1
|
Zhang S, Wang X, Gao X, Chen X, Li L, Li G, Liu C, Miao Y, Wang R, Hu K. Radiopharmaceuticals and their applications in medicine. Signal Transduct Target Ther 2025; 10:1. [PMID: 39747850 PMCID: PMC11697352 DOI: 10.1038/s41392-024-02041-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/30/2024] [Accepted: 10/28/2024] [Indexed: 01/04/2025] Open
Abstract
Radiopharmaceuticals involve the local delivery of radionuclides to targeted lesions for the diagnosis and treatment of multiple diseases. Radiopharmaceutical therapy, which directly causes systematic and irreparable damage to targeted cells, has attracted increasing attention in the treatment of refractory diseases that are not sensitive to current therapies. As the Food and Drug Administration (FDA) approvals of [177Lu]Lu-DOTA-TATE, [177Lu]Lu-PSMA-617 and their complementary diagnostic agents, namely, [68Ga]Ga-DOTA-TATE and [68Ga]Ga-PSMA-11, targeted radiopharmaceutical-based theranostics (radiotheranostics) are being increasingly implemented in clinical practice in oncology, which lead to a new era of radiopharmaceuticals. The new generation of radiopharmaceuticals utilizes a targeting vector to achieve the accurate delivery of radionuclides to lesions and avoid off-target deposition, making it possible to improve the efficiency and biosafety of tumour diagnosis and therapy. Numerous studies have focused on developing novel radiopharmaceuticals targeting a broader range of disease targets, demonstrating remarkable in vivo performance. These include high tumor uptake, prolonged retention time, and favorable pharmacokinetic properties that align with clinical standards. While radiotheranostics have been widely applied in tumor diagnosis and therapy, their applications are now expanding to neurodegenerative diseases, cardiovascular diseases, and inflammation. Furthermore, radiotheranostic-empowered precision medicine is revolutionizing the cancer treatment paradigm. Diagnostic radiopharmaceuticals play a pivotal role in patient stratification and treatment planning, leading to improved therapeutic outcomes in targeted radionuclide therapy. This review offers a comprehensive overview of the evolution of radiopharmaceuticals, including both FDA-approved and clinically investigated agents, and explores the mechanisms of cell death induced by radiopharmaceuticals. It emphasizes the significance and future prospects of theranostic-based radiopharmaceuticals in advancing precision medicine.
Collapse
Grants
- 82372002 National Natural Science Foundation of China (National Science Foundation of China)
- 0104002 Beijing Nova Program
- L248087; L234044 Natural Science Foundation of Beijing Municipality (Beijing Natural Science Foundation)
- Nonprofit Central Research Institute Fund of the Chinese Academy of Medical Sciences (No. 2022-RC350-04), the CAMS Innovation Fund for Medical Sciences (Nos. 2021-I2M-1-026, 2022-I2M-2-002-2, and 2021-I2M-3-001), the National Key Research and Development Program of China (No. 2022YFE0111700),the Fundamental Research Funds for the Central Universities (Nos. 3332023044 and 3332023151), the CIRP Open Fund of Radiation Protection Laboratories (No. ZHYLYB2021005), and the China National Nuclear Corporation Young Talent Program.
- Fundamental Research Funds for the Central Universities,Nos. 3332023044
- Fundamental Research Funds for the Central Universities,Nos. 3332023151
- he Nonprofit Central Research Institute Fund of Chinese Academy of Medical Sciences,No. 2022-RC350-04;the CAMS Innovation Fund for Medical Sciences,Nos. 2021-I2M-1-026, 2022-I2M-2-002-2, and 2021-I2M-3-001;the National Key Research and Development Program of China,No. 2022YFE0111700
Collapse
Affiliation(s)
- Siqi Zhang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Xingkai Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Xin Gao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Xueyao Chen
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Linger Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Guoqing Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Can Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Yuan Miao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China
| | - Rui Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China.
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences & Research Unit of Peptide Science, Chinese Academy of Medical Sciences, Lanzhou University, 2019RU066, 730000, Lanzhou, China.
| | - Kuan Hu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 100050, Beijing, China.
| |
Collapse
|
2
|
Li Z, Jiang Y, Ruan Q, Yin G, Han P, Duan X, Zhang J. Synthesis and Evaluation of 99mTc-Labeled DPro-Gly-Containing Tracers Targeting PSMA. Mol Pharm 2024; 21:5305-5314. [PMID: 39298677 DOI: 10.1021/acs.molpharmaceut.4c00799] [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: 09/22/2024]
Abstract
The specific expression of prostate-specific membrane antigen (PSMA) makes it an ideal target for the diagnosis and treatment of prostate cancer. Currently, many 99mTc-labeled PSMA-targeted tracers have been developed. However, the high renal uptake of these 99mTc-labeled tracers is a common problem that limits their clinical application. In this work, the ligand (EUKPG) using DPro-Gly as the linker was synthesized and three 99mTc-labeled complexes ([99mTc]Tc-EUKPG-EDDA, [99mTc]Tc-EUKPG-TPPTS, [99mTc]Tc-EUKPG-TPPMS) with different coligands were prepared and evaluated. Among them, [99mTc]Tc-EUKPG-EDDA showed the most favorable pharmacokinetic properties, with significantly reduced uptake in the kidney (14.04 ± 0.23% ID/g), rapid clearance and low uptake in nontarget organs, thus making it to exhibit high tumor-to-background ratios (tumor/blood: 7.47, tumor/muscle: 12.65). Affinity studies have shown that it has high specificity for PSMA both in vivo and in vitro. Therefore, [99mTc]Tc-EUKPG-EDDA has great potential as a promising molecular tracer to target PSMA for tumor imaging.
Collapse
Affiliation(s)
- Zuojie Li
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, College of Chemistry, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Beijing Normal University, Beijing 100875, P. R. China
| | - Yuhao Jiang
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, College of Chemistry, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Beijing Normal University, Beijing 100875, P. R. China
- Key Laboratory of Beam Technology of the Ministry of Education, College of Physics and Astronomy, Beijing Normal University, Beijing 100875, P. R. China
| | - Qing Ruan
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, College of Chemistry, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Beijing Normal University, Beijing 100875, P. R. China
- Key Laboratory of Beam Technology of the Ministry of Education, College of Physics and Astronomy, Beijing Normal University, Beijing 100875, P. R. China
| | - Guangxing Yin
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, College of Chemistry, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Beijing Normal University, Beijing 100875, P. R. China
| | - Peiwen Han
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, College of Chemistry, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Beijing Normal University, Beijing 100875, P. R. China
| | - Xiaojiang Duan
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, P. R. China
| | - Junbo Zhang
- Key Laboratory of Radiopharmaceuticals of the Ministry of Education, College of Chemistry, NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Beijing Normal University, Beijing 100875, P. R. China
| |
Collapse
|
3
|
Kur K, Papadopoulou E, Nidam M, Fertouk M, Binyamini O, Friedlander Barenboim S, Lazarovitci TS, Domachevsky L, Yarom N. The diagnostic value of prostate-specific membrane antigen PET-CT in differentiating medication-related osteonecrosis of the jaw and metastasis to the jawbone. Dentomaxillofac Radiol 2024; 53:497-500. [PMID: 39087565 DOI: 10.1093/dmfr/twae034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 03/22/2024] [Accepted: 07/12/2024] [Indexed: 08/02/2024] Open
Abstract
INTRODUCTION Medication-related osteonecrosis of the jaw (MRONJ) and jaw metastasis might share similar clinical and radiographic characteristics, with both demonstrating F-18 fluorodeoxyglucose (FDG) uptake on PET-CT. Prostate-specific membrane antigen (PSMA) PET-CT is used to demonstrate prostate cancer dissemination. Unlike FDG PET-CT, PSMA PET-CT is more specific to cancer than to inflammation. Therefore, we hypothesized that it might be a useful tool to differentiate between MRONJ and jaw metastasis. METHODS All files of prostate cancer patients diagnosed with MRONJ and with available PSMA PET-CT studies were retrieved. A similar number of solid cancer patients with MRONJ and with available FDG PET-CT studies served as a second study group. All studies were reviewed by 2 blinded co-investigators (L.D. and M.F.). RESULTS Seventeen patients who underwent PSMA PET-CT (24 studies) and 15 patients who underwent FDG PET-CT (29 studies) met the inclusion criteria. All patients with FDG PET-CT studies showed pathological uptake at the site of MRONJ in at least one of their studies versus only 23.5% of patients in the PSMA PET-CT group (P < .001). FDG PET-CT studies showed pathological uptake in 89.6% of the studies compared with only 20.8% in the PSMA PET-CT group (P < .001). The mean standardized uptake value (SUVmax) and the mean uptake volume in the FDG PET-CT group were significantly higher compared with the PSMA PET-CT group (P < .001 and P < .005, respectively). The interclass correlation coefficient for all parameters was higher than 0.95. CONCLUSIONS PSMA PET-CT is useful to differentiate between MRONJ and jaw metastasis.
Collapse
Affiliation(s)
- Karin Kur
- Oral Medicine Unit, Sheba Medical Center, Tel-Hashomer 5265601, Israel
| | - Erofili Papadopoulou
- Department of Oral Medicine & Pathology and Hospital Dentistry, School of Dentistry, National and Kapodistrian University of Athens, Athens 11527, Greece
| | - Meital Nidam
- Nuclear Medicine Institute, Sheba Medical Center, Tel-Hashomer 5265601, Israel
| | - Michal Fertouk
- Nuclear Medicine Institute, Sheba Medical Center, Tel-Hashomer 5265601, Israel
| | - Omer Binyamini
- The Maurice and Gabriela Goldschleger School of Dental Medicine, Faculty of Health and Medical Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | | | - Towy Sorel Lazarovitci
- Department of Oral and Maxillofacial Surgery, Sheba Medical Center, Tel-Hashomer 5265601, Israel
| | - Liran Domachevsky
- Nuclear Medicine Institute, Sheba Medical Center, Tel-Hashomer 5265601, Israel
- Faculty of Health and Medical Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Noam Yarom
- Oral Medicine Unit, Sheba Medical Center, Tel-Hashomer 5265601, Israel
- The Maurice and Gabriela Goldschleger School of Dental Medicine, Faculty of Health and Medical Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
4
|
Muglia VF, Laschena L, Pecoraro M, de Lion Gouvea G, Colli LM, Panebianco V. Imaging assessment of prostate cancer recurrence: advances in detection of local and systemic relapse. Abdom Radiol (NY) 2024:10.1007/s00261-024-04412-7. [PMID: 39254707 DOI: 10.1007/s00261-024-04412-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 09/11/2024]
Abstract
Prostate cancer (PCa) relapse, defined either by persistent PSA levels (after RP) or biochemical recurrence (BCR), is a common occurrence. The imaging evaluation of patients experiencing PCa relapse has undergone significant advancements in the past decade, notably with the introduction of new Positron Emission Tomography (PET) tracers such as Prostate-specific membrane antigen (PSMA), and the progress in functional Magnetic Resonance Imaging (MRI). This article will explore the role of traditional imaging, the evolution of MRI towards the development of the Prostate Magnetic Resonance Imaging for Local Recurrence Reporting (PI-RR) scoring system, and how next-generation imaging is enhancing diagnostic accuracy in the setting of PCa relapse, which is essential for adopting personalized strategies that may ultimately impact outcomes.
Collapse
Affiliation(s)
- Valdair Francisco Muglia
- Department of Medical Images, Oncology and Hematology, Ribeirao Preto Medical School, University of Sao Paulo, Hospital Clinicas Ribeirao Preto - Av. Bandeirantes 3900. Campus Monte Alegre -USP, Sao Paulo, 14049-900, Brazil.
| | - Ludovica Laschena
- Department of Radiological Sciences, Oncology and Pathology, Sapienza University, Rome, Italy
| | - Martina Pecoraro
- Department of Radiological Sciences, Oncology and Pathology, Sapienza University, Rome, Italy
| | - Gabriel de Lion Gouvea
- Department of Medical Images, Oncology and Hematology, Ribeirao Preto Medical School, University of Sao Paulo, Hospital Clinicas Ribeirao Preto - Av. Bandeirantes 3900. Campus Monte Alegre -USP, Sao Paulo, 14049-900, Brazil
| | - Leandro Machado Colli
- Department of Medical Images, Oncology and Hematology, Ribeirao Preto Medical School, University of Sao Paulo, Hospital Clinicas Ribeirao Preto - Av. Bandeirantes 3900. Campus Monte Alegre -USP, Sao Paulo, 14049-900, Brazil
| | - Valeria Panebianco
- Department of Radiological Sciences, Oncology and Pathology, Sapienza University, Rome, Italy
| |
Collapse
|
5
|
Andrew J, Ezra-Manicum AL, Witika BA. Developments in radionanotheranostic strategies for precision diagnosis and treatment of prostate cancer. EJNMMI Radiopharm Chem 2024; 9:62. [PMID: 39180599 PMCID: PMC11344754 DOI: 10.1186/s41181-024-00295-7] [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: 06/13/2024] [Accepted: 08/13/2024] [Indexed: 08/26/2024] Open
Abstract
BACKGROUND Prostate Cancer (PCa) is the second most diagnosed urological cancer among men worldwide. Conventional methods used for diagnosis of PCa have several pitfalls which include lack of sensitivity and specificity. On the other hand, traditional treatment of PCa poses challenges such as long-term side effects and the development of multidrug resistance (MDR). MAIN BODY Hence, there is a need for novel PCa agents with the potential to lessen the burden of these adverse effects on patients. Nanotechnology has emerged as a promising approach to support both early diagnosis and effective treatment of tumours by ensuring precise delivery of the drug to the targeted site of the disease. Most cancer-related biological processes occur on the nanoscale hence application of nanotechnology has been greatly appreciated and implemented in the management and therapeutics of cancer. Nuclear medicine plays a significant role in the non-invasive diagnosis and treatment of PCa using appropriate radiopharmaceuticals. This review aims to explore the different radiolabelled nanomaterials to enhance the specific delivery of imaging and therapeutic agents to cancer cells. Thereafter, the review appraises the advantages and disadvantages of these modalities and then discusses and outlines the benefits of radiolabelled nanomaterials in targeting cancerous prostatic tumours. Moreover, the nanoradiotheranostic approaches currently developed for PCa are discussed and finally the prospects of combining radiopharmaceuticals with nanotechnology in improving PCa outcomes will be highlighted. CONCLUSION Nanomaterials have great potential, but safety and biocompatibility issues remain. Notwithstanding, the combination of nanomaterials with radiotherapeutics may improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Jubilee Andrew
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria, 0208, South Africa
| | - Amanda-Lee Ezra-Manicum
- Department of Chemistry, Faculty of Science, Tshwane University of Technology (Arcadia Campus), Pretoria, South Africa
| | - Bwalya Angel Witika
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria, 0208, South Africa.
| |
Collapse
|
6
|
Inaki A, Mizokami A, Wakabayashi H, Izumi K, Kadono Y, Toyama T, Takahara S, Murayama T, Kinuya S. Evaluation of 68Ga-PSMA-11 PET/CT: a Phase 1 clinical study in Japanese patients with primary, recurrent, or suspected recurrent prostate cancer. Ann Nucl Med 2024; 38:587-595. [PMID: 38750331 PMCID: PMC11281955 DOI: 10.1007/s12149-024-01931-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 04/09/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND Prostate-specific membrane antigen (PSMA)-targeted radiopharmaceuticals allow whole-body imaging to detect prostate cancer (PC). Positron emission tomography imaging using gallium-68 (68Ga)-PSMA-11 has been shown to have a favorable safety and tolerability profile and high diagnostic performance. The study evaluates the safety and pharmacokinetics of 68Ga-PSMA-11 in Japanese patients with primary, recurrent, or suspected recurrent prostate cancer. METHODS This single arm study enrolled Japanese patients with primary PC (n = 3), suspected recurrent PC following radical prostatectomy (n = 4), or suspected recurrent PC following radical radiotherapy (n = 3). All patients received a single intravenous dose of 68Ga-PSMA-11 2.0 MBq/kg (±10%) followed by PSMA PET imaging and safety and pharmacokinetic evaluations. Based on the blood concentrations of 68Ga-PSMA-11 and the radioactivity distribution rate in each organ/tissue, the absorbed doses in major organs/tissues and the whole-body effective dose were calculated by the Medical Internal Radiation Dose method. RESULTS Ten patients were enrolled. Mean age was 73.3 ± 4.8 years, and median prostate-specific antigen was 8.250 ng/mL. Five patients (50%) experienced a total of 6 adverse events, and no grade ≥ 2 adverse events or serious adverse events were reported. No clinically significant changes in vital signs, haematology parameters, or blood chemistry or ECG abnormalities were observed. The estimated whole body effective dose of 68Ga-PSMA-11 (mean ± standard deviation) was 2.524 × 10-2 ± 2.546 × 10-3 mSv/MBq. Time to maximum concentration (1.16 × 10-4 ± 1.3 × 10-5% ID/mL) in whole blood was 2.15 ± 0.33 min. CONCLUSIONS 68Ga-PSMA-11 has a favourable safety and tolerability profile in Japanese patients with primary, recurrent, or suspected recurrent prostate cancer, which is comparable to previous observations in other populations.
Collapse
Affiliation(s)
- Anri Inaki
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-Machi, Kanazawa-Shi, Ishikawa, 920-8641, Japan.
| | - Atsushi Mizokami
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Hospital, 13-1 Takara-Machi, Kanazawa-Shi, Ishikawa, 920-8641, Japan
| | - Hiroshi Wakabayashi
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-Machi, Kanazawa-Shi, Ishikawa, 920-8641, Japan
| | - Kouji Izumi
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Hospital, 13-1 Takara-Machi, Kanazawa-Shi, Ishikawa, 920-8641, Japan
| | - Yoshifumi Kadono
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Hospital, 13-1 Takara-Machi, Kanazawa-Shi, Ishikawa, 920-8641, Japan
| | - Tadashi Toyama
- Innovative Clinical Research Center, Kanazawa University Hospital, 13-1 Takara-Machi, Kanazawa-Shi, Ishikawa, 920-8641, Japan
| | - Shizuko Takahara
- Innovative Clinical Research Center, Kanazawa University Hospital, 13-1 Takara-Machi, Kanazawa-Shi, Ishikawa, 920-8641, Japan
| | - Toshinori Murayama
- Innovative Clinical Research Center, Kanazawa University Hospital, 13-1 Takara-Machi, Kanazawa-Shi, Ishikawa, 920-8641, Japan
- Department of Clinical Development, Kanazawa University Hospital, 13-1 Takara-Machi, Kanazawa-Shi, Ishikawa, 920-8641, Japan
| | - Seigo Kinuya
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-Machi, Kanazawa-Shi, Ishikawa, 920-8641, Japan
| |
Collapse
|
7
|
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
|
8
|
Xiao L, Fang Z, Tang Y, Sun Y, Zhu Z, Li J, Zhou M, Yang N, Zheng K, Hu S. Evaluation of gastrin-releasing peptide receptor, prostate-specific membrane antigen, and neurotensin receptor 1 as potential biomarkers for accurate prostate cancer stratified diagnosis. EJNMMI Res 2024; 14:55. [PMID: 38880858 PMCID: PMC11180645 DOI: 10.1186/s13550-024-01116-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 06/02/2024] [Indexed: 06/18/2024] Open
Abstract
BACKGROUND Studies on single-target PET imaging of gastrin-releasing peptide receptor (GRPR), prostate-specific membrane antigen (PSMA), or neurotensin receptor 1(NTR1) have been reported. However, the performance of these three targets in the progression of PCa remains unclear. Our study aims to compare the expression of GRPR, PSMA, and NTR1 in patients with prostatic intraepithelial neoplasia (PIN), prostate cancer (PCa), and lymph node metastasis. We synthesized molecular probes targeting the markers to achieve a non-invasive precise detection of PCa patients with PET/CT imaging. METHODS In this study, the expression of GRPR, PSMA, and NTR1 was evaluated by immunohistochemistry in 34 PIN, 171 PCa, and 22 lymph node metastasis tissues of patients. The correlation between their expression and the clinicopathological parameters of PCa patients was assessed. Sixteen PCa patients with different Gleason scores (GS) underwent dual-tracer (68Ga-NOTA-RM26 and 68Ga-NOTA-PSMA617) PET/CT. RESULTS In the PIN stage, the expression of GRPR was significantly higher than that of PSMA and NTR1 (P < 0.001), while NTR1 expression was significantly higher than PSMA and GRPR expression in primary PCa (P = 0.001). High PSMA expression in PCa patients was associated with shorter progression-free survival (P = 0.037) and overall survival (P = 0.035). PCa patients with high GS had higher tumor uptake of 68Ga-NOTA-PSMA617 than those with low GS (P = 0.001), while PCa patients with low GS had higher tumor uptake of 68Ga-NOTA-RM26 than those with high GS (P = 0.001). CONCLUSIONS This study presents three novel biomarkers (PSMA, GRPR, and NTR1) as imaging agents for PET/CT, and may offer a promising approach for non-invasive precise detection and Gleason grade prediction of PCa patients.
Collapse
Affiliation(s)
- Ling Xiao
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha City, 410008, Hunan Province, P.R. China
| | - Zhihui Fang
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha City, 410008, Hunan Province, P.R. China
- Department of Nuclear Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yongxiang Tang
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha City, 410008, Hunan Province, P.R. China
| | - Yanyan Sun
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, 450000, China
| | - Zehua Zhu
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha City, 410008, Hunan Province, P.R. China
| | - Jian Li
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha City, 410008, Hunan Province, P.R. China
| | - Ming Zhou
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha City, 410008, Hunan Province, P.R. China
| | - Nengan Yang
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha City, 410008, Hunan Province, P.R. China
| | - Kai Zheng
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha City, 410008, Hunan Province, P.R. China
| | - Shuo Hu
- Department of Nuclear Medicine, Xiangya Hospital, Central South University, No.87 Xiangya Road, Changsha City, 410008, Hunan Province, P.R. China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
9
|
Zhang J, Kang F, Wang X, Chen X, Yang X, Yang Z, Wang J. Recent Advances in Radiotracers Targeting Novel Cancer-Specific Biomarkers in China: A Brief Overview. J Nucl Med 2024; 65:38S-45S. [PMID: 38719241 DOI: 10.2967/jnumed.123.266314] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/23/2024] [Indexed: 07/16/2024] Open
Abstract
Radiopharmaceuticals play a critical role in nuclear medicine, providing novel tools for specifically delivering radioisotopes for the diagnosis and treatment of cancers. As the starting point for developing radiopharmaceuticals, cancer-specific biomarkers are important and receive worldwide attention. This field in China is currently experiencing a rapid expansion, with multiple radiotracers targeting novel targets being developed and translated into clinical studies. This review provides a brief overview of the exploration of novel imaging targets, preclinical evaluation of their targeting ligands, and translational research in China from 2020 to 2023, for detecting cancer, guiding targeted therapy, and visualizing the immune microenvironment. We believe that China will play an even more important role in the development of nuclear medicine in the world in the future.
Collapse
Affiliation(s)
- Jingming Zhang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
- Department of Nuclear Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Fei Kang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiao Wang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| | - Xuejiao Chen
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
| | - Xing Yang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, China
- Department of Central Laboratory, Peking University First Hospital, Beijing, China
- Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Beijing, China
- International Cancer Institute, Peking University Health Science Center, Beijing, China; and
| | - Zhi Yang
- Key Laboratory for Research and Evaluation of Radiopharmaceuticals (National Medical Products Administration), Beijing, China;
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital and Institute, Beijing, China
| | - Jing Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China;
| |
Collapse
|
10
|
Pantel AR, Bae SW, Li EJ, O'Brien SR, Manning HC. PET Imaging of Metabolism, Perfusion, and Hypoxia: FDG and Beyond. Cancer J 2024; 30:159-169. [PMID: 38753750 PMCID: PMC11101148 DOI: 10.1097/ppo.0000000000000716] [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: 05/18/2024]
Abstract
ABSTRACT Imaging glucose metabolism with [18F]fluorodeoxyglucose positron emission tomography has transformed the diagnostic and treatment algorithms of numerous malignancies in clinical practice. The cancer phenotype, though, extends beyond dysregulation of this single pathway. Reprogramming of other pathways of metabolism, as well as altered perfusion and hypoxia, also typifies malignancy. These features provide other opportunities for imaging that have been developed and advanced into humans. In this review, we discuss imaging metabolism, perfusion, and hypoxia in cancer, focusing on the underlying biology to provide context. We conclude by highlighting the ability to image multiple facets of biology to better characterize cancer and guide targeted treatment.
Collapse
Affiliation(s)
- Austin R Pantel
- From the Department of Radiology, University of Pennsylvania, Philadelphia, PA
| | - Seong-Woo Bae
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Elizabeth J Li
- From the Department of Radiology, University of Pennsylvania, Philadelphia, PA
| | - Sophia R O'Brien
- From the Department of Radiology, University of Pennsylvania, Philadelphia, PA
| | - H Charles Manning
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
11
|
Mendoza-Ávila M, Esparza-Pérez H, Castillo-López JA, Rodea-Montero ER. Agreement between PSMA-RADS and E-PSMA systems in classifying [ 18F]PSMA-1007 PET/CT lesions among prostate cancer patients: exploring the correlation between lesion size and uptake. Front Med (Lausanne) 2024; 11:1368093. [PMID: 38545506 PMCID: PMC10967023 DOI: 10.3389/fmed.2024.1368093] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 02/19/2024] [Indexed: 01/03/2025] Open
Abstract
PURPOSE To determine the agreement between the PSMA-RADS and E-PSMA standardized reporting systems in the classification of [18F]PSMA-1007-uptaking lesions identified on PET/CT scan in patients with prostate cancer (PCa) and post-prostatectomy with suspected recurrent disease (local recurrence, regional nodal involvement and distant metastases), based on biochemical recurrence, while also exploring the correlation between lesion size and tracer uptake. MATERIALS AND METHODS A retrospective cross-sectional study of 32 post-prostatectomy PCa patients who had suspected recurrent disease based on biochemical recurrence post-prostatectomy (prostate-specific antigen values that are 0.2 ng/mL or higher) underwent [18F]PSMA-1007 PET/CT scan. The recurrent disease PCa lesions were characterized and subsequently classified using two standardized reporting systems (PSMA-RADS and E-PSMA). The lesions were grouped based on anatomical site, their size and SUVmax were compared using Kruskal-Wallis test with Dunn-Bonferroni post hoc tests. Spearman correlation coefficients were calculated between the size of the lesions and their SUVmax of the radiotracer [18F]PSMA-1007 for all the lesions and when grouped by anatomical site. Additionally, the agreement between lesion classifications was assessed using Cohen's kappa index. RESULTS Only 32 (69.98 ± 8.27, men) patients met the inclusion criteria, a total of 149 lesions with avid uptake of [18F]PSMA-1007 were identified. Positive correlation (r = 0.516, p < 0.001) was observed between the size of the metastatic prostate cancer lymph node lesions and their [18F]PSMA-1007 uptake. Substantial agreement was noted between the PSMA-RADS and E-PSMA classification system scores among all lesions (κ = 0.70, p < 0.001), with notable discrepancies primarily among lymph node lesions. CONCLUSION Our findings revealed a positive correlation between the size of the metastatic prostate cancer lymph node lesions and [18F]PSMA-1007 uptake, and although there was substantial agreement between the PSMA-RADS and E-PSMA classification systems, there were discrepancies mainly among the lymph node lesions.
Collapse
Affiliation(s)
- Miguel Mendoza-Ávila
- Department of Radiology, Hospital Regional de Alta Especialidad del Bajío, León, Mexico
- Faculty of Medicine, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Hiram Esparza-Pérez
- Department of Nuclear Medicine, Hospital Regional de Alta Especialidad del Bajío, León, Mexico
| | | | - Edel Rafael Rodea-Montero
- Department of Research, Hospital Regional de Alta Especialidad del Bajío, León, Mexico
- UPIIG, Instituto Politécnico Nacional, Silao de la Victoria, Mexico
| |
Collapse
|
12
|
Xie W, Ravi P, Buyse M, Halabi S, Kantoff P, Sartor O, Soule H, Clarke N, Dignam J, James N, Fizazi K, Gillessen S, Mottet N, Murphy L, Parulekar W, Sandler H, Tombal B, Williams S, Sweeney CJ. Validation of metastasis-free survival as a surrogate endpoint for overall survival in localized prostate cancer in the era of docetaxel for castration-resistant prostate cancer. Ann Oncol 2024; 35:285-292. [PMID: 38061427 PMCID: PMC10922430 DOI: 10.1016/j.annonc.2023.11.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/20/2023] [Accepted: 11/29/2023] [Indexed: 01/21/2024] Open
Abstract
BACKGROUND Prior work from the Intermediate Clinical Endpoints in Cancer of the Prostate (ICECaP) consortium (ICECaP-1) demonstrated that metastasis-free survival (MFS) is a valid surrogate for overall survival (OS) in localized prostate cancer (PCa). This was based on data from patients treated predominantly before 2004, prior to docetaxel being available for the treatment of metastatic castrate-resistant prostate cancer (mCRPC). We sought to validate surrogacy in a more contemporary era (ICECaP-2) with greater availability of docetaxel and other systemic therapies for mCRPC. PATIENTS AND METHODS Eligible trials for ICECaP-2 were those providing individual patient data (IPD) after publication of ICECaP-1 and evaluating adjuvant/salvage therapy for localized PCa, and which collected MFS and OS data. MFS was defined as distant metastases or death from any cause, and OS was defined as death from any cause. Surrogacy was evaluated using a meta-analytic two-stage validation model, with an R2 ≥ 0.7 defined a priori as clinically relevant. RESULTS A total of 15 164 IPD from 14 trials were included in ICECaP-2, with 70% of patients treated after 2004. The median follow-up was 8.3 years and the median postmetastasis survival was 3.1 years in ICECaP-2, compared with 1.9 years in ICECaP-1. For surrogacy condition 1, Kendall's tau was 0.92 for MFS with OS at the patient level, and R2 from weighted linear regression (WLR) of 8-year OS on 5-year MFS was 0.73 (95% confidence interval 0.53-0.82) at the trial level. For condition 2, R2 was 0.83 (95% confidence interval 0.64-0.89) from WLR of log[hazard ratio (HR)]-OS on log(HR)-MFS. The surrogate threshold effect on OS was an HR(MFS) of 0.81. CONCLUSIONS MFS remained a valid surrogate for OS in a more contemporary era, where patients had greater access to docetaxel and other systemic therapies for mCRPC. This supports the use of MFS as the primary outcome measure for ongoing adjuvant trials in localized PCa.
Collapse
Affiliation(s)
- W Xie
- Dana-Farber Cancer Institute, Boston, USA
| | - P Ravi
- Dana-Farber Cancer Institute, Boston, USA
| | - M Buyse
- International Drug Development Institute, Louvain-la-Neuve; I-BioStat, Hasselt University, Hasselt, Belgium
| | | | | | | | - H Soule
- Prostate Cancer Foundation, Santa Monica, USA
| | - N Clarke
- The Christie NHS Foundation Trust, Manchester, UK
| | - J Dignam
- University of Chicago, Chicago, USA
| | - N James
- The Institute of Cancer Research & The Royal Marsden NHS Foundation Trust, London, UK
| | - K Fizazi
- Institut Gustave Roussy, University of Paris Saclay, Villejuif, France
| | - S Gillessen
- Oncology Institute of Southern Switzerland, EOC, Bellinzona; Università della Svizzera Italiana, Lugano, Switzerland
| | - N Mottet
- Mutualite Francoise Loire, St Etienne, France
| | - L Murphy
- Medical Research Council at UCL, London, UK
| | - W Parulekar
- Queens University, Kingston, Ontario, Canada
| | - H Sandler
- Cedars-Sinai Medical Center, Los Angeles, USA
| | - B Tombal
- Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - S Williams
- Peter MacCallum Cancer Centre, Melbourne
| | - C J Sweeney
- South Australian Immunogenomics Cancer Institute, University of Adelaide, Adelaide, Australia.
| |
Collapse
|
13
|
Hu C, Qiao X, Hu C, Cao C, Wang X, Bao J. The practical clinical role of machine learning models with different algorithms in predicting prostate cancer local recurrence after radical prostatectomy. Cancer Imaging 2024; 24:23. [PMID: 38326860 PMCID: PMC10848341 DOI: 10.1186/s40644-024-00667-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 01/21/2024] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND The detection of local recurrence for prostate cancer (PCa) patients following radical prostatectomy (RP) is challenging and can influence the treatment plan. Our aim was to construct and verify machine learning models with three different algorithms based on post-operative mpMRI for predicting local recurrence of PCa after RP and explore their potential clinical value compared with the Prostate Imaging for Recurrence Reporting (PI-RR) score of expert-level radiologists. METHODS A total of 176 patients were retrospectively enrolled and randomly divided into training (n = 123) and testing (n = 53) sets. The PI-RR assessments were performed by two expert-level radiologists with access to the operative histopathological and pre-surgical clinical results. The radiomics models to predict local recurrence were built by utilizing three different algorithms (i.e., support vector machine [SVM], linear discriminant analysis [LDA], and logistic regression-least absolute shrinkage and selection operator [LR-LASSO]). The combined model integrating radiomics features and PI-RR score was developed using the most effective classifier. The classification performances of the proposed models were assessed by receiver operating characteristic (ROC) curve analysis. RESULTS There were no significant differences between the training and testing sets concerning age, prostate-specific antigen (PSA), Gleason score, T-stage, seminal vesicle invasion (SVI), perineural invasion (PNI), and positive surgical margins (PSM). The radiomics model based on LR-LASSO exhibited superior performance than other radiomics models, with an AUC of 0.858 in the testing set; the PI-RR yielded an AUC of 0.833, and there was no significant difference between the best radiomics model and the PI-RR score. The combined model achieved the best predictive performance with an AUC of 0.924, and a significant difference was observed between the combined model and PI-RR score. CONCLUSIONS Our radiomics model is an effective tool to predict PCa local recurrence after RP. By integrating radiomics features with the PI-RR score, our combined model exhibited significantly better predictive performance of local recurrence than expert-level radiologists' PI-RR assessment.
Collapse
Affiliation(s)
- Chenhan Hu
- Department of Radiology, the First Affiliated Hospital of Soochow University, 188#, Shizi Road, Suzhou, 215006, China
| | - Xiaomeng Qiao
- Department of Radiology, the First Affiliated Hospital of Soochow University, 188#, Shizi Road, Suzhou, 215006, China
| | - Chunhong Hu
- Department of Radiology, the First Affiliated Hospital of Soochow University, 188#, Shizi Road, Suzhou, 215006, China
| | - Changhao Cao
- Department of Radiology, the First Affiliated Hospital of Soochow University, 188#, Shizi Road, Suzhou, 215006, China
| | - Ximing Wang
- Department of Radiology, the First Affiliated Hospital of Soochow University, 188#, Shizi Road, Suzhou, 215006, China.
| | - Jie Bao
- Department of Radiology, the First Affiliated Hospital of Soochow University, 188#, Shizi Road, Suzhou, 215006, China.
| |
Collapse
|
14
|
Houshmand S, Lawhn-Heath C, Behr S. PSMA PET imaging in the diagnosis and management of prostate cancer. Abdom Radiol (NY) 2023; 48:3610-3623. [PMID: 37493837 PMCID: PMC10682054 DOI: 10.1007/s00261-023-04002-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/27/2023]
Abstract
Prostate cancer is the second leading cause of cancer-related deaths in men in the United States. Imaging techniques such as CT, MRI, and bone scans have traditionally been used for diagnosis and staging. Molecular imaging modalities targeting the prostate-specific membrane antigen (PSMA) have recently gained attention due to their high affinity and accuracy. PSMA PET has been combined with other modalities such as multiparametric MRI for better diagnostic and prognostic performance. PSMA imaging has been studied at different clinical settings with a wide range of disease aggressiveness. In this review we will explore the role of PSMA PET in high-risk prostate cancer staging, biochemical recurrence, and castration-resistant prostate cancer. The primary focus of this review article is to examine the latest developments in the use of PSMA imaging and emphasize the clinical situations where its effectiveness has been demonstrated to significantly impact the treatment of prostate cancer. In addition, we will touch upon the potential future advancements of PSMA PET imaging and its evolving significance in the management of prostate cancer.
Collapse
Affiliation(s)
- Sina Houshmand
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA.
| | - Courtney Lawhn-Heath
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| | - Spencer Behr
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
15
|
Huls SJ, Burkett B, Ehman E, Lowe VJ, Subramaniam RM, Kendi AT. Clinical practice in prostate PET imaging. Ther Adv Med Oncol 2023; 15:17588359231213618. [PMID: 38028142 PMCID: PMC10666681 DOI: 10.1177/17588359231213618] [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: 06/12/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Positron emission tomography (PET) imaging in prostate cancer has advanced significantly in the past decade with prostate cancer targeted radiopharmaceuticals now playing a growing role in diagnosis, staging, and treatment. This narrative review focuses on the most commonly used PET radiopharmaceuticals in the USA: prostate-specific membrane antigen (PSMA), fluciclovine, and choline. 18F-fluorodeoxyglucose (FDG) is used in many other malignancies, but rarely in prostate cancer. Previous literature is discussed regarding each radiopharmaceutical's utility in the settings of screening/diagnosis, initial staging, biochemical recurrence, advanced disease, and evaluation prior to targeted radiopharmaceutical therapy and radiation therapy. PET imaging has demonstrated utility over traditional imaging in various scenarios; however, there are few head-to-head studies comparing PET radiopharmaceuticals. PSMA radiopharmaceuticals are the newest tracers developed and have unique properties and uses, especially at low prostate-specific antigen (PSA) levels. However, each PET radiopharmaceutical has different properties which can affect image interpretation. Choline and fluciclovine have minimal urinary activity, whereas PSMA agents can have high urinary activity which may affect locoregional disease evaluation. Of the three radiopharmaceuticals, only PSMA is approved for both diagnostic and therapeutic indications with 177Lu-PSMA. A variety of diagnostic PET radiotracers for prostate cancer allows for increased flexibility, especially in the setting of supply chain and medication shortages. For the time being, keeping a diverse group of PET radiopharmaceuticals for prostate cancer is justifiable.
Collapse
Affiliation(s)
- Sean J. Huls
- Department of Radiology, Mayo Clinic, 200 First Street SW, Rochester MN 55905, USA
| | - Brian Burkett
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Eric Ehman
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Val J. Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Rathan M. Subramaniam
- Department of Medicine, University of Otago Medical School, Dunedin, New Zealand
- Department of Radiology, Duke University, Durham, NC, USA
| | - A. Tuba Kendi
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
16
|
Zhang J, Kang F, Gao J, Jiao J, Quan Z, Ma S, Li Y, Guo S, Li Z, Jing Y, Zhang K, Yang F, Han D, Wen W, Zhang J, Ren J, Wang J, Guo H, Qin W. A Prostate-Specific Membrane Antigen PET-Based Approach for Improved Diagnosis of Prostate Cancer in Gleason Grade Group 1: A Multicenter Retrospective Study. J Nucl Med 2023; 64:1750-1757. [PMID: 37652543 DOI: 10.2967/jnumed.122.265001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 07/11/2023] [Indexed: 09/02/2023] Open
Abstract
The preoperative Gleason grade group (GG) from transrectal ultrasound-guided prostate biopsy is crucial for treatment decisions but may underestimate the postoperative GG and miss clinically significant prostate cancer (csPCa), particularly in patients with biopsy GG1. In such patients, an SUVmax of at least 12 has 100% specificity for detecting csPCa. In patients with an SUVmax of less than 12, we aimed to develop a model to improve the diagnostic accuracy of csPCa. Methods: The study retrospectively included 56 prostate cancer patients with transrectal ultrasound-guided biopsy GG1 and an SUVmax of less than 12 from 2 tertiary hospitals. All [68Ga]Ga-PSMA-HBED-CC PET scans were centrally reviewed in Xijing Hospital. A deep learning model was used to evaluate the overlap of SUVmax (size scale, 3 cm) and the level of Gleason pattern (size scale, 500 μm). A diagnostic model was developed using the PRIMARY score and SUVmax, and its discriminative performance and clinical utility were compared with other methods. The 5-fold cross-validation (repeated 1,000 times) was used for internal validation. Results: In patients with GG1 and an SUVmax of less than 12, significant prostate-specific membrane antigen (PSMA) histochemical score (H-score) H-score overlap occurred among benign gland, Gleason pattern 3, and Gleason pattern 4 lesions, causing SUVmax overlap between csPCa and non-csPCa. The model of 10 × PRIMARY score + 2 × SUVmax exhibited a higher area under the curve (AUC, 0.8359; 95% CI, 0.7233-0.9484) than that found using only the SUVmax (AUC, 0.7353; P = 0.048) or PRIMARY score (AUC, 0.7257; P = 0.009) for the cohort and a higher AUC (0.8364; 95% CI, 0.7114-0.9614) than that found using only the Prostate Imaging Reporting and Data System (PI-RADS) score of 5-4 versus 3-1 (AUC, 0.7036; P = 0.149) and the PI-RADS score of 5-3 versus 2-1 (AUC, 0.6373; P = 0.014) for a subgroup. The model reduced the misdiagnosis of the PI-RADS score of 5-4 versus 3-1 by 78.57% (11/14) and the PI-RADS score of 5-3 versus 2-1 by 77.78% (14/18). The internal validation showed that the mean 5-fold cross-validated AUC was 0.8357 (95% CI, 0.8357-0.8358). Conclusion: We preliminarily suggest that the model of 10 × PRIMARY score + 2 × SUVmax may enhance the diagnostic accuracy of csPCa in patients with biopsy GG1 and an SUVmax of less than 12 by maximizing PSMA information use, reducing the misdiagnosis of the PI-RADS score, and thereby aiding in making appropriate treatment decisions.
Collapse
Affiliation(s)
- Jingliang Zhang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Fei Kang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jie Gao
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, China
| | - Jianhua Jiao
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zhiyong Quan
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Shuaijun Ma
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yu Li
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Shikuan Guo
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zeyu Li
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yuming Jing
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Keying Zhang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Fa Yang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Donghui Han
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Weihong Wen
- Institute of Medical Research, Northwestern Polytechnical University, Xi'an, China
| | - Jing Zhang
- Department of Pathology, Xijing Hospital, Fourth Military Medical University, Xi'an, China; and
| | - Jing Ren
- Department of Radiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jing Wang
- Department of Nuclear Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hongqian Guo
- Department of Urology, Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, Nanjing, China
| | - Weijun Qin
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, China;
| |
Collapse
|
17
|
Abusalem M, Martiniova L, Soebianto S, DePalatis L, Ravizzini G. Current Status of Radiolabeled Monoclonal Antibodies Targeting PSMA for Imaging and Therapy. Cancers (Basel) 2023; 15:4537. [PMID: 37760506 PMCID: PMC10526399 DOI: 10.3390/cancers15184537] [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: 06/27/2023] [Revised: 08/21/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Prostate cancer (PCa) is one of the most prevalent cancer diagnoses among men in the United States and in several other developed countries. The prostate specific membrane antigen (PSMA) has been recognized as a promising molecular target in PCa, which has led to the development of specific radionuclide-based tracers for imaging and radiopharmaceuticals for PSMA targeted therapy. These compounds range from small molecule ligands to monoclonal antibodies (mAbs). Monoclonal antibodies play a crucial role in targeting cancer cell-specific antigens with a high degree of specificity while minimizing side effects to normal cells. The same mAb can often be labeled in different ways, such as with radionuclides suitable for imaging with Positron Emission Tomography (β+ positrons), Gamma Camera Scintigraphy (γ photons), or radiotherapy (β- electrons, α-emitters, or Auger electrons). Accordingly, the use of radionuclide-based PSMA-targeting compounds in molecular imaging and therapeutic applications has significantly grown in recent years. In this article, we will highlight the latest developments and prospects of radiolabeled mAbs that target PSMA for the detection and treatment of prostate cancer.
Collapse
Affiliation(s)
- Mohammed Abusalem
- Department of Nuclear Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lucia Martiniova
- Department of Experimental Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sarita Soebianto
- Department of Nuclear Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Louis DePalatis
- BioDevelopment Solutions, LLC, 226 Becker Circle, Johnstown, CO 80534, USA
| | - Gregory Ravizzini
- Department of Nuclear Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
18
|
Fu L, Mu Z, Zhou J, Qing M, Bai L. "Gold-plated" PCN-222(Fe) and superconductive carbon black-based sandwich-type immunosensor for detecting CYFRA21-1. J Mater Chem B 2023; 11:8262-8270. [PMID: 37578169 DOI: 10.1039/d3tb01245j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Cytokeratin 19 fragment antigen 21-1 (CYFRA21-1) is a protein fragment dissolved in the blood after apoptosis of lung epithelial cells, which is a predictive biomarker for the diagnosis of non-small cell lung cancer (NSCLC). Detection of serum CYFRA21-1 has a significant clinical value in diagnosis, monitoring and prognosis of NSCLC. Herein, a novel electrochemical immunosensor was constructed for the sensitive detection of CYFRA21-1. First, superconductive carbon black (KB) functionalized polyethyleneimine (PEI)-gold nanoparticles (AuNPs) were covered on the surface of methylene blue (MB) and used as substrate materials to immobilize the CYFRA21-1 antibody. Then, target CYFRA21-1 was successfully detected using an electrochemical immunosensor through specific recognition of antigen and antibody. The zirconium-based metal organic framework of PCN-222(Fe) with a large pore size and three-dimensional (3D) structure can absorb abundant AuNPs through strong electrostatic interaction, which enhances the conductive properties of PCN-222(Fe) and prevents the self-aggregation of AuNPs. However, PCN-222(Fe) with peroxidase-like activity can catalyze the generation of hydroxyl free radicals (˙OH) from H2O2, which oxidized MB, leading to a decrease in the current signal. The signal response to the degradation of MB was recorded using differential pulse voltammetry (DPV). This indirect method of immunosensor offered a new strategy to address the limitations imposed by the poor conductivity of PCN-222(Fe), further enabling the amplification of the signal through the oxidative degradation of MB. Compared with traditional electrochemical immunosensors, this method has the advantages of a stable current signal and good reproducibility, providing a promising reference for the broad application of PCN-222(Fe) in electrochemical biosensors.
Collapse
Affiliation(s)
- Lin Fu
- Chongqing Research Center for Pharmaceutical Engineering, College of Pharmacy, Chongqing Medical University, Chongqing 400016, P. R. China.
| | - Zhaode Mu
- Research Center for Pharmacodynamic Evaluation Engineering Technology of Chongqing, College of Pharmacy, Chongqing Medical University, Chongqing 400016, P. R. China.
| | - Jing Zhou
- Chongqing Research Center for Pharmaceutical Engineering, College of Pharmacy, Chongqing Medical University, Chongqing 400016, P. R. China.
| | - Min Qing
- Research Center for Pharmacodynamic Evaluation Engineering Technology of Chongqing, College of Pharmacy, Chongqing Medical University, Chongqing 400016, P. R. China.
| | - Lijuan Bai
- Chongqing Research Center for Pharmaceutical Engineering, College of Pharmacy, Chongqing Medical University, Chongqing 400016, P. R. China.
| |
Collapse
|
19
|
Nabavi N, Mahdavi SR, Ardalan MA, Chamanara M, Mosaed R, Lara A, Bastos D, Harsini S, Askari E, Velho PI, Bagheri H. Bipolar Androgen Therapy: When Excess Fuel Extinguishes the Fire. Biomedicines 2023; 11:2084. [PMID: 37509723 PMCID: PMC10377678 DOI: 10.3390/biomedicines11072084] [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: 06/06/2023] [Revised: 07/12/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Androgen deprivation therapy (ADT) remains the cornerstone of advanced prostate cancer treatment. However, the progression towards castration-resistant prostate cancer is inevitable, as the cancer cells reactivate androgen receptor signaling and adapt to the castrate state through autoregulation of the androgen receptor. Additionally, the upfront use of novel hormonal agents such as enzalutamide and abiraterone acetate may result in long-term toxicities and may trigger the selection of AR-independent cells through "Darwinian" treatment-induced pressure. Therefore, it is crucial to develop new strategies to overcome these challenges. Bipolar androgen therapy (BAT) is one such approach that has been devised based on studies demonstrating the paradoxical inhibitory effects of supraphysiologic testosterone on prostate cancer growth, achieved through a variety of mechanisms acting in concert. BAT involves rapidly alternating testosterone levels between supraphysiological and near-castrate levels over a period of a month, achieved through monthly intramuscular injections of testosterone plus concurrent ADT. BAT is effective and well-tolerated, improving quality of life and potentially re-sensitizing patients to previous hormonal therapies after progression. By exploring the mechanisms and clinical evidence for BAT, this review seeks to shed light on its potential as a promising new approach to prostate cancer treatment.
Collapse
Affiliation(s)
- Nima Nabavi
- Nuclear Medicine Research Center, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran
- Radiation Sciences Research Center, AJA University of Medical Sciences, Tehran 14117-18541, Iran
| | - Seied Rabi Mahdavi
- Department of Medical Physics, Radiation Biology Research Center, Iran University of Medical Sciences, Tehran 14117-18541, Iran
| | - Mohammad Afshar Ardalan
- Department of Internal Medicine, School of Medicine, AJA University of Medical Sciences, Tehran 14117-18541, Iran
| | - Mohsen Chamanara
- Department of Pharmacology, School of Medicine, AJA University of Medical Sciences, Tehran 14117-18541, Iran
| | - Reza Mosaed
- Department of Clinical Pharmacy, School of Medicine, AJA University of Medical Sciences, Tehran 14117-18541, Iran
| | - Aline Lara
- Hospital Sírio-Libanês, São Paulo 01308-050, Brazil
- Hospital do Câncer UOPECCAN, Cascavel 85806-300, Brazil
| | - Diogo Bastos
- Oncology Department, Hospital Sirio-Libanês, São Paulo 01308-050, Brazil
| | - Sara Harsini
- BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
| | - Emran Askari
- Nuclear Medicine Research Center, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran
| | - Pedro Isaacsson Velho
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD 21231, USA
- Hospital Moinhos de Vento, Porto Alegre 90035-000, Brazil
| | - Hamed Bagheri
- Radiation Sciences Research Center, AJA University of Medical Sciences, Tehran 14117-18541, Iran
- School of Medicine, AJA University of Medical Sciences, Tehran 14118-13389, Iran
| |
Collapse
|
20
|
Goh V. Genitourinary Imaging in 2040. Radiology 2023; 307:e230223. [PMID: 37249430 PMCID: PMC10315527 DOI: 10.1148/radiol.230223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 05/31/2023]
Affiliation(s)
- Vicky Goh
- From the Department of Cancer Imaging, School of Biomedical
Engineering and Imaging Sciences, King’s College London, SE1 7EH,
United Kingdom; and Department of Radiology, Guy’s & St
Thomas’ NHS Foundation Trust, Level 1, Lambeth Wing, St Thomas’
Hospital, Westminster Bridge Rd, London, United Kingdom
| |
Collapse
|
21
|
Sayar E, Patel RA, Coleman IM, Roudier MP, Zhang A, Mustafi P, Low JY, Hanratty B, Ang LS, Bhatia V, Adil M, Bakbak H, Quigley DA, Schweizer MT, Hawley JE, Kollath L, True LD, Feng FY, Bander NH, Corey E, Lee JK, Morrissey C, Gulati R, Nelson PS, Haffner MC. Reversible epigenetic alterations mediate PSMA expression heterogeneity in advanced metastatic prostate cancer. JCI Insight 2023; 8:e162907. [PMID: 36821396 PMCID: PMC10132157 DOI: 10.1172/jci.insight.162907] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 02/21/2023] [Indexed: 02/24/2023] Open
Abstract
Prostate-specific membrane antigen (PSMA) is an important cell surface target in prostate cancer. There are limited data on the heterogeneity of PSMA tissue expression in metastatic castration-resistant prostate cancer (mCRPC). Furthermore, the mechanisms regulating PSMA expression (encoded by the FOLH1 gene) are not well understood. Here, we demonstrate that PSMA expression is heterogeneous across different metastatic sites and molecular subtypes of mCRPC. In a rapid autopsy cohort in which multiple metastatic sites per patient were sampled, we found that 13 of 52 (25%) cases had no detectable PSMA and 23 of 52 (44%) cases showed heterogeneous PSMA expression across individual metastases, with 33 (63%) cases harboring at least 1 PSMA-negative site. PSMA-negative tumors displayed distinct transcriptional profiles with expression of druggable targets such as MUC1. Loss of PSMA was associated with epigenetic changes of the FOLH1 locus, including gain of CpG methylation and loss of histone 3 lysine 27 (H3K27) acetylation. Treatment with histone deacetylase (HDAC) inhibitors reversed this epigenetic repression and restored PSMA expression in vitro and in vivo. Collectively, these data provide insights into the expression patterns and regulation of PSMA in mCRPC and suggest that epigenetic therapies - in particular, HDAC inhibitors - can be used to augment PSMA levels.
Collapse
Affiliation(s)
- Erolcan Sayar
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Radhika A. Patel
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Ilsa M. Coleman
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Martine P. Roudier
- Department of Urology, University of Washington (UW), Seattle, Washington, USA
| | - Ailin Zhang
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Pallabi Mustafi
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Jin-Yih Low
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Brian Hanratty
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Lisa S. Ang
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Vipul Bhatia
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Mohamed Adil
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Hasim Bakbak
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - David A. Quigley
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California, USA
| | - Michael T. Schweizer
- Division of Medical Oncology, Department of Medicine, UW, Seattle, Washington, USA
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Jessica E. Hawley
- Division of Medical Oncology, Department of Medicine, UW, Seattle, Washington, USA
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Lori Kollath
- Department of Urology, University of Washington (UW), Seattle, Washington, USA
| | - Lawrence D. True
- Department of Laboratory Medicine and Pathology, UW, Seattle, Washington, USA
| | - Felix Y. Feng
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California, USA
| | - Neil H. Bander
- Department of Urology, Weill Cornell Medicine, New York, New York, USA
| | - Eva Corey
- Department of Urology, University of Washington (UW), Seattle, Washington, USA
| | - John K. Lee
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Urology, Weill Cornell Medicine, New York, New York, USA
| | - Colm Morrissey
- Department of Urology, University of Washington (UW), Seattle, Washington, USA
| | - Roman Gulati
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Peter S. Nelson
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Urology, University of Washington (UW), Seattle, Washington, USA
- Division of Medical Oncology, Department of Medicine, UW, Seattle, Washington, USA
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Laboratory Medicine and Pathology, UW, Seattle, Washington, USA
| | - Michael C. Haffner
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Laboratory Medicine and Pathology, UW, Seattle, Washington, USA
| |
Collapse
|
22
|
Park MY, Park KJ, Kim MH, Kim JK. Focal nodular enhancement on DCE MRI of the prostatectomy bed: radiologic-pathologic correlations and prognostic value. Eur Radiol 2023; 33:2985-2994. [PMID: 36350389 DOI: 10.1007/s00330-022-09241-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 10/01/2022] [Accepted: 10/13/2022] [Indexed: 11/10/2022]
Abstract
OBJECTIVES To determine the concordance of dynamic contrast-enhanced (DCE) imaging findings with clinico-pathologic characteristics and their prognostic impact for predicting biochemical recurrence (BCR) in patients who underwent radical prostatectomy (RP) for prostate cancer. METHODS This retrospective study included patients who underwent MRI within 1 year after RP between November 2019 and October 2020. DCE findings and their concordance with the presence and location of positive surgical margin (PSM) were assessed using RP specimens. Kaplan-Meier and logistic regression analyses were used to evaluate the prognostic impact of DCE findings for BCR. RESULTS Among the 272 men (mean age ± standard deviation, 66.6 ± 7.4 years), focal nodular enhancement was more frequently observed in those with PSM compared to those with negative margin (85.4% versus 14.6%; p < 0.001). The sites of focal nodular enhancement were 72.9% (35/48) concordant with the PSM locations. Focal nodular enhancement was associated with a higher Gleason score, higher preoperative PSA (≥ 10 ng/mL), higher Gleason grade at the surgical margin, and non-limited margin involvement (p = 0.002, 0.006, 0.032, and 0.001, respectively). In patients without BCR at the time of MRI, focal nodular enhancement was associated with a shorter time to BCR (p < 0.001) and a significant factor predicting 1-year BCR in both univariate (odds ratio = 8.4 [95% CI: 2.5-28.3]; p = 0.001) and multivariate (odds ratio = 5.49 [1.56-19.3]; p = 0.008) analyses. CONCLUSIONS Focal nodular enhancement on post-prostatectomy MRI was associated with adverse clinico-pathologic characteristics of high risk for recurrence and can be a predictor for 1-year BCR in patients undergoing RP. KEY POINTS • Focal nodular enhancement (PI-RR DCE score ≥ 4) was 72.9% (35/48) concordant with the site of positive resection margin by radiologic-histologic correlation. • Focal nodular enhancement (PI-RR DCE score ≥ 4) was associated with higher Gleason score ( ≥ 8), preoperative PSA ( > 10 ng/mL), and Gleason grade 4 or 5 at the surgical margin and non-limited margin involvement (p ≤ 0.032). • In patients without BCR at the time of MRI, focal nodular enhancement was a significant factor predicting 1-year BCR (odds ratio = 5.49; 95% CI: 1.56-19.3; p = 0.008).
Collapse
Affiliation(s)
- Mi Yeon Park
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 86 Asanbyeongwon-Gil, Songpa-Gu, Seoul, 05505, Korea
| | - Kye Jin Park
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 86 Asanbyeongwon-Gil, Songpa-Gu, Seoul, 05505, Korea.
| | - Mi-Hyun Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 86 Asanbyeongwon-Gil, Songpa-Gu, Seoul, 05505, Korea
| | - Jeong Kon Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 86 Asanbyeongwon-Gil, Songpa-Gu, Seoul, 05505, Korea
| |
Collapse
|
23
|
Gillessen S, Bossi A, Davis ID, de Bono J, Fizazi K, James ND, Mottet N, Shore N, Small E, Smith M, Sweeney C, Tombal B, Antonarakis ES, Aparicio AM, Armstrong AJ, Attard G, Beer TM, Beltran H, Bjartell A, Blanchard P, Briganti A, Bristow RG, Bulbul M, Caffo O, Castellano D, Castro E, Cheng HH, Chi KN, Chowdhury S, Clarke CS, Clarke N, Daugaard G, De Santis M, Duran I, Eeles R, Efstathiou E, Efstathiou J, Ngozi Ekeke O, Evans CP, Fanti S, Feng FY, Fonteyne V, Fossati N, Frydenberg M, George D, Gleave M, Gravis G, Halabi S, Heinrich D, Herrmann K, Higano C, Hofman MS, Horvath LG, Hussain M, Jereczek-Fossa BA, Jones R, Kanesvaran R, Kellokumpu-Lehtinen PL, Khauli RB, Klotz L, Kramer G, Leibowitz R, Logothetis CJ, Mahal BA, Maluf F, Mateo J, Matheson D, Mehra N, Merseburger A, Morgans AK, Morris MJ, Mrabti H, Mukherji D, Murphy DG, Murthy V, Nguyen PL, Oh WK, Ost P, O'Sullivan JM, Padhani AR, Pezaro C, Poon DMC, Pritchard CC, Rabah DM, Rathkopf D, Reiter RE, Rubin MA, Ryan CJ, Saad F, Pablo Sade J, Sartor OA, Scher HI, Sharifi N, Skoneczna I, Soule H, Spratt DE, Srinivas S, Sternberg CN, Steuber T, Suzuki H, Sydes MR, Taplin ME, Tilki D, Türkeri L, Turco F, Uemura H, Uemura H, Ürün Y, Vale CL, van Oort I, Vapiwala N, Walz J, Yamoah K, Ye D, Yu EY, Zapatero A, Zilli T, Omlin A. Management of Patients with Advanced Prostate Cancer. Part I: Intermediate-/High-risk and Locally Advanced Disease, Biochemical Relapse, and Side Effects of Hormonal Treatment: Report of the Advanced Prostate Cancer Consensus Conference 2022. Eur Urol 2023; 83:267-293. [PMID: 36494221 PMCID: PMC7614721 DOI: 10.1016/j.eururo.2022.11.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 11/08/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Innovations in imaging and molecular characterisation and the evolution of new therapies have improved outcomes in advanced prostate cancer. Nonetheless, we continue to lack high-level evidence on a variety of clinical topics that greatly impact daily practice. To supplement evidence-based guidelines, the 2022 Advanced Prostate Cancer Consensus Conference (APCCC 2022) surveyed experts about key dilemmas in clinical management. OBJECTIVE To present consensus voting results for select questions from APCCC 2022. DESIGN, SETTING, AND PARTICIPANTS Before the conference, a panel of 117 international prostate cancer experts used a modified Delphi process to develop 198 multiple-choice consensus questions on (1) intermediate- and high-risk and locally advanced prostate cancer, (2) biochemical recurrence after local treatment, (3) side effects from hormonal therapies, (4) metastatic hormone-sensitive prostate cancer, (5) nonmetastatic castration-resistant prostate cancer, (6) metastatic castration-resistant prostate cancer, and (7) oligometastatic and oligoprogressive prostate cancer. Before the conference, these questions were administered via a web-based survey to the 105 physician panel members ("panellists") who directly engage in prostate cancer treatment decision-making. Herein, we present results for the 82 questions on topics 1-3. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Consensus was defined as ≥75% agreement, with strong consensus defined as ≥90% agreement. RESULTS AND LIMITATIONS The voting results reveal varying degrees of consensus, as is discussed in this article and shown in the detailed results in the Supplementary material. The findings reflect the opinions of an international panel of experts and did not incorporate a formal literature review and meta-analysis. CONCLUSIONS These voting results by a panel of international experts in advanced prostate cancer can help physicians and patients navigate controversial areas of clinical management for which high-level evidence is scant or conflicting. The findings can also help funders and policymakers prioritise areas for future research. Diagnostic and treatment decisions should always be individualised based on patient and cancer characteristics (disease extent and location, treatment history, comorbidities, and patient preferences) and should incorporate current and emerging clinical evidence, therapeutic guidelines, and logistic and economic factors. Enrolment in clinical trials is always strongly encouraged. Importantly, APCCC 2022 once again identified important gaps (areas of nonconsensus) that merit evaluation in specifically designed trials. PATIENT SUMMARY The Advanced Prostate Cancer Consensus Conference (APCCC) provides a forum to discuss and debate current diagnostic and treatment options for patients with advanced prostate cancer. The conference aims to share the knowledge of international experts in prostate cancer with health care providers and patients worldwide. At each APCCC, a panel of physician experts vote in response to multiple-choice questions about their clinical opinions and approaches to managing advanced prostate cancer. This report presents voting results for the subset of questions pertaining to intermediate- and high-risk and locally advanced prostate cancer, biochemical relapse after definitive treatment, advanced (next-generation) imaging, and management of side effects caused by hormonal therapies. The results provide a practical guide to help clinicians and patients discuss treatment options as part of shared multidisciplinary decision-making. The findings may be especially useful when there is little or no high-level evidence to guide treatment decisions.
Collapse
Affiliation(s)
- Silke Gillessen
- Oncology Institute of Southern Switzerland, EOC, Bellinzona, Switzerland; Università della Svizzera Italiana, Lugano, Switzerland.
| | - Alberto Bossi
- Genitourinary Oncology, Prostate Brachytherapy Unit, Gustave Roussy, Paris, France
| | - Ian D Davis
- Monash University and Eastern Health, Victoria, Australia
| | - Johann de Bono
- The Institute of Cancer Research, London, UK; Royal Marsden Hospital, London, UK
| | - Karim Fizazi
- Institut Gustave Roussy, University of Paris Saclay, Villejuif, France
| | | | | | - Neal Shore
- Carolina Urologic Research Center, Myrtle Beach, SC, USA; Urology/Surgical Oncology, GenesisCare, Myrtle Beach, SC, USA
| | - Eric Small
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | - Mathew Smith
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Christopher Sweeney
- Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | - Ana M Aparicio
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Andrew J Armstrong
- Duke Cancer Institute Center for Prostate and Urologic Cancers, Durham, NC, USA
| | | | - 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
| | - Pierre Blanchard
- Département de Radiothérapie, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Alberto Briganti
- Unit of Urology/Division of Oncology, URI, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, Milan, Italy
| | - Rob G Bristow
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, 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
| | - Daniel Castellano
- Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Elena Castro
- Institute of Biomedical Research in Málaga (IBIMA), Málaga, Spain
| | - Heather H Cheng
- Fred Hutchinson Cancer Center, University of Washington, Seattle, WA, USA
| | - Kim N Chi
- BC Cancer, Vancouver Prostate Centre, University of British Columbia, Vancouver, British Columbia, 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
| | - Gedske Daugaard
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Maria De Santis
- Department of Urology, Charité Universitätsmedizin, Berlin, Germany; Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Ignacio Duran
- Department of Medical Oncology, Hospital Universitario Marques de Valdecilla, IDIVAL, Santander, Cantabria, Spain
| | - Ros Eeles
- The Institute of Cancer Research and Royal Marsden NHS Foundation Trust, London, 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, Alakahia, Port Harcourt, Nigeria
| | | | - Stefano Fanti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Felix Y Feng
- University of California San Francisco, San Francisco, CA, USA
| | - Valerie Fonteyne
- Department of Radiation-Oncology, Ghent University Hospital, Ghent, Belgium
| | - Nicola Fossati
- Department of Urology, Ospedale Regionale di Lugano, Civico USI - Università della Svizzera Italiana, Lugano, Switzerland
| | - Mark Frydenberg
- Department of Surgery, Prostate Cancer Research Program, Monash University, Melbourne, Australia; Department of Anatomy & Developmental Biology, Faculty of Nursing, Medicine & Health Sciences, Monash University, Melbourne, Australia
| | - Daniel George
- Department of Medicine, Duke Cancer Institute, Duke University, Durham, NC, USA; Department of Surgery, Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Martin Gleave
- Urological Sciences, Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Gwenaelle Gravis
- Department of Medical Oncology, Institut Paoli Calmettes, Aix-Marseille Université, Marseille, France
| | - Susan Halabi
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Daniel Heinrich
- Department of Oncology and Radiotherapy, Innlandet Hospital Trust, Gjøvik, Norway
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | - Celestia Higano
- University of British Columbia, Vancouver, British Columbia, Canada
| | - 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
| | - Lisa G Horvath
- Chris O'Brien Lifehouse, Camperdown, NSW, Australia; Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, Australia; The University of Sydney, Sydney, NSW, Australia
| | - Maha Hussain
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Barbara Alicja Jereczek-Fossa
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Department of Radiotherapy, European Institute of Oncology (IEO) IRCCS, Milan, Italy
| | - Robert Jones
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | | | - Pirkko-Liisa Kellokumpu-Lehtinen
- Faculty of Medicine and Health Technology, Tampere University and Tampere Cancer Center, Tampere, Finland; Research, Development and Innovation Center, Tampere University Hospital, Tampere, Finland
| | - Raja B Khauli
- Department of Urology and the Naef K. Basile Cancer Institute (NKBCI), American University of Beirut Medical Center, Beirut, Lebanon
| | - Laurence Klotz
- Division of Urology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Gero Kramer
- Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Raya Leibowitz
- Oncology Institute, Shamir Medical Center, Be'er Ya'akov, Israel; Faculty of Medicine, Tel-Aviv University, Israel
| | - Christopher J Logothetis
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; University of Athens Alexandra Hospital, Athens, Greece
| | - Brandon A Mahal
- Department of Radiation Oncology, University of Miami Sylvester Cancer Center, Miami, FL, USA
| | - Fernando Maluf
- Beneficiência Portuguesa de São Paulo, São Paulo, SP, Brasil; Departamento de Oncologia, Hospital Israelita Albert Einstein, São Paulo, SP, Brazil
| | - Joaquin Mateo
- Department of Medical Oncology and Prostate Cancer Translational Research Group, Vall d'Hebron Institute of Oncology (VHIO) and Vall d'Hebron University Hospital, Barcelona, Spain
| | - David Matheson
- Faculty of Education, Health and Wellbeing, Walsall Campus, Walsall, UK
| | - Niven Mehra
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Axel Merseburger
- Department of Urology, University Hospital Schleswig-Holstein, Luebeck, Germany
| | - 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
| | - Deborah Mukherji
- Clemenceau Medical Center, Dubai, United Arab Emirates; Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Declan G Murphy
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Australia
| | | | - Paul L Nguyen
- Department of Radiation Oncology, Brigham and Women's Hospital and Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - 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, Northern Ireland
| | - Anwar R Padhani
- Mount Vernon Cancer Centre and Institute of Cancer Research, London, UK
| | - Carmel Pezaro
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Darren M C Poon
- Comprehensive Oncology Centre, Hong Kong Sanatorium & Hospital, Hong Kong; The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Colin C Pritchard
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Danny M Rabah
- Cancer Research Chair and Department of Surgery, College of Medicine, King Saud University, Riyadh, Saudi Arabia; Department of Urology, KFSHRC, Riyadh, Saudi Arabia
| | - Dana Rathkopf
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Mark A 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, Quebec, Canada
| | | | | | - 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
| | - Nima Sharifi
- Department of Hematology and Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland, OH, USA; Department of Cancer Biology, GU Malignancies Research Center, Cleveland Clinic Lerner Research Institute, Cleveland, OH, USA
| | - Iwona Skoneczna
- Rafal Masztak Grochowski Hospital, Maria Sklodowska Curie National Research Institute of Oncology, Warsaw, Poland
| | - Howard Soule
- Prostate Cancer Foundation, Santa Monica, CA, USA
| | - Daniel E Spratt
- University Hospitals Seidman Cancer Center, Cleveland, OH, USA
| | - Sandy Srinivas
- Division of Medical Oncology, Stanford University Medical Center, Stanford, CA, 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
| | - Thomas Steuber
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | | | - Matthew R Sydes
- MRC Clinical Trials Unit at UCL, Institute of Clinical Trials and Methodology, University College London, London, UK
| | - Mary-Ellen Taplin
- Department of Medical Oncology, 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
| | - Fabio Turco
- Oncology Institute of Southern Switzerland, EOC, Bellinzona, Switzerland
| | - Hiroji Uemura
- Yokohama City University Medical Center, Yokohama, Japan
| | - Hirotsugu Uemura
- Department of Urology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Yüksel Ürün
- Department of Medical Oncology, Ankara University School of Medicine, Ankara, Turkey; Ankara University Cancer Research Institute, Ankara, Turkey
| | - Claire L Vale
- University College London, MRC Clinical Trials Unit at UCL, London, UK
| | - Inge van Oort
- Radboud University Medical Center, Nijmegen, The Netherlands
| | - Neha Vapiwala
- Department of Radiation Oncology, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Jochen Walz
- Department of Urology, Institut Paoli-Calmettes Cancer Centre, Marseille, France
| | - 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
| | - Evan Y Yu
- Department of Medicine, Division of Oncology, University of Washington and Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Almudena Zapatero
- Department of Radiation Oncology, Hospital Universitario de La Princesa, Health Research Institute, Madrid, Spain
| | - Thomas Zilli
- Radiation Oncology, Oncology Institute of Southern Switzerland, EOC, Bellinzona, Switzerland; Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Aurelius Omlin
- Onkozentrum Zurich, University of Zurich and Tumorzentrum Hirslanden Zurich, Switzerland
| |
Collapse
|
24
|
Parihar AS, Hofman MS, Iravani A. 177Lu-Prostate-specific Membrane Antigen Radioligand Therapy in Patients with Metastatic Castration-resistant Prostate Cancer. Radiology 2023; 306:e220859. [PMID: 36125377 DOI: 10.1148/radiol.220859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
A 76-year-old man with metastatic castration-resistant prostate carcinoma progressing with antiandrogen and taxane therapy was treated with lutetium 177 prostate-specific membrane antigen (PSMA)-617 and showed marked biochemical and imaging response, with improvement in clinical status and osseous pain. A summary of nuclear medicine theranostics with emphasis on PSMA targeting agents is presented.
Collapse
Affiliation(s)
- Ashwin Singh Parihar
- From the Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4525 Scott Ave, Suite 3433, MIR East Building, St Louis, MO 63110 (A.S.P., A.I.); and Department of Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia (M.S.H.)
| | - Michael S Hofman
- From the Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4525 Scott Ave, Suite 3433, MIR East Building, St Louis, MO 63110 (A.S.P., A.I.); and Department of Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia (M.S.H.)
| | - Amir Iravani
- From the Mallinckrodt Institute of Radiology, Washington University School of Medicine, 4525 Scott Ave, Suite 3433, MIR East Building, St Louis, MO 63110 (A.S.P., A.I.); and Department of Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, Australia (M.S.H.)
| |
Collapse
|
25
|
Kunert JP, Müller M, Günther T, Stopper L, Urtz-Urban N, Beck R, Wester HJ. Synthesis and preclinical evaluation of novel 99mTc-labeled PSMA ligands for radioguided surgery of prostate cancer. EJNMMI Res 2023; 13:2. [PMID: 36645586 PMCID: PMC9842843 DOI: 10.1186/s13550-022-00942-7] [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: 07/28/2022] [Accepted: 10/15/2022] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Radioguided surgery (RGS) has recently emerged as a valuable new tool in the management of recurrent prostate cancer (PCa). After preoperative injection of a 99mTc-labeled prostate-specific membrane antigen (PSMA) inhibitor, radioguided intraoperative identification and resection of lesions is facilitated by means of suitable γ-probes. First clinical experiences show the feasibility of RGS and suggest superiority over conventional lymph node dissection in recurrent PCa. However, commonly used [99mTc]Tc-PSMA-I&S exhibits slow whole-body clearance, thus hampering optimal tumor-to-background ratios (TBR) during surgery. We therefore aimed to develop novel 99mTc-labeled, PSMA-targeted radioligands with optimized pharmacokinetic profile to increase TBR at the time of surgery. METHODS Three 99mTc-labeled N4-PSMA ligands were preclinically evaluated and compared to [99mTc]Tc-PSMA-I&S. PSMA affinity (IC50) and internalization were determined on LNCaP cells. Lipophilicity was assessed by means of the distribution coefficient logD7.4 and an ultrafiltration method was used to determine binding to human plasma proteins. Biodistribution studies and static µSPECT/CT-imaging were performed at 6 h p.i. on LNCaP tumor-bearing CB17-SCID mice. RESULTS The novel N4-PSMA tracers were readily labeled with [99mTc]TcO4- with RCP > 95%. Comparable and high PSMA affinity was observed for all [99mTc]Tc-N4-PSMA-ligands. The ligands showed variable binding to human plasma and medium to low lipophilicity (logD7.4 - 2.6 to - 3.4), both consistently decreased compared to [99mTc]Tc-PSMA-I&S. Biodistribution studies revealed comparable tumor uptake among all [99mTc]Tc-N4-PSMA-ligands and [99mTc]Tc-PSMA-I&S, while clearance from most organs was superior for the novel tracers. Accordingly, increased TBR were achieved. [99mTc]Tc-N4-PSMA-12 showed higher TBR than [99mTc]Tc-PSMA-I&S for blood and all evaluated tissue. In addition, a procedure suitable for routine clinical production of [99mTc]Tc-N4-PSMA-12 was established. Labeling with 553 ± 187 MBq was achieved with RCP of 98.5 ± 0.6% (n = 10). CONCLUSION High tumor accumulation and favorable clearance from blood and non-target tissue make [99mTc]Tc-N4-PSMA-12 an attractive tracer for RGS, possibly superior to currently established [99mTc]Tc-PSMA-I&S. Its GMP-production according to a method presented here and first clinical investigations with this novel radioligand is highly recommended.
Collapse
Affiliation(s)
- Jan-Philip Kunert
- Chair of Pharmaceutical Radiochemistry, Department of Chemistry, Technical University of Munich (TUM), Walther-Meißner-Str 3, 85748, Garching, Germany.
| | - Max Müller
- grid.6936.a0000000123222966Chair of Pharmaceutical Radiochemistry, Department of Chemistry, Technical University of Munich (TUM), Walther-Meißner-Str 3, 85748 Garching, Germany
| | - Thomas Günther
- grid.6936.a0000000123222966Chair of Pharmaceutical Radiochemistry, Department of Chemistry, Technical University of Munich (TUM), Walther-Meißner-Str 3, 85748 Garching, Germany
| | - León Stopper
- grid.6936.a0000000123222966Chair of Pharmaceutical Radiochemistry, Department of Chemistry, Technical University of Munich (TUM), Walther-Meißner-Str 3, 85748 Garching, Germany
| | - Nicole Urtz-Urban
- grid.6936.a0000000123222966Chair of Pharmaceutical Radiochemistry, Department of Chemistry, Technical University of Munich (TUM), Walther-Meißner-Str 3, 85748 Garching, Germany
| | - Roswitha Beck
- grid.6936.a0000000123222966Chair of Pharmaceutical Radiochemistry, Department of Chemistry, Technical University of Munich (TUM), Walther-Meißner-Str 3, 85748 Garching, Germany
| | - Hans-Jürgen Wester
- grid.6936.a0000000123222966Chair of Pharmaceutical Radiochemistry, Department of Chemistry, Technical University of Munich (TUM), Walther-Meißner-Str 3, 85748 Garching, Germany
| |
Collapse
|
26
|
Roberts MJ, Maurer T, Perera M, Eiber M, Hope TA, Ost P, Siva S, Hofman MS, Murphy DG, Emmett L, Fendler WP. Using PSMA imaging for prognostication in localized and advanced prostate cancer. Nat Rev Urol 2023; 20:23-47. [PMID: 36473945 DOI: 10.1038/s41585-022-00670-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [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
|
27
|
Zhang J, Lu T, Lu S, Ma S, Han D, Zhang K, Xu C, Liu S, Gan L, Wu X, Yang F, Wen W, Qin W. Single-cell analysis of multiple cancer types reveals differences in endothelial cells between tumors and normal tissues. Comput Struct Biotechnol J 2022; 21:665-676. [PMID: 36659929 PMCID: PMC9826920 DOI: 10.1016/j.csbj.2022.12.049] [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: 10/09/2022] [Revised: 12/28/2022] [Accepted: 12/29/2022] [Indexed: 12/31/2022] Open
Abstract
Endothelial cells (ECs) play an important role in tumor progression. Currently, the main target of anti-angiogenic therapy is the vascular endothelial growth factor (VEGF) pathway. Some patients do benefit from anti-VEGF/VEGFR therapy; however, a large number of patients do not have response or acquire drug resistance after treatment. Moreover, anti-VEGF/VEGFR therapy may lead to nephrotoxicity and cardiovascular-related side effects due to its action on normal ECs. Therefore, it is necessary to identify targets that are specific to tumor ECs and could be applied to various cancer types. We integrated single-cell RNA sequencing data from six cancer types and constructed a multi-cancer EC atlas to decode the characteristic of tumor ECs. We found that tip-like ECs mainly exist in tumor tissues but barely exist in normal tissues. Tip-like ECs are involved in the promotion of tumor angiogenesis and inhibition on anti-tumor immune responses. Moreover, tumor cells, myeloid cells, and pericytes are the main sources of pro-angiogenic factors. High proportion of tip-like ECs is associated with poor prognosis in multiple cancer types. We also identified that prostate-specific membrane antigen (PSMA) is a specific marker for tip-like ECs in all the cancer types we studied. In summary, we demonstrate that tip-like ECs are the main differential EC subcluster between tumors and normal tissues. Tip-like ECs may promote tumor progression through promoting angiogenesis while inhibiting anti-tumor immune responses. PSMA was a specific marker for tip-like ECs, which could be used as a potential target for the diagnosis and treatment of non-prostate cancers.
Collapse
Key Words
- BRCA, Breast invasive carcinoma
- CESC, Cervical squamous cell carcinoma and endocervical adenocarcinoma
- CRC, Colorectal cancer
- ECs, Endothelial cells
- Endothelial cells
- GC, Gastric cancer
- HNSC, Head and Neck squamous cell carcinoma
- KICH, Kidney chromophobe
- KIRC, Kidney renal clear cell carcinoma
- KIRP, Kidney renal papillary cell carcinoma
- LC, Lung cancer
- LIHC, Liver hepatocellular carcinoma
- LUAD, Lung adenocarcinoma
- LUSC, Lung squamous cell carcinoma
- OV, Ovarian serous cystadenocarcinoma
- OVC, Ovarian cancer
- PAAD, Pancreatic adenocarcinoma
- PDAC, Pancreatic ductal adenocarcinoma
- PRAD, Prostate adenocarcinoma
- PSMA, Prostate-specific membrane antigen
- RCC, Renal cell carcinoma
- READ, Rectum adenocarcinoma
- STAD, Stomach adenocarcinoma
- Single-cell RNA sequencing
- TME, Tumor microenvironment
- Tumor microenvironment
- VEGF, Vascular endothelial growth factor
- scRNA-seq, Single-cell RNA sequencing
Collapse
Affiliation(s)
- Jiayu Zhang
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Tong Lu
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Shiqi Lu
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| | - Shuaijun Ma
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Donghui Han
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Keying Zhang
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Chao Xu
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Shaojie Liu
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Lunbiao Gan
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| | - Xinjie Wu
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China
| | - Fa Yang
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi’an, China,Correspondence to: Department of Urology, Xijing Hospital, Air Force Medical University, 127 Changle West Road, Xi'an, China.
| | - Weihong Wen
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an, China,Correspondence to: Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Xi'an, China.
| | - Weijun Qin
- Department of Urology, Xijing Hospital, Air Force Medical University, Xi’an, China,Correspondence to: Department of Urology, Xijing Hospital, Air Force Medical University, 127 Changle West Road, Xi'an, China.
| |
Collapse
|
28
|
Imber BS, O’Dwyer E, Lobaugh S, McBride SM, Hopkins M, Kollmeier M, Gorovets D, Brennan V, Pike LR, Gewanter R, Mychalczak B, Zhang Z, Schöder H, Zelefsky MJ. Failure Patterns by PSMA PET for Recurrent Prostate Cancer after Prostatectomy and Salvage Radiation. Urology 2022; 170:146-153. [PMID: 36115426 PMCID: PMC10576466 DOI: 10.1016/j.urology.2022.08.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/17/2022] [Accepted: 08/22/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To characterize patterns of failure using prostate-specific membrane antigen positron emission tomography (PSMA PET) after radical prostatectomy (RP) and salvage radiotherapy (SRT). METHODS Patients with rising PSA post-RP+SRT underwent 68Ga-HBED-iPSMA PET/CT on a single-arm, prospective imaging trial (NCT03204123). Scans were centrally reviewed with pattern-of-failure analysis by involved site. Positive scans were classified using 3 failure categories: pelvic nodal, extra-pelvic nodal or distant non-nodal. Associations with failure categories were analyzed using cumulative incidence and generalized logits regression. RESULTS We included 133 men who received SRT a median of 20 months post-RP; 56% received SRT to the prostatic fossa alone, while 44% received pelvic SRT. PSMA PET/CT was performed a median of 48 months post-SRT. Overall, 31% of PSMA PET/CT scans were negative, 2% equivocal and 67% had at least 1 positive site. Scan detection was significantly associated with PSA level prior to PSMA PET/CT. Analysis of 89 positive scans demonstrated pelvic nodal (53%) was the most common relapse and fossa relapse was low (9%). Overall, positive scans were pelvic (n = 35, 26%), extra-pelvic nodal (n = 26, 20%) or distant non-nodal failure (n = 28, 21%), and 70% of positive scans were oligorecurrent. We observed similar cumulative incidence for all failure categories and relatively few clinicodemographic associations. Men treated with pelvic SRT had reduced odds of pelvic failure versus exclusive fossa treatment. CONCLUSION Pelvic, extra-pelvic nodal, and distant non-nodal failures occur with similar incidence post-SRT. Regional nodal relapse is relatively common, especially with fossa-only SRT. A high oligorecurrence rate suggests a potentially important role for PSMA-guided focal therapies.
Collapse
Affiliation(s)
- Brandon S. Imber
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Elisabeth O’Dwyer
- Molecular Imaging and Therapeutics, Department of Radiology, Weill Cornell Medical College, New York, NY 10065
| | - Stephanie Lobaugh
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Sean M. McBride
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Margaret Hopkins
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Marisa Kollmeier
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Daniel Gorovets
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Victoria Brennan
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Luke R.G. Pike
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Richard Gewanter
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Borys Mychalczak
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Zhigang Zhang
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| | - Heiko Schöder
- Department of Radiology, Molecular Imaging and Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael J. Zelefsky
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| |
Collapse
|
29
|
Ayesa SL, Murphy A. Positron emission tomography: Evolving modalities, radiopharmaceuticals and professional collaboration. J Med Radiat Sci 2022; 69:415-418. [DOI: 10.1002/jmrs.629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022] Open
Affiliation(s)
- Sally L. Ayesa
- Department of Medical Imaging & Nuclear Medicine Gosford & Wyong Hospitals Gosford New South Wales Australia
- Department of Nuclear Medicine Royal North Shore Hospital St Leonards New South Wales Australia
- School of Medicine University of Sydney Campderdown New South Wales Australia
| | - Andrew Murphy
- Department of Medical Imaging Princess Alexandra Hospital Woolloongabba QLD Australia
- School of Clinical Sciences Faculty of Health Queensland University of Technology Brisbane QLD Australia
| |
Collapse
|
30
|
Ulaner GA, Thomsen B, Bassett J, Torrey R, Cox C, Lin K, Patel T, Techasith T, Mauguen A, Rowe SP, Lindenberg L, Mena E, Choyke P, Yoshida J. 18F-DCFPyL PET/CT for Initially Diagnosed and Biochemically Recurrent Prostate Cancer: Prospective Trial with Pathologic Confirmation. Radiology 2022; 305:419-428. [PMID: 35852431 PMCID: PMC9619197 DOI: 10.1148/radiol.220218] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 05/11/2022] [Accepted: 05/17/2022] [Indexed: 08/26/2023]
Abstract
Background Prostate-specific membrane antigen (PSMA) PET is standard for newly diagnosed high-risk and biochemically recurrent (BCR) prostate cancer. Although studies suggest high specificity of 2-(3-{1-carboxy-5-[(6-[(18)F]fluoro-pyridine-3-carbonyl)-amino]-pentyl}-ureido)-pentanedioic acid (DCFPyL) for targeting PSMA, false-positive findings have been identified and most studies lack histologic confirmation of malignancy. Purpose To estimate the positive predictive value (PPV) of DCFPyL PET/CT by providing histopathologic proof for DCFPyL-avid lesions suspected of being distant metastases at initial diagnosis and recurrence in BCR prostate cancer. Materials and Methods In this prospective trial, men with newly diagnosed high-risk prostate cancer (sample 1) or BCR prostate cancer and negative findings at conventional CT and/or bone scanning (sample 2) were enrolled between January and December 2021. All men underwent DCFPyL PET/CT. Suspected distant metastases and/or recurrences were biopsied. PPV was calculated. Results A total of 92 men with newly diagnosed prostate cancer (median age, 70 years; IQR, 64-75 years) (sample 1) and 92 men with BCR prostate cancer (median age, 71 years; IQR, 66-75 years) (sample 2) were enrolled. In sample 1, 25 of the 92 men (27%) demonstrated DCFPyL-avid lesions suspicious for distant metastases. Biopsy was performed in 23 of the 25 men (92%), with 17 of the 23 (74%) biopsies positive for malignancy and six (26%) benign. Of the six benign biopsies, three were solitary rib foci and three were solitary pelvic bone foci. In sample 2, 57 of the 92 men (62%) demonstrated DCFPyL-avid lesions suspicious for recurrence. Biopsy was performed in 37 of the 57 men (65%), with 33 of the 37 (89%) biopsies positive for malignancy and four (11%) benign. Of the four benign biopsies, two were subcentimeter pelvic nodes and/or nodules, one was a rib, and one was a pelvic bone focus. Conclusion PET/CT with 2-(3-{1-carboxy-5-[(6-[(18)F]fluoro-pyridine-3-carbonyl)-amino]-pentyl}-ureido)-pentanedioic acid (DCFPyL) had a high biopsy-proven positive predictive value for distant metastases in newly diagnosed prostate cancer (74%) and for recurrence sites in men with biochemical recurrence (89%). However, there were DCFPyL-avid false-positive findings (particularly in ribs and pelvic bones). Solitary DCFPyL avidity in these locations should not be presumed as malignant. Biopsy may still be needed prior to therapy decisions. ClinicalTrials.gov registration no. NCT04700332 © RSNA, 2022 See also the editorial by Zukotynski and Kuo in this issue.
Collapse
Affiliation(s)
- Gary A. Ulaner
- From the Departments of Molecular Imaging and Therapy (G.A.U., B.T.),
Urology (J.B., R.T., J.Y.), Radiation Oncology (C.C., K.L.), and Radiology
(T.P., T.T.), Hoag Family Cancer Institute, 16105 Sand Canyon Ave, Irvine, CA
92618; Departments of Radiology and Translational Genomics, University of
Southern California, Los Angeles, Calif (G.A.U.); Department of Epidemiology and
Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY (A.M.); The
Russell H. Morgan Department of Radiology and Radiological Science, Johns
Hopkins University School of Medicine, Baltimore, Md (S.P.R.); and Molecular
Imaging Branch, National Cancer Institute, National Institutes of Health,
Bethesda, Md (L.L., E.M., P.C.)
| | - Beth Thomsen
- From the Departments of Molecular Imaging and Therapy (G.A.U., B.T.),
Urology (J.B., R.T., J.Y.), Radiation Oncology (C.C., K.L.), and Radiology
(T.P., T.T.), Hoag Family Cancer Institute, 16105 Sand Canyon Ave, Irvine, CA
92618; Departments of Radiology and Translational Genomics, University of
Southern California, Los Angeles, Calif (G.A.U.); Department of Epidemiology and
Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY (A.M.); The
Russell H. Morgan Department of Radiology and Radiological Science, Johns
Hopkins University School of Medicine, Baltimore, Md (S.P.R.); and Molecular
Imaging Branch, National Cancer Institute, National Institutes of Health,
Bethesda, Md (L.L., E.M., P.C.)
| | - Jeffrey Bassett
- From the Departments of Molecular Imaging and Therapy (G.A.U., B.T.),
Urology (J.B., R.T., J.Y.), Radiation Oncology (C.C., K.L.), and Radiology
(T.P., T.T.), Hoag Family Cancer Institute, 16105 Sand Canyon Ave, Irvine, CA
92618; Departments of Radiology and Translational Genomics, University of
Southern California, Los Angeles, Calif (G.A.U.); Department of Epidemiology and
Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY (A.M.); The
Russell H. Morgan Department of Radiology and Radiological Science, Johns
Hopkins University School of Medicine, Baltimore, Md (S.P.R.); and Molecular
Imaging Branch, National Cancer Institute, National Institutes of Health,
Bethesda, Md (L.L., E.M., P.C.)
| | - Robert Torrey
- From the Departments of Molecular Imaging and Therapy (G.A.U., B.T.),
Urology (J.B., R.T., J.Y.), Radiation Oncology (C.C., K.L.), and Radiology
(T.P., T.T.), Hoag Family Cancer Institute, 16105 Sand Canyon Ave, Irvine, CA
92618; Departments of Radiology and Translational Genomics, University of
Southern California, Los Angeles, Calif (G.A.U.); Department of Epidemiology and
Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY (A.M.); The
Russell H. Morgan Department of Radiology and Radiological Science, Johns
Hopkins University School of Medicine, Baltimore, Md (S.P.R.); and Molecular
Imaging Branch, National Cancer Institute, National Institutes of Health,
Bethesda, Md (L.L., E.M., P.C.)
| | - Craig Cox
- From the Departments of Molecular Imaging and Therapy (G.A.U., B.T.),
Urology (J.B., R.T., J.Y.), Radiation Oncology (C.C., K.L.), and Radiology
(T.P., T.T.), Hoag Family Cancer Institute, 16105 Sand Canyon Ave, Irvine, CA
92618; Departments of Radiology and Translational Genomics, University of
Southern California, Los Angeles, Calif (G.A.U.); Department of Epidemiology and
Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY (A.M.); The
Russell H. Morgan Department of Radiology and Radiological Science, Johns
Hopkins University School of Medicine, Baltimore, Md (S.P.R.); and Molecular
Imaging Branch, National Cancer Institute, National Institutes of Health,
Bethesda, Md (L.L., E.M., P.C.)
| | - Kevin Lin
- From the Departments of Molecular Imaging and Therapy (G.A.U., B.T.),
Urology (J.B., R.T., J.Y.), Radiation Oncology (C.C., K.L.), and Radiology
(T.P., T.T.), Hoag Family Cancer Institute, 16105 Sand Canyon Ave, Irvine, CA
92618; Departments of Radiology and Translational Genomics, University of
Southern California, Los Angeles, Calif (G.A.U.); Department of Epidemiology and
Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY (A.M.); The
Russell H. Morgan Department of Radiology and Radiological Science, Johns
Hopkins University School of Medicine, Baltimore, Md (S.P.R.); and Molecular
Imaging Branch, National Cancer Institute, National Institutes of Health,
Bethesda, Md (L.L., E.M., P.C.)
| | - Trushar Patel
- From the Departments of Molecular Imaging and Therapy (G.A.U., B.T.),
Urology (J.B., R.T., J.Y.), Radiation Oncology (C.C., K.L.), and Radiology
(T.P., T.T.), Hoag Family Cancer Institute, 16105 Sand Canyon Ave, Irvine, CA
92618; Departments of Radiology and Translational Genomics, University of
Southern California, Los Angeles, Calif (G.A.U.); Department of Epidemiology and
Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY (A.M.); The
Russell H. Morgan Department of Radiology and Radiological Science, Johns
Hopkins University School of Medicine, Baltimore, Md (S.P.R.); and Molecular
Imaging Branch, National Cancer Institute, National Institutes of Health,
Bethesda, Md (L.L., E.M., P.C.)
| | - Tust Techasith
- From the Departments of Molecular Imaging and Therapy (G.A.U., B.T.),
Urology (J.B., R.T., J.Y.), Radiation Oncology (C.C., K.L.), and Radiology
(T.P., T.T.), Hoag Family Cancer Institute, 16105 Sand Canyon Ave, Irvine, CA
92618; Departments of Radiology and Translational Genomics, University of
Southern California, Los Angeles, Calif (G.A.U.); Department of Epidemiology and
Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY (A.M.); The
Russell H. Morgan Department of Radiology and Radiological Science, Johns
Hopkins University School of Medicine, Baltimore, Md (S.P.R.); and Molecular
Imaging Branch, National Cancer Institute, National Institutes of Health,
Bethesda, Md (L.L., E.M., P.C.)
| | - Audrey Mauguen
- From the Departments of Molecular Imaging and Therapy (G.A.U., B.T.),
Urology (J.B., R.T., J.Y.), Radiation Oncology (C.C., K.L.), and Radiology
(T.P., T.T.), Hoag Family Cancer Institute, 16105 Sand Canyon Ave, Irvine, CA
92618; Departments of Radiology and Translational Genomics, University of
Southern California, Los Angeles, Calif (G.A.U.); Department of Epidemiology and
Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY (A.M.); The
Russell H. Morgan Department of Radiology and Radiological Science, Johns
Hopkins University School of Medicine, Baltimore, Md (S.P.R.); and Molecular
Imaging Branch, National Cancer Institute, National Institutes of Health,
Bethesda, Md (L.L., E.M., P.C.)
| | - Steven P. Rowe
- From the Departments of Molecular Imaging and Therapy (G.A.U., B.T.),
Urology (J.B., R.T., J.Y.), Radiation Oncology (C.C., K.L.), and Radiology
(T.P., T.T.), Hoag Family Cancer Institute, 16105 Sand Canyon Ave, Irvine, CA
92618; Departments of Radiology and Translational Genomics, University of
Southern California, Los Angeles, Calif (G.A.U.); Department of Epidemiology and
Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY (A.M.); The
Russell H. Morgan Department of Radiology and Radiological Science, Johns
Hopkins University School of Medicine, Baltimore, Md (S.P.R.); and Molecular
Imaging Branch, National Cancer Institute, National Institutes of Health,
Bethesda, Md (L.L., E.M., P.C.)
| | - Liza Lindenberg
- From the Departments of Molecular Imaging and Therapy (G.A.U., B.T.),
Urology (J.B., R.T., J.Y.), Radiation Oncology (C.C., K.L.), and Radiology
(T.P., T.T.), Hoag Family Cancer Institute, 16105 Sand Canyon Ave, Irvine, CA
92618; Departments of Radiology and Translational Genomics, University of
Southern California, Los Angeles, Calif (G.A.U.); Department of Epidemiology and
Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY (A.M.); The
Russell H. Morgan Department of Radiology and Radiological Science, Johns
Hopkins University School of Medicine, Baltimore, Md (S.P.R.); and Molecular
Imaging Branch, National Cancer Institute, National Institutes of Health,
Bethesda, Md (L.L., E.M., P.C.)
| | - Esther Mena
- From the Departments of Molecular Imaging and Therapy (G.A.U., B.T.),
Urology (J.B., R.T., J.Y.), Radiation Oncology (C.C., K.L.), and Radiology
(T.P., T.T.), Hoag Family Cancer Institute, 16105 Sand Canyon Ave, Irvine, CA
92618; Departments of Radiology and Translational Genomics, University of
Southern California, Los Angeles, Calif (G.A.U.); Department of Epidemiology and
Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY (A.M.); The
Russell H. Morgan Department of Radiology and Radiological Science, Johns
Hopkins University School of Medicine, Baltimore, Md (S.P.R.); and Molecular
Imaging Branch, National Cancer Institute, National Institutes of Health,
Bethesda, Md (L.L., E.M., P.C.)
| | - Peter Choyke
- From the Departments of Molecular Imaging and Therapy (G.A.U., B.T.),
Urology (J.B., R.T., J.Y.), Radiation Oncology (C.C., K.L.), and Radiology
(T.P., T.T.), Hoag Family Cancer Institute, 16105 Sand Canyon Ave, Irvine, CA
92618; Departments of Radiology and Translational Genomics, University of
Southern California, Los Angeles, Calif (G.A.U.); Department of Epidemiology and
Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY (A.M.); The
Russell H. Morgan Department of Radiology and Radiological Science, Johns
Hopkins University School of Medicine, Baltimore, Md (S.P.R.); and Molecular
Imaging Branch, National Cancer Institute, National Institutes of Health,
Bethesda, Md (L.L., E.M., P.C.)
| | - Jeffrey Yoshida
- From the Departments of Molecular Imaging and Therapy (G.A.U., B.T.),
Urology (J.B., R.T., J.Y.), Radiation Oncology (C.C., K.L.), and Radiology
(T.P., T.T.), Hoag Family Cancer Institute, 16105 Sand Canyon Ave, Irvine, CA
92618; Departments of Radiology and Translational Genomics, University of
Southern California, Los Angeles, Calif (G.A.U.); Department of Epidemiology and
Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY (A.M.); The
Russell H. Morgan Department of Radiology and Radiological Science, Johns
Hopkins University School of Medicine, Baltimore, Md (S.P.R.); and Molecular
Imaging Branch, National Cancer Institute, National Institutes of Health,
Bethesda, Md (L.L., E.M., P.C.)
| |
Collapse
|
31
|
Li M, Zelchan R, Orlova A. The Performance of FDA-Approved PET Imaging Agents in the Detection of Prostate Cancer. Biomedicines 2022; 10:biomedicines10102533. [PMID: 36289795 PMCID: PMC9599369 DOI: 10.3390/biomedicines10102533] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 10/01/2022] [Accepted: 10/06/2022] [Indexed: 11/23/2022] Open
Abstract
Positron emission tomography (PET) incorporated with X-ray computed tomography (PET/CT) or magnetic resonance imaging (PET/MRI) is increasingly being used as a diagnostic tool for prostate cancer (PCa). In this review, we describe and evaluate the clinical performance of some Food and Drug Administration (FDA)-approved agents used for visualizing PCa: [18F]FDG, [11C]choline, [18F]FACBC, [68Ga]Ga-PSMA-11, [18F]DCFPyL, and [18F]-NaF. We carried out a comprehensive literature search based on articles published from 1 January 2010 to 1 March 2022. We selected English language articles associated with the discovery, preclinical study, clinical study, and diagnostic performance of the imaging agents for the evaluation. Prostate-specific membrane antigen (PSMA)-targeted imaging agents demonstrated superior diagnostic performance in both primary and recurrent PCa, compared with [11C]choline and [18F]FACBC, both of which target dividing cells and are used especially in patients with low prostate-specific antigen (PSA) values. When compared to [18F]-NaF (which is suitable for the detection of bone metastases), PSMA-targeted agents were also capable of detecting lesions in the lymph nodes, soft tissues, and bone. However, a limitation of PSMA-targeted imaging was the heterogeneity of PSMA expression in PCa, and consequently, a combination of two PET tracers was proposed to overcome this obstacle. The preliminary studies indicated that the use of PSMA-targeted scanning is more cost efficient than conventional imaging modalities for high-risk PCa patients. Furthering the development of imaging agents that target PCa-associated receptors and molecules could improve PET-based diagnosis of PCa.
Collapse
Affiliation(s)
- Mei Li
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden
- Liaoning Medical Device Test Institute, Shenyang 110171, China
| | - Roman Zelchan
- Department of Nuclear Medicine, Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 5 Kooperativny St., 634009 Tomsk, Russia
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
| | - Anna Orlova
- Department of Medicinal Chemistry, Uppsala University, 751 23 Uppsala, Sweden
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia
- Science for Life Laboratory, Uppsala University, 752 37 Uppsala, Sweden
- Correspondence:
| |
Collapse
|
32
|
Huang YT, Tseng NC, Chen YK, Huang KH, Lin HY, Huang YY, Hwang TIS, Ou YC. The Detection Performance of 18 F-Prostate-Specific Membrane Antigen-1007 PET/CT in Primary Prostate Cancer : A Systemic Review and Meta-analysis. Clin Nucl Med 2022; 47:755-762. [PMID: 35452013 DOI: 10.1097/rlu.0000000000004228] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
BACKGROUND Multiple tools are now available to determine the requirement for a biopsy to diagnose prostate cancer, and PET/CT with radiolabeled prostate-specific membrane antigen (PSMA)-targeting radiotracers has been recommended for detecting primary prostate cancer. Particularly, the radiotracer 18 F-PSMA-1007 was found to be more favorable for primary tumors compared with other PSMA-targeting radiotracers because of its low clearance via the urinary tract and better image resolution. Thus, we performed a systematic review and meta-analysis to more accurately evaluate the detection performance of 18 F-PSMA-1007 PET/CT in primary prostate cancer patients. METHODS An update on the databases of PubMed/MEDLINE, EMBASE, and Cochrane Library for comprehensive literature search was performed on September 30, 2021. The pooling detection rate was calculated on a per-patient basis. The pooling median of the SUV max was analyzed from the included studies. Furthermore, the positive predictive value of 18 F-PSMA-1007 PET/CT with pathologic lesions was analyzed using the criterion standard. RESULTS Twelve studies (540 patients total) were included in the meta-analysis. The overall pooling detection rate of 18 F-PSMA-1007 per patient was 94%, and the pooling median of SUV max located at the intraprostate tumor was 16 (range, 3.7-77.7). The positive predictive value of 18 F-PSMA-1007 per lesion with histopathological validation was 0.90, detecting regional lymph node metastasis was 0.94, and detecting localized prostatic tumors was 0.84. CONCLUSIONS In the current meta-analysis, we revealed the excellent performance of 18 F-PSMA-1007 to detect localized prostatic tumor lesions and regional lymph node metastasis. Moreover, the uptake of localized tumors in primary prostate cancer was nearly liver uptake and may be considered a suspicious malignancy if it was equal to or greater than the liver uptake.
Collapse
|
33
|
Positron Emission Tomography Radiopharmaceuticals in Differentiated Thyroid Cancer. Molecules 2022; 27:molecules27154936. [PMID: 35956886 PMCID: PMC9370596 DOI: 10.3390/molecules27154936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/01/2022] [Accepted: 08/01/2022] [Indexed: 11/29/2022] Open
Abstract
Differentiated thyroid cancer (DTC), arising from thyroid follicular epithelial cells, is the most common type of thyroid cancer. Despite the well-known utilization of radioiodine treatment in DTC, i.e., iodine-131, radioiodine imaging in DTC is typically performed with iodine-123 and iodine-131, with the current hybrid scanner performing single photon emission tomography/computed tomography (SPECT/CT). Positron emission tomography/computed tomography (PET/CT) provides superior visualization and quantification of functions at the molecular level; thus, lesion assessment can be improved compared to that of SPECT/CT. Various types of cancer, including radioiodine-refractory DTC, can be detected by 2-[18F]fluoro-2-deoxy-D-glucose ([18F]FDG), the most well-known and widely used PET radiopharmaceutical. Several other PET radiopharmaceuticals have been developed, although some are limited in availability despite their potential clinical utilizations. This article aims to summarize PET radiopharmaceuticals in DTC, focusing on molecular pathways and applications.
Collapse
|
34
|
Wang Y, Galante JR, Haroon A, Wan S, Afaq A, Payne H, Bomanji J, Adeleke S, Kasivisvanathan V. The future of PSMA PET and WB MRI as next-generation imaging tools in prostate cancer. Nat Rev Urol 2022; 19:475-493. [PMID: 35789204 DOI: 10.1038/s41585-022-00618-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2022] [Indexed: 11/09/2022]
Abstract
Radiolabelled prostate-specific membrane antigen (PSMA)-based PET-CT has been shown in numerous studies to be superior to conventional imaging in the detection of nodal or distant metastatic lesions. 68Ga-PSMA PET-CT is now recommended by many guidelines for the detection of biochemically relapsed disease after radical local therapy. PSMA radioligands can also function as radiotheranostics, and Lu-PSMA has been shown to be a potential new line of treatment for metastatic castration-resistant prostate cancer. Whole-body (WB) MRI has been shown to have a high diagnostic performance in the detection and monitoring of metastatic bone disease. Prospective, randomized, multicentre studies comparing 68Ga-PSMA PET-CT and WB MRI for pelvic nodal and metastatic disease detection are yet to be performed. Challenges for interpretation of PSMA include tracer trapping in non-target tissues and also urinary excretion of tracers, which confounds image interpretation at the vesicoureteral junction. Additionally, studies have shown how long-term androgen deprivation therapy (ADT) affects PSMA expression and could, therefore, reduce tracer uptake and visibility of PSMA+ lesions. Furthermore, ADT of short duration might increase PSMA expression, leading to the PSMA flare phenomenon, which makes the accurate monitoring of treatment response to ADT with PSMA PET challenging. Scan duration, detection of incidentalomas and presence of metallic implants are some of the major challenges with WB MRI. Emerging data support the wider adoption of PSMA PET and WB MRI for diagnosis, staging, disease burden evaluation and response monitoring, although their relative roles in the standard-of-care management of patients are yet to be fully defined.
Collapse
Affiliation(s)
- Yishen Wang
- School of Clinical Medicine, University of Cambridge, Cambridge, UK. .,Barking, Havering and Redbridge University Hospitals NHS Trust, Romford, UK.
| | - Joao R Galante
- Department of Oncology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Athar Haroon
- Department of Nuclear Medicine, Barts Health NHS Trust, London, UK
| | - Simon Wan
- Institute of Nuclear Medicine, University College London, London, UK
| | - Asim Afaq
- Institute of Nuclear Medicine, University College London, London, UK.,Department of Radiology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Heather Payne
- Department of Oncology, University College London Hospitals, London, UK
| | - Jamshed Bomanji
- Institute of Nuclear Medicine, University College London, London, UK
| | - Sola Adeleke
- Department of Oncology, Guy's and St Thomas' NHS Foundation Trust, London, UK.,School of Cancer & Pharmaceutical Sciences, King's College London, London, UK
| | - Veeru Kasivisvanathan
- Division of Surgery & Interventional Science, University College London, London, UK.,Department of Urology, University College London Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
35
|
18F-PSMA-11 Versus 68Ga-PSMA-11 Positron Emission Tomography/Computed Tomography for Staging and Biochemical Recurrence of Prostate Cancer: A Prospective Double-blind Randomised Cross-over Trial. Eur Urol 2022; 82:501-509. [PMID: 35690515 DOI: 10.1016/j.eururo.2022.05.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 04/08/2022] [Accepted: 05/13/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND Fluorine-18 (18F)-labelled prostate-specific membrane antigen (PSMA) offers several advantages over gallium-68 (68Ga) in terms of costs, yield, transport/distribution, and image resolution. OBJECTIVE This trial investigates the new radiotracer 18F-PSMA-11 via a prospective, intraindividual crossover design. The trial was powered for noninferiority of 18F-PSMA-11 over 68Ga-PSMA-11 positron emission tomography (PET)/computed tomography (CT) in terms of the number of positive PET scans. Secondary endpoints were as follows: (1) superiority of 18F-PSMA-11 over 68Ga-PSMA-11 with respect to the number of positive PET scans, the total number of suspicious prostate cancer lesions, and the miPSMA expression score of corresponding lesions; (2) correlation of the PET/CT images with available follow-up data for 18F-PSMA-11 and 68Ga-PSMA-11; and (3) assessment of the interobserver variability. DESIGN, SETTING, AND PARTICIPANTS Prostate cancer patients (primary or biochemical recurrence) were randomised in a double-blind crossover design whereby each patient received both 18F-PSMA-11 and 68Ga-PSMA-11 PET/CT. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS All scans were reviewed and scored by three independent experienced nuclear physicians following the proposed guideline for the interpretation of PSMA-ligand PET/CT, as described by Eiber et al. RESULTS AND LIMITATIONS: In total, 82 patients were included for scan analyses. The primary endpoint was met: per patient, the proportions of positive scans rated by the three readers were 67%/67%, 65%/65%, and 73%/70% for 18F-PSMA-11/68Ga-PSMA-11 PET/CT. The miPSMA expression score was higher for 18F-PSMA-11 than for 68Ga-PSMA-11 for the reference reader. Follow-up data showed identical estimated sensitivity for both the 18F-PSMA-11 and the 68Ga-PSMA-11 scan (0.92, 0.83, and 0.92 for the three readers). A fair to good agreement among readers (at patient level) was obtained, which was demonstrated by a Light's kappa value of 0.59 for both tracers. CONCLUSIONS The tracer 18F-PSMA-11 is noninferior to68Ga-PSMA-11. Superiority of 18F-PSMA-11 was limited to the miPSMA expression score, given by the reference reader. Inter-rater agreement was fair to good, and equal for both radiotracers. PATIENT SUMMARY In this study, we compared two radiotracers: 18F-PSMA-11 and 68Ga-PSMA-11. We proved that 18F-PSMA-11 is not inferior to 68Ga-PSMA-11 for detecting prostate cancer and thus can be used as an alternative. Possible superiority of this tracer should be further investigated in specific subpopulations.
Collapse
|
36
|
Jadvar H, Colletti PM. Molecular Imaging Assessment of Androgen Deprivation Therapy in Prostate Cancer. PET Clin 2022; 17:389-397. [PMID: 35662493 DOI: 10.1016/j.cpet.2022.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Hormonal therapy has long been recognized as a mainstay treatment for prostate cancer. New generation imaging agents have provided unprecedented opportunities at all phases along the natural history of prostate cancer. We review the literature on the effect of androgens and androgen deprivation therapy on prostate tumor at its various biological phases using the new generation molecular imaging agents in conjunction with positron emission tomography.
Collapse
Affiliation(s)
- Hossein Jadvar
- Division of Nuclear Medicine, Department of Radiology, USC Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Kenneth Norris Jr. Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA.
| | - Patrick M Colletti
- Division of Nuclear Medicine, Department of Radiology, USC Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
37
|
Farolfi A, Mei R, Ali S, Castellucci P. Theragnostics in prostate cancer. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF... 2022; 65:333-341. [PMID: 35133097 DOI: 10.23736/s1824-4785.21.03419-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Prostate-specific membrane antigen (PSMA) is a molecular target for both imaging diagnostics and therapeutics, i.e., a theragnostics target. There has been a growing body of evidence supporting PSMA theragnostics approaches in the management of prostate cancer (PCa) for tailored precision medicine. Tumor characterization through PSMA-ligand PET imaging is crucial for assessing the molecular signature and eligibility for PSMA radioligand therapy. Recent U.S. Food and Drug Administration (FDA) approval of two new drug applications for PSMA PET imaging contribute to reinforce PSMA as an oncologic blockbuster. Additionally, relevant progress in the PSMA treatment has been made in the last five years. [177Lu]Lu-PSMA-617 radioligand therapy for patients with progressive PSMA-avid metastatic castration-resistant PCa (mCRPC) significantly increased overall survival and radiographic progression-free survival, according to the results of an international, prospective, open label, multicenter, randomized, phase III study (VISION trial). The objective of this comprehensive review is to highlight the recent advances in PCa theragnostics, focusing on actual clinical applications and future perspectives.
Collapse
Affiliation(s)
- Andrea Farolfi
- Division of Nuclear Medicine, IRCCS University Hospital of Bologna, Bologna, Italy -
| | - Riccardo Mei
- Division of Nuclear Medicine, IRCCS University Hospital of Bologna, Bologna, Italy
| | - Sakaria Ali
- Department of Pediatrics, University College London Hospital, London, UK
| | - Paolo Castellucci
- Division of Nuclear Medicine, IRCCS University Hospital of Bologna, Bologna, Italy
| |
Collapse
|
38
|
The Emerging Role of Next-Generation Imaging in Prostate Cancer. Curr Oncol Rep 2022; 24:33-42. [DOI: 10.1007/s11912-021-01156-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2021] [Indexed: 12/23/2022]
|
39
|
Langbein T, Wang H, Rauscher I, Krönke M, Knorr K, Wurzer A, Schwamborn K, Maurer T, Horn T, Haller B, Wester HJ, Eiber M. Utility of 18F-rhPSMA-7.3 positron emission tomography for imaging of primary prostate cancer and pre-operative efficacy in N-staging of unfavorable intermediate to very high-risk patients validated by histopathology. J Nucl Med 2022; 63:1334-1342. [PMID: 34992154 DOI: 10.2967/jnumed.121.263440] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/28/2021] [Indexed: 11/16/2022] Open
Abstract
18F-rhPSMA-7.3, the lead compound of a new class of radiohybrid prostate-specific membrane antigen (rhPSMA) ligands, is currently in phase III trials for prostate cancer (PCa) imaging. Here, we describe our experience in primary PCa staging. Methods: We retrospectively identified 279 patients with primary PCa who underwent 18F-rhPSMA-7.3 PET/CT (staging cohort). A subset of patients (83/279) subsequently underwent prostatectomy with lymph node (LN) dissection without prior treatment (efficacy cohort). Distribution of tumor lesions was determined for the staging cohort and stratified by National Comprehensive Cancer Network (NCCN) risk score. Involvement of pelvic LN was assessed retrospectively by 3 blinded independent central readers, and a majority rule was used for analysis. Standard surgical fields were rated on a five-point scale independently for PET and for morphological imaging. Results were compared to histopathological findings on a patient-, right vs. -left, and template-basis. Results: For the staging cohort 18F-rhPSMA-7.3 PET was positive in 275/279 (98.6%), 106/279 (38.0%), 46/279 (16.5%), 65/279 (23.3%) and 5/279 (1.8%) patients for local, pelvic nodal, extrapelvic nodal, metastatic bone, and visceral metastatic disease. In the efficacy cohort, LN metastases were present in 24/83 patients (29%), located in 48/420 (11%) resected templates and in 33/166 (19.9%) hemi-pelvic templates in histopathology. Based on majority vote results, the patient-level sensitivity, specificity and accuracy for pelvic nodal metastases were 66.7% (95%CI, 44.7-83.6%), 96.6% (95%CI, 87.3-99.4%) and 88.0% (95%CI, 78.5-93.8%) for 18F-rhPSMA-7.3 PET and 37.5% (95%CI, 19.6-59.2%), 91.5% (95%CI, 80.6-96.8%) and 75.9% (95%CI, 65.0-84.3%) for morphological imaging, respectively. 18F-rhPSMA-7.3 showed higher interobserver agreement than morphological imaging (patient-level Fleiss' κ=0.54; 95%CI, 0.47-0.62 vs. 0.24; 95%CI, 0.17-0.31). A mean standardized uptake value ratio of 6.6 (95%CI, 5.2-8.1) documented a high image contrast between local tumors and adjacent low urinary tracer retention. Conclusion: 18F-rhPSMA-7.3 PET offers superior diagnostic performance to morphological imaging for primary N-staging of newly diagnosed PCa, shows lower inter-reader variation, and offers good distinction between primary tumor and bladder background activity. With increasing NCCN risk group an increasing frequency of extra-prostatic tumor lesions was observed.
Collapse
Affiliation(s)
- Thomas Langbein
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Nuclear Medicine, Germany
| | - Hui Wang
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Nuclear Medicine, Germany
| | - Isabel Rauscher
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Nuclear Medicine, Germany
| | - Markus Krönke
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Nuclear Medicine, Germany
| | - Karina Knorr
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Nuclear Medicine, Germany
| | | | - Kristina Schwamborn
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Institute of Pathology, Germany
| | - Tobias Maurer
- Martini-Klinik and Department of Urology, University Hospital Hamburg-Eppendorf, Germany
| | - Thomas Horn
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Urology, Germany
| | - Bernhard Haller
- Technical University of Munich, School of Medicine, Institute of Medical Informatics, Statistics and Epidemiology, Germany
| | | | - Matthias Eiber
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Nuclear Medicine, Germany
| |
Collapse
|
40
|
Yousefirizi F, Pierre Decazes, Amyar A, Ruan S, Saboury B, Rahmim A. AI-Based Detection, Classification and Prediction/Prognosis in Medical Imaging:: Towards Radiophenomics. PET Clin 2021; 17:183-212. [PMID: 34809866 DOI: 10.1016/j.cpet.2021.09.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Artificial intelligence (AI) techniques have significant potential to enable effective, robust, and automated image phenotyping including the identification of subtle patterns. AI-based detection searches the image space to find the regions of interest based on patterns and features. There is a spectrum of tumor histologies from benign to malignant that can be identified by AI-based classification approaches using image features. The extraction of minable information from images gives way to the field of "radiomics" and can be explored via explicit (handcrafted/engineered) and deep radiomics frameworks. Radiomics analysis has the potential to be used as a noninvasive technique for the accurate characterization of tumors to improve diagnosis and treatment monitoring. This work reviews AI-based techniques, with a special focus on oncological PET and PET/CT imaging, for different detection, classification, and prediction/prognosis tasks. We also discuss needed efforts to enable the translation of AI techniques to routine clinical workflows, and potential improvements and complementary techniques such as the use of natural language processing on electronic health records and neuro-symbolic AI techniques.
Collapse
Affiliation(s)
- Fereshteh Yousefirizi
- Department of Integrative Oncology, BC Cancer Research Institute, 675 West 10th Avenue, Vancouver, British Columbia V5Z 1L3, Canada.
| | - Pierre Decazes
- Department of Nuclear Medicine, Henri Becquerel Centre, Rue d'Amiens - CS 11516 - 76038 Rouen Cedex 1, France; QuantIF-LITIS, Faculty of Medicine and Pharmacy, Research Building - 1st floor, 22 boulevard Gambetta, 76183 Rouen Cedex, France
| | - Amine Amyar
- QuantIF-LITIS, Faculty of Medicine and Pharmacy, Research Building - 1st floor, 22 boulevard Gambetta, 76183 Rouen Cedex, France; General Electric Healthcare, Buc, France
| | - Su Ruan
- QuantIF-LITIS, Faculty of Medicine and Pharmacy, Research Building - 1st floor, 22 boulevard Gambetta, 76183 Rouen Cedex, France
| | - Babak Saboury
- Department of Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD, USA; Department of Computer Science and Electrical Engineering, University of Maryland, Baltimore County, Baltimore, MD, USA; Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Arman Rahmim
- Department of Integrative Oncology, BC Cancer Research Institute, 675 West 10th Avenue, Vancouver, British Columbia V5Z 1L3, Canada; Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada; Department of Physics, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
41
|
Ng TSC, Gao X, Salari K, Zlatev DV, Heidari P, Kamran SC. Incorporating PSMA-Targeting Theranostics Into Personalized Prostate Cancer Treatment: a Multidisciplinary Perspective. Front Oncol 2021; 11:722277. [PMID: 34395293 PMCID: PMC8355555 DOI: 10.3389/fonc.2021.722277] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 07/12/2021] [Indexed: 01/12/2023] Open
Abstract
Recent developments in prostate-specific membrane antigen (PSMA) targeted diagnostic imaging and therapeutics (theranostics) promise to advance the management of primary, biochemically recurrent, and metastatic prostate cancer. In order to maximize the clinical impact of PSMA-targeted theranostics, a coordinated approach between the clinical stakeholders involved in prostate cancer management is required. Here, we present a vision for multidisciplinary use of PSMA theranostics from the viewpoints of nuclear radiology, medical oncology, urology, and radiation oncology. We review the currently available and forthcoming PSMA-based imaging and therapeutics and examine current and potential impacts on prostate cancer management from early localized disease to advanced treatment-refractory disease. Finally, we highlight the clinical and research opportunities related to PSMA-targeted theranostics and describe the importance of multidisciplinary collaboration in this space.
Collapse
Affiliation(s)
- Thomas S C Ng
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Xin Gao
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Keyan Salari
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Dimitar V Zlatev
- Department of Urology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Pedram Heidari
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Sophia C Kamran
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
42
|
PSMA Expression Predicts Early Biochemical Response in Patients with Metastatic Castration-Resistant Prostate Cancer under 177Lu-PSMA-617 Radioligand Therapy. Cancers (Basel) 2021; 13:cancers13122938. [PMID: 34208246 PMCID: PMC8230748 DOI: 10.3390/cancers13122938] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 05/30/2021] [Accepted: 06/09/2021] [Indexed: 01/02/2023] Open
Abstract
Simple Summary Prostate-specific membrane antigen (PSMA) is a promising target for both imaging and radioligand therapies (RLT) for men with prostate cancer. However, not all patients respond to RLT and some even progress early in the treatment course. We aimed to identify parameters to forecast which patients will achieve therapy response prior to commencing RLT. Therefore, we tested whether the tumor volume, the level of PSMA expression or a combination of both in metastases derived from PSMA-targeted molecular imaging prior to RLT can inform the treating physician whether a patient will respond to RLT. Compared to tumor volume, the level of PSMA-expression can better identify patients responding to RLT early in the treatment course. Abstract 177Lu-Prostate-specific membrane antigen (PSMA)-radioligand therapy (RLT) is a promising treatment option in patients with metastatic castration-resistant prostate cancer (mCRPC). We aimed to determine the predictive value of pretherapeutic PSMA-ligand positron emission tomography (PET) and established clinical parameters for early biochemical response after two cycles of RLT. In total, 71 mCRPC patients who had undergone PET/computed tomography (CT) with 68Ga-PSMA-11 prior to two cycles of 177Lu-PSMA-617 RLT were included. Malignant lesions on pretherapeutic PET/CTs were manually segmented and average maximum PSMA expression (maximum standardized uptake values, SUVmax), whole-body PSMA-tumor volume (TV), and whole-body total lesion (TL)-PSMA were calculated. We then tested the predictive performance of these parameters for early biochemical response (defined as prostate-sepcific antigen (PSA) decrease of ≥50% according to PCWG2) after two cycles of RLT, relative to established clinical parameters. Early PSA response was observed in 34/71 patients. PSA change after two cycles of RLT correlated with pretherapeutic SUVmax (r = −0.49; p < 0.001), but not with PSMA-TV (r = 0.02; p = 0.89) or TL-PSMA (r = −0.15; p = 0.22). A cut-off of 19.8 for SUVmax and 75.5 years for age was defined by receiver operating characteristics and revealed a significant outcome difference for early biochemical response between patients with adversely low vs. high PSMA expression and low vs. high age (p < 0.001). Multivariate analysis identified SUVmax (HR, 7.94, p = 0.001) and age (HR, 8.05, p = 0.002) as independent predictors for PSA response early in the treatment course. Thus, high age and high PSMA expression in patients scheduled for RLT identify patients with early biochemical response. This study provides a rationale for further prospective studies exploring PET-guided treatment intensification in selected patients.
Collapse
|
43
|
A bicentric retrospective analysis of clinical utility of 18F-fluciclovine PET in biochemically recurrent prostate cancer following primary radiation therapy: is it helpful in patients with a PSA rise less than the Phoenix criteria? Eur J Nucl Med Mol Imaging 2021; 48:4463-4471. [PMID: 34091713 DOI: 10.1007/s00259-021-05415-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 05/12/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE 18F-Fluciclovine PET imaging has been increasingly used in the restaging of prostate cancer patients with biochemical recurrence (BCR); however, its clinical utility in patients with low prostate-specific antigen (PSA) levels following primary radiation therapy has not been well-studied. This study aims to determine the detection rate and diagnostic accuracy of 18F-fluciclovine PET and the patterns of prostate cancer recurrence in patients with rising PSA after initial radiation therapy, particularly in patients with PSA levels below the accepted Phoenix definition of BCR (PSA nadir +2 ng/mL). METHODS This retrospective study included patients from two tertiary institutions who underwent 18F-fluciclovine PET scans for elevated PSA level following initial external beam radiation therapy, brachytherapy, and/or proton therapy. Logistic regression and receiver operating characteristic (ROC) curve analyses were performed to determine the diagnostic accuracy of 18F-fluciclovine PET and associations of PSA kinetic parameters with 18F-fluciclovine PET outcome. RESULTS One hundred patients were included in this study. The overall detection rate on a patient-level was 79% (79/100). 18F-Fluciclovine PET was positive in 62% (23/37) of cases with PSA below the Phoenix criteria. The positive predictive value of 18F-fluciclovine PET was 89% (95% CI: 80-94%). In patients with PSA below the Phoenix criteria, the PSA velocity had the highest predictive value of 18F-fluciclovine PET outcome. PSA doubling time (PSADT) and PSA velocity were associated with the presence of extra-pelvic metastatic disease. CONCLUSION 18F-Fluciclovine PET can identify recurrent disease at low PSA level and PSA rise below accepted Phoenix criteria in patients with suspected BCR after primary radiation therapy, particularly in patients with low PSADT or high PSA velocity. In patients with low PSADT or high PSA velocity, there is an increased probability of extra-pelvic metastases. Therefore, these patients are more likely to benefit from PET/CT or PET/MRI than pelvic MRI alone.
Collapse
|
44
|
Jadvar H. Competitive Advantage of PSMA Theranostics in Prostate Cancer. Radiology 2021; 299:261-263. [PMID: 33788590 PMCID: PMC8103912 DOI: 10.1148/radiol.2021210348] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 12/14/2022]
Affiliation(s)
- Hossein Jadvar
- From the Division of Nuclear Medicine, Department of Radiology, Keck School of Medicine, University of Southern California, 2250 Alcazar St, CSC 102, Los Angeles, CA 90033
| |
Collapse
|