate Cancer Network Amsterdam, Amsterdam, The Netherlands
| | - Henk G van der Poel
- Department of Urology, Antoni van Leeuwenhoek Nederlands Kanker Instituut, Prostate Cancer Network Amsterdam, Amsterdam, The Netherlands.,Department of Urology, Amsterdam University Medical Center, Prostate Cancer Network Amsterdam, VU University, Amsterdam, The Netherlands
| | - Marcel P M Stokkel
- Department of Nuclear Medicine, Antoni van Leeuwenhoek Nederlands Kanker Instituut, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands
| | - Wouter V Vogel
- Department of Nuclear Medicine, Antoni van Leeuwenhoek Nederlands Kanker Instituut, Plesmanlaan 121, 1066 CX, Amsterdam, the Netherlands.,Department of Radiation Oncology, Antoni van Leeuwenhoek Nederlands Kanker Instituut, Amsterdam, The Netherlands
| |
Collapse
|
39
|
Gillessen S, Armstrong A, Attard G, Beer TM, Beltran H, Bjartell A, Bossi A, Briganti A, Bristow RG, Bulbul M, Caffo O, Chi KN, Clarke CS, Clarke N, Davis ID, de Bono JS, Duran I, Eeles R, Efstathiou E, Efstathiou J, Ekeke ON, Evans CP, Fanti S, Feng FY, Fizazi K, Frydenberg M, George D, Gleave M, Halabi S, Heinrich D, Higano C, Hofman MS, Hussain M, James N, Jones R, Kanesvaran R, Khauli RB, Klotz L, Leibowitz R, Logothetis C, Maluf F, Millman R, Morgans AK, Morris MJ, Mottet N, Mrabti H, Murphy DG, Murthy V, Oh WK, Ost P, O'Sullivan JM, Padhani AR, Parker C, Poon DMC, Pritchard CC, Rabah DM, Rathkopf D, Reiter RE, Rubin M, Ryan CJ, Saad F, Sade JP, Sartor O, Scher HI, Shore N, Skoneczna I, Small E, Smith M, Soule H, Spratt DE, Sternberg CN, Suzuki H, Sweeney C, Sydes MR, Taplin ME, Tilki D, Tombal B, Türkeri L, Uemura H, Uemura H, van Oort I, Yamoah K, Ye D, Zapatero A, Omlin A. Management of Patients with Advanced Prostate Cancer: Report from the Advanced Prostate Cancer Consensus Conference 2021. Eur Urol 2022; 82:115-141. [PMID: 35450732 DOI: 10.1016/j.eururo.2022.04.002] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/01/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Innovations in treatments, imaging, and molecular characterisation in advanced prostate cancer have improved outcomes, but various areas of management still lack high-level evidence to inform clinical practice. The 2021 Advanced Prostate Cancer Consensus Conference (APCCC) addressed some of these questions to supplement guidelines that are based on level 1 evidence. OBJECTIVE To present the voting results from APCCC 2021. DESIGN, SETTING, AND PARTICIPANTS The experts identified three major areas of controversy related to management of advanced prostate cancer: newly diagnosed metastatic hormone-sensitive prostate cancer (mHSPC), the use of prostate-specific membrane antigen ligands in diagnostics and therapy, and molecular characterisation of tissue and blood. A panel of 86 international prostate cancer experts developed the programme and the consensus questions. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS The panel voted publicly but anonymously on 107 pre-defined questions, which were developed by both voting and non-voting panel members prior to the conference following a modified Delphi process. RESULTS AND LIMITATIONS The voting reflected the opinions of panellists and did not incorporate a standard literature review or formal meta-analysis. The answer options for the consensus questions received varying degrees of support from panellists, as reflected in this article and the detailed voting results reported in the Supplementary material. CONCLUSIONS These voting results from a panel of experts in advanced prostate cancer can help clinicians and patients to navigate controversial areas of management for which high-level evidence is scant. However, diagnostic and treatment decisions should always be individualised according to patient characteristics, such as the extent and location of disease, prior treatment(s), comorbidities, patient preferences, and treatment recommendations, and should also incorporate current and emerging clinical evidence and logistic and economic constraints. Enrolment in clinical trials should be strongly encouraged. Importantly, APCCC 2021 once again identified salient questions that merit evaluation in specifically designed trials. PATIENT SUMMARY The Advanced Prostate Cancer Consensus Conference is a forum for discussing current diagnosis and treatment options for patients with advanced prostate cancer. An expert panel votes on predefined questions focused on the most clinically relevant areas for treatment of advanced prostate cancer for which there are gaps in knowledge. The voting results provide a practical guide to help clinicians in discussing treatment options with patients as part of shared decision-making.
Collapse
Affiliation(s)
- Silke Gillessen
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland; Universita della Svizzera Italiana, Lugano, Switzerland; University of Berne, Berne, Switzerland; Division of Cancer Sciences, University of Manchester, Manchester, UK.
| | - Andrew Armstrong
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, USA
| | - Gert Attard
- University College London Cancer Institute, London, UK
| | - Tomasz M Beer
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Himisha Beltran
- Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Anders Bjartell
- Department of Urology, Skåne University Hospital, Malmö, Sweden
| | - Alberto Bossi
- Genitourinary Oncology, Prostate Brachytherapy Unit, Gustave Roussy, Paris, France
| | - Alberto Briganti
- Unit of Urology/Division of Oncology, Urological Research Institute, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, Milan, Italy
| | - Robert G Bristow
- Division of Cancer Sciences, University of Manchester, Manchester, UK; Christie NHS Trust and CRUK Manchester Institute and Cancer Centre, Manchester, UK
| | - Muhammad Bulbul
- Division of Urology, Department of Surgery, American University of Beirut Medical Center, Beirut, Lebanon
| | - Orazio Caffo
- Department of Medical Oncology, Santa Chiara Hospital, Trento, Italy
| | - Kim N Chi
- BC Cancer, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Caroline S Clarke
- Research Department of Primary Care & Population Health, Royal Free Campus, University College London, London, UK
| | - Noel Clarke
- The Christie and Salford Royal Hospitals, Manchester, UK
| | - Ian D Davis
- Monash University and Eastern Health, Victoria, Australia
| | - Johann S de Bono
- The Institute of Cancer Research, Royal Marsden NHS Foundation Trust, Sutton, UK
| | - Ignacio Duran
- Department of Medical Oncology, Hospital Universitario Marques de Valdecilla, IDIVAL, Santander, Spain
| | - Ros Eeles
- The Institute of Cancer Research, Royal Marsden NHS Foundation Trust, Sutton, UK
| | | | - Jason Efstathiou
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, MA, USA
| | - Onyeanunam Ngozi Ekeke
- Department of Surgery, University of Port Harcourt Teaching Hospital, Port Harcourt, Nigeria
| | | | - Stefano Fanti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Felix Y Feng
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California-San Francisco, San Francisco, CA, USA
| | - Karim Fizazi
- Institut Gustave Roussy, University of Paris Saclay, Villejuif, France
| | - Mark Frydenberg
- Department of Surgery, Prostate Cancer Research Program, Monash University, Melbourne, Australia
| | - Dan George
- Departments of Medicine and Surgery, Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Martin Gleave
- Urological Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, Canada
| | - Susan Halabi
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Daniel Heinrich
- Department of Oncology and Radiotherapy, Innlandet Hospital Trust, Gjøvik, Norway
| | | | - Michael S Hofman
- Prostate Cancer Theranostics and Imaging Centre of Excellence, Department of Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre and Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Maha Hussain
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Nick James
- The Institute of Cancer Research, Royal Marsden NHS Foundation Trust, Sutton, UK
| | - Robert Jones
- Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | | | - Raja B Khauli
- Department of Urology and the Naef K. Basile Cancer Institute, American University of Beirut Medical Center, Beirut, Lebanon
| | - Laurence Klotz
- Division of Urology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Raya Leibowitz
- Oncology Institute, Shamir Medical Center and Faculty of Medicine, Tel-Aviv University, Tel Aviv, Israel
| | - Chris Logothetis
- Department of Genitourinary Medical Oncology, David H. Koch Centre, MD Anderson Cancer Centre, Houston, TX, USA; Department of Clinical Therapeutics, University of Athens Alexandra Hospital, Athens, Greece
| | - Fernando Maluf
- Beneficiência Portuguesa de São Paulo, São Paulo, SP, Brazil; Departamento de Oncologia, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | | | - Alicia K Morgans
- Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael J Morris
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Hind Mrabti
- National Institute of Oncology, Mohamed V University, Rabat, Morocco
| | - Declan G Murphy
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | | | - William K Oh
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, The Tisch Cancer Institute, New York, NY, USA
| | - Piet Ost
- Department of Radiation Oncology, Iridium Netwerk, Antwerp, Belgium; Department of Human Structure and Repair, Ghent University, Ghent, Belgium
| | - Joe M O'Sullivan
- Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Northern Ireland Cancer Centre, Belfast City Hospital, Belfast, UK
| | - Anwar R Padhani
- The Institute of Cancer Research, Royal Marsden NHS Foundation Trust, Sutton, UK; Mount Vernon Cancer Centre, London, UK
| | - Chris Parker
- The Institute of Cancer Research, Royal Marsden NHS Foundation Trust, Sutton, UK
| | - Darren M C Poon
- Comprehensive Oncology Centre, Hong Kong Sanatorium & Hospital, The Chinese University of Hong Kong, Hong Kong
| | - Colin C Pritchard
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Danny M Rabah
- The Cancer Research Chair, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Dana Rathkopf
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rob E Reiter
- University of California-Los Angeles, Los Angeles, CA, USA
| | - Mark Rubin
- Bern Center for Precision Medicine and Department for Biomedical Research, Bern, Switzerland
| | - Charles J Ryan
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Fred Saad
- Centre Hospitalier de Université de Montréal, Montreal, Canada
| | - Juan P Sade
- Instituto Alexander Fleming, Buenos Aires, Argentina
| | | | - Howard I Scher
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Neal Shore
- Carolina Urologic Research Center, Myrtle Beach, SC, USA
| | - Iwona Skoneczna
- Rafal Masztak Grochowski Hospital and Maria Sklodowska Curie National Research Institute of Oncology, Warsaw, Poland
| | - Eric Small
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California-San Francisco, San Francisco, CA, USA
| | - Matthew Smith
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Howard Soule
- Prostate Cancer Foundation, Santa Monica, CA, USA
| | - Daniel E Spratt
- University Hospitals Seidman Cancer Center, Cleveland, OH, USA
| | - Cora N Sternberg
- Englander Institute for Precision Medicine, Weill Cornell Medicine Division of Hematology and Oncology, Meyer Cancer Center, New York Presbyterian Hospital, New York, NY, USA
| | | | - Christopher Sweeney
- Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Matthew R Sydes
- MRC Clinical Trials Unit, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Mary-Ellen Taplin
- Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Derya Tilki
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Department of Urology, Koc University Hospital, Istanbul, Turkey
| | | | - Levent Türkeri
- Department of Urology, M.A. Aydınlar Acıbadem University, Altunizade Hospital, Istanbul, Turkey
| | - Hiroji Uemura
- Yokohama City University Medical Center, Yokohama, Japan
| | - Hirotsugu Uemura
- Department of Urology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Inge van Oort
- Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kosj Yamoah
- Department of Radiation Oncology & Cancer Epidemiology, H. Lee Moffitt Cancer Center & Research Institute, University of South Florida, Tampa, FL, USA
| | - Dingwei Ye
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Almudena Zapatero
- Department of Radiation Oncology, Hospital Universitario de La Princesa, Health Research Institute, Madrid, Spain
| | - Aurelius Omlin
- Department of Medical Oncology and Haematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland
| |
Collapse
|
40
|
Eshghi A, Covington MF, Eshghi N, Kuo PH. Utility of PET to Appropriately Select Patients for PSMA-Targeted Theranostics. Clin Nucl Med 2022; 47:488-495. [PMID: 35384904 DOI: 10.1097/rlu.0000000000004196] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
ABSTRACT The majority of aggressive prostate cancers overexpress the transmembrane protein prostate-specific membrane antigen (PSMA). PSMA is, therefore, an attractive target for drug development. Over the last decade, numerous PSMA-targeted radiopharmaceuticals for imaging and therapy have been developed and investigated in theranostic combination. PSMA-targeted radiopharmaceuticals for imaging have been primarily developed for PET. PSMA PET provides whole-body evaluation of the degree of PSMA expression on tumors and potentially provides a method to better select patients for PSMA-targeted therapy. Numerous PSMA-targeted therapeutic agents using β- or α-particle emitters are under study in clinical trials. In particular, the β-particle-emitting radioisotope 177Lu bound to PSMA-targeted small molecules have ongoing and completed late-stage clinical trials in metastatic castration-resistant prostate cancer. To define the most appropriate patient group for PSMA-targeted therapeutics, multiple studies have investigated PSMA and FDG PET/CT to establish PET parameters as predictive and prognostic biomarkers. This article discusses recent clinical trials that examine the optimal use of PET for the selection of patients for PSMA-targeted therapeutics and provides an integrative overview of choice of PET tracer(s), targeting molecule, therapeutic radioisotope, nonradioactive therapy, and cancer type (prostate or nonprostate).
Collapse
Affiliation(s)
- Anna Eshghi
- From the College of Osteopathic Medicine, Kansas City University, Joplin, MO
| | - Matthew F Covington
- Department of Radiology and Imaging Sciences, University of Utah, Center for Quantitative Cancer Imaging, Huntsman Cancer Institute, Salt Lake City, UT
| | - Naghmehossadat Eshghi
- Department of Radiology, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Phillip H Kuo
- Departments of Medical Imaging, Medicine, and Biomedical Engineering, University of Arizona, Tucson, AZ
| |
Collapse
|
41
|
Liu Y, Zhang X, Liu J, Zhang J, Xu B. Prospective intraindividual comparison of 18F-PSMA-7Q and 18F-DCFPyL PET/CT in patients with newly diagnosed prostate cancer. Nucl Med Commun 2022; 43:725-730. [PMID: 35560134 DOI: 10.1097/mnm.0000000000001564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Fluorine 18 (18F)-2-(3-{1-Carboxy-5-[(6-[(18)F]fluoro-pyridine-3-carbonyl)-amino]-pentyl}-ureido)-pentanedioic acid (DCFPyL) is an early 18F-labeled prostate-specific membrane antigen (PSMA) targeted PET tracer that has shown promise in the diagnostic workup of prostate cancer and was recently approved by the US Food and Drug Administration. 18F-PSMA-7Q is a novel 18F-labeled PSMA-ligand PET tracer designed and synthesized by our team. This study compared the tracer-specific positron emission tomography/computed tomography (PET/CT) characteristics of 18F-PSMA-7Q with those of 18F-DCFPyL in patients with newly diagnosed prostate cancer. METHODS Ten patients received similar doses of 18F-DCFPyL and 18F-PSMA-7Q 48 h apart and were imaged 1 h after injection on the same PET/CT scanner. Normal-organ biodistribution and tumor uptake were quantified using maximum and mean standardized uptake values (SUVmax and SUVmean), and all lesions were assigned a molecular imaging PSMA (miPSMA) score based on Prostate Cancer Molecular Imaging Standardized Evaluation criteria. RESULTS Seventeen lesions were detected in the 10 patients by both 18F-DCFPyL and 18F-PSMA-7Q. No statistically significant difference was observed when comparing the SUVmax and SUVmean of 18F-DCFPyL and 18F-PSMA-7Q in the lesions and parotid gland. The κ value for the miPSMA scores of the lesions between the two tracers was 0.907, indicating excellent agreement. CONCLUSION 18F-PSMA-7Q can be used in clinical research as reliably as 18F-DCFPyL. The limited urinary excretion of 18F-PSMA-7Q may represent a potential advantage over 18F-DCFPyL for detection of lesions in the pelvis, which need to be verified by further studies.
Collapse
Affiliation(s)
- Yachao Liu
- Department of Nuclear Medicine, Chinese PLA General Hospital, Beijing, China
| | | | | | | | | |
Collapse
|
42
|
Voter AF, Werner RA, Pienta KJ, Gorin MA, Pomper MG, Solnes LB, Rowe SP. Piflufolastat F-18 ( 18F-DCFPyL) for PSMA PET imaging in prostate cancer. Expert Rev Anticancer Ther 2022; 22:681-694. [DOI: 10.1080/14737140.2022.2081155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Andrew F. Voter
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Transitional Year Residency Program, Aurora St. Luke’s Medical Center, Advocate Aurora Health, Milwaukee, WI, USA
| | - Rudolf A. Werner
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Kenneth J. Pienta
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael A. Gorin
- Urology Associates and UPMC Western Maryland, Cumberland, MD, USA
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Martin G. Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lilja B. Solnes
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - 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
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
43
|
Basuli F, Phelps TE, Zhang X, Woodroofe CC, Roy J, Choyke PL, Swenson RE, Jagoda EM. Fluorine-18 Labeled Urea-Based Ligands Targeting Prostate-Specific Membrane Antigen (PSMA) with Increased Tumor and Decreased Renal Uptake. Pharmaceuticals (Basel) 2022; 15:597. [PMID: 35631423 PMCID: PMC9144807 DOI: 10.3390/ph15050597] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/07/2022] [Accepted: 05/11/2022] [Indexed: 12/22/2022] Open
Abstract
High expression of prostate-specific membrane antigen (PSMA) in prostate cancers prompted the development of the PSMA-targeted PET-imaging agent [18F]DCFPyL, which was recently approved by the FDA. Fluorine-18-labeled Lys-Urea-Glu-based oxime derivatives of [18F]DCFPyL were prepared for the comparison of their in vitro and in vivo properties to potentially improve kidney clearance and tumor targeting. The oxime radiotracers were produced by condensation of an aminooxy functionalized PSMA-inhibitor Lys-Urea-Glu scaffold with fluorine-18-labeled aldehydes. The radiochemical yields were between 15-42% (decay uncorrected) in 50-60 min. In vitro saturation and competition binding assays with human prostate cancer cells transfected with PSMA, PC3(+), indicated similar high nM binding affinities to PSMA for all radiotracers. In vivo biodistribution studies with positive control PC3(+) tumor xenografts showed that the kidneys had the highest uptake followed by tumors at 60 min. The PC3(+) tumor uptake was blocked with non-radioactive DCFPyL, and PC3(-) tumor xenograft (negative control) tumor uptake was negligible indicating that PSMA targeting was preserved. The most lipophilic tracer, [18F]2a, displayed comparable tumor-targeting to [18F]DCFPyL and a desirable alteration in pharmacokinetics and metabolism, resulting in significantly lower kidney uptake with a shift towards hepatobiliary clearance and increased liver uptake.
Collapse
Affiliation(s)
- Falguni Basuli
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; (X.Z.); (C.C.W.); (R.E.S.)
| | - Tim E. Phelps
- Molecular Imaging Branch, National Cancer Institute, Bethesda, MD 20892, USA; (T.E.P.); (J.R.); (P.L.C.); (E.M.J.)
| | - Xiang Zhang
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; (X.Z.); (C.C.W.); (R.E.S.)
| | - Carolyn C. Woodroofe
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; (X.Z.); (C.C.W.); (R.E.S.)
| | - Jyoti Roy
- Molecular Imaging Branch, National Cancer Institute, Bethesda, MD 20892, USA; (T.E.P.); (J.R.); (P.L.C.); (E.M.J.)
| | - Peter L. Choyke
- Molecular Imaging Branch, National Cancer Institute, Bethesda, MD 20892, USA; (T.E.P.); (J.R.); (P.L.C.); (E.M.J.)
| | - Rolf E. Swenson
- Chemistry and Synthesis Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; (X.Z.); (C.C.W.); (R.E.S.)
| | - Elaine M. Jagoda
- Molecular Imaging Branch, National Cancer Institute, Bethesda, MD 20892, USA; (T.E.P.); (J.R.); (P.L.C.); (E.M.J.)
| |
Collapse
|
44
|
Fedrigo R, Kadrmas DJ, Edem PE, Fougner L, Klyuzhin IS, Petric MP, Bénard F, Rahmim A, Uribe C. Quantitative evaluation of PSMA PET imaging using a realistic anthropomorphic phantom and shell-less radioactive epoxy lesions. EJNMMI Phys 2022; 9:2. [PMID: 35032234 PMCID: PMC8761183 DOI: 10.1186/s40658-021-00429-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 12/20/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Positron emission tomography (PET) with prostate specific membrane antigen (PSMA) have shown superior performance in detecting metastatic prostate cancers. Relative to [18F]fluorodeoxyglucose ([18F]FDG) PET images, PSMA PET images tend to visualize significantly higher-contrast focal lesions. We aim to evaluate segmentation and reconstruction algorithms in this emerging context. Specifically, Bayesian or maximum a posteriori (MAP) image reconstruction, compared to standard ordered subsets expectation maximization (OSEM) reconstruction, has received significant interest for its potential to reach convergence with minimal noise amplifications. However, few phantom studies have evaluated the quantitative accuracy of such reconstructions for high contrast, small lesions (sub-10 mm) that are typically observed in PSMA images. In this study, we cast 3 mm-16-mm spheres using epoxy resin infused with a long half-life positron emitter (sodium-22; 22Na) to simulate prostate cancer metastasis. The anthropomorphic Probe-IQ phantom, which features a liver, bladder, lungs, and ureters, was used to model relevant anatomy. Dynamic PET acquisitions were acquired and images were reconstructed with OSEM (varying subsets and iterations) and BSREM (varying β parameters), and the effects on lesion quantitation were evaluated. RESULTS The 22Na lesions were scanned against an aqueous solution containing fluorine-18 (18F) as the background. Regions-of-interest were drawn with MIM Software using 40% fixed threshold (40% FT) and a gradient segmentation algorithm (MIM's PET Edge+). Recovery coefficients (RCs) (max, mean, peak, and newly defined "apex"), metabolic tumour volume (MTV), and total tumour uptake (TTU) were calculated for each sphere. SUVpeak and SUVapex had the most consistent RCs for different lesion-to-background ratios and reconstruction parameters. The gradient-based segmentation algorithm was more accurate than 40% FT for determining MTV and TTU, particularly for lesions [Formula: see text] 6 mm in diameter (R2 = 0.979-0.996 vs. R2 = 0.115-0.527, respectively). CONCLUSION An anthropomorphic phantom was used to evaluate quantitation for PSMA PET imaging of metastatic prostate cancer lesions. BSREM with β = 200-400 and OSEM with 2-5 iterations resulted in the most accurate and robust measurements of SUVmean, MTV, and TTU for imaging conditions in 18F-PSMA PET/CT images. SUVapex, a hybrid metric of SUVmax and SUVpeak, was proposed for robust, accurate, and segmentation-free quantitation of lesions for PSMA PET.
Collapse
Affiliation(s)
- Roberto Fedrigo
- Department of Integrative Oncology, BC Cancer Research Institute, 675 W 10th Avenue, Vancouver, BC, V5Z1L3, Canada
- Department of Physics and Astronomy, University of British Columbia, 325-6224 Agricultural Road, Vancouver, BC, V6T1Z1, Canada
| | - Dan J Kadrmas
- Department of Radiology and Imaging Sciences, University of Utah, 201 Presidents' Cir, Salt Lake City, UT, 84112, USA
| | - Patricia E Edem
- Functional Imaging, BC Cancer, 600 W 10th Avenue, Vancouver, BC, V5Z4E6, Canada
| | - Lauren Fougner
- Functional Imaging, BC Cancer, 600 W 10th Avenue, Vancouver, BC, V5Z4E6, Canada
| | - Ivan S Klyuzhin
- Department of Integrative Oncology, BC Cancer Research Institute, 675 W 10th Avenue, Vancouver, BC, V5Z1L3, Canada
- Department of Physics and Astronomy, University of British Columbia, 325-6224 Agricultural Road, Vancouver, BC, V6T1Z1, Canada
| | - M Peter Petric
- Functional Imaging, BC Cancer, 600 W 10th Avenue, Vancouver, BC, V5Z4E6, Canada
| | - François Bénard
- Department of Integrative Oncology, BC Cancer Research Institute, 675 W 10th Avenue, Vancouver, BC, V5Z1L3, Canada
- Department of Physics and Astronomy, University of British Columbia, 325-6224 Agricultural Road, Vancouver, BC, V6T1Z1, Canada
- Department of Molecular Oncology, BC Cancer Research Institute, 675 W 10th Avenue, Vancouver, BC, V5Z1L3, Canada
| | - Arman Rahmim
- Department of Integrative Oncology, BC Cancer Research Institute, 675 W 10th Avenue, Vancouver, BC, V5Z1L3, Canada
- Department of Physics and Astronomy, University of British Columbia, 325-6224 Agricultural Road, Vancouver, BC, V6T1Z1, Canada
- Department of Radiology, University of British Columbia, 675 W 10th Avenue, Vancouver, BC, V5Z1L3, Canada
| | - Carlos Uribe
- Functional Imaging, BC Cancer, 600 W 10th Avenue, Vancouver, BC, V5Z4E6, Canada.
- Department of Radiology, University of British Columbia, 675 W 10th Avenue, Vancouver, BC, V5Z1L3, Canada.
| |
Collapse
|
45
|
Emmett L. Side effects of therapy with radiolabelled prostate specific membrane antigen (PSMA). Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00150-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
|
46
|
Gühne F, Radke S, Winkens T, Kühnel C, Greiser J, Seifert P, Drescher R, Freesmeyer M. Differences in Distribution and Detection Rate of the [ 68Ga]Ga-PSMA Ligands PSMA-617, -I&T and -11-Inter-Individual Comparison in Patients with Biochemical Relapse of Prostate Cancer. Pharmaceuticals (Basel) 2021; 15:ph15010009. [PMID: 35056066 PMCID: PMC8779232 DOI: 10.3390/ph15010009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/04/2022] Open
Abstract
The biochemical relapse of prostate cancer is diagnostically challenging but of high clinical impact for subsequent patient treatment. PET/CT with radiolabeled PSMA ligands outperforms conventional diagnostic methods in the detection of tumor recurrence. Several radiopharmaceuticals were and are available for use. The aim of this study was to investigate whether the routinely applied [68Ga]Ga-PSMA ligands PSMA-617, -I&T and -11 (HBED-CC) differ in physiological and pathological distribution, or in tumor detection rate. A retrospective evaluation of 190 patients (39 patients received PSMA-617, 68 patients PSMA-I&T and 83 patients PSMA-11) showed significant differences in tracer accumulation within all organs examined. The low retention within the compartments blood pool, bone and muscle tissue is a theoretical advantage of PSMA-11. Evaluation of tumor lesion uptake and detection rate did not reveal superiority of one of the three radiopharmaceuticals, neither in the whole population, nor in particularly challenging subgroups like patients with very low PSA levels. We conclude that all three [68Ga]Ga-PSMA ligands are equally feasible in this clinically important scenario, and may replace each other in case of unavailability or production restrictions.
Collapse
|
47
|
Kiraga Ł, Kucharzewska P, Paisey S, Cheda Ł, Domańska A, Rogulski Z, Rygiel TP, Boffi A, Król M. Nuclear imaging for immune cell tracking in vivo – Comparison of various cell labeling methods and their application. Coord Chem Rev 2021. [DOI: 10.1016/j.ccr.2021.214008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
48
|
Abstract
The role of PET imaging with 11C-choline and 18F-fluciclovine in evaluating patients with prostate cancer (PCa) has become more important over the years and has been incorporated into the NCCN guidelines. A new generation of PET radiotracers targeting the prostate-specific membrane antigen (PSMA) is widely used outside the United States to evaluate patients with primary PCa and PCa recurrence. PET imaging influences treatment planning and demonstrates a significantly higher disease detection rate than conventional imaging such as computed tomography and MR imaging. Early data indicate that using PET radiotracers such as 18F-fluciclovine and PSMA improves patient outcomes. 68-Ga-PSMA-11 and 18F-DCFPyL-PET/CT were recently approved by the US Food & Drug Administration (FDA) for clinical use. Other PSMA radiotracers, including fluorinated variants, will likely gain FDA approval in the not-too-distant future.
Collapse
|
49
|
[ 68Ga]Ga-PSMA-11: The First FDA-Approved 68Ga-Radiopharmaceutical for PET Imaging of Prostate Cancer. Pharmaceuticals (Basel) 2021; 14:ph14080713. [PMID: 34451810 PMCID: PMC8401928 DOI: 10.3390/ph14080713] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/15/2021] [Accepted: 07/17/2021] [Indexed: 12/12/2022] Open
Abstract
For the positron emission tomography (PET) imaging of prostate cancer, radiotracers targeting the prostate-specific membrane antigen (PSMA) are nowadays used in clinical practice. Almost 10 years after its discovery, [68Ga]Ga-PSMA-11 has been approved in the United States by the Food and Drug Administration (FDA) as the first 68Ga-radiopharmaceutical for the PET imaging of PSMA-positive prostate cancer in 2020. This radiopharmaceutical combines the peptidomimetic Glu-NH-CO-NH-Lys(Ahx)-HBED-CC with the radionuclide 68Ga, enabling specific imaging of tumor cells expressing PSMA. Such a targeting approach may also be used for therapy planning as well as potentially for the evaluation of treatment response.
Collapse
|
50
|
Abstract
Piflufolastat F 18 (PYLARIFY®) is an 18F-labelled diagnostic imaging agent that has been developed by Progenics Pharmaceuticals Inc., a Lantheus company, for positron emission tomography (PET) that targets prostate-specific membrane antigen (PSMA). Piflufolastat F 18 was approved in the USA on 27 May 2021 for PET of PSMA positive lesions in men with prostate cancer with suspected metastasis who are candidates for initial definitive therapy or with suspected recurrence based on elevated serum prostate specific antigen (PSA) level. This article summarizes the milestones in the development of piflufolastat F 18 leading to this approval as a radioactive diagnostic agent in prostate cancer.
Collapse
Affiliation(s)
- Susan J Keam
- Springer Nature, Private Bag 65901, Mairangi Bay, Auckland, 0754, New Zealand.
| |
Collapse
|