1
|
Dall’ Armellina S, Aghakhanyan G, Rizzo A, Fanni SC, Aringhieri G, Faggioni L, Cioni D, Neri E, Volterrani D, Morbelli S. PSMA-targeted PET imaging for brain metastases from non-prostatic solid tumors: a systematic review. Front Oncol 2025; 15:1553505. [PMID: 40165900 PMCID: PMC11955466 DOI: 10.3389/fonc.2025.1553505] [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: 01/03/2025] [Accepted: 02/28/2025] [Indexed: 04/02/2025] Open
Abstract
Introduction Prostate-Specific Membrane Antigen (PSMA) is a transmembrane glycoprotein initially identified in prostate cancer (PCa) but also expressed in the neovasculature of various solid tumors. Recently, PSMA PET has emerged as a promising tool for detecting brain metastases (BMs) from non-prostatic cancers, offering diagnostic capabilities in addition to conventional imaging. This systematic review evaluates the role of PSMA-targeted radiopharmaceuticals in imaging BMs, highlighting their comparative diagnostic performance and exploring their potential for theranostic applications. Methods A systematic review of the literature was conducted following PRISMA guidelines. Studies evaluating the diagnostic accuracy of PSMA PET imaging in identifying brain metastases (BMs) from non-prostatic solid tumors were included. Both full research articles and case reports were considered to capture the breadth of current evidence. The methodological quality of the included studies was assessed using the QUADAS-2 tool, and data were synthesized qualitatively. Results The review includes 23 studies reporting on 77 BMs from diverse primary malignancies, including lung, breast, salivary gland, thyroid, kidney, and melanoma. PSMA PET demonstrated high tumor-to-background ratios (TBR), enabling superior detection of BMs compared to conventional imaging modalities such as contrast-enhanced MRI and [18F]FDG PET. In post-radiotherapy cases, PSMA PET effectively differentiated radionecrosis from tumor recurrence. Moreover, PSMA PET demonstrated superior sensitivity in detecting thyroid metastases compared to traditional scintigraphy methods, highlighting its potential in cases where standard techniques yield inconclusive results. Conclusions PSMA PET imaging shows significant promise in improving the diagnosis and management of BMs from non-prostatic cancers. While its theranostic applications remain underexplored, initial findings suggest promising avenues for integrating PSMA PET into personalized neuro-oncology care. Future studies should focus on standardizing imaging protocols, exploring PSMA PET utility in diverse tumor subtypes, and validating its role in clinical decision-making to maximize its impact on patient outcomes.
Collapse
Affiliation(s)
- Sara Dall’ Armellina
- Nuclear Medicine Unit, Service Area Department, Azienda Socio-Sanitaria Territoriale (ASST)-Rhodense P.O. Bollate, Milan, Italy
| | - Gayane Aghakhanyan
- Department of Translational Research and of New Surgical and Medical Technology, Nuclear Medicine Unit, University of Pisa, Pisa, Italy
| | - Alessio Rizzo
- Department of Nuclear Medicine, Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia - Istituto di Ricovero e Cura a Carattere Scientifico (FPO-IRCCS), Turin, Italy
| | - Salvatore C. Fanni
- Department of Translational Research and of New Surgical and Medical Technology, Academic Radiology, University of Pisa, Pisa, Italy
| | - Giacomo Aringhieri
- Department of Translational Research and of New Surgical and Medical Technology, Academic Radiology, University of Pisa, Pisa, Italy
| | - Lorenzo Faggioni
- Department of Translational Research and of New Surgical and Medical Technology, Academic Radiology, University of Pisa, Pisa, Italy
| | - Dania Cioni
- Department of Translational Research and of New Surgical and Medical Technology, Academic Radiology, University of Pisa, Pisa, Italy
| | - Emanuele Neri
- Department of Translational Research and of New Surgical and Medical Technology, Academic Radiology, University of Pisa, Pisa, Italy
| | - Duccio Volterrani
- Department of Translational Research and of New Surgical and Medical Technology, Nuclear Medicine Unit, University of Pisa, Pisa, Italy
| | - Silvia Morbelli
- Department of Medical Sciences, University of Turin, Turin, Italy
- Azienda Ospedaliero-Universitaria Città della Salute e della Scienza di Torino, Nuclear Medicine Unit, Turin, Italy
| |
Collapse
|
2
|
Na R, Chen Z, Liu Y, Chen Q, Yang Q, Qiu Y, Wang T, Song L, Wu S, Huang W, Sun X, Xian S, Kang L. Diagnostic and prognostic role of 18F-FDG PET/CT for sarcomatoid differentiation in renal cell carcinoma. EJNMMI Res 2025; 15:11. [PMID: 39971809 PMCID: PMC11839549 DOI: 10.1186/s13550-025-01206-w] [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: 11/14/2024] [Accepted: 02/09/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Sarcomatoid differentiation is an invasive dedifferentiated feature of tumor and associated with poor prognosis in renal cell carcinoma (RCC) patients. This study aimed to evaluate the utility of 18F-FDG PET/CT in predicting sarcomatoid differentiation in RCC and its potential prognostic value. RESULTS This retrospective study assessed newly diagnosed sarcomatoid differentiation renal cell carcinoma (SDRCC) patients who were staged using 18F-FDG PET/CT. Patients were categorized into high-grade sarcomatoid differentiation RCC (HG-SDRCC), low-grade sarcomatoid differentiation RCC (LG-SDRCC), and non-sarcomatoid differentiation RCC (non-SDRCC). The maximum standardized uptake value (SUVmax), mean standardized uptake value (SUVmean), metabolic tumor volume (MTV), and total lesion glycolysis (TLG) were compared. Overall survival (OS) and disease-free survival (DFS) were analyzed. SUVmax, MTV, TLG, and SUVmean values were significantly higher in SDRCC compared to non-SDRCC (P < 0.05). Additionally, SUVmax, TLG, and SUVmean were significantly higher in HG-SDRCC compared to non-HG-SDRCC (P < 0.05). ROC curves revealed that SUVmax and SUVmean were effective for distinguishing HG-SDRCC from non-HG-SDRCC. The log-rank test identified SUVmax > 11, MTV > 95, TLG > 500, SUVmean > 5.2, invasion of peripheral tissue and/or organs, and metastasis as risk factors for SDRCC patients. Multivariate Cox proportional hazards model analyses indicated that TLG > 500 was a risk factor for poor DFS, while SUVmax > 11 and SUVmean > 5.2 were risk factors for poor OS. CONCLUSIONS 18F-FDG PET/CT can effectively differentiate HG-SDRCC with more aggressive malignancy. The prognostic model developed in this study demonstrates that metabolic parameters, particularly TLG for DFS and SUVmax/SUVmean for OS, serve as more robust predictors of patient outcomes than the degree of sarcomatoid differentiation.
Collapse
Affiliation(s)
- Ritai Na
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Str., Xicheng District, Beijing, China
| | - Zhao Chen
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Str., Xicheng District, Beijing, China
| | - Yongshun Liu
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Str., Xicheng District, Beijing, China
| | - Qianrui Chen
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Str., Xicheng District, Beijing, China
| | - Qi Yang
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Str., Xicheng District, Beijing, China
| | - Yongkang Qiu
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Str., Xicheng District, Beijing, China
| | - Tianyao Wang
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Str., Xicheng District, Beijing, China
| | - Lele Song
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Str., Xicheng District, Beijing, China
| | - Sitong Wu
- Department of Interventional Radiology and Vascular Surgery, Peking University First Hospital, Beijing, China
| | - Wenpeng Huang
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Str., Xicheng District, Beijing, China
| | - Xinyao Sun
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Str., Xicheng District, Beijing, China
| | - Shaozhong Xian
- Department of Urology, Beijing Luhe Hospital, Capital Medical University, No. 82 Xinhua South Str., Tongzhou District, Beijing, China.
| | - Lei Kang
- Department of Nuclear Medicine, Peking University First Hospital, No. 8 Xishiku Str., Xicheng District, Beijing, China.
| |
Collapse
|
3
|
Le Rhun E, Albert NL, Hüllner M, Franceschi E, Galldiks N, Karschnia P, Minniti G, Weiss T, Preusser M, Ellingson BM, Weller M. Targeted radionuclide therapy for patients with central nervous system metastasis: Overlooked potential? Neuro Oncol 2024; 26:S229-S241. [PMID: 39351771 PMCID: PMC11631097 DOI: 10.1093/neuonc/noae192] [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: 12/12/2024] Open
Abstract
Targeted radionuclide therapy is an emerging therapeutic concept for metastatic cancer that can be considered if a tumor can be delineated by nuclear medicine imaging and also targeted based on the expression of a particular target (thera-nostics). This mode of treatment can also compete with or supplement conventional radiotherapy, for example, if MRI does not fully capture the extent of the disease, including microscopic metastases. Targeted radionuclide therapy for patients with thyroid cancer, with certain somatostatin receptor 2-expressing tumors and with prostate-specific membrane antigen-expressing prostate cancer is approved, and numerous approaches of targeted radionuclide therapy for patients with metastatic cancer are in development (eg, using fibroblast activation protein as a target). Although brain metastases are rare in cancers with approved targeted radionuclide therapies, there is no a priori reason to assume that such treatments would not be effective against brain metastases if the targets are expressed and not shielded by the blood-brain barrier. Here, we discuss the current state of the art and opportunities of targeted radionuclide therapies for patients with brain and leptomeningeal metastases.
Collapse
Affiliation(s)
- Emilie Le Rhun
- Department of Medical Oncology and Hematology, University Hospital and University of Zurich, Zurich, Switzerland
| | - Nathalie L Albert
- Department of Nuclear Medicine, Ludwig Maximilians-University of Munich, Munich, Germany
| | - Martin Hüllner
- Department of Nuclear Medicine, University Hospital and University of Zurich, Zurich, Switzerland
| | - Enrico Franceschi
- Nervous System Medical Oncology Department, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy
| | - Norbert Galldiks
- Center for Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne, and Duesseldorf, Germany
- Institute of Neuroscience and Medicine (INM-3), Research Center Juelich, Juelich, Germany
- Department of Neurology, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Philipp Karschnia
- German Cancer Consortium, Partner Site Munich, Munich, Germany
- Department of Neurosurgery, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Giuseppe Minniti
- IRCCS Istituto Neurologico Mediterraneo Neuromed, Pozzilli, Italy
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University, Rome, Italy
| | - Tobias Weiss
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| | - Matthias Preusser
- Department of Medicine I, Division of Oncology, and Comprehensive Cancer Center Vienna, Medical University of Vienna, Vienna, Austria
| | - Benjamin M Ellingson
- UCLA Brain Tumor Imaging Laboratory (BTIL), Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center, University Hospital and University of Zurich, Zurich, Switzerland
| |
Collapse
|
4
|
Wu Q, Shao H, Zhai W, Huang G, Liu J, Calais J, Wei W. Molecular imaging of renal cell carcinomas: ready for prime time. Nat Rev Urol 2024:10.1038/s41585-024-00962-z. [PMID: 39543358 DOI: 10.1038/s41585-024-00962-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2024] [Indexed: 11/17/2024]
Abstract
The clinical diagnosis of renal cell carcinoma (RCC) is constantly evolving. Diagnostic imaging of RCC relying on enhanced computed tomography (CT) and magnetic resonance imaging (MRI) is commonly used for renal mass characterization and assessment of tumour thrombosis, whereas pathology is the gold standard for establishing diagnosis. However, molecular imaging is rapidly improving the clinical management of RCC, particularly clear-cell RCC. Molecular imaging aids in the non-invasive visualization and characterization of specific biomarkers such as carbonic anhydrase IX and CD70 within the tumours, which help to assess tumour heterogeneity and status. Target-specific molecular imaging of RCCs will substantially improve the diagnostic landscape of RCC and will further facilitate clinical decision-making regarding initial staging and re-staging, monitoring of recurrence and metastasis, patient stratification and selection, and the prediction and evaluation of treatment responses.
Collapse
Affiliation(s)
- Qianyun Wu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hongda Shao
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Zhai
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Jeremie Calais
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA.
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
5
|
Sadaghiani MS, Baskaran S, Gorin MA, Rowe SP, Provost JC, Teslenko I, Bilyk R, An H, Sheikhbahaei S. Utility of PSMA PET/CT in Staging and Restaging of Renal Cell Carcinoma: A Systematic Review and Metaanalysis. J Nucl Med 2024; 65:1007-1012. [PMID: 38782453 PMCID: PMC11218724 DOI: 10.2967/jnumed.124.267417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
Prostate-specific membrane antigen (PSMA) is expressed in the neovasculature of multiple solid tumors, including renal cell carcinoma (RCC). Studies have demonstrated promising results on the utility of PSMA-targeted PET/CT imaging in RCC. This report aims to provide a systematic review and metaanalysis on the utility and detection rate of PSMA PET/CT imaging in staging or evaluation of primary RCC and restaging of metastatic or recurrent RCC. Methods: Searches were performed in PubMed, Embase, and abstract proceedings (last updated, August 2023). Studies that provided a lesion-level detection rate of PSMA radiotracers in staging or restaging of RCC were included in the metaanalysis. The overall pooled detection rate with a 95% CI was estimated, and subgroup analysis was performed when feasible. Results: Nine studies comprising 152 patients (133 clear cell RCC [ccRCC], 19 other RCC subtypes) were included in the metaanalysis. The pooled detection rate of PSMA PET/CT in evaluation of primary or metastatic RCC was estimated to be 0.83 (95% CI, 0.67-0.92). Subgroup analysis showed a pooled PSMA detection rate of 0.74 (95% CI, 0.57-0.86) in staging or evaluation of primary RCC lesions and 0.87 (95% CI, 0.73-0.95) in restaging of metastatic or recurrent RCC. Analysis based on the type of radiotracer showed a pooled detection rate of 0.85 (95% CI, 0.62-0.95) for 68Ga-based PSMA tracers and 0.92 (95% CI, 0.76-0.97) for 18F-DCFPyL PET/CT. Furthermore, in metastatic ccRCC, the available data support a significantly higher detection rate for 18F-DCFPyL PET/CT than for conventional imaging modalities (2 studies). Conclusion: Our preliminary results show that PSMA PET/CT could be a promising alternative imaging modality for evaluating RCC, particularly metastatic ccRCC. Large prospective studies are warranted to confirm clinical utility in the staging and restaging of RCC.
Collapse
Affiliation(s)
- Moe S Sadaghiani
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Michael A Gorin
- Milton and Carroll Petrie Department of Urology, Icahn School of Medicine at Mount Sinai, New York, New York; and
| | - Steven P Rowe
- Department of Radiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | | | | | | | - Hong An
- Lantheus, Bedford, Massachusetts
| | - Sara Sheikhbahaei
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, Maryland;
| |
Collapse
|
6
|
Ovruchesky E, Pan E, Guer M, Elliott A, Siva S, Ravi P, McGregor B, Bagrodia A, Derweesh I, Barata P, Heath EI, Antonarakis ES, Darabi S, Hoon DSB, Mortazavi A, Choueiri TK, Nabhan C, Wei S, McKay RR. Characterization of FOLH1 Expression in Renal Cell Carcinoma. Cancers (Basel) 2024; 16:1855. [PMID: 38791934 PMCID: PMC11119455 DOI: 10.3390/cancers16101855] [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: 04/09/2024] [Revised: 04/26/2024] [Accepted: 05/11/2024] [Indexed: 05/26/2024] Open
Abstract
PURPOSE Given the emergence of PSMA-targeted diagnostic agents and therapeutics, we sought to investigate patterns of FOLH1 expression in RCC and their impacts on RCC outcomes. METHODS We conducted a pooled multi-institutional analysis of patients with RCC having undergone DNA and RNA next-generation sequencing. FOLH1-high/low expression was defined as the ≥75th/<25th percentile of RNA transcripts per million (TPM). Angiogenic, T-effector, and myeloid expression signatures were calculated using previously defined gene sets. Kaplan-Meier estimates were calculated from the time of tissue collection or therapy start. RESULTS We included 1,724 patients in the analysis. FOLH1 expression was significantly higher in clear cell (71%) compared to non-clear cell RCC tumors (19.0 versus 3.3 TPM, p < 0.001) and varied by specimen site (45% primary kidney/55% metastasis, 13.6 versus 9.9 TPM, p < 0.001). FOLH1 expression was correlated with angiogenic gene expression (Spearman = 0.76, p < 0.001) and endothelial cell abundance (Spearman = 0.76, p < 0.001). While OS was similar in patients with FOLH1-high versus -low ccRCC, patients with FOLH1-high clear cell tumors experienced a longer time on cabozantinib treatment (9.7 versus 4.6 months, respectively, HR 0.57, 95% CI 0.35-0.93, p < 0.05). CONCLUSIONS We observed differential patterns of FOLH1 expression based on histology and tumor site in RCC. FOLH1 was correlated with angiogenic gene expression, increased OS, and a longer duration of cabozantinib treatment.
Collapse
Affiliation(s)
- Eric Ovruchesky
- Division of Urologic Oncology, Department of Oncology, Moores Cancer Center, University of California San Diego, San Diego, CA 92037, USA
| | - Elizabeth Pan
- Division of Urologic Oncology, Department of Oncology, Moores Cancer Center, University of California San Diego, San Diego, CA 92037, USA
| | - Melis Guer
- Division of Urologic Oncology, Department of Oncology, Moores Cancer Center, University of California San Diego, San Diego, CA 92037, USA
| | - Andrew Elliott
- Department of Clinical and Translational Research, Caris Life Sciences, Inc., Phoenix, AZ 85040, USA
| | - Shankar Siva
- Division of Radiation Oncology and Cancer Imaging, Department of Oncology, Peter MacCallum Cancer Centre, the University of Melbourne, Melbourne, VIC 3052, Australia
| | - Praful Ravi
- The Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Bradley McGregor
- The Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Aditya Bagrodia
- Division of Urologic Oncology, Department of Oncology, Moores Cancer Center, University of California San Diego, San Diego, CA 92037, USA
| | - Ithaar Derweesh
- Division of Urologic Oncology, Department of Oncology, Moores Cancer Center, University of California San Diego, San Diego, CA 92037, USA
| | - Pedro Barata
- Department of Hematology and Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH 44106, USA
| | - Elisabeth I. Heath
- Department of Oncology, Wayne State University, Karmanos Cancer Institute, Detroit, MI 48201, USA
| | - Emmanuel S. Antonarakis
- Department of Hematology and Oncology, University of Minnesota, Masonic Cancer Center, Minneapolis, MN 55455, USA
| | - Sourat Darabi
- Clinical Genomics, Hoag Family Cancer Institute, Newport Beach, CA 92663, USA
| | - Dave S. B. Hoon
- Department of Translational Molecular Medicine, Saint John’s Cancer Institute, Providence Health Systems, Santa Monica, CA 90404, USA
| | - Amir Mortazavi
- Division of Medical Oncology, Department of Internal Medicine, College of Medicine, The Ohio State University, The Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Toni K. Choueiri
- The Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Chadi Nabhan
- Department of Clinical and Translational Research, Caris Life Sciences, Inc., Phoenix, AZ 85040, USA
| | - Shuanzeng Wei
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Rana R. McKay
- Division of Urologic Oncology, Department of Oncology, Moores Cancer Center, University of California San Diego, San Diego, CA 92037, USA
| |
Collapse
|
7
|
Oldan JD, Schroeder JA, Hoffman-Censits J, Rathmell WK, Milowsky MI, Solnes LB, Nimmagadda S, Gorin MA, Khandani AH, Rowe SP. PET/Computed Tomography Transformation of Oncology: Kidney and Urinary Tract Cancers. PET Clin 2024; 19:197-206. [PMID: 38199916 DOI: 10.1016/j.cpet.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Renal cell carcinoma (RCC) and urothelial carcinoma (UC) are two of the most common genitourinary malignancies. 2-deoxy-2-[18F]fluoro-d-glucose (18F-FDG) can play an important role in the evaluation of patients with RCC and UC. In addition to the clinical utility of 18F-FDG PET to evaluate for metastatic RCC or UC, the shift in molecular imaging to focus on specific ligand-receptor interactions should provide novel diagnostic and therapeutic opportunities in genitourinary malignancies. In combination with the rise of artificial intelligence, our ability to derive imaging biomarkers that are associated with treatment selection, response assessment, and overall patient prognostication will only improve.
Collapse
Affiliation(s)
- Jorge D Oldan
- Molecular Imaging and Therapeutics, Department of Radiology, University of North Carolina, Chapel Hill, NC, USA
| | - Jennifer A Schroeder
- Molecular Imaging and Therapeutics, Department of Radiology, University of North Carolina, Chapel Hill, NC, USA
| | - Jean Hoffman-Censits
- Department of Medical Oncology and Urology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - W Kimryn Rathmell
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Matthew I Milowsky
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Lilja B Solnes
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sridhar Nimmagadda
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael A Gorin
- Milton and Carroll Petrie Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Amir H Khandani
- Molecular Imaging and Therapeutics, Department of Radiology, University of North Carolina, Chapel Hill, NC, USA
| | - Steven P Rowe
- Molecular Imaging and Therapeutics, Department of Radiology, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
8
|
David C, Muhammad A, Cristian U, Ben T, Arun A, Lewis A, Lavinia S, Marlon P, Shankar S. SABR for oligometastatic renal cell carcinoma. Clin Transl Radiat Oncol 2024; 45:100739. [PMID: 38380117 PMCID: PMC10877104 DOI: 10.1016/j.ctro.2024.100739] [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: 11/08/2023] [Revised: 01/25/2024] [Accepted: 01/27/2024] [Indexed: 02/22/2024] Open
Abstract
Stereotactic ablative body radiotherapy (SABR) aims to accurately deliver a higher than conventional dose of radiotherapy to a well-defined target tumour incorporating advanced immobilisation and imaging techniques. SABR is an emerging treatment option for primary kidney cancer especially when surgery is contraindicated. Increasingly, SABR is being incorporated into the management of low-volume stage IV kidney cancers to delay the need for systemic therapy or to prolong the duration of ongoing systemic treatment. This review will evaluate the evidence and limitations of SABR for oligometastatic renal cell carcinoma.
Collapse
Affiliation(s)
- Chang David
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Ali Muhammad
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Udovicich Cristian
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Tran Ben
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Azad Arun
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Au Lewis
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Spain Lavinia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Perera Marlon
- Division of Surgical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Siva Shankar
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
9
|
Singhal T, Singh P, Parida GK, Agrawal K. Role of PSMA-targeted PET-CT in renal cell carcinoma: a systematic review and meta-analysis. Ann Nucl Med 2024; 38:176-187. [PMID: 38340144 DOI: 10.1007/s12149-024-01904-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/08/2024] [Indexed: 02/12/2024]
Abstract
Fluoro-deoxy glucose positron emission tomography/computed tomography (PET/CT), the workhorse of nuclear medicine, has limited utility for renal cell carcinoma (RCC), particularly clear cell variant. Thus, various other tracers have been tried for evaluation of RCC. One of the most promising targets for radiotracers is prostate-specific membrane antigen (PSMA) expressed in abundance in carcinoma-associated neo-vasculature. Thus, we tried to review and analyse the role of PSMA-targeted PET/CT in evaluation of RCC. Databases like PubMed, EMBASE and SCOPUS were searched for original studies published on PSMA-targeted PET/CT in RCC till 30 September 2023. Revised Tool for the Quality Assessment of Diagnostic Accuracy Studies-2 (QUADAS-2) checklist was used to assess the included studies. Pooled sensitivity and specificity were calculated and represented with 95% confidence intervals (95%CI). Heterogeneity in the studies was assessed by I-square index. Pooled sensitivity and specificity of PSMA-targeted PET/CT for detection of local disease estimates were 87.2% (95%CI: 77-94%) and 100% (95%CI: 92.9-100%), respectively. Pooled sensitivity and specificity for detection of local recurrent disease are 100% (95%CI: 71.5-100%) and 100% (95%CI: 89.4-100%), respectively. Pooled sensitivity and specificity for detection of metastatic disease are 92% (95%CI: 86.2-96%) and 96.9% (95%CI: 83.8-99.9%), respectively. Pooled sensitivity of PSMA-targeted PET/CT for detection of clear cell renal cell carcinoma (ccRCC) and non-ccRCC are 94.7% (95%CI: 88-98.3%) and 75% (95%CI: 35-96.8%), respectively. PSMA-targeted PET-CT demonstrated better diagnostic efficacy for the detection of recurrent RCC. Whilst for staging RCC, it had higher specificity but lower sensitivity. Thus, it can serve as a non-invasive adjuvant tool to conventional imaging in the evaluation of staging of RCC, particularly clear cell variant.
Collapse
Affiliation(s)
- Tejasvini Singhal
- Department of Nuclear Medicine, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Parneet Singh
- Department of Nuclear Medicine, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Girish Kumar Parida
- Department of Nuclear Medicine, All India Institute of Medical Sciences, Bhubaneswar, India.
| | - Kanhaiyalal Agrawal
- Department of Nuclear Medicine, All India Institute of Medical Sciences, Bhubaneswar, India
| |
Collapse
|
10
|
Aggarwal P, Singh H, Das CK, Mavuduru RS, Kakkar N, Lal A, Gorsi U, Kumar R, Mittal BR. Potential role of 68Ga-PSMA PET/CT in metastatic renal cell cancer: A prospective study. Eur J Radiol 2024; 170:111218. [PMID: 38007857 DOI: 10.1016/j.ejrad.2023.111218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 11/28/2023]
Abstract
PURPOSE Prostate-specific membrane antigen (PSMA), in addition to its utility in prostate cancer, is also an angiogenic imaging marker for hypervascular tumors like renal cell carcinoma (RCC). Our study aims to assess the potential role of 68Ga-PSMA-11 positron emission tomography (PET)/CT in metastatic RCC and compare it with contrast-enhanced computed tomography (CECT). METHODS Biopsy-proven RCC patients with known or suspected distant metastases who underwent 68Ga-PSMA-11 PET/CT for staging/restaging were prospectively recruited. Those patients who had undergone 18F-FDG PET/CT within six weeks of 68Ga-PSMA PET/CT were also included retrospectively for comparative analysis. A patient-based and lesion-based analysis was done to compare the lesion detection rates of CECT, 68Ga-PSMA-11 PET and 18F-FDG PET. PET-based quantitative parameters were also compared between both the PET modalities. Impact of baseline parameters on survival was assessed using Cox regression analysis. A p-value of < 0.05 was considered significant. RESULTS Thirty-seven patients with median age 60 years ± 13 years (range = 26-76 years) were included in the study. Twenty-seven patients had clear cell (cc) RCC, six had papillary RCC (pRCC), and one each had an eosinophilic variant of ccRCC, collecting duct RCC, translocation RCC and poorly differentiated RCC. 68Ga-PSMA-11 PET performed better in detecting marrow and equivocal bone lesions and worse in detecting liver lesions compared to CECT. 68Ga-PSMA-11 PET-based angiogenic tumor burden estimation using Total Lesion-PSMA (TL-PSMA) and PSMA-Total volume (PSMA-TV) had a prognostic impact on the survival of patients. 68Ga-PSMA-11 PET also detected more lesions and showed significantly higher SUVmax than 18F-FDG PET. CONCLUSION 68Ga-PSMA-11 PET/CT performs better than CECT and 18F-FDG PET/CT in metastatic evaluation and has prognostic value in the management of clear cell RCC.
Collapse
Affiliation(s)
- Piyush Aggarwal
- Department of Nuclear Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Harmandeep Singh
- Department of Nuclear Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India.
| | - Chandan Krushna Das
- Department of Clinical Hematology and Medical Oncology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | | | - Nandita Kakkar
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Anupam Lal
- Department of Radiodiagnosis, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Ujjwal Gorsi
- Department of Radiodiagnosis, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Rajender Kumar
- Department of Nuclear Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| | - Bhagwant Rai Mittal
- Department of Nuclear Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh 160012, India
| |
Collapse
|
11
|
Voter AF, Werner RA, Savas H, Gafita A, Ross AE, Gorin MA, Solnes LB, Pomper MG, Rowe SP, Sheikhbahaei S. A Practical Guide to the Pearls and Pitfalls of PSMA PET Imaging. Semin Nucl Med 2024; 54:119-131. [PMID: 37980186 DOI: 10.1053/j.semnuclmed.2023.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/30/2023] [Accepted: 11/01/2023] [Indexed: 11/20/2023]
Abstract
Prostate-specific membrane antigen (PSMA)-targeted PET agents have revolutionized the care of patients with prostate cancer, supplanting traditional methods of imaging prostate cancer, and improving the selection and delivery of therapies. This has led to a rapid expansion in both the number of PSMA PET scans performed and the imaging specialists required to interpret those scans. To aid those imagers and clinicians who are new to the interpretation of PSMA PET, this review provides an overview of the interpretation of PSMA PET/CT imaging and pearls for overcoming commonly encountered pitfalls. We discuss the physiologic distribution of the clinically available PSMA-targeted radiotracers, the commonly encountered patterns of prostate cancer spread, as well as the benign and malignant mimics of prostate cancer. Additionally, we review the standardized PSMA PET reporting systems and the role of PSMA in selecting appropriate patients for PSMA-targeted therapies.
Collapse
Affiliation(s)
- Andrew F Voter
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Rudolf A Werner
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Hatice Savas
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Andrei Gafita
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ashley E Ross
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Michael A Gorin
- Milton and Carroll Petrie Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Lilja B Solnes
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Martin G Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Steven P Rowe
- Department of Radiology, University of North Carolina, Chapel Hill, NC.
| | - Sara Sheikhbahaei
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
12
|
Urso L, Bauckneht M, Albano D, Chondrogiannis S, Grassetto G, Lanfranchi F, Dondi F, Fornarini G, Lazzeri M, Evangelista L. The evolution of PET imaging in renal, bladder, upper urinary tract urothelial, testicular and penile carcinoma - Today's impact, tomorrow's potential. Expert Rev Med Devices 2024; 21:55-72. [PMID: 38072680 DOI: 10.1080/17434440.2023.2293919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 12/07/2023] [Indexed: 02/05/2024]
Abstract
INTRODUCTION The advancement of hybrid PET/CT or PET/MRI imaging for non-prostate genitourinary cancers has not experienced the rapid progress of prostate cancer. Nevertheless, these neoplasms are aggressive and reliable imaging plays a pivotal role in enhancing patients' quality of life and prognosis. AREAS COVERED the main evidence regarding [18F]FDG and non-[18F]FDG PET/CT or PET/MRI in non-prostate uro-oncological malignancies are summarized and discussed. Moreover, potential future directions concerning PET imaging in these neoplasms are debated, with the aim to stimulate future research projects covering these fields. EXPERT OPINION In Renal Cell Carcinoma (RCC), [18F]FDG PET/CT demonstrates varying efficacy in staging, restaging, and prognostic stratification, but PSMA PET/CT is emerging as a potential game-changer, particularly in advanced, high-grade aggressive clear cell RCC. [18F]FDG PET/CT may see an increased use in N and M-staging of bladder cancer, as well as for detecting recurrence and response to neoadjuvant chemotherapy. Preliminary data regarding [68Ga]-FAPI also looks promising in this context. [18F]FDG PET/MRI could be useful for the T-staging of bladder cancer, while upper tract urothelial carcinoma still lacks of molecular imaging literature reports. In testicular and penile cancer [18F]FDG PET/CT has demonstrated its usefulness in several clinical settings, although experiences with non-[18F]FDG radiotracers are lacking.
Collapse
Affiliation(s)
- Luca Urso
- Department of Nuclear Medicine - PET/CT Center, S. Maria Della Misericordia Hospital, Rovigo, Italy
| | - Matteo Bauckneht
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico, San Martino, Genova, Italy
- Department of Health Sciences (DISSAL), University of Genoa, Genova, Italy
| | - Domenico Albano
- Nuclear Medicine Department, ASST Spedali Civili of Brescia, University of Brescia, Brescia, Italy
| | - Sotirios Chondrogiannis
- Department of Nuclear Medicine - PET/CT Center, S. Maria Della Misericordia Hospital, Rovigo, Italy
| | - Gaia Grassetto
- Department of Nuclear Medicine - PET/CT Center, S. Maria Della Misericordia Hospital, Rovigo, Italy
| | - Francesco Lanfranchi
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico, San Martino, Genova, Italy
- Department of Health Sciences (DISSAL), University of Genoa, Genova, Italy
| | - Francesco Dondi
- Nuclear Medicine Department, ASST Spedali Civili of Brescia, University of Brescia, Brescia, Italy
| | - Giuseppe Fornarini
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico, San Martino, Genova, Italy
| | - Massimo Lazzeri
- Department of Urology, IRCCS Humanitas Clinical and Research Hospital, Rozzano, Italy
| | - Laura Evangelista
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Nuclear Medicine Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| |
Collapse
|
13
|
Bui VN, Unterrainer LM, Brendel M, Kunte SC, Holzgreve A, Allmendinger F, Bartenstein P, Klauschen F, Unterrainer M, Staehler M, Ledderose S. PSMA-Expression Is Highly Associated with Histological Subtypes of Renal Cell Carcinoma: Potential Implications for Theranostic Approaches. Biomedicines 2023; 11:3095. [PMID: 38002095 PMCID: PMC10668989 DOI: 10.3390/biomedicines11113095] [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: 09/23/2023] [Revised: 10/13/2023] [Accepted: 10/19/2023] [Indexed: 11/26/2023] Open
Abstract
In renal cell carcinoma (RCC), accurate imaging methods are required for treatment planning and response assessment to therapy. In addition, there is an urgent need for new therapeutic options, especially in metastatic RCC. One way to combine diagnostics and therapy in a so-called theranostic approach is the use of radioligands directed against surface antigens. For instance, radioligands against prostate-specific membrane antigen (PSMA) have already been successfully used for diagnosis and radionuclide therapy of metastatic prostate cancer. Recent studies have demonstrated that PSMA is expressed not only in prostate cancer but also in the neovasculature of several solid tumors, which has raised hopes to use PSMA-guided theranostic approaches in other tumor entities, too. However, data on PSMA expression in different histopathological subtypes of RCC are sparse. Because a better understanding of PSMA expression in RCC is critical to assess which patients would benefit most from theranostic approaches using PSMA-targeted ligands, we investigated the expression pattern of PSMA in different subtypes of RCC on protein level. Immunohistochemical staining for PSMA was performed on formalin-fixed, paraffin-embedded archival material of major different histological subtypes of RCC (clear cell RCC (ccRCC)), papillary RCC (pRCC) and chromophobe RCC (cpRCC). The extent and intensity of PSMA staining were scored semi-quantitatively and correlated with the histological RCC subtypes. Group comparisons were calculated with the Kruskal-Wallis test. In all cases, immunoreactivity was detected only in the tumor-associated vessels and not in tumor cells. Staining intensity was the strongest in ccRCC, followed by cpRCC and pRCC. ccRCC showed the most diffuse staining pattern, followed by cpRCC and pRCC. Our results provide a rationale for PSMA-targeted theranostic approaches in ccRCC and cpRCC.
Collapse
Affiliation(s)
- Vinh Ngoc Bui
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (M.B.); (S.C.K.); (F.A.); (P.B.); (M.U.)
| | - Lena M. Unterrainer
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (M.B.); (S.C.K.); (F.A.); (P.B.); (M.U.)
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (M.B.); (S.C.K.); (F.A.); (P.B.); (M.U.)
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Sophie C. Kunte
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (M.B.); (S.C.K.); (F.A.); (P.B.); (M.U.)
| | - Adrien Holzgreve
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (M.B.); (S.C.K.); (F.A.); (P.B.); (M.U.)
| | - Fabian Allmendinger
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (M.B.); (S.C.K.); (F.A.); (P.B.); (M.U.)
| | - Peter Bartenstein
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (M.B.); (S.C.K.); (F.A.); (P.B.); (M.U.)
| | | | - Marcus Unterrainer
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (M.B.); (S.C.K.); (F.A.); (P.B.); (M.U.)
- Die RADIOLOGIE, 80331 Munich, Germany
| | - Michael Staehler
- Department of Urology, LMU University Hospital, LMU Munich, 81377 Munich, Germany;
| | - Stephan Ledderose
- Institute of Pathology, LMU Munich, 81377 Munich, Germany; (F.K.); (S.L.)
| |
Collapse
|
14
|
Baraban EG, Ged Y, Singla N, Allaf ME, Gorin MA, Markowski MC, Rowe SP, Argani P. Vascular Expression of Prostate-specific Membrane Antigen (PSMA) in MiTF Family Translocation Renal Cell Carcinoma and Related Neoplasms. Appl Immunohistochem Mol Morphol 2023; 31:544-549. [PMID: 37471632 DOI: 10.1097/pai.0000000000001142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 06/07/2023] [Indexed: 07/22/2023]
Abstract
Multiple studies have demonstrated prostate-specific membrane antigen (PSMA) expression in the neo-vasculature of non-prostate tumors including clear cell renal cell carcinoma (ccRCC). However, PSMA expression in rare renal tumors including MiTF family translocation renal cell carcinoma has not been previously characterized. We examined PSMA expression by immunohistochemistry in a series of MiTF family translocation renal cell carcinomas as well as in several genetically related tumors including alveolar soft part sarcoma and PEComas with TFE3 rearrangements. PSMA expression was also studied in several cases of ccRCC and papillary RCC. Overall, PSMA immunohistochemistry was performed in 61 samples from 58 patients. Vascular PSMA expression was seen with the highest frequency in ccRCC [88% (14/16)] (38% focal, 50% diffuse). Translocation RCC (tRCC) demonstrated the second highest frequency of PSMA expression [71% (22/28)] (57% focal, 14% diffuse), followed by alveolar soft part sarcoma [50% (4/8)] (38% focal, 12% diffuse). No PSMA expression was seen in PEComas with TFE3 rearrangement (0/3) or papillary RCC (0/6). PSMA expression was only present in tumor-associated neo-vasculature. A patient with oligometastatic tRCC underwent 68 Ga-PSMA-11 PET imaging which detected multiple putative metastatic lesions not detected on conventional computed tomography imaging performed 2 weeks prior, supporting the potential utility of PSMA imaging in tRCC. These findings have potential implications for the utility of PSMA guided diagnostic and therapeutic agents in both common and uncommon renal cell carcinoma subtypes as well as genetically related mesenchymal neoplasms.
Collapse
Affiliation(s)
| | | | - Nirmish Singla
- The James Buchanan Brady Urological Institute and Department of Urology
| | - Mohammad E Allaf
- The James Buchanan Brady Urological Institute and Department of Urology
| | - Michael A Gorin
- Milton and Carroll Petrie, Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY
| | | | - Steven P Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD
| | | |
Collapse
|
15
|
Haj-Mirzaian A, Mahmood U, Heidari P. Targeted Molecular Imaging as a Biomarker in Urologic Oncology. Urol Clin North Am 2023; 50:115-131. [PMID: 36424076 PMCID: PMC10133841 DOI: 10.1016/j.ucl.2022.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Urologic malignancies constitute a large portion of annually diagnosed cancers. Timely diagnosis, accurate staging, and assessment of tumor heterogeneity are essential to devising the best treatment strategy for individual patients. The high sensitivity of molecular imaging allows for early and sensitive detection of lesions that were not readily detectable using conventional imaging techniques. Moreover, molecular imaging enables the interrogation of molecular processes used in targeted cancer therapies and predicts cancer response to treatment. Here we review the current advancements in molecular imaging of urologic cancers, including prostatic, vesical, renal testicular, and ureteral cancers.
Collapse
Affiliation(s)
- Arvin Haj-Mirzaian
- Department of Radiology, Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, 55 Fruit St, Wht 427, Boston, MA 02114, USA; Center for Precision Imaging, Athinoula A Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 55 Fruit St, Wht 427, Boston, MA 02114, USA
| | - Umar Mahmood
- Department of Radiology, Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, 55 Fruit St, Wht 427, Boston, MA 02114, USA; Center for Precision Imaging, Athinoula A Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 55 Fruit St, Wht 427, Boston, MA 02114, USA.
| | - Pedram Heidari
- Department of Radiology, Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, 55 Fruit St, Wht 427, Boston, MA 02114, USA; Center for Precision Imaging, Athinoula A Martinos Center for Biomedical Imaging, Massachusetts General Hospital, 55 Fruit St, Wht 427, Boston, MA 02114, USA
| |
Collapse
|
16
|
Posada Calderon L, Eismann L, Reese SW, Reznik E, Hakimi AA. Advances in Imaging-Based Biomarkers in Renal Cell Carcinoma: A Critical Analysis of the Current Literature. Cancers (Basel) 2023; 15:cancers15020354. [PMID: 36672304 PMCID: PMC9856305 DOI: 10.3390/cancers15020354] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/31/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Cross-sectional imaging is the standard diagnostic tool to determine underlying biology in renal masses, which is crucial for subsequent treatment. Currently, standard CT imaging is limited in its ability to differentiate benign from malignant disease. Therefore, various modalities have been investigated to identify imaging-based parameters to improve the noninvasive diagnosis of renal masses and renal cell carcinoma (RCC) subtypes. MRI was reported to predict grading of RCC and to identify RCC subtypes, and has been shown in a small cohort to predict the response to targeted therapy. Dynamic imaging is promising for the staging and diagnosis of RCC. PET/CT radiotracers, such as 18F-fluorodeoxyglucose (FDG), 124I-cG250, radiolabeled prostate-specific membrane antigen (PSMA), and 11C-acetate, have been reported to improve the identification of histology, grading, detection of metastasis, and assessment of response to systemic therapy, and to predict oncological outcomes. Moreover, 99Tc-sestamibi and SPECT scans have shown promising results in distinguishing low-grade RCC from benign lesions. Radiomics has been used to further characterize renal masses based on semantic and textural analyses. In preliminary studies, integrated machine learning algorithms using radiomics proved to be more accurate in distinguishing benign from malignant renal masses compared to radiologists' interpretations. Radiomics and radiogenomics are used to complement risk classification models to predict oncological outcomes. Imaging-based biomarkers hold strong potential in RCC, but require standardization and external validation before integration into clinical routines.
Collapse
Affiliation(s)
- Lina Posada Calderon
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Lennert Eismann
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Stephen W. Reese
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ed Reznik
- Computational Oncology, Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Abraham Ari Hakimi
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Correspondence:
| |
Collapse
|
17
|
Rizzo A, Racca M, Dall’Armellina S, Rescigno P, Banna GL, Albano D, Dondi F, Bertagna F, Annunziata S, Treglia G. The Emerging Role of PET/CT with PSMA-Targeting Radiopharmaceuticals in Clear Cell Renal Cancer: An Updated Systematic Review. Cancers (Basel) 2023; 15:355. [PMID: 36672305 PMCID: PMC9857064 DOI: 10.3390/cancers15020355] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Recent articles proposed the employment of positron emission tomography/computed tomography (PET/CT) with prostate-specific membrane antigen (PSMA)-targeting radiopharmaceuticals in clear cell renal cell carcinoma (ccRCC). METHODS The authors performed a comprehensive literature search of studies on the performance of PET/CT with PSMA-targeting radiopharmaceuticals in ccRCC. Original articles concerning this imaging examination were included in newly diagnosed ccRCC patients and ccRCC patients with disease recurrence. RESULTS A total of sixteen papers concerning the diagnostic performance of PSMA-targeted PET/CT in ccRCC (331 patients) were included in this systematic review. The included articles demonstrated an excellent detection rate of PSMA-targeting PET/CT in ccRCC. CONCLUSIONS PSMA-targeted PET/CT seems promising in detecting ccRCC lesions as well as in discriminating the presence of aggressive phenotypes. Prospective multicentric studies are warranted to strengthen the role of PSMA-targeting PET/CT in ccRCC.
Collapse
Affiliation(s)
- Alessio Rizzo
- Department of Nuclear Medicine, Candiolo Cancer Institute, FPO–IRCCS, 10060 Turin, Italy
| | - Manuela Racca
- Department of Nuclear Medicine, Candiolo Cancer Institute, FPO–IRCCS, 10060 Turin, Italy
| | - Sara Dall’Armellina
- Nuclear Medicine Unit, Department of Medical Sciences, AOU Città della Salute e della Scienza, University of Turin, 10126 Turin, Italy
| | - Pasquale Rescigno
- Department of Oncology, Candiolo Cancer Institute, FPO-IRCCS, 10060 Turin, Italy
| | | | - Domenico Albano
- Division of Nuclear Medicine, Università degli Studi di Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Francesco Dondi
- Division of Nuclear Medicine, Università degli Studi di Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Francesco Bertagna
- Division of Nuclear Medicine, Università degli Studi di Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Salvatore Annunziata
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Giorgio Treglia
- Clinic of Nuclear Medicine, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, 6501 Bellinzona, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| |
Collapse
|
18
|
PSMA Expression in Solid Tumors beyond the Prostate Gland: Ready for Theranostic Applications? J Clin Med 2022; 11:jcm11216590. [PMID: 36362824 PMCID: PMC9657217 DOI: 10.3390/jcm11216590] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
In the past decades, the expanding use of prostate-specific membrane antigen (PSMA) imaging for prostate cancer has led to the incidental detection of a lot of extra-prostatic malignancies showing an increased uptake of PSMA. Due to these incidental findings, the increasing amount of immunohistochemistry studies and the deeper knowledge of the mechanisms of expression of this antigen, it is now clear that “PSMA” is a misnomer, since it is not specific to the prostate gland. Nevertheless, this lack of specificity could represent an interesting opportunity to bring new insights on the biology of PSMA and its sites of expression to image and treat new conditions, particularly several cancers. In this review, we will describe the main extra-prostatic cancers that exhibit PSMA expression and that can be studied with PSMA-based positron emission tomography–computed tomography (PET/CT) as an additional or alternative tool to conventional imaging. In particular, we will focus on cancers in which a radioligand therapy with 177lutetium has been attempted, aiming to provide an overview of the possible future theragnostic applications of PSMA.
Collapse
|
19
|
Impact of Prostate-specific Membrane Antigen Positron Emission Tomography/Computed Tomography in the Management of Oligometastatic Renal Cell Carcinoma. EUR UROL SUPPL 2022; 44:60-68. [PMID: 36185587 PMCID: PMC9520507 DOI: 10.1016/j.euros.2022.08.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2022] [Indexed: 11/21/2022] Open
Abstract
Background Prostate-specific membrane antigen (PSMA) is overexpressed in the neovasculature of renal cell carcinoma (RCC). However, there remains limited evidence regarding the use of PSMA positron emission tomography/computed tomography (PET/CT) in RCC. Objective To assess the impact of PSMA PET/CT in the management of metastatic RCC. Design, setting, and participants This was a retrospective review of patients who underwent PSMA PET/CT from 2014 to 2020 for restaging or suspected metastatic RCC in a tertiary academic setting. Outcome measurements and statistical analysis Management plans before and after PSMA PET/CT were recorded. Impact was classified as high (change of treatment intent, modality, or site), medium (change in treatment method), or low. Secondary outcomes included the patient-level detection rate, PSMA PET/CT parameters, sensitivity, and comparison to CT and, if available, fluorodeoxyglucose (FDG) PET/CT. Results and limitations Sixty-one patients met the inclusion criteria, of whom 54 (89%) had clear cell RCC. PSMA-positive disease was detected in 51 patients (84%). For 30 patients (49%) there was a change in management due to PSMA PET/CT (high impact, 29 patients, 48%). In 15 patients (25%), more metastases were detected on PSMA PET/CT than on CT. The sensitivity of combined PSMA PET/CT and diagnostic CT was 91% (95% confidence interval 77–98%). In a subcohort of 40 patients, the detection rate was 88% for PSMA and 75% for FDG PET/CT (p = 0.17). The maximum standardised uptake value (SUVmax) was higher for PSMA than for FDG PET/CT (15.2 vs 8.0; p = 0.02). Limitations include selection bias due to the retrospective design, and a lack of corresponding histopathology for all patients. Conclusions PSMA PET/CT is a promising imaging modality in metastatic RCC and led to a change in management in 49% of patients. PSMA PET/CT detected additional metastases compared to CT in 25% of patients and registered a significantly higher SUVmax than FDG PET/CT. Prospective studies are required to further define its role. Patient summary We report on a group of patients undergoing a new type of imaging for suspected advanced kidney cancer, called PSMA PET/CT. This imaging changed the management plan in 49% of the patients. PSMA PET/CT detected metastases in 84% of our patients and detected more metastases than computed tomography imaging in 25%.
Collapse
|
20
|
Rizzo A, Dall’Armellina S, Pizzuto DA, Perotti G, Zagaria L, Lanni V, Treglia G, Racca M, Annunziata S. PSMA Radioligand Uptake as a Biomarker of Neoangiogenesis in Solid Tumours: Diagnostic or Theragnostic Factor? Cancers (Basel) 2022; 14:4039. [PMID: 36011032 PMCID: PMC9406909 DOI: 10.3390/cancers14164039] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/10/2022] [Accepted: 08/17/2022] [Indexed: 01/10/2023] Open
Abstract
Due to its overexpression on the surface of prostate cancer cells, prostate-specific membrane antigen (PSMA) is a relatively novel effective target for molecular imaging and radioligand therapy (RLT) in prostate cancer. Recent studies reported that PSMA is expressed in the neovasculature of various types of cancer and regulates tumour cell invasion as well as tumour angiogenesis. Several authors explored the role of diagnostic and therapeutic PSMA radioligands in various malignancies. In this narrative review, we describe the current status of the literature on PSMA radioligands' application in solid tumours other than prostate cancer to explore their potential role as diagnostic or therapeutic agents, with particular regard to the relevance of PSMA radioligand uptake as neoangiogenetic biomarker. Hence, a comprehensive review of the literature was performed to find relevant articles on the applications of PSMA radioligands in non-prostate solid tumours. Data on the general, methodological and clinical aspects of all included studies were collected. Forty full-text papers were selected for final review, 8 of which explored PSMA radioligand PET/CT performances in gliomas, 3 in salivary gland malignancies, 6 in thyroid cancer, 2 in breast cancer, 16 in renal cell carcinoma and 5 in hepatocellular carcinoma. In the included studies, PSMA radioligand PET showed promising performance in patients with non-prostate solid tumours. Further studies are needed to better define its potential role in oncological patients management, especially in those undergoing antineoangiogenic therapies, and to assess the efficacy of PSMA-RLT in this clinical context.
Collapse
Affiliation(s)
- Alessio Rizzo
- Department of Nuclear Medicine, Candiolo Cancer Institute, FPO—IRCCS, 10060 Turin, Italy
| | - Sara Dall’Armellina
- Nuclear Medicine Unit, Department of Medical Sciences, AOU Città della Salute e della Scienza, University of Turin, 10134 Turin, Italy
| | - Daniele Antonio Pizzuto
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Germano Perotti
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Luca Zagaria
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Valerio Lanni
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Giorgio Treglia
- Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, 6501 Bellinzona, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| | - Manuela Racca
- Department of Nuclear Medicine, Candiolo Cancer Institute, FPO—IRCCS, 10060 Turin, Italy
| | - Salvatore Annunziata
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| |
Collapse
|
21
|
Abstract
Renal cell carcinoma (RCC) is the sixth most common cancer among men and the ninth among women, and its prognosis is closely correlated with metastasis. Targeted therapy and immunotherapy are the main adjuvant treatments for advanced RCC and require early diagnosis, precise assessment, and prediction of the therapeutic responses. Current conventional imaging methods of RCC only provide structural information rather than biological processes. Noninvasive diagnostic tools are therefore needed to image RCC early and accurately at the molecular level. Nuclear medicine imaging combines the high sensitivity of radionuclides with the high resolution of structural imaging to visualize the metabolic processes and specific targets of RCC for more accurate and reliable diagnosis, staging, prognosis prediction, and response assessment. This review summarizes the most recent applications of nuclear medicine receptor imaging and metabolic imaging in RCC and highlights future development perspectives in the field.
Collapse
Affiliation(s)
- Qianyun Wu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200217, China
| |
Collapse
|
22
|
Prostate-Specific Membrane Antigen as Target for Neuroimaging of Central Nervous System Tumors. Mol Imaging 2022; 2022:5358545. [PMID: 35517711 PMCID: PMC9042374 DOI: 10.1155/2022/5358545] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/24/2022] [Indexed: 12/18/2022] Open
Abstract
Introduction. Positron emission tomography (PET) imaging with prostate-specific membrane antigen- (PSMA-) binding tracers has been found incidentally to demonstrate uptake in CNS tumors. Following the encouraging findings of several such case reports, there is a growing interest in the potential application of PSMA-targeted PET imaging for diagnostics, theranostics, and monitoring of CNS tumors. This is a systematic literature review on PSMA-binding tracers in CNS tumors. Methods. A PubMed search was conducted, including preclinical and clinical reports. One hundred and twelve records were identified, and after screening, 56 were included in the final report. Results. Tissue studies demonstrated PSMA expression in tumor vascular endothelial cells, without expression in normal brain tissue, though the extent and intensity of staining varied by anti-PSMA antibody and methodology. Most included studies reported on gliomas, which showed strong PSMA ligand uptake and more favorable tumor to background ratios than other PET tracers. There are also case reports demonstrating PSMA ligand uptake in prostate cancer brain metastases, nonprostate cancer brain metastases, and meningiomas. We also review the properties of the various PSMA-binding radiotracers available. Therapeutic and theranostic applications of PSMA-binding tracers have been studied, including labeled alpha- and beta-ray emitting isotopes, as well as PSMA targeting in directing MRI-guided focused ultrasound. Conclusions. There is a potential application for PSMA-targeted PET in neuro-oncology as a combination of diagnostic and therapeutic use, as a theranostic modality for managing CNS tumors. Further research is needed regarding the mechanism(s) of PSMA expression in CNS tumors and its differential performance by tumor type.
Collapse
|
23
|
Muselaers S, Erdem S, Bertolo R, Ingels A, Kara Ö, Pavan N, Roussel E, Pecoraro A, Marchioni M, Carbonara U, Marandino L, Amparore D, Campi R, on behalf of the European Association of Urology (EAU) Young Academic Urologists (YAU) Renal Cancer Working Group. PSMA PET/CT in Renal Cell Carcinoma: An Overview of Current Literature. J Clin Med 2022; 11:jcm11071829. [PMID: 35407435 PMCID: PMC8999609 DOI: 10.3390/jcm11071829] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 03/23/2022] [Indexed: 11/23/2022] Open
Abstract
Although the vast majority of prostate-specific membrane antigen (PSMA) positron emission tomography (PET) imaging occurs in the field of prostate cancer, PSMA is also highly expressed on the cell surface of the microvasculature of several other solid tumors, including renal cell carcinoma (RCC). This makes it a potentially interesting imaging target for the staging and monitoring of RCC. The objective of this review is to provide an overview of the current evidence regarding the use of PSMA PET/Computed Tomography in RCC patients.
Collapse
Affiliation(s)
- Stijn Muselaers
- Department of Urology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Correspondence: ; Tel.: +31-243-613-735
| | - Selcuk Erdem
- Division of Urologic Oncology, Department of Urology, Faculty of Medicine, Istanbul University, Istanbul 34452, Turkey;
| | - Riccardo Bertolo
- Department of Urology, San Carlo Di Nancy Hospital, 00165 Rome, Italy;
| | - Alexandre Ingels
- Department of Urology, University Hospital Henri Mondor, APHP, 94000 Créteil, France;
| | - Önder Kara
- Department of Urology, School of Medicine, Kocaeli University, İzmit 41001, Turkey;
| | - Nicola Pavan
- Urology Clinic, Department of Medical, Surgical and Health Science, University of Trieste, 34127 Trieste, Italy;
| | - Eduard Roussel
- Department of Urology, University Hospitals Leuven, 3000 Leuven, Belgium;
| | - Angela Pecoraro
- Department of Urology, San Luigi Gonzaga Hospital, University of Turin, Orbassano, 10124 Turin, Italy; (A.P.); (D.A.)
| | - Michele Marchioni
- Department of Medical, Oral and Biotechnological Sciences, G. d’Annunzio University of Chieti, 66100 Chieti, Italy;
| | - Umberto Carbonara
- Department of Emergency and Organ Transplantation-Urology, Andrology and Kidney Transplantation, University of Bari, 70121 Bari, Italy;
| | - Laura Marandino
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, 20132 Milan, Italy;
| | - Daniele Amparore
- Department of Urology, San Luigi Gonzaga Hospital, University of Turin, Orbassano, 10124 Turin, Italy; (A.P.); (D.A.)
| | - Riccardo Campi
- Unit of Urological Robotic Surgery and Renal Transplantation, Careggi Hospital, University of Florence, 50121 Florence, Italy;
| | | |
Collapse
|
24
|
Non-prostate cancer tumours: incidence on 18F-DCFPyL PSMA PET/CT and uptake characteristics in 1445 patients. Eur J Nucl Med Mol Imaging 2022; 49:3277-3288. [PMID: 35254481 PMCID: PMC9250467 DOI: 10.1007/s00259-022-05721-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 02/07/2022] [Indexed: 12/21/2022]
Abstract
Purpose With increasing use of PSMA PET/CT in the staging and restaging of prostate cancer (PCa), the identification of non-prostate cancer tumours (NPCaT) has become an increasing clinical dilemma. Atypical presentations of PSMA expression in prostate cancer and expression in NPCaT are not well established. Understanding the normal and abnormal distribution of PSMA expression is essential in preparing clinically relevant reports and in guiding multidisciplinary discussion and decisions. Methods Retrospective review of 1445 consecutive 18F-DCFPyL PSMA PET/CT studies by experienced radiologists and nuclear medicine physicians. Lesions indeterminate for PCa were identified. Correlation was made with patient records, biopsy results, and dedicated imaging. Lesions were then categorized into four groups: 1. Confirmed prostate cancer, metastases, 2. NPCaT 3. Benign, and 4. Indeterminate lesions. Results 68/1445 patients had lesions atypical for prostate cancer metastases. These comprised 8/68 (11.8%) atypical prostate cancer metastases, 17/68 (25.0%) NPCaT, 29/68 (42.6%) indeterminate, and 14/68 (20.6%) benign. In the context of the entire cohort, these are adjusted to 8/1445 (0.6%), 17/1445 (1.2%), 29/1445 (2.0%), and 14/1445 (1.0%) respectively. With the exception of Renal Cell Carcinoma (RCC), NPCaT demonstrated no or low PSMA expression. A similar trend was also observed for indeterminate and benign lesions. Conversely, most atypical PCa metastases demonstrated intermediate or high PSMA expression. Conclusion 18F-DCFPyL PSMA PET/CT detection of NPCaT is low. Lesions demonstrating intermediate to high PSMA expression were exclusively prostate cancer metastases, aside from RCC, and lesions detected in organs with high background expression. Supplementary Information The online version contains supplementary material available at 10.1007/s00259-022-05721-z.
Collapse
|
25
|
Urso L, Castello A, Rocca GC, Lancia F, Panareo S, Cittanti C, Uccelli L, Florimonte L, Castellani M, Ippolito C, Frassoldati A, Bartolomei M. Role of PSMA-ligands imaging in Renal Cell Carcinoma management: current status and future perspectives. J Cancer Res Clin Oncol 2022; 148:1299-1311. [PMID: 35217902 PMCID: PMC9114025 DOI: 10.1007/s00432-022-03958-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 02/14/2022] [Indexed: 12/17/2022]
Abstract
Background Renal masses detection is continually increasing worldwide, with Renal Cell Carcinoma (RCC) accounting for approximately 90% of all renal cancers and remaining one of the most aggressive urological malignancies. Despite improvements in cancer management, accurate diagnosis and treatment strategy of RCC by computed tomography (CT) and magnetic resonance imaging (MRI) are still challenging. Prostate-Specific Membrane Antigen (PSMA) is known to be highly expressed on the endothelial cells of the neovasculature of several solid tumors other than prostate cancer, including RCC. In this context, recent preliminary studies reported a promising role for positron emission tomography (PET)/CT with radiolabeled molecules targeting PSMA, in alternative to fluorodeoxyglucose (FDG) in RCC patients. Purpose The aim of our review is to provide an updated overview of current evidences and major limitations regarding the use of PSMA PET/CT in RCC. Methods A literature search, up to 31 December 2021, was performed using the following electronic databases: PubMed, SCOPUS, Web of Science, and Google Scholar. Results The findings of this review suggest that PSMA PET/CT could represent a valid imaging option for diagnosis, staging, and therapy response evaluation in RCC, particularly in clear cell RCC. Conclusions Further studies are needed for this “relatively” new imaging modality to consolidate its indications, timing, and practical procedures.
Collapse
Affiliation(s)
- Luca Urso
- Department of Translational Medicine, University of Ferrara, Via Aldo Moro 8, 44124, Ferrara, Italy.,Nuclear Medicine Unit, Oncological Medical and Specialists Department, University Hospital of Ferrara, Ferrara, Italy
| | - Angelo Castello
- Department of Nuclear Medicine, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Federica Lancia
- Oncological Medical and Specialists Department, Oncology Unit, University Hospital of Ferrara, Ferrara, Italy
| | - Stefano Panareo
- Nuclear Medicine Unit, Oncology and Haematology Department, University Hospital of Modena, Modena, Italy
| | - Corrado Cittanti
- Department of Translational Medicine, University of Ferrara, Via Aldo Moro 8, 44124, Ferrara, Italy. .,Nuclear Medicine Unit, Oncological Medical and Specialists Department, University Hospital of Ferrara, Ferrara, Italy.
| | - Licia Uccelli
- Department of Translational Medicine, University of Ferrara, Via Aldo Moro 8, 44124, Ferrara, Italy.,Nuclear Medicine Unit, Oncological Medical and Specialists Department, University Hospital of Ferrara, Ferrara, Italy
| | - Luigia Florimonte
- Department of Nuclear Medicine, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Massimo Castellani
- Department of Nuclear Medicine, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Carmelo Ippolito
- Urology Unit, Surgical Department, University Hospital of Ferrara, Ferrara, Italy
| | - Antonio Frassoldati
- Oncological Medical and Specialists Department, Oncology Unit, University Hospital of Ferrara, Ferrara, Italy
| | - Mirco Bartolomei
- Nuclear Medicine Unit, Oncological Medical and Specialists Department, University Hospital of Ferrara, Ferrara, Italy
| |
Collapse
|
26
|
DePalatis L, Martiniova L, de Almeida Graff T, Ravizzini G. Applications of PSMA-PET in tumors other than prostate cancer. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00116-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
27
|
Oflas M, Ozluk Y, Sanli O, Ozkan ZG, Kuyumcu S. 68Ga-PSMA Uptake Patterns of Clear Cell Renal Carcinoma Across Different Histopathological Subtypes. Clin Nucl Med 2022; 47:e45-e46. [PMID: 34284482 DOI: 10.1097/rlu.0000000000003814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
ABSTRACT 68Ga-prostate-specific membrane antigen (PSMA) PET/CT is a well-established imaging modality in patients with prostate cancer; however, PSMA expression is also reported in the tumor-associated neovasculature, including but not limited to hepatocellular carcinoma, breast cancer, and renal cell carcinoma. Herein, we present 2 patients diagnosed with different histopathological subtypes of renal cell carcinoma who underwent 68Ga-PSMA PET/CT before surgery. Both cases have different PSMA expression characteristics and are presented along with pathological findings.
Collapse
Affiliation(s)
| | | | - Oner Sanli
- Urology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | | | | |
Collapse
|
28
|
Werner RA, Pomper MG, Buck AK, Rowe SP, Higuchi T. SPECT and PET Radiotracers in Renal Imaging. Semin Nucl Med 2022; 52:406-418. [DOI: 10.1053/j.semnuclmed.2021.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 12/06/2021] [Indexed: 12/24/2022]
|
29
|
Currie GM, Trifunovic M, Liu J, Kim S, Gurney H. 18F-DCFPyL PET/CT in Metastatic Renal Cell Carcinoma. J Nucl Med Technol 2021; 50:282-285. [PMID: 34750233 DOI: 10.2967/jnmt.121.262799] [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: 06/25/2021] [Accepted: 09/10/2021] [Indexed: 11/16/2022] Open
Abstract
Targeted molecular imaging with positron emission tomography (PET) utilize chemical ligands is a peptide that specifically targets a receptor of interest. Prostate specific membrane antigen (PSMA) is substantially upregulated in prostate cancer but is also expressed in the neovascular tissue of several malignancies, including renal cell carcinoma (RCC). Radiolabeled peptide targets for PSMA may be helpful in detecting metastatic RCC lesions. This teaching case provides an insight incidental detection of RCC metastatic disease with PSMA targeted PET and explores potential in deliberate evaluation of RCC with PSMA targeted tracers.
Collapse
Affiliation(s)
| | - Marko Trifunovic
- Macquarie Medical Imaging, Macquarie University Hospital, Australia
| | - Jui Liu
- Macquarie Medical Imaging, Macquarie University Hospital, Australia
| | - Sang Kim
- Medical Oncology, Macquarie University, Australia
| | | |
Collapse
|
30
|
Filizoglu N, Cetin IA, Kissa TN, Niftaliyeva K, Ones T. 68Ga-PSMA PET/CT to Distinguish Brain Metastasis of Renal Cell Carcinoma From Radiation Necrosis After Stereotactic Radiosurgery. Clin Nucl Med 2021; 46:913-914. [PMID: 34284481 DOI: 10.1097/rlu.0000000000003820] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT Renal cell carcinoma (RCC) is the most common primary malignancy of the kidney. Approximately 35% of patients of RCC presents with distant metastasis at initial evaluation. CT and MRI are the mainstay imaging modalities. Distinguishing radiation necrosis from tumor progression after stereotactic radiosurgery is challenging. Herein, we present a case of a 43-year-old man with RCC who was treated with stereotactic radiosurgery for brain metastases. We want to emphasize the potential value of 68Ga-PSMA PET/CT imaging in the differential diagnosis and follow-up of tumor progression from radiation necrosis in RCC patients.
Collapse
Affiliation(s)
| | - Ilknur Alsan Cetin
- Radiation Oncology, Marmara University Pendik Training and Research Hospital, Istanbul, Turkey
| | | | | | - Tunc Ones
- From the Departments of Nuclear Medicine
| |
Collapse
|
31
|
Klinkhammer BM, Lammers T, Mottaghy FM, Kiessling F, Floege J, Boor P. Non-invasive molecular imaging of kidney diseases. Nat Rev Nephrol 2021; 17:688-703. [PMID: 34188207 PMCID: PMC7612034 DOI: 10.1038/s41581-021-00440-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2021] [Indexed: 02/05/2023]
Abstract
In nephrology, differential diagnosis or assessment of disease activity largely relies on the analysis of glomerular filtration rate, urinary sediment, proteinuria and tissue obtained through invasive kidney biopsies. However, currently available non-invasive functional parameters, and most serum and urine biomarkers, cannot capture intrarenal molecular disease processes specifically. Moreover, although histopathological analyses of kidney biopsy samples enable the visualization of pathological morphological and molecular alterations, they only provide information about a small part of the kidney and do not allow longitudinal monitoring. These limitations not only hinder understanding of the dynamics of specific disease processes in the kidney, but also limit the targeting of treatments to active phases of disease and the development of novel targeted therapies. Molecular imaging enables non-invasive and quantitative assessment of physiological or pathological processes by combining imaging technologies with specific molecular probes. Here, we discuss current preclinical and clinical molecular imaging approaches in nephrology. Non-invasive visualization of the kidneys through molecular imaging can be used to detect and longitudinally monitor disease activity and can therefore provide companion diagnostics to guide clinical trials, as well as the safe and effective use of drugs.
Collapse
Affiliation(s)
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany
- Department of Pharmaceutics, Utrecht University, Utrecht, Netherlands
- Department of Targeted Therapeutics, University of Twente, Enschede, Netherlands
| | - Felix M Mottaghy
- Department of Nuclear Medicine, University Hospital RWTH, Aachen, Germany
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, Netherlands
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany
- Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany
| | - Jürgen Floege
- Department of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany
| | - Peter Boor
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany.
- Department of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany.
- Electron Microscopy Facility, RWTH Aachen University Hospital, Aachen, Germany.
| |
Collapse
|
32
|
Comparison of 18F-DCFPyL and 18F-FDG PET/computed tomography for the restaging of clear cell renal cell carcinoma: preliminary results of 15 patients. Nucl Med Commun 2021; 41:1299-1305. [PMID: 32941403 DOI: 10.1097/mnm.0000000000001285] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
OBJECTIVES This study aimed to compare the diagnostic performance of F-DCFPyL and 2-deoxy-2-[F]fluoro-D-glucose (F-FDG PET/computed tomography in the restaging of clear cell renal cell carcinoma after nephrectomy. METHODS In this retrospective study, a total of 15 patients with suspected local recurrence of clear cell renal cell carcinoma or metastasis after surgery underwent both F-DCFPyL and F-FDG PET/computed tomography. A systematic comparison of the maximum standardized uptake value and the target to background ratio was carried out between the lesions detected by the two tracers. RESULTS A total of 42 lesions were detected either by F-DCFPyL PET/computed tomography or by F-FDG PET/computed tomography. F-DCFPyL PET/computed tomography, but not F-FDG PET/computed tomography, accurately distinguished the two local recurrence from four postoperative changes. The remaining 36 lesions were soft tissue (14) and bone lesions (22); all 36 lesions were detected by F-DCFPyL PET/computed tomography while only 10 (10/14) soft tissue lesions and 12 (12/22) bone lesions were detected by F-FDG PET/computed tomography. The higher detection rate of soft tissue lesions using F-DCFPyL PET/computed tomography was not statistically significant (P = 0.125); however, F-DCFPyL PET/computed tomography was statistically better (P = 0.002) at detecting bone lesions. The average maximum standardized uptake value and target to background ratio of F-DCFPyL were significantly higher than that of F-FDG for soft tissue lesions (maximum standardized uptake value P = 0.005; target to background ratio P = 0.028) and bone lesions (maximum standardized uptake value P = 0.001; target to background ratio P = 0.001). CONCLUSIONS Our preliminary results indicated that F-DCFPyL PET/computed tomography is superior to F-FDG PET/computed tomography for the detection of local recurrence at both the surgical site and in bone metastasis while the tracers are comparable in the detection of soft tissue metastases.
Collapse
|
33
|
Abstract
Piflufolastat F 18 (PYLARIFY®) is an 18F-labelled diagnostic imaging agent that has been developed by Progenics Pharmaceuticals Inc., a Lantheus company, for positron emission tomography (PET) that targets prostate-specific membrane antigen (PSMA). Piflufolastat F 18 was approved in the USA on 27 May 2021 for PET of PSMA positive lesions in men with prostate cancer with suspected metastasis who are candidates for initial definitive therapy or with suspected recurrence based on elevated serum prostate specific antigen (PSA) level. This article summarizes the milestones in the development of piflufolastat F 18 leading to this approval as a radioactive diagnostic agent in prostate cancer.
Collapse
Affiliation(s)
- Susan J Keam
- Springer Nature, Private Bag 65901, Mairangi Bay, Auckland, 0754, New Zealand.
| |
Collapse
|
34
|
Gühne F, Seifert P, Theis B, Steinert M, Freesmeyer M, Drescher R. PSMA-PET/CT in Patients with Recurrent Clear Cell Renal Cell Carcinoma: Histopathological Correlations of Imaging Findings. Diagnostics (Basel) 2021; 11:diagnostics11071142. [PMID: 34201583 PMCID: PMC8304877 DOI: 10.3390/diagnostics11071142] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 11/16/2022] Open
Abstract
PET/CT with prostate-specific membrane antigen (PSMA)-targeted tracers has been used in the diagnosis and staging of patients with clear cell renal cell carcinoma (ccRCC). For ccRCC primary tumors, PET parameters were shown to predict histologic grade and features. The aim of this study was to correlate PSMA PET/CT with histopathological findings in patients with metastatic recurrence of ccRCC. Patients with ccRCC who underwent PSMA-targeted PET/CT and subsequent histopathological evaluation of suspicious lesions were included. Specimens underwent immunohistochemical marking. Lesion diameter, volume and tracer uptake were correlated with the extent and intensity of molecular PSMA expression and with clinical findings. Twelve PET-positive lesions of nine patients were evaluated. Eleven ccRCC metastases and one prostate carcinoma were detected histopathologically. Molecular PSMA expression was detected in all lesions, which intensity and distribution did not correlate with PET parameters. PSMA-targeted PET/CT is a feasible tool for the evaluation of patients with ccRCC but cannot reliably predict histologic features of metastases. PSMA may also be expressed in malignant lesions other than ccRCC, leading to incidental detection of these tumors.
Collapse
Affiliation(s)
- Falk Gühne
- Clinic of Nuclear Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; (F.G.); (P.S.); (R.D.)
| | - Philipp Seifert
- Clinic of Nuclear Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; (F.G.); (P.S.); (R.D.)
| | - Bernhard Theis
- Pathology Division, Institute of Forensic Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany;
| | - Matthias Steinert
- Department of Thoracic Surgery, Leipzig University Hospital, Liebigstraße 18, 04103 Leipzig, Germany;
| | - Martin Freesmeyer
- Clinic of Nuclear Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; (F.G.); (P.S.); (R.D.)
- Correspondence: ; Tel.: +49-3641-9329801
| | - Robert Drescher
- Clinic of Nuclear Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; (F.G.); (P.S.); (R.D.)
| |
Collapse
|
35
|
PSMA radioligand therapy for solid tumors other than prostate cancer: background, opportunities, challenges, and first clinical reports. Eur J Nucl Med Mol Imaging 2021; 48:4350-4368. [PMID: 34120192 PMCID: PMC8566635 DOI: 10.1007/s00259-021-05433-w] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 05/24/2021] [Indexed: 12/11/2022]
Abstract
In the past decade, a growing body of literature has reported promising results for prostate-specific membrane antigen (PSMA)-targeted radionuclide imaging and therapy in prostate cancer. First clinical studies evaluating the efficacy of [177Lu]Lu-PSMA radioligand therapy (PSMA-RLT) demonstrated favorable results in prostate cancer patients. [177Lu]Lu-PSMA is generally well tolerated due to its limited side effects. While PSMA is highly overexpressed in prostate cancer cells, varying degrees of PSMA expression have been reported in other malignancies as well, particularly in the tumor-associated neovasculature. Hence, it is anticipated that PSMA-RLT could be explored for other solid cancers. Here, we describe the current knowledge of PSMA expression in other solid cancers and define a perspective towards broader clinical implementation of PSMA-RLT. This review focuses specifically on salivary gland cancer, glioblastoma, thyroid cancer, renal cell carcinoma, hepatocellular carcinoma, lung cancer, and breast cancer. An overview of the (pre)clinical data on PSMA immunohistochemistry and PSMA PET/CT imaging is provided and summarized. Furthermore, the first clinical reports of non-prostate cancer patients treated with PSMA-RLT are described.
Collapse
|
36
|
Golan S, Aviv T, Groshar D, Yakimov M, Zohar Y, Prokocimer Y, Nadu A, Baniel J, Domachevsky L, Bernstine H. Dynamic 68Ga-PSMA-11 PET/CT for the Primary Evaluation of Localized Renal Mass: A Prospective Study. J Nucl Med 2020; 62:773-778. [PMID: 33097628 DOI: 10.2967/jnumed.120.251272] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/23/2020] [Indexed: 01/29/2023] Open
Abstract
The potential role of prostate-specific membrane antigen (PSMA) PET/CT in non-prostate cancer tumors has shown promising results. We examined the performance of dynamic 68Ga-PSMA-11 PET/CT (DPSMA) for the evaluation of localized renal mass. Methods: A prospective case series of patients with a newly diagnosed renal mass who were referred for surgery was examined. DPSMA was performed in a standardized manner before surgery. The final surgical histology served as the standard of reference. PSMA expression in the tumor vasculature was assessed and staining intensity was scored. Tracer uptake and PSMA expression were compared between benign and malignant tissue. Results: Of 29 enhancing renal masses evaluated in 27 patients, 24 (83%) were malignant lesions. The median SUVmean of benign and malignant lesions was 2.3 (interquartile range [IQR], 2.2-2.7) and 6.8 (IQR, 4.2-10.1), respectively (P = 0.009). Median SUVmax of benign and malignant lesions was 3.8 (IQR, 3.3-4.5) and 9.4 (IQR, 5.4-15.8), respectively (P = 0.015). The median washout coefficient (K 2) was significantly lower in malignant lesions than in benign lesions (0.17 vs. 0.70, P = 0.02). Positive PSMA staining was found in 20 of 24 malignant lesions and in 2 of 5 benign lesions (P = 0.04). Conclusion: This pilot study demonstrated DPSMA uptake and kinetics in localized renal masses. Increased 68Ga-PSMA-11 tracer uptake and intratumoral retention correlate with PSMA expression in malignant renal tumors compared with benign renal masses, supporting further assessment of DPSMA as a potential tool for evaluating localized renal masses.
Collapse
Affiliation(s)
- Shay Golan
- Department of Urology, Rabin Medical Center, Petach Tikva, Israel .,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tzach Aviv
- Department of Urology, Rabin Medical Center, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - David Groshar
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Nuclear Medicine Rabin Medical Center, Petach Tikva, Israel
| | - Maxim Yakimov
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Pathology, Rabin Medical Center, Petach Tikva, Israel
| | - Yaniv Zohar
- Department of Pathology, Rambam Health Care Campus, Haifa, Israel; and
| | - Yoad Prokocimer
- Department of Urology, Rabin Medical Center, Petach Tikva, Israel
| | - Andrei Nadu
- Department of Urology, Rabin Medical Center, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jack Baniel
- Department of Urology, Rabin Medical Center, Petach Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Liran Domachevsky
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Nuclear Medicine, The Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Hanna Bernstine
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Nuclear Medicine Rabin Medical Center, Petach Tikva, Israel
| |
Collapse
|
37
|
Farolfi A, Koschel S, Murphy DG, Fanti S. PET imaging in urology: a rapidly growing successful collaboration. Curr Opin Urol 2020; 30:623-627. [PMID: 32701721 DOI: 10.1097/mou.0000000000000800] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW To discuss and highlight the recent findings in urological oncology focusing on nuclear medicine advances on imaging and therapy. RECENT FINDINGS Testicular tumors: F-FDG as the standard positron emission tomography (PET) tracer with proven good accuracy in detecting metastatic testicular cancer; urothelial cancer: good accuracy of F-FDG PET in detecting distant metastases but poor results in detecting local disease; prostate cancer: prostate-specific membrane antigen (PSMA) is a promising target for imaging prostate cancer with unprecedented accuracy in both staging and restaging and prospective studies were recently published. In castration-resistant prostate cancer, PSMA-targeting radionuclide therapy is showing potential as a curative possibility (e.g. using Lu-PSMA); renal cell cancer (RCC): besides FDG other PET radiotracers are under clinical evaluation (e.g. antibody-based molecular imaging, metabolic radiotracers and PSMA-based tracers). PSMA-based imaging may have applications in staging clear-cell RCC and in the selection and in the antiangiogenic treatment's response assessment. Possible role of PSMA-targeting radionuclide therapy? SUMMARY In urological oncology, the use of F-FDG has been limited by a generally low tumor uptake and physiological F-FDG excretion by the urinary system. Other radiotracers are increasing the urologist's portfolio allowing imaging of several biochemical pathways. Theragnostic possibilities are also under investigation thanks to PSMA-based tracers.
Collapse
Affiliation(s)
- Andrea Farolfi
- Nuclear Medicine Unit, University of Bologna, S. Orsola Hospital, Italy
| | - Samantha Koschel
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne
| | - Declan G Murphy
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Victoria, Australia
| | - Stefano Fanti
- Nuclear Medicine Unit, University of Bologna, S. Orsola Hospital, Italy
| |
Collapse
|
38
|
Ahn T, Roberts MJ, Abduljabar A, Joshi A, Perera M, Rhee H, Wood S, Vela I. A Review of Prostate-Specific Membrane Antigen (PSMA) Positron Emission Tomography (PET) in Renal Cell Carcinoma (RCC). Mol Imaging Biol 2020; 21:799-807. [PMID: 30617728 DOI: 10.1007/s11307-018-01307-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Metastatic renal cell carcinoma (mRCC) is a disease that portends poor prognosis despite an increasing number of novel systemic treatment options including new targeted therapies and immunotherapy. Ablative intervention directed at oligometastatic RCC has demonstrated survival benefit. Consequently, developing techniques for improved staging of mRCC on contemporary imaging modalities including X-ray computed tomography (CT), magnetic resonance imaging (MRI) and/or bone scan (BS) is a clinical priority. This is relevant for metastatic deposits too small to characterize or lymph nodes within physiological normality. Prostate-specific membrane antigen (PSMA) is a type II transmembrane glycoprotein highly expressed on prostate cancer epithelial cells. Recently, small molecules targeting the PSMA receptor, linked to radioactive isotopes have been developed for use with positron emission tomography (PET). Despite its nomenclature, PSMA has also been found to be expressed in the neovasculature of non-prostate cancers such as renal cell carcinoma (RCC) and hence PSMA PET/CT imaging has been proposed as an alternative staging modality. Preliminary small studies involving the use of PSMA PET/CT imaging in mRCC have been encouraging with evidence of improved staging sensitivity which has directly led to change in management in some cases. Given these early encouraging reports, we performed a comprehensive narrative review on the available evidence, including the scientific basis for PSMA expression in RCC, the role of PSMA PET/CT imaging with potential clinical implications in mRCC, its limitations and future opportunities.
Collapse
Affiliation(s)
- Thomas Ahn
- Department of Urology, Greenslopes Private Hospital, Brisbane, Queensland, Australia. .,Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.
| | - Matthew J Roberts
- Department of Urology, Greenslopes Private Hospital, Brisbane, Queensland, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Department of Urology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Aous Abduljabar
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Andre Joshi
- Department of Urology, Townsville Hospital, Brisbane, Queensland, Australia.,Centre for Clinical Research, The University of Queensland, Brisbane, Queensland, Australia.,Centre for Kidney Disease Research, Translational Research Institute, Brisbane, Australia
| | - Marlon Perera
- Department of Urology, Princess Alexandra Hospital, Brisbane, Queensland, Australia.,Department of Surgery, Austin Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Handoo Rhee
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Department of Urology, Princess Alexandra Hospital, Brisbane, Queensland, Australia.,Centre for Clinical Research, The University of Queensland, Brisbane, Queensland, Australia
| | - Simon Wood
- Department of Urology, Princess Alexandra Hospital, Brisbane, Queensland, Australia.,Centre for Kidney Disease Research, Translational Research Institute, Brisbane, Australia
| | - Ian Vela
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Department of Urology, Princess Alexandra Hospital, Brisbane, Queensland, Australia.,Australian Prostate Cancer Research Center Queensland, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| |
Collapse
|
39
|
Abstract
PURPOSE OF REVIEW Worldwide, over 400 000 new cases of kidney cancer were diagnosed and over 175 000 deaths anticipated in 2018. It is ranked as the 14th most common cancer in women and 9th most common in men. Imaging is important for initial detection, staging, and monitoring to assist treatment planning, but conventional anatomic imaging is limited. Although functional PET/CT has proven helpful in the diagnosis and management of many cancers, its value in renal cell carcinoma (RCC) is still in evolution. RECENT FINDINGS FDG is probably the most useful radiotracer in RCC, although CAIX imaging can be helpful in clear cell RCC. Current research on PET imaging agents in RCC including 89Zr bevacizumab, 89Zr geruntuximab, 18F fluoro-L-thymidine (FLT), prostate-specific membrane antigen (PSMA), 11C choline, 18F sodium fluoride (NaF), and18F fluorodeoxyglucose (FDG) have shown some interesting results for detection and prognosis of the disease. SUMMARY Many innovative radiotracers have been tested in RCC, but robust differentiation of primary disease from normal parenchyma remains elusive for almost all of them. The metastatic setting and response to therapy for this cancer are more favorable PET applications. Continued research in promising molecular tracers will hopefully advance both diagnostic and therapeutic strategies to ultimately eradicate RCC.
Collapse
|
40
|
Siva S, Udovicich C, Tran B, Zargar H, Murphy DG, Hofman MS. Expanding the role of small-molecule PSMA ligands beyond PET staging of prostate cancer. Nat Rev Urol 2020; 17:107-118. [PMID: 31937920 DOI: 10.1038/s41585-019-0272-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2019] [Indexed: 12/25/2022]
Abstract
Prostate-specific membrane antigen (PSMA) positron emission tomography (PET) is rapidly being established as arguably the leading contemporary imaging modality in the management of prostate cancer. Outside of its conventional use in the de novo staging of localized disease and detection of biochemical recurrence, additional applications for the use of PSMA PET are emerging. Uptake of PSMA tracers in other genitourinary malignancies, particularly renal cell carcinoma, has led to new fields of investigation. Therapeutic delivery of radiolabelled PSMA small molecules has shown considerable promise in advanced prostate cancer. The ability to use the same molecule for imaging and therapy - theranostics - enables a highly personalized approach. PSMA PET can also have a considerable influence in the selection and guidance of radiotherapy fields for high-risk and recurrent disease. Intriguingly, changes in intensity of PSMA uptake during systemic therapy might provide early response assessment or novel insight into the biological responses of genitourinary malignancies to treatment. An evolving range of radiolabelled PSMA radiopharmaceuticals is emerging in the multiple facets of modern clinical practice.
Collapse
Affiliation(s)
- Shankar Siva
- Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia. .,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia.
| | - Cristian Udovicich
- Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Ben Tran
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia.,Division of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia.,Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
| | - Homi Zargar
- Department of Urology, Royal Melbourne Hospital, Melbourne, Victoria, Australia.,Department of Surgery, University of Melbourne, Melbourne, Victoria, Australia
| | - Declan G Murphy
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia.,Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Michael S Hofman
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia.,Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, Australia
| |
Collapse
|
41
|
Werner RA, Derlin T, Lapa C, Sheikbahaei S, Higuchi T, Giesel FL, Behr S, Drzezga A, Kimura H, Buck AK, Bengel FM, Pomper MG, Gorin MA, Rowe SP. 18F-Labeled, PSMA-Targeted Radiotracers: Leveraging the Advantages of Radiofluorination for Prostate Cancer Molecular Imaging. Theranostics 2020; 10:1-16. [PMID: 31903102 PMCID: PMC6929634 DOI: 10.7150/thno.37894] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 09/11/2019] [Indexed: 12/22/2022] Open
Abstract
Prostate-specific membrane antigen (PSMA)-targeted PET imaging for prostate cancer with 68Ga-labeled compounds has rapidly become adopted as part of routine clinical care in many parts of the world. However, recent years have witnessed the start of a shift from 68Ga- to 18F-labeled PSMA-targeted compounds. The latter imaging agents have several key advantages, which may lay the groundwork for an even more widespread adoption into the clinic. First, facilitated delivery from distant suppliers expands the availability of PET radiopharmaceuticals in smaller hospitals operating a PET center but lacking the patient volume to justify an onsite 68Ge/68Ga generator. Thus, such an approach meets the increasing demand for PSMA-targeted PET imaging in areas with lower population density and may even lead to cost-savings compared to in-house production. Moreover, 18F-labeled radiotracers have a higher positron yield and lower positron energy, which in turn decreases image noise, improves contrast resolution, and maximizes the likelihood of detecting subtle lesions. In addition, the longer half-life of 110 min allows for improved delayed imaging protocols and flexibility in study design, which may further increase diagnostic accuracy. Moreover, such compounds can be distributed to sites which are not allowed to produce radiotracers on-site due to regulatory issues or to centers without access to a cyclotron. In light of these advantageous characteristics, 18F-labeled PSMA-targeted PET radiotracers may play an important role in both optimizing this transformative imaging modality and making it widely available. We have aimed to provide a concise overview of emerging 18F-labeled PSMA-targeted radiotracers undergoing active clinical development. Given the wide array of available radiotracers, comparative studies are needed to firmly establish the role of the available 18F-labeled compounds in the field of molecular PCa imaging, preferably in different clinical scenarios.
Collapse
Affiliation(s)
- Rudolf A. Werner
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Constantin Lapa
- Department of Nuclear Medicine, University Hospital Würzburg, Germany
| | - Sara Sheikbahaei
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Takahiro Higuchi
- Department of Nuclear Medicine, University Hospital Würzburg, Germany
- Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Frederik L. Giesel
- Department of Nuclear Medicine, University Hospital Heidelberg, INF 400, 69120 Heidelberg, Germany
| | - Spencer Behr
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA
| | - Alexander Drzezga
- Department of Nuclear Medicine, University Hospital Cologne, Germany
| | - Hiroyuki Kimura
- Department of Analytical and Bioinorganic Chemistry, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Andreas K. Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Germany
| | - Frank M. Bengel
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Martin G. Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael A. Gorin
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Steven P. Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
42
|
Pozzessere C, Bassanelli M, Ceribelli A, Rasul S, Li S, Prior JO, Cicone F. Renal Cell Carcinoma: the Oncologist Asks, Can PSMA PET/CT Answer? Curr Urol Rep 2019; 20:68. [PMID: 31605269 DOI: 10.1007/s11934-019-0938-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW To critically review the potential clinical applications of prostate-specific membrane antigen (PSMA) radioactive ligands in renal cell carcinoma (RCC). RECENT FINDINGS Radioactive probes targeting PSMA hold promise in several malignancies in addition to prostate cancer, owing to the expression of PSMA by tumor neovasculature. The majority of clear cell RCCs (ccRCC), the most malignant RCC subtype, express PSMA on tumor-associated neovasculature. The endothelium of less aggressive RCC subtypes is PSMA positive in a lower, but still significant percentage of cases. PSMA might therefore represent an interesting theragnostic target in RCC. The preliminary data available suggest a potential role for PSMA-targeting radiopharmaceuticals in complementing conventional imaging for staging ccRCC patients at risk of nodal involvement and oligometastatic disease. Additional applications of PSMA imaging may be the selection and the response assessment of patients receiving anti-angiogenic treatments. The effectiveness of PSMA-targeting radionuclide therapy should also be investigated.
Collapse
Affiliation(s)
- Chiara Pozzessere
- Department of Radiology, AUSL Toscana Centro San Giuseppe Hospital, Viale Boccaccio 20, 50053, Empoli, Italy.
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland.
| | - Maria Bassanelli
- Division of Medical Oncology, San Camillo De Lellis Hospital, Rieti, Italy
| | - Anna Ceribelli
- Division of Medical Oncology, San Camillo De Lellis Hospital, Rieti, Italy
| | - Sazan Rasul
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Shuren Li
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - John O Prior
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Francesco Cicone
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
43
|
Van de Wiele C, Sathekge M, de Spiegeleer B, de Jonghe PJ, Beels L, Maes A. PSMA-Targeting Positron Emission Agents for Imaging Solid Tumors Other Than Non-Prostate Carcinoma: A Systematic Review. Int J Mol Sci 2019; 20:E4886. [PMID: 31581638 PMCID: PMC6801742 DOI: 10.3390/ijms20194886] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/30/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022] Open
Abstract
Despite its name, prostate-specific membrane antigen (PSMA) has been shown using immunohistochemistry (IHC) to also be over-expressed in the tumor neovasculature of a wide variety of solid tumors other than prostate carcinoma. Accordingly, positron-emitting radiolabeled small molecules targeting PSMA, initially developed for positron emission tomography in prostate carcinomas, are currently being explored for their staging and restaging potential as an alternative imaging modality in other solid tumor types where 18-F-fluorodeoxyglucose (FDG)-PET imaging has low diagnostic accuracy. In this paper, the currently available literature in this field is reviewed. Preliminary, mainly retrospective studies are encouraging, with evidence of improved diagnostic sensitivity and specificity in clear cell renal carcinoma, glioma, and hepatocellular carcinoma, leading to a change in patient management in several patients. However, the results published thus far warrant confirmation by larger prospective studies additionally assessing the longitudinal impact on patient outcomes.
Collapse
Affiliation(s)
- Christophe Van de Wiele
- Department of Nuclear Medicine, AZ Groeninge, 8500 Kortrijk, Belgium.
- Department of Nuclear Medicine and Radiology, University Ghent, 9000 GHent, Belgium.
| | - Mike Sathekge
- Department of Nuclear Medicine, University of Pretoria, 0001 Pretoria, South-Africa.
| | - Bart de Spiegeleer
- Laboratory of Drug Quality and Registration, University Ghent, 9000 Ghent, Belgium.
| | | | - Laurence Beels
- Department of Nuclear Medicine, AZ Groeninge, 8500 Kortrijk, Belgium.
| | - Alex Maes
- Department of Nuclear Medicine, AZ Groeninge, 8500 Kortrijk, Belgium.
- Department of Imaging and Pathology, KULAK, University of Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
44
|
Meyer AR, Carducci MA, Denmeade SR, Markowski MC, Pomper MG, Pierorazio PM, Allaf ME, Rowe SP, Gorin MA. Improved identification of patients with oligometastatic clear cell renal cell carcinoma with PSMA-targeted 18F-DCFPyL PET/CT. Ann Nucl Med 2019; 33:617-623. [PMID: 31147927 PMCID: PMC9774684 DOI: 10.1007/s12149-019-01371-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 05/21/2019] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Complete surgical resection of metastatic sites has been shown to prolong survival in select patients with oligometastatic RCC. This treatment strategy is dependent upon the accurate characterization of a patient's extent of disease. The objective of this study was to explore the utility of PSMA-targeted 18F-DCFPyL PET/CT in patients with presumed oligometastatic clear cell RCC. METHODS This is a subset analysis of a prospective study in which patients with RCC were imaged with 18F-DCFPyL PET/CT (ClinicalTrials.gov identifier NCT02687139). In the present analysis, patients with oligometastatic clear cell RCC, defined as ≤ 3 metastatic lesions on conventional imaging, were evaluated. 18F-DCFPyL PET/CT scans were reviewed for sites of disease and compared to conventional imaging. RESULTS The final cohort included 14 patients with oligometastatic clear cell RCC. Conventional imaging revealed 21 metastatic lesions and 3 primary tumors. 18F-DCFPyL PET/CT detected 29 sites of metastatic disease and 3 primary tumors. Of the 21 metastatic lesions detected on conventional imaging, 17 (81.0%) had radiotracer uptake. Additionally, all 3 primary tumors had radiotracer uptake. In 4 (28.6%) patients a total of 12 more lesions were identified on 18F-DCFPyL PET/CT than conventional imaging. Notably, 3 (21.4%) patients were no longer considered oligometastatic. The detection rates of conventional imaging and 18F-DCFPyL PET/CT for identifying sites of disease were 66.7% and 88.9%, respectively. CONCLUSIONS PSMA-targeted PET/CT appears to aid in the identification of patients with oligometastatic clear cell RCC. If borne out in future studies, this suggests that PSMA-targeted imaging has the potential to help select candidates for metastasis-directed therapy.
Collapse
Affiliation(s)
- Alexa R. Meyer
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Park 213, Baltimore, MD 21287, USA
| | - Michael A. Carducci
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Park 213, Baltimore, MD 21287, USA,Department of Oncology, Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University of Medicine, Baltimore, MD, USA
| | - Samuel R. Denmeade
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Park 213, Baltimore, MD 21287, USA,Department of Oncology, Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University of Medicine, Baltimore, MD, USA
| | - Mark C. Markowski
- Department of Oncology, Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University of Medicine, Baltimore, MD, USA
| | - Martin G. Pomper
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Park 213, Baltimore, MD 21287, USA,Department of Oncology, Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University of Medicine, Baltimore, MD, USA,The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Philip M. Pierorazio
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Park 213, Baltimore, MD 21287, USA,Department of Oncology, Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University of Medicine, Baltimore, MD, USA
| | - Mohamad E. Allaf
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Park 213, Baltimore, MD 21287, USA,Department of Oncology, Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University of Medicine, Baltimore, MD, USA
| | - Steven P. Rowe
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Park 213, Baltimore, MD 21287, USA,The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael A. Gorin
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, 600 North Wolfe Street, Park 213, Baltimore, MD 21287, USA,Department of Oncology, Sydney Kimmel Comprehensive Cancer Center, Johns Hopkins University of Medicine, Baltimore, MD, USA,The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
45
|
Werner RA, Thackeray JT, Pomper MG, Bengel FM, Gorin MA, Derlin T, Rowe SP. Recent Updates on Molecular Imaging Reporting and Data Systems (MI-RADS) for Theranostic Radiotracers-Navigating Pitfalls of SSTR- and PSMA-Targeted PET/CT. J Clin Med 2019; 8:E1060. [PMID: 31331016 PMCID: PMC6678732 DOI: 10.3390/jcm8071060] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/05/2019] [Accepted: 07/17/2019] [Indexed: 12/12/2022] Open
Abstract
The theranostic concept represents a paradigmatic example of personalized treatment. It is based on the use of radiolabeled compounds which can be applied for both diagnostic molecular imaging and subsequent treatment, using different radionuclides for labelling. Clinically relevant examples include somatostatin receptor (SSTR)-targeted imaging and therapy for the treatment of neuroendocrine tumors (NET), as well as prostate-specific membrane antigen (PSMA)-targeted imaging and therapy for the treatment of prostate cancer (PC). As such, both classes of radiotracers can be used to triage patients for theranostic endoradiotherapy using positron emission tomography (PET). While interpreting PSMA- or SSTR-targeted PET/computed tomography scans, the reader has to navigate certain pitfalls, including (I.) varying normal biodistribution between different PSMA- and SSTR-targeting PET radiotracers, (II.) varying radiotracer uptake in numerous kinds of both benign and malignant lesions, and (III.) resulting false-positive and false-negative findings. Thus, two novel reporting and data system (RADS) classifications for PSMA- and SSTR-targeted PET imaging (PSMA- and SSTR-RADS) have been recently introduced under the umbrella term molecular imaging reporting and data systems (MI-RADS). Notably, PSMA- and SSTR-RADS are structured in a reciprocal fashion, i.e., if the reader is familiar with one system, the other system can readily be applied. Learning objectives of the present case-based review are as follows: (I.) the theranostic concept for the treatment of NET and PC will be briefly introduced, (II.) the most common pitfalls on PSMA- and SSTR-targeted PET/CT will be identified, (III.) the novel framework system for theranostic radiotracers (MI-RADS) will be explained, applied to complex clinical cases and recent studies in the field will be highlighted. Finally, current treatment strategies based on MI-RADS will be proposed, which will demonstrate how such a generalizable framework system truly paves the way for clinically meaningful molecular imaging-guided treatment of either PC or NET. Thus, beyond an introduction of MI-RADS, the present review aims to provide an update of recently published studies which have further validated the concept of structured reporting systems in the field of theranostics.
Collapse
Affiliation(s)
- Rudolf A Werner
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany
- Johns Hopkins School of Medicine, The Russell H Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Baltimore, MD 21287, USA
| | - James T Thackeray
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Martin G Pomper
- Johns Hopkins School of Medicine, The Russell H Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Baltimore, MD 21287, USA
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Michael A Gorin
- Johns Hopkins School of Medicine, The Russell H Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Baltimore, MD 21287, USA
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Steven P Rowe
- Johns Hopkins School of Medicine, The Russell H Morgan Department of Radiology and Radiological Science, Division of Nuclear Medicine and Molecular Imaging, Baltimore, MD 21287, USA.
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
46
|
Graybiel CE, Flavell RR, Wang ZJ, Behr SC. Molecular Imaging of Renal Malignancy: A Review. CURRENT RADIOLOGY REPORTS 2019. [DOI: 10.1007/s40134-019-0320-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
47
|
Nakamoto Y, Ishimori T, Shimizu Y, Sano K, Togashi K. Clinical utility of 68Ga-DOTATOC positron emission tomography/computed tomography for recurrent renal cell carcinoma. Eur J Nucl Med Mol Imaging 2019; 46:1524-1530. [PMID: 30847538 DOI: 10.1007/s00259-019-04298-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 02/25/2019] [Indexed: 12/11/2022]
Abstract
PURPOSE Positron emission tomography (PET)/computed tomography (CT) with 68Ga-labelled 1,4,7,10-tetraazacyclododecane-N,N',N″,N″'-tetraacetic acid-D-Phe1-Tyr3-octreotide (DOTATOC) has been accepted as a diagnostic imaging tool especially for patients with neuroendocrine tumours. However, its clinical usefulness for restaging of renal cell carcinoma (RCC) has not been fully investigated. This retrospective study was performed to elucidate the clinical value of PET/CT using 68Ga-DOTATOC in patients with known or suspected recurrent RCC. METHODS We analysed 25 consecutive patients who underwent DOTATOC-PET/CT scans after surgery for RCC (23 clear cell, 1 papillary, 1 unclassified). PET/CT findings were reviewed and the detection rate was calculated on a patient and lesion basis. The detectability was compared in patients who also underwent PET/CT scans with 18F-fluorodeoxyglucose (FDG). Histopathological findings or clinical follow-up were used as the reference standard. RESULTS Based on the final diagnosis, 76 recurrent or metastatic lesions were confirmed in this population. Of these lesions, 66 lesions in 22 patients were positive by DOTATOC-PET/CT. The patient-based and lesion-based sensitivity was 88% (22/25) and 87% (66/76), respectively. Twelve patients underwent both DOTATOC-PET/CT and FDG-PET/CT. The lesion-based sensitivity of DOTATOC was 74% (20/27), while that of FDG was 59% (16/27). Eight lesions were identified only by DOTATOC, but four lesions from papillary RCC were detected only by FDG. CONCLUSION Our data indicate that DOTATOC-PET/CT would be useful for detecting recurrent foci in patients with clear cell RCC. DOTATOC-PET/CT and FDG-PET/CT are considered to have complementary roles.
Collapse
Affiliation(s)
- Yuji Nakamoto
- Department of Diagnostic Imaging and Nuclear Medicine, Kyoto University Graduate School of Medicine, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Takayoshi Ishimori
- Department of Diagnostic Imaging and Nuclear Medicine, Kyoto University Graduate School of Medicine, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yoichi Shimizu
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Kohei Sano
- Laboratory of Biophysical Chemistry, Kobe Pharmaceutical University, Kobe, Japan
| | - Kaori Togashi
- Department of Diagnostic Imaging and Nuclear Medicine, Kyoto University Graduate School of Medicine, 54 Shogoinkawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|