1
|
Hirasawa H, Taketomi-Takahashi A, Katsumata N, Higuchi T, Sekine Y, Suzuki K, Kaneko Y, Hiromura K, Fukushima Y, Tsushima Y. Efficacy of 18F-Fluorodeoxyglucose positron emission tomography/computed tomography for detecting renal cell carcinoma in patients with end-stage renal disease. Jpn J Radiol 2024; 42:1178-1186. [PMID: 38795287 PMCID: PMC11442643 DOI: 10.1007/s11604-024-01593-5] [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: 01/23/2024] [Accepted: 05/11/2024] [Indexed: 05/27/2024]
Abstract
PURPOSE Dialysis patients are at an increased risk of developing renal cell carcinoma (RCC); however, differentiating between RCC and benign cysts can sometimes be difficult using modalities, such as computed tomography (CT) and ultrasonography. 18F-Fluorodeoxyglucose positron emission tomography (FDG-PET)/CT efficiently detects malignant tumors; however, physiological accumulation of FDG in the kidney limits its efficacy in detecting renal tumors. However, in patients with severely impaired renal function, the renal accumulation of FDG is decreased, possibly improving the detection of renal malignancies in this patient population. This study evaluated the usefulness of FDG-PET/CT as a screening tool for detecting RCC in patients with end-stage renal disease. MATERIALS AND METHODS This prospective study recruited 150 participants from 2012 to 2016 who were on dialysis or underwent renal transplantation and were on dialysis until transplantation. FDG-PET/CT was performed to screen for RCC. Three radiologists independently evaluated the images. No protocol was defined for the additional management of positive examinations, leaving decisions to the discretion of each participant. Negative examinations were observed until the end of 2019. RESULTS In total, 150 participants (mean age, 58 ± 13 years; 105 men) underwent FDG-PET/CT. Twenty patients (13.4%) were diagnosed as positive. Fifteen patients underwent additional examinations and/or procedures, and RCC was found in seven patients. Of the four patients who underwent surgical resection, the pathological results were clear cell RCC in one, papillary RCC in one, and acquired cystic disease-associated RCC in two. Two participants were diagnosed with RCC on bone biopsy, and one was diagnosed on dynamic CT but opted for observation. The sensitivity, specificity, and negative predictive value were 100%, 93.9%, and 100%, respectively. CONCLUSION FDG-PET/CT was useful for detecting RCC in patients with end-stage renal disease. Our findings show the potential use of FDG-PET/CT as a screening tool for RCC in this patient population.
Collapse
Affiliation(s)
- Hiromi Hirasawa
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, 3-39-22 Showa, Maebashi, Gunma, 371-8511, Japan.
| | - Ayako Taketomi-Takahashi
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, 3-39-22 Showa, Maebashi, Gunma, 371-8511, Japan
| | - Natsumi Katsumata
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, 3-39-22 Showa, Maebashi, Gunma, 371-8511, Japan
| | - Tetsuya Higuchi
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, 3-39-22 Showa, Maebashi, Gunma, 371-8511, Japan
| | - Yoshitaka Sekine
- Department of Urology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Kazuhiro Suzuki
- Department of Urology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yoriaki Kaneko
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Keiju Hiromura
- Department of Nephrology and Rheumatology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yasuhiro Fukushima
- Department of Applied Medical Imaging, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yoshito Tsushima
- Department of Diagnostic Radiology and Nuclear Medicine, Gunma University Graduate School of Medicine, 3-39-22 Showa, Maebashi, Gunma, 371-8511, Japan
| |
Collapse
|
2
|
Singh R, Thotakura AK, Alati S, Lisok A, Jiang Z, Merino VF, Minn I, Yadav S, Markowski MC, Ged Y, Pavlovich CP, Singla N, Solnes LB, Gorin MA, Pomper MG, Rowe SP, Banerjee SR. Performance of PSMA-targeted radiotheranostics in an experimental model of renal cell carcinoma. Front Oncol 2024; 14:1432286. [PMID: 39324008 PMCID: PMC11423292 DOI: 10.3389/fonc.2024.1432286] [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: 05/13/2024] [Accepted: 07/29/2024] [Indexed: 09/27/2024] Open
Abstract
Introduction Renal cell carcinoma (RCC) represents cancer originating from the renal epithelium and accounts for > 90% of cancers in the kidney. Prostate-specific membrane antigen (PSMA) is overexpressed in tumor-associated neovascular endothelial cells of many solid tumors, including metastatic RCC. Although studied in several small clinical studies, PSMA-based imaging and therapy have not been pursued rigorously in preclinical RCC. This study aimed to evaluate the preclinical performance of PSMA-based radiotheranostic agents in a relevant murine model. Methods A PSMA-overexpressing murine cell line, PSMA+ RENCA, was developed by lentiviral transduction. PSMA-based theranostic agents, 68Ga-L1/177Lu-L1/225Ac-L1, were synthesized in high radiochemical yield and purity following our reported methods. Immunocompetent BALB/c mice were used for flank and orthotopic tumor inoculation. 68Ga-L1 was evaluated in small animal PET/CT imaging in flank and PET/MR imaging in orthotopic models. Cell viability studies were conducted for 177Lu-L1 and 225Ac-L1. Proof-of-concept treatment studies were performed using 225Ac-L1 (0, 37 kBq, 2 kBq × 37 kBq, 1 week apart) using PSMA+ RENCA in the flank model. Results Cellular uptake of 68Ga-L1, 177Lu-L1, and 225Ac-L1 confirmed the specificity of the agents to PSMA+ RENCA cells rather than to RENCA (wt) cells, which are low in PSMA expression. The uptake in PSMA+ RENCA cells at 1 h for 68Ga-L1 (49.0% incubated dose [ID] ± 3.6%ID/million cells), 177Lu-L1 (22.1%ID ± 0.5%ID)/million cells), and 225Ac-L1 (4.1% ± 0.2% ID)/million cells), respectively, were higher than the RENCA (wt) cells (~ 1%ID-2%ID/million cells). PET/CT images displayed > 7-fold higher accumulation of 68Ga-L1 in PSMA+ RENCA compared to RENCA (wt) in flank implantation at 1 h. A twofold higher accumulation of 68Ga-L1 was observed in orthotopic tumors than in normal kidneys during 1-3 h postinjection. High lung uptake was observed with 68Ga-L1 PET/MR imaging 3 weeks after orthotopic implantation of PSMA+ RENCA due to spontaneous lung metastases. The imaging data were further confirmed by immunohistochemical characterization. 225Ac-L1 (0-37 kBq) displayed a dose-dependent reduction of cell proliferation in the PSMA+ RENCA cells after 48 h incubation; ~ 40% reduction in the cells with treated 37 kBq compared to vehicle (p < 0.001); however, no effect was observed with 177Lu-L1 (0-3700 kBq) up to 144 h postinoculation, suggesting lower efficacy of β-particle-emitting radiations in cellular studies compared to α-particle-emitting 225Ac-L1. Animals treated with 225Ac-L1 at 1 week posttumor inoculation in flank models displayed significant tumor growth delay (p < 0.03) and longer median survival of 21 days and 24 days for the treatment groups 37 kBq and 2 kBq × 37 kBq, respectively, compared to the vehicle group (12 days). Conclusion The results suggest that a theranostic strategy targeting PSMA, employing PET and α-emitting radiopharmaceuticals, enabled tumor growth control and enhanced survival in a relevant immunocompetent murine model of RCC. These studies provide the rationale for clinical studies of PSMA-targeted theranostic agents in patients with RCC.
Collapse
Affiliation(s)
- Rajan Singh
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, United States
| | - Anand K. Thotakura
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, United States
| | - Suresh Alati
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, United States
| | - Alla Lisok
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, United States
| | - Zirui Jiang
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, United States
| | - Vanessa F. Merino
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, United States
| | - Il Minn
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, United States
| | - Santosh Yadav
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, United States
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center,
Baltimore, MD, United States
| | - Mark C. Markowski
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center,
Baltimore, MD, United States
- Department of Urology, Brady Urological Institute, Johns Hopkins University, Baltimore, MD, United States
| | - Yasser Ged
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center,
Baltimore, MD, United States
- Department of Urology, Brady Urological Institute, Johns Hopkins University, Baltimore, MD, United States
| | - Christian P. Pavlovich
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center,
Baltimore, MD, United States
- Department of Urology, Brady Urological Institute, Johns Hopkins University, Baltimore, MD, United States
| | - Nirmish Singla
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center,
Baltimore, MD, United States
- Department of Urology, Brady Urological Institute, Johns Hopkins University, Baltimore, MD, United States
| | - Lilja B. Solnes
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, United States
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center,
Baltimore, MD, United States
| | - Michael A. Gorin
- The Milton and Carroll Petrie Department of Urology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Martin G. Pomper
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center,
Baltimore, MD, United States
- Department of Radiology, UT Southwestern Medical Center, Dallas, TX, United States
| | - Steven P. Rowe
- Department of Radiology, University of North Carolina, Chapel Hill, NC, United States
| | - Sangeeta Ray Banerjee
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University, Baltimore, MD, United States
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center,
Baltimore, MD, United States
| |
Collapse
|
3
|
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
|
4
|
Ribeiro AMB, Borin AS, Mello GDRD, Chojniak R. Bone Metastasis from Renal Cancer Coinciding with the Same Anatomical Position as a Vertebral Hemangioma: A Collision Lesion Case Report. World J Nucl Med 2024; 23:135-140. [PMID: 38933074 PMCID: PMC11199034 DOI: 10.1055/s-0044-1779747] [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] [Indexed: 06/28/2024] Open
Abstract
Collisions lesions are rare neoplasms where two histologically distinct tumors coexist in the same organ or anatomical site. Vertebral hemangiomas (VHs) are the most common lesions involving the vertebral bodies and imaging findings of typical and atypical hemangiomas, variant forms of hemangioma such as aggressive hemangiomas are well known, but collision lesions involving VHs are extremely rare. This article presents a case report of a 73-year-old male patient diagnosed with clear cell renal cancer in a rare presentation of a bone metastasis coinciding with the same anatomical position as a VH (collision lesion). This required a multidisciplinary approach involving various diagnostic techniques to determine the best therapeutic management.
Collapse
Affiliation(s)
| | - Amanda Silles Borin
- Department of Nuclear Medicine A.C.Camargo Cancer Center, São Paulo, SP, Brazil
| | | | - Rubens Chojniak
- Department of Radiology, A.C.Camargo Cancer Center, São Paulo, SP, Brazil
| |
Collapse
|
5
|
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] [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
|
6
|
Tariq A, Pearce A, Rhee H, Kyle S, Raveenthiran S, Pelecanos A, Gan CL, Goh JC, Wong D, McBean R, Marsh P, Goodman S, Dunglison N, Esler R, Navaratnam A, Yaxley JW, Thomas P, Pattison DA, Roberts MJ. The Role of Prostate-specific Membrane Antigen Positron Emission Tomography/Computed Tomography in Primary Staging of Selected Renal Tumours: Initial Experience in a Multicentre Cohort. Eur Urol Focus 2024:S2405-4569(23)00296-1. [PMID: 38195354 DOI: 10.1016/j.euf.2023.12.004] [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: 07/31/2023] [Revised: 12/02/2023] [Accepted: 12/18/2023] [Indexed: 01/11/2024]
Abstract
BACKGROUND Accurate primary staging of renal cancer with conventional imaging is challenging. Prostate-specific membrane antigen (PSMA) positron emission tomography/computed tomography (PET/CT) may serve to improve the accuracy of renal cancer staging. OBJECTIVE To determine clinicopathological and management differences for primary renal cancer staged with PSMA PET/CT in comparison to conventional imaging. DESIGN, SETTING, AND PARTICIPANTS We conducted a retrospective cohort study of PSMA PET/CT scans performed for primary staging of renal cancer and incidental renal lesions at three sites in Brisbane, Australia between June 2015 and June 2020. Clinical characteristics, imaging, and histopathology were reviewed. OUTCOME MEASUREMENTS AND STATISTICAL ANALYSIS Clinicopathological and management differences according to staging modality (PSMA PET/CT, conventional imaging) were assessed. Descriptive statistics were used to report demographics and clinical parameters. Nonparametric methods were used for statistical analysis. Fisher's exact test was used for comparison of small-cell size categorical variables. RESULTS AND LIMITATIONS From a total of 120 PSMA PET/CT scans, 61 were included (52 staging, 9 incidental) for predominantly males (74%) with a mean age of 65.1 yr (standard deviation 12.0). Most primary lesions (40/51) were clear-cell renal cell carcinoma (ccRCC; 98% PSMA-avid), eight were non-ccRCC (75% PSMA-avid), and three were non-RCC (oncocytoma; 67% PSMA-avid). PSMA PET identified a greater number of presumed metastatic lesions than conventional imaging (195 vs 160). A management change was observed for 32% of patients (20% major, 12% minor). Limitations include the retrospective design and selection bias, lack of blinding to PSMA reporting, and the use of different PSMA radiotracers. CONCLUSIONS PSMA PET/CT detected more metastases than conventional imaging and most renal cancers were PSMA-avid, resulting in a management change for one-third of the patients. PATIENT SUMMARY We looked at a newer type of scan called PSMA PET/CT for first staging of kidney cancer. We found that this detects more metastasis and helps in decisions on changes in treatment for some patients. This type of imaging is a useful addition to conventional scans in tricky cases and may help in better selection of suitable treatments, but more studies are required.
Collapse
Affiliation(s)
- Arsalan Tariq
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Australia; Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Adam Pearce
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Australia; Faculty of Medicine, The University of Queensland, Brisbane, Australia; Wesley Urology Clinic, The Wesley Hospital, Brisbane, Australia
| | - Handoo Rhee
- Faculty of Medicine, The University of Queensland, Brisbane, Australia; Department of Urology, Princess Alexandra Hospital, Brisbane, Australia
| | - Samuel Kyle
- Department of Medical Imaging, Royal Brisbane and Women's Hospital, Brisbane, Australia; Department of Nuclear Medicine, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - Sheliyan Raveenthiran
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Australia; Department of Urology, Redcliffe Hospital, Redcliffe, Australia
| | - Anita Pelecanos
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Chun Loo Gan
- Faculty of Medicine, The University of Queensland, Brisbane, Australia; Department of Medical Oncology, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - Jeffrey C Goh
- Faculty of Medicine, The University of Queensland, Brisbane, Australia; Department of Medical Oncology, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - David Wong
- Department of Radiology & Nuclear Medicine, The Wesley Hospital, Brisbane, Australia
| | - Rhiannon McBean
- Department of Radiology & Nuclear Medicine, The Wesley Hospital, Brisbane, Australia
| | - Phillip Marsh
- Department of Medical Imaging, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - Steven Goodman
- Department of Nuclear Medicine, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - Nigel Dunglison
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Australia; Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Rachel Esler
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Australia; Wesley Urology Clinic, The Wesley Hospital, Brisbane, Australia
| | - Anojan Navaratnam
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Australia; Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - John W Yaxley
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Australia; Faculty of Medicine, The University of Queensland, Brisbane, Australia; Wesley Urology Clinic, The Wesley Hospital, Brisbane, Australia
| | - Paul Thomas
- Faculty of Medicine, The University of Queensland, Brisbane, Australia; Department of Nuclear Medicine, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - David A Pattison
- Faculty of Medicine, The University of Queensland, Brisbane, Australia; Department of Nuclear Medicine, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - Matthew J Roberts
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Australia; Faculty of Medicine, The University of Queensland, Brisbane, Australia; Department of Urology, Redcliffe Hospital, Redcliffe, Australia; UQ Centre for Clinical Research, Faculty of Medicine, University of Queensland, Brisbane, Australia.
| |
Collapse
|
7
|
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
|
8
|
Pathmanathan S, Tariq A, Pearce A, Rhee H, Kyle S, Raveenthiran S, Wong D, McBean R, Marsh P, Goodman S, Dhiantravan N, Esler R, Dunglison N, Navaratnam A, Yaxley J, Thomas P, Pattison DA, Goh JC, Gan CL, Roberts MJ. Clinical impact of Prostate-Specific Membrane Antigen Positron Emission Tomography (PET) on intensification or deintensification of advanced renal cell carcinoma management. Eur J Nucl Med Mol Imaging 2023; 51:295-303. [PMID: 37592084 PMCID: PMC10684606 DOI: 10.1007/s00259-023-06380-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 08/02/2023] [Indexed: 08/19/2023]
Abstract
PURPOSE There is an emerging role of the use of Prostate-Specific Membrane Antigen (PSMA) Positron Emission Tomography (PET) in renal cell carcinoma. Herein, we report our experience in use of PSMA PET in recurrent or metastatic renal cell carcinoma (RCC). METHODS A retrospective analysis of all patients who underwent PSMA PET for suspected recurrent or de-novo metastatic RCC between 2015 and 2020 at three institutions was performed. The primary outcome was change in management (intensification or de-intensification) following PSMA PET scan. Secondary outcomes included histopathological correlation of PSMA avid sites, comparison of sites of disease on PSMA PET to diagnostic CT and time to systemic treatment. RESULTS
Collapse
Affiliation(s)
- Shivanshan Pathmanathan
- Department of Medical Oncology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Arsalan Tariq
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, 4029, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Adam Pearce
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, 4029, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- Wesley Urology Clinic, The Wesley Hospital, Brisbane, Queensland, Australia
| | - Handoo Rhee
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- Department of Urology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Samuel Kyle
- Department of, Medical Imaging, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
- Department of Nuclear Medicine, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Sheliyan Raveenthiran
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, 4029, Australia
- Department of Urology, Redcliffe Hospital, Brisbane, Queensland, Australia
| | - David Wong
- I-MED Radiology, The Wesley Hospital, Brisbane, Queensland, Australia
| | - Rhiannon McBean
- I-MED Radiology, The Wesley Hospital, Brisbane, Queensland, Australia
| | - Phillip Marsh
- Department of Diagnostic Radiology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Steven Goodman
- Department of Nuclear Medicine, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Nattakorn Dhiantravan
- Department of Nuclear Medicine, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Rachel Esler
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, 4029, Australia
- Wesley Urology Clinic, The Wesley Hospital, Brisbane, Queensland, Australia
| | - Nigel Dunglison
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, 4029, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Anojan Navaratnam
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, 4029, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - John Yaxley
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, 4029, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- Wesley Urology Clinic, The Wesley Hospital, Brisbane, Queensland, Australia
| | - Paul Thomas
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- Department of Nuclear Medicine, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - David A Pattison
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
- Department of Nuclear Medicine, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Jeffrey C Goh
- Department of Medical Oncology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Chun Loo Gan
- Department of Medical Oncology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Matthew J Roberts
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, 4029, Australia.
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.
- Department of Urology, Redcliffe Hospital, Brisbane, Queensland, Australia.
- Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
9
|
Sadowski EA, Lees B, McMillian AB, Kusmirek JE, Cho SY, Barroilhet LM. Distribution of prostate specific membrane antigen (PSMA) on PET-MRI in patients with and without ovarian cancer. Abdom Radiol (NY) 2023; 48:3643-3652. [PMID: 37261441 DOI: 10.1007/s00261-023-03957-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 06/02/2023]
Abstract
OBJECTIVES Ovarian cancer is the most lethal cancer and future research needs to focus on the early detection and exploration of new therapeutic agents. The objectives of this proof-of-concept study are to assess the feasibility of PSMA 18F-DCFPyl PET/MR imaging for detecting ovarian cancer and to evaluate the PSMA distribution in patients with and without ovarian cancer. METHODS This prospective pilot proof-of-concept study in patients with and without ovarian cancers occurred between October 2017 and January 2020. Patients were recruited from gynecologic oncology or hereditary ovarian cancer clinics, and underwent surgical removal of the uterus and ovaries for gynecologic indications. PSMA 18F-DCFPyl PET/MRI was obtained prior to standard of care surgery. RESULTS Fourteen patients were scanned: four patients with normal ovaries, six patients with benign ovarian lesions, and four patients with malignant ovarian lesions. Tracer uptake in normal ovaries (SUVmax = 2.8 ± 0.4) was greater than blood pool (SUVmax = 1.8 ± 0.5, p < 0.0001). Tracer uptake in benign ovarian lesions (2.2 ± 1.0) did not differ significantly from blood pool (p = 0.331). Tracer uptake in ovarian cancer (SUVmax = 7.8 ± 3.8) was greater than blood pool (p < 0.0001), normal ovaries (p = 0.0014), and benign ovarian lesions (p = 0.005). CONCLUSION PET/MR imaging detected PSMA uptake in ovarian cancer, with little to no uptake in benign ovarian findings. These results are encouraging and further studies in a larger patient cohort would be useful to help determine the extent and heterogeneity of PSMA uptake in ovarian cancer patients.
Collapse
Affiliation(s)
- Elizabeth A Sadowski
- Departments of Radiology, Obstetrics and Gynecology, University of Wisconsin School of Medicine and Public Health, 600 Highland Ave, E3/372, Madison, WI, 53792-3252, USA.
| | - Brittany Lees
- Atrium Health Levine Cancer Institute, 1021 Morehead Medical Drive, Suite 2100, Charlotte, NC, 28204, USA
| | - Alan B McMillian
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, 1111 Highland Avenue, Rm 1139, Madison, WI, 53705, USA
| | - Joanna E Kusmirek
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, 600 Highland Ave., E3/372, Madison, WI, 53792-3252, USA
| | - Steve Y Cho
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, 600 Highland Ave., E3/372, Madison, WI, 53792-3252, USA
| | - Lisa M Barroilhet
- Departments of Radiology, Obstetrics and Gynecology, University of Wisconsin School of Medicine and Public Health, 600 Highland Ave, E3/372, Madison, WI, 53792-3252, USA
| |
Collapse
|
10
|
Gasparro D, Scarlattei M, Silini EM, Migliari S, Baldari G, Cervati V, Graziani T, Campanini N, Maestroni U, Ruffini L. High Prognostic Value of 68Ga-PSMA PET/CT in Renal Cell Carcinoma and Association with PSMA Expression Assessed by Immunohistochemistry. Diagnostics (Basel) 2023; 13:3082. [PMID: 37835825 PMCID: PMC10572927 DOI: 10.3390/diagnostics13193082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/05/2023] [Accepted: 09/23/2023] [Indexed: 10/15/2023] Open
Abstract
In oligo-metastatic renal cell carcinoma (RCC), neither computed tomography (CT) nor bone scan is sensitive enough to detect small tumor deposits hampering early treatment and potential cure. Prostate-specific membrane antigen (PSMA) is a transmembrane glycoprotein expressed in the neo-vasculature of numerous malignant neoplasms, including RCC, that can be targeted by positron emission tomography (PET) using PSMA-targeting radioligands. Our aim was to investigate whether PSMA-expression patterns of renal cancer in the primary tumor or metastatic lesions on immunohistochemistry (IHC) are associated with PET/CT findings using [68Ga]-PSMA-HBED-CC (PSMA-PET/CT). We then analyzed the predictive and prognostic role of the PSMA-PET/CT signal. In this retrospective single-center study we included patients with renal cancer submitted to PSMA-PET/CT for staging or restaging, with tumor specimens available for PSMA-IHC. Clinical information (age, tumor type, and grade) and IHC results from the primary tumor or metastases were collected. The intensity of PSMA expression at IHC was scored into four categories: 0: none; 1: weak; 2: moderate; 3: strong. PSMA expression was also graded according to the proportion of vessels involved (PSMA%) into four categories: 0: none; 1: 1-25%; 2: 25-50%; 3: >50%. The intensity of PSMA expression and PSMA% were combined in a three-grade score: 0-2 absent or mildly positive, 3-4 moderately positive, and 5-6 strongly positive. PSMA scores were used for correlation with PSMA-PET/CT results. Results: IHC and PET scans were available for the analysis in 26 patients (22 ccRCC, 2 papillary RCC, 1 chromophobe, 1 "not otherwise specified" RCC). PSMA-PET/CT was positive in 17 (65%) and negative in 9 patients (35%). The mean and median SUVmax in the target lesion were 34.1 and 24.9, respectively. Reporter agreement was very high for both distant metastasis location and local recurrence (kappa 1, 100%). PSMA-PET detected more lesions than conventional imaging and revealed unknown metastases in 4 patients. Bone involvement, extension, and lesion number were greater than in the CT scan (median lesion number on PET/CT 3.5). The IHC PSMA score was concordant in primary tumors and metastases. All positive PSMA-PET/CT results (15/22 ccRCC, 1 papillary cancer type II, and 1 chromofobe type) were revealed in tumors with strong or moderate PSMA combined scores (3-4 and 5-6). In ccRCC tissue samples, PSMA expression was strong to moderate in 20/22 cases. The SUVmax values correlated to the intensity of PSMA expression which were assessed using IHC (p = 0.01), especially in the ccRCC subgroup (p = 0.009). Median survival was significantly higher in patients with negative PSMA-PET/CT (48 months) compared to patients with a positive scan (24 months, p= 0.001). SUVmax ≥ 7.4 provides discrimination of patients with a poor prognosis. Results of PSMA-PET/CT changed treatment planning. Conclusions: in renal cancer, positive PSMA-PET/CT is strongly correlated to the intensity of PSMA expression on immunohistochemistry in both ccRCC and chromophobe cancer. PSMA-PET/CT signal predicts a poor prognosis confirming its potential as an aggressiveness biomarker and providing paramount additional information influencing patient management.
Collapse
Affiliation(s)
- Donatello Gasparro
- Oncology Division, Azienda Ospedaliero-Universitaria di Parma, 43126 Parma, Italy;
| | - Maura Scarlattei
- Nuclear Medicine Division, Azienda Ospedaliero-Universitaria di Parma, 43126 Parma, Italy; (M.S.); (G.B.); (V.C.); (T.G.); (L.R.)
| | - Enrico Maria Silini
- Pathology Division, Azienda Ospedaliero-Universitaria di Parma, 43126 Parma, Italy; (E.M.S.); (N.C.)
- Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy
| | - Silvia Migliari
- Nuclear Medicine Division, Azienda Ospedaliero-Universitaria di Parma, 43126 Parma, Italy; (M.S.); (G.B.); (V.C.); (T.G.); (L.R.)
| | - Giorgio Baldari
- Nuclear Medicine Division, Azienda Ospedaliero-Universitaria di Parma, 43126 Parma, Italy; (M.S.); (G.B.); (V.C.); (T.G.); (L.R.)
| | - Veronica Cervati
- Nuclear Medicine Division, Azienda Ospedaliero-Universitaria di Parma, 43126 Parma, Italy; (M.S.); (G.B.); (V.C.); (T.G.); (L.R.)
| | - Tiziano Graziani
- Nuclear Medicine Division, Azienda Ospedaliero-Universitaria di Parma, 43126 Parma, Italy; (M.S.); (G.B.); (V.C.); (T.G.); (L.R.)
| | - Nicoletta Campanini
- Pathology Division, Azienda Ospedaliero-Universitaria di Parma, 43126 Parma, Italy; (E.M.S.); (N.C.)
- Department of Medicine and Surgery, University of Parma, 43121 Parma, Italy
| | - Umberto Maestroni
- Urology Division, Azienda Ospedaliero-Universitaria di Parma, 43126 Parma, Italy;
| | - Livia Ruffini
- Nuclear Medicine Division, Azienda Ospedaliero-Universitaria di Parma, 43126 Parma, Italy; (M.S.); (G.B.); (V.C.); (T.G.); (L.R.)
| |
Collapse
|
11
|
Wang JH, Kiess AP. PSMA-targeted therapy for non-prostate cancers. Front Oncol 2023; 13:1220586. [PMID: 37645427 PMCID: PMC10461313 DOI: 10.3389/fonc.2023.1220586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/25/2023] [Indexed: 08/31/2023] Open
Abstract
Radioligand therapy (RLT) agents are demonstrating a crucial role in the clinical approach to aggressive malignancies such as metastatic castrate-resistant prostate cancer (m-CRPC). With the recent FDA approval of prostate-specific membrane antigen (PSMA)-targeted RLT for m-CRPC, the field has broadened its gaze to explore other cancers that express PSMA in the tumor parenchyma or tumor neovasculature. In this review article, we discuss current progress in the clinical use of PSMA RLTs in non-prostate cancers such salivary gland cancers, renal cell carcinoma, high grade glioma, and soft tissue sarcoma. We highlight early reports in small case series and clinical trials indicating promise for PSMA-targeted RLT and highlighting the importance of identifying patient cohorts who may most benefit from these interventions. Further study is indicated in non-prostate cancers investigating PSMA RLT dosimetry, PSMA PET/CT imaging as a biomarker, and assessing PSMA RLT safety and efficacy in these cancers.
Collapse
Affiliation(s)
- Jarey H. Wang
- Department of Radiation Oncology and Molecular Radiation Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| | - Ana P. Kiess
- Department of Radiation Oncology and Molecular Radiation Sciences, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
12
|
Zhu W, Li X, Zheng G, Bai C, Ji Z, Zhang H, Xing H, Zhang Y, Huo L. Preclinical and pilot clinical evaluation of a small-molecule carbonic anhydrase IX targeting PET tracer in clear cell renal cell carcinoma. Eur J Nucl Med Mol Imaging 2023; 50:3116-3125. [PMID: 37246998 DOI: 10.1007/s00259-023-06248-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/24/2023] [Indexed: 05/30/2023]
Abstract
PURPOSE Clear cell renal cell carcinoma (ccRCC) highly expresses carbonic anhydrase IX (CAIX). The purpose of this study was to evaluate 68Ga-NY104, a small-molecule CAIX-targeting PET agent, in tumor models of ccRCC and patients diagnosed with confirmed, or suspicious, ccRCC. METHODS The in vivo and ex vivo biodistribution of 68Ga-NY104 was investigated in CAIX-positive OS-RC-2 xenograft-bearing models. The binding of the tracer was further validated using autoradiography for human ccRCC samples. In addition, three patients with confirmed or suspicious ccRCC were studied. RESULTS NY104 can be labeled with high radiochemical yield and purity. It quickly cleared through kidney with α-half-life of 0.15 h. Discernible uptake is noted in the heart, lung, liver, stomach, and kidney. The OS-RC-2 xenograft demonstrated intense uptake 5 min after injection and gradually increased until 3 h after injection with ID%/g of 29.29 ± 6.82. Significant binding was detected using autoradiography on sections of human ccRCC tumor. In the three patients studied, 68Ga-NY104 was well-tolerated and no adverse events were reported. Substantial accumulation was observed in both primary and metastatic lesions in patient 1 and 2 with SUVmax of 42.3. Uptake in the stomach, pancreas, intestine, and choroid plexus was noted. The lesion in third patient was correctly diagnosed as non-metastatic for negative 68Ga-NY104 uptake. CONCLUSION 68Ga-NY104 can efficiently and specifically bind to CAIX. Given the pilot nature of our study, future clinical studies are warranted to evaluate 68Ga-NY104 for detection of CAIX-positive lesions in patients with ccRCC. TRIAL REGISTRATION The clinical evaluation part of this study was retrospectively registered at ClinicalTrial.gov (NCT05728515) as NYPILOT on 6 Feb, 2023.
Collapse
Affiliation(s)
- Wenjia Zhu
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Xiaoyuan Li
- Department of Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Guoyang Zheng
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Chunmei Bai
- Department of Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Zhigang Ji
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Haiqiong Zhang
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Haiqun Xing
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China
| | - Yushi Zhang
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Li Huo
- Department of Nuclear Medicine, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Shuaifuyuan, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
13
|
Saad J, Shanmugasundaram R, Wong V, Le K, Arianayagam M, Roberts MJ. 68Ga-PSMA PET/CT false positive liver metastasis prior to radical prostatectomy. ANZ J Surg 2023; 93:1029-1030. [PMID: 36317600 DOI: 10.1111/ans.18051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 08/13/2022] [Accepted: 08/30/2022] [Indexed: 04/14/2023]
Affiliation(s)
- Jeremy Saad
- Nepean Urology Research Group, Nepean Hospital, Kingswood, New South Wales, Australia
| | - Ramesh Shanmugasundaram
- Nepean Urology Research Group, Nepean Hospital, Kingswood, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Veronica Wong
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Department of Nuclear Medicine, Nepean Hospital, Kingswood, New South Wales, Australia
| | - Ken Le
- Department of Nuclear Medicine, Nepean Hospital, Kingswood, New South Wales, Australia
| | - Mohan Arianayagam
- Nepean Urology Research Group, Nepean Hospital, Kingswood, New South Wales, Australia
| | - Matthew J Roberts
- Nepean Urology Research Group, Nepean Hospital, Kingswood, New South Wales, Australia
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Centre for Clinical Research, University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
14
|
Wang B, Deng Y, Xu Q, Gao J, Shen H, He X, Ding Q, Wang F, Guo H. Exploration of 68Ga-labelled prostate-specific membrane antigen-11 PET/CT parameters for identifying PBRM1 status in primary clear cell renal cell carcinoma. Clin Radiol 2023; 78:e417-e424. [PMID: 36805287 DOI: 10.1016/j.crad.2023.01.003] [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: 08/30/2022] [Revised: 12/26/2022] [Accepted: 01/10/2023] [Indexed: 02/04/2023]
Abstract
AIM To investigate the predictive value of 68Ga-labelled prostate-specific membrane antigen-11 (68Ga-PSMA-11) integrated positron-emission tomography (PET)/computed tomography (CT) in PBRM1-deficient clear cell renal cell carcinoma (ccRCC). MATERIALS AND METHODS A total of 41 patients with ccRCC, were enrolled retrospectively and underwent 68Ga-PSMA-11 PET/CT preoperatively. Radiological parameters, including CT attenuation value and maximum standard uptake value (SUVmax), were derived. Immunohistochemical and multiple immunofluorescences staining were performed to evaluate the PBRM1 status and immune response. The predictive value of imaging factors was analysed using a receiver operator characteristic curve analysis. Univariate and multivariate logistic regression analyses were used to investigate the relationship between clinical and radiological variables and PBRM1 status. RESULTS A total of 41 patients were included in this study, with 14 patients having PBRM1-deficient status. The tumour diameter on imaging and SUVmax differed significantly in patients with different PBRM1 expression statuses and no difference in CT attenuation was identified. Univariate and multivariate logistic regression analyses showed SUVmax was an obvious predictor for identification of PBRM1-deficient tumours. In addition, PBRM1-deficient tumours tended to be accompanied by greater cytotoxic T-cell infiltration, although most of them were in an exhausted state. CONCLUSIONS 68Ga-PSMA-11 PET/CT could be used to discriminate invasive PBRM1-deficient ccRCC.
Collapse
Affiliation(s)
- B Wang
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Y Deng
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Q Xu
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - J Gao
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - H Shen
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - X He
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Q Ding
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Xuzhou Medical University, Nanjing, Jiangsu, China
| | - F Wang
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - H Guo
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
15
|
Li Y, Zheng R, Zhang Y, Huang C, Tian L, Liu R, Liu Y, Zhang Z, Han H, Zhou F, He L, Dong P. Special issue "The advance of solid tumor research in China": 68Ga-PSMA-11 PET/CT for evaluating primary and metastatic lesions in different histological subtypes of renal cell carcinoma. Int J Cancer 2023; 152:42-50. [PMID: 35751420 PMCID: PMC9796964 DOI: 10.1002/ijc.34189] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 05/25/2022] [Accepted: 05/27/2022] [Indexed: 01/07/2023]
Abstract
Conventional imaging examinations are not sensitive enough for the early detection of recurrent or metastatic lesions in renal cell carcinoma (RCC) patients. We aimed to explore the role of 68 Ga-prostate specific membrane antigen (PSMA)-11 positron emission tomography (PET)/computed tomography (CT) in the detection of primary and metastatic lesions in such patients. We retrospectively analyzed 50 RCC patients who underwent 68 Ga-PSMA-11 PET/CT from November 2017 to December 2020. We observed a higher median accuracy and tumor-to-background maximum standard uptake value (SUVmax ) ratio (TBR) of 68 Ga-PSMA-11 PET/CT in clear cell RCC (ccRCC; 96.57% and 6.00, respectively) than in non-clear cell RCC (ncRCC; 82.05% and 2.99, respectively). The accuracies in detecting lesions in the renal region, bone, lymph nodes and lungs in ccRCC were 100.00%, 95.00%, 98.08% and 75.00%, respectively, and those in the renal region, bone and lymph nodes in ncRCC were 100.00%, 86.67% and 36.36%, respectively. The median TBRs of the lesions from the above locations were 0.38, 10.96, 6.69 and 13.71, respectively, in ccRCC and 0.13, 4.02 and 0.73, respectively, in ncRCC. The PSMA score evaluated with immunohistochemistry was correlated with the SUVmax (P = .046) in RCC. Higher PSMA scores were observed in ccRCC than in ncRCC (P = .031). 68 Ga-PSMA-11 PET/CT resulted in changes in clinical management in 12.9% (4/31) of cases because of the discovery of new metastases not detected with conventional imaging. These results indicate that 68 Ga-PSMA-11 PET/CT is a promising method for the detection of metastatic lesions in ccRCC, especially for those in the bone and lymph nodes.
Collapse
Affiliation(s)
- Yilin Li
- Department of Radiation OncologySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineGuangzhouPeople's Republic of China
| | - Rongliang Zheng
- Department of Nuclear MedicineSun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineGuangzhouPeople's Republic of China
| | - Yijun Zhang
- Department of PathologySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineGuangzhouPeople's Republic of China
| | - Chaoyun Huang
- Department of PathologySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineGuangzhouPeople's Republic of China
| | - Li Tian
- Department of RadiologySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineGuangzhouPeople's Republic of China
| | - Ruiqi Liu
- Department of Radiation OncologySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineGuangzhouPeople's Republic of China
| | - Yang Liu
- Department of Radiation OncologySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineGuangzhouPeople's Republic of China
| | - Zhiling Zhang
- Department of UrologySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineGuangzhouPeople's Republic of China
| | - Hui Han
- Department of UrologySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineGuangzhouPeople's Republic of China
| | - Fangjian Zhou
- Department of UrologySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineGuangzhouPeople's Republic of China
| | - Liru He
- Department of Radiation OncologySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineGuangzhouPeople's Republic of China
| | - Pei Dong
- Department of UrologySun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer MedicineGuangzhouPeople's Republic of China
| |
Collapse
|
16
|
Cimadamore A, Caliò A, Marandino L, Marletta S, Franzese C, Schips L, Amparore D, Bertolo R, Muselaers S, Erdem S, Ingels A, Pavan N, Pecoraro A, Kara Ö, Roussel E, Carbonara U, Campi R, Marchioni M. Hot topics in renal cancer pathology: implications for clinical management. Expert Rev Anticancer Ther 2022; 22:1275-1287. [PMID: 36377655 DOI: 10.1080/14737140.2022.2145952] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
INTRODUCTION The updated European Association of Urology (EAU) Guidelines issued a weak recommendation for adjuvant pembrolizumab for patients with high-risk operable clear cell Renal Cell Carcinoma (ccRCC). High risk of recurrence was defined, as per protocol-criteria, as T2 with nuclear grade 4 or sarcomatoid differentiation, T3 or higher, regional lymph node metastasis, or stage M1 with no evidence of disease. Considering the heterogeneous population included in the recommendation, it has been questioned if adjuvant pembrolizumab may lead to overtreatment of some patients as well as undertreatment of patients with worse prognosis. AREAS COVERED In this review, we discuss the issues related to the assessment of pathological features required to identify those patients harboring a high-risk tumor, highlighting the issue related to interobserver variability and discuss the currently available prognostic scoring systems in ccRCC. EXPERT OPINION PPathologist assessment of prognostic features suffers from interobserver variability which may depend on gross sampling and the pathologist's expertise. The presence of clear cell feature is not sufficient criteria by itself to define ccRCC since clear cell can be also found in other histotypes. Application of molecular biomarkers may be useful tools in the near future to help clinicians identify patients harboring tumors with worse prognosis.
Collapse
Affiliation(s)
- Alessia Cimadamore
- Institute of Pathological Anatomy, Department of Medical Area, University of UdineUdineItaly
| | - Anna Caliò
- Department of Diagnostic and Public Health, University of Verona, Verona, Italy
| | - Laura Marandino
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Stefano Marletta
- Department of Diagnostic and Public Health, University of Verona, Verona, Italy
| | - Carmine Franzese
- Department of Urology, Polytechnic University of Marche, Ancona, Italy
| | - Luigi Schips
- Department of Medical, Oral and Biotechnological Science, "Ss. Annunziata" Hospital Urology Unit, "G. d'Annunzio" University of Chieti and Pescara, Chieti, Italy
| | - Daniele Amparore
- Division of Urology, Department of Oncology, School of Medicine, San Luigi Hospital, University of Turin, Orbassano, Italy
| | | | - Stijn Muselaers
- Department of Urology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Selcuk Erdem
- Division of Urologic Oncology, Department of Urology, Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Alexandre Ingels
- Department of Urology, University Hospital Henri Mondor, Créteil, France
| | - Nicola Pavan
- Urology Clinic, Department of Medical, Surgical and Health Science, University of Trieste, Trieste, Italy
| | - Angela Pecoraro
- Division of Urology, Department of Oncology, School of Medicine, San Luigi Hospital, University of Turin, Orbassano, Italy
| | - Önder Kara
- Department of Urology, Kocaeli University School of Medicine, Izmit, Turkey
| | - Eduard Roussel
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - Umberto Carbonara
- Department of Emergency and Organ Transplantation-Urology, Andrology and Kidney Transplantation Unit, University of Bari, Bari, Italy
| | - Riccardo Campi
- Unit of Urological Robotic Surgery and Renal Transplantation, Careggi Hospital, University of Florence, Florence, Italy
| | - Michele Marchioni
- Department of Medical, Oral and Biotechnological Science, "Ss. Annunziata" Hospital Urology Unit, "G. d'Annunzio" University of Chieti and Pescara, Chieti, Italy
| | | |
Collapse
|
17
|
Raghubar AM, Roberts MJ, Wood S, Healy HG, Kassianos AJ, Mallett AJ. Cellular milieu in clear cell renal cell carcinoma. Front Oncol 2022; 12:943583. [PMID: 36313721 PMCID: PMC9614096 DOI: 10.3389/fonc.2022.943583] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 09/21/2022] [Indexed: 11/13/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is globally the most prevalent renal cancer. The cells of origin in ccRCC have been identified as proximal tubular epithelial cells (PTEC); however, the transcriptomic pathways resulting in the transition from normal to malignant PTEC state have remained unclear. Immunotherapy targeting checkpoints have revolutionized the management of ccRCC, but a sustained clinical response is achieved in only a minority of ccRCC patients. This indicates that our understanding of the mechanisms involved in the malignant transition and resistance to immune checkpoint therapy in ccRCC is unclear. This review examines recent single-cell transcriptomics studies of ccRCC to clarify the transition of PTEC in ccRCC development, and the immune cell types, states, and interactions that may limit the response to targeted immune therapy, and finally suggests stromal cells as key drivers in recurrent and locally invasive ccRCC. These and future single-cell transcriptomics studies will continue to clarify the cellular milieu in the ccRCC microenvironment, thus defining actional clinical, therapeutic, and prognostic characteristics of ccRCC.
Collapse
Affiliation(s)
- Arti M. Raghubar
- Kidney Health Service, Royal Brisbane and Women’s Hospital, Herston, QLD, Australia
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Health Support Queensland, Herston, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Anatomical Pathology, Pathology Queensland, Health Support Queensland, Herston, QLD, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - Matthew J. Roberts
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Department of Urology, Royal Brisbane and Women’s Hospital, Brisbane, QLD, Australia
- Department of Urology, Redcliffe Hospital, Redcliffe, QLD, Australia
- Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia
| | - Simon Wood
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Department of Urology, Princess Alexandra Hospital, Brisbane, QLD, Australia
| | - Helen G. Healy
- Kidney Health Service, Royal Brisbane and Women’s Hospital, Herston, QLD, Australia
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Health Support Queensland, Herston, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Andrew J. Kassianos
- Kidney Health Service, Royal Brisbane and Women’s Hospital, Herston, QLD, Australia
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Health Support Queensland, Herston, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Andrew J. Mallett
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
- College of Medicine & Dentistry, James Cook University, Townsville, QLD, Australia
- Department of Renal Medicine, Townsville University Hospital, Townsville, QLD, Australia
- *Correspondence: Andrew J. Mallett,
| |
Collapse
|
18
|
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: 20] [Impact Index Per Article: 10.0] [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
|
19
|
PET-CT in Clinical Adult Oncology-IV. Gynecologic and Genitourinary Malignancies. Cancers (Basel) 2022; 14:cancers14123000. [PMID: 35740665 PMCID: PMC9220973 DOI: 10.3390/cancers14123000] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/06/2022] [Accepted: 06/08/2022] [Indexed: 01/04/2023] Open
Abstract
Simple Summary Positron emission tomography (PET), typically combined with computed tomography (CT), has become a critical advanced imaging technique in oncology. With concurrently acquired positron emission tomography and computed tomography (PET-CT), a radioactive molecule (radiotracer) is injected in the bloodstream and localizes to sites of tumor because of specific cellular features of the tumor that accumulate the targeting radiotracer. The CT scan provides information to allow better visualization of radioactivity from deep or dense structures and to provide detailed anatomic information. PET-CT has a variety of applications in oncology, including staging, therapeutic response assessment, restaging and surveillance. This series of six review articles provides an overview of the value, applications, and imaging interpretive strategies for PET-CT in the more common adult malignancies. The fourth report in this series provides a review of PET-CT imaging in gynecologic and genitourinary malignancies. Abstract Concurrently acquired positron emission tomography and computed tomography (PET-CT) is an advanced imaging modality with diverse oncologic applications, including staging, therapeutic assessment, restaging and longitudinal surveillance. This series of six review articles focuses on providing practical information to providers and imaging professionals regarding the best use and interpretative strategies of PET-CT for oncologic indications in adult patients. In this fourth article of the series, the more common gynecological and adult genitourinary malignancies encountered in clinical practice are addressed, with an emphasis on Food and Drug Administration (FDA)-approved and clinically available radiopharmaceuticals. The advent of new FDA-approved radiopharmaceuticals for prostate cancer imaging has revolutionized PET-CT imaging in this important disease, and these are addressed in this report. However, [18F]F-fluoro-2-deoxy-d-glucose (FDG) remains the mainstay for PET-CT imaging of gynecologic and many other genitourinary malignancies. This information will serve as a guide for the appropriate role of PET-CT in the clinical management of gynecologic and genitourinary cancer patients for health care professionals caring for adult cancer patients. It also addresses the nuances and provides guidance in the accurate interpretation of FDG PET-CT in gynecological and genitourinary malignancies for imaging providers, including radiologists, nuclear medicine physicians and their trainees.
Collapse
|
20
|
Specified iron oxide nanoparticles by PSMA-11 as a promising nanomolecular imaging probe for early detection of prostate cancer. APPLIED NANOSCIENCE 2022. [DOI: 10.1007/s13204-022-02507-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
21
|
Tariq A, McGeorge S, Pearce A, Rhee H, Wood S, Kyle S, Marsh P, Raveenthiran S, Wong D, McBean R, Westera J, Dunglison N, Esler R, Navaratnam A, Yaxley J, Thomas P, Pattison DA, Roberts MJ. Characterization of tumor thrombus in renal cell carcinoma with prostate specific membrane antigen (PSMA) positron emission tomography (PET)/computed tomography (CT). Urol Oncol 2022; 40:276.e1-276.e9. [DOI: 10.1016/j.urolonc.2022.03.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 03/06/2022] [Accepted: 03/12/2022] [Indexed: 02/04/2023]
|
22
|
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
|
23
|
Sergeeva O, Zhang Y, Julian W, Sasikumar A, Awadallah A, Kenyon J, Shi W, Sergeev M, Huang S, Sexton S, Iyer R, Xin W, Avril N, Chan ER, Lee Z. Imaging of Tumor-Associated Vascular Prostate-Specific Membrane Antigen in Woodchuck Model of Hepatocellular Carcinoma. GASTRO HEP ADVANCES 2022; 1:631-639. [PMID: 35844243 PMCID: PMC9280909 DOI: 10.1016/j.gastha.2022.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
BACKGROUND AND AIMS Radiolabeled short peptide ligands targeting prostate-specific membrane antigen (PSMA) were developed initially for imaging and treatment of prostate cancers. While many nonprostate solid tumors including hepatocellular carcinoma (HCC) express little PSMA, their neovasculature expresses a high level of PSMA, which is avid for Gallium-68-labeled PSMA-targeting radio-ligand (68Ga-PSMA-11) for positron emission tomography (PET). However, the lack of a spontaneous animal model of tumor-associated vascular PSMA overexpression has hindered the development and assessment of PSMA-targeting radioligands for imaging and therapy of the nonprostatic cancers. We identified detectable indigenous PSMA expression on tumor neovascular endothelia in a naturally occurring woodchuck model of HCC. METHODS Molecular docking was performed with 3 bait PSMA ligands and compared between human and woodchuck PSMA. Initially, PET images were acquired dynamically after intravenously injecting 37 MBq (1.0 mCi) of 68Ga-PSMA-11 into woodchuck models of HCC. Subsequently, 10-minute static PET scans were conducted for other animals 1-hour after injection due to HCC and liver background uptake stabilization at 30-45 minutes after injection. Liver tissue samples were harvested after imaging, fresh-frozen for quantitative reverse transcription polymerase chain reaction and western blot for validation, or fixed for histology for correlation. RESULTS Our preclinical studies confirmed the initial clinical findings of 68Ga-PSMA-11 uptake in HCC. The agents (ligands and antibodies) developed against human PSMA were found to be reactive against the woodchuck PSMA. CONCLUSION This animal model offers a unique opportunity for investigating the biogenesis of tumor-associated vascular PSMA, its functional role(s), and potentials for future treatment strategies targeting tumor vascular PSMA using already developed PSMA-targeting agents.
Collapse
Affiliation(s)
- Olga Sergeeva
- Radiology, Case Western Reserve University, Cleveland, Ohio
| | - Yifan Zhang
- Radiology, Case Western Reserve University, Cleveland, Ohio
| | - Willian Julian
- Radiology, Case Western Reserve University, Cleveland, Ohio
| | - Arun Sasikumar
- Nuclear Medicine, St. Gregorios International Cancer Care Centre, Parumala, Kerala, India
| | - Amad Awadallah
- Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | | | - Wuxian Shi
- Center for Proteomics and Bioinformatics, Case Western Reserve University, Cleveland, Ohio
| | - Maxim Sergeev
- Radiology, University Hospitals Clevel and Medical Center, Cleveland, Ohio
| | - Steve Huang
- Nuclear Medicine, Cleveland Clinic, Cleveland, Ohio
| | - Sandra Sexton
- Medical Oncology, Roswell Park Cancer Institute, Buffalo, New York
| | - Renuka Iyer
- Medical Oncology, Roswell Park Cancer Institute, Buffalo, New York
| | - Wei Xin
- Pathology, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Norbert Avril
- Radiology, University Hospitals Clevel and Medical Center, Cleveland, Ohio
| | - Ernest Ricky Chan
- Institute for Computational Biology, Case Western Reserve University, Cleveland, Ohio
| | - Zhenghong Lee
- Radiology, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
24
|
Gao J, Meng L, Xu Q, Zhao X, Deng Y, Fu Y, Guo S, He K, Shi J, Wang F, Zhang S, Guo H. 68Ga-PSMA-11 PET/CT Parameter Correlates with Pathological VEGFR-2/PDGFR-β Expression in Renal Cell Carcinoma Patients. Mol Imaging Biol 2022; 24:759-768. [PMID: 35451707 DOI: 10.1007/s11307-022-01725-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/14/2022] [Accepted: 03/21/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND Response prediction is necessary for renal cell carcinoma (RCC) tumors. We aim to evaluate parameters derived from 68 Ga-PSMA-11 PET/CT images for prediction of pathological VEGFR-2/PDGFR-β expression of primary RCC tumors. METHODS Forty-eight RCC patients were retrospectively enrolled with preoperative 68 Ga-PSMA-11 PET/CT scan and surgical specimen. Radiological parameters including tumor diameter, mean CT value, and maximal standard uptake value (SUVmax) were derived from PET/CT images and pathological VEGFR-2/PDGFR-β/PSMA expression were identified with immunohistochemistry. Mann-Whitney U test was performed for continuous variables and the chi-square test for categorical variables. ROC was used for determining the effectiveness of preoperative parameters in differentiating VEGFR-2/PDGFR-β expression. Univariate and multivariate logistic regression analyses were performed for significant parameters to predict VEGFR-2 & PDGFR-β co-expression. RESULTS Of the 48 tumors, 25 (52.1%) harbored positive VEGFR-2 expression, 28 (58.3%) harbored positive PDGFR-β expression, and 24 (50%) were both VEGFR-2 positive and PDGFR-β positive. SUVmax significantly differed by subgroups of VEGFR-2/PDGFR-β expression (both P < 0.001). SUVmax demonstrated superior performance for differentiating VEGFR-2 & PDGFR-β co-expression (positive vs. negative), with area under the curve 0.87 (95% CI 0.78-0.96, P < 0.001), sensitivity 93% and specificity 78%. Moreover, SUVmax was identified as the significant predictor for VEGFR-2 & PDGFR-β co-expression (odds ratio 4.01, 95% CI 1.99-8.08, P < 0.001). Concordant with radiological findings with 68 Ga-PSMA-11 PET/CT, pathological PSMA staining intensity was significantly higher in both VEGFR-2-positive tumor and PDGFR-β-positive tumor (P = 0.009 and P < 0.001, respectively). CONCLUSION 68 Ga-PSMA-11 PET/CT could effectively identify pathological VEGFR-2/PDGFR-β expression of primary RCC tumors, which may help with selection of mRCC patients suitable for TKIs treatment.
Collapse
Affiliation(s)
- Jie Gao
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu Province, China
| | - Longxiyu Meng
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu Province, China
| | - Qinfeng Xu
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Jiangsu, China
| | - Xiaozhi Zhao
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu Province, China
| | - Yongming Deng
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu Province, China
| | - Yao Fu
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Jiangsu, China
| | - Suhan Guo
- School of Artificial Intelligence, Nanjing University, Jiangsu, China
| | - Kuiqiang He
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu Province, China
| | - Jiong Shi
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Jiangsu, China
| | - Feng Wang
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Jiangsu, China
| | - Shiwei Zhang
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu Province, China.
| | - Hongqian Guo
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu Province, China.
| |
Collapse
|
25
|
Tariq A, Raman V, Alexander H, Roberts MJ, Thomas P. Pituitary Metastasis of Renal Cell Carcinoma Characterized by 18F-Prostate-Specific Membrane Antigen-1007 PET/CT. Clin Nucl Med 2022; 47:e346-e347. [PMID: 35044961 DOI: 10.1097/rlu.0000000000004055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
ABSTRACT Renal cell carcinoma (RCC) is one of the most common oncological diagnoses worldwide. Accurate staging and restaging imaging continue to be a priority in clinical practice as 20% to 30% of patients present with advanced metastatic disease at diagnosis, and 20% to 30% develop recurrence after surgical resection. We present a case of a 71-year-old man presenting with an isolated pituitary RCC metastasis accurately defined on 18F-prostate-specific membrane antigen-1007 masquerading as a macroadenoma on conventional imaging (CT and MRI). This demonstrates the potential utility of 18F-prostate-specific membrane antigen-1007 in characterizing RCC brain metastasis.
Collapse
|
26
|
Meng L, Zhang S, Gao J, Xu Q, Fu Y, Zhou YH, Wang F, Guo H. [ 68Ga]Ga-PSMA-11 PET/CT has potential application in predicting tumor HIF-2α expression and therapeutic response to HIF-2α antagonists in patients with RCC. Eur Radiol 2022; 32:6545-6553. [PMID: 35357538 DOI: 10.1007/s00330-022-08738-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To evaluate the efficacy of parameters derived from [68Ga]Ga-PSMA-11 PET/CT images in predicting pathological HIF-2α expression in primary tumors among patients with renal cell carcinoma (RCC). METHODS Fifty-three RCC patients with preoperative [68Ga]Ga-PSMA-11 PET/CT scans and complete surgical specimens were retrospectively enrolled in this study. Radiographic parameters were obtained from PET/CT images, and immunohistochemistry was used to measure the expression of HIF-2α and PSMA. Continuous variables and categorical variables were analyzed by the Mann-Whitney U test and chi-square test, respectively. ROC analysis was used to test the efficacy of several preoperative parameters in identifying pathological HIF-2α expression. Univariable logistic regression analyses were performed for significant parameters to predict pathological HIF-2α expression in RCC. RESULTS Of the 53 tumors, 29 (54.7%) had high expression of HIF-2α. The SUVmax was significantly different in the HIF-2α expression subgroups (p < 0.001). SUVmax emerged as the most significant parameter to differentiate HIF-2α expression subgroups (high vs. low), with the AUC of 0.93 (95% CI 0.85-1.00, p < 0.001), sensitivity of 90%, and specificity of 88%. Furthermore, SUVmax was confirmed as the most significant predictor of HIF-2α expression level by univariable logistic regression model analysis (odds ratio 1.39, 95% CI 1.17-1.65, p < 0.001). Consistent with the radiographic results of [68Ga]Ga-PSMA-11 PET/CT, the staining intensity of pathological PSMA was significantly higher in HIF-2α-high-expressing tumors (p = 0.003). CONCLUSIONS [68Ga]Ga-PSMA-11 PET/CT was superior in identifying pathological HIF-2α expression in primary tumors of RCC patients, demonstrating its potential application in predicting responses to HIF-2α antagonists. KEY POINTS • [68Ga]Ga-PSMA-11 PET/CT could potentially predict the HIF-2α expression of primary tumors among patients with RCC. • SUVmaxof [68Ga]Ga-PSMA-11 PET/CT was the most significant predictor of HIF-2α expression level. • This probability could help predict the therapeutic response of patients with RCC to HIF-2α antagonists.
Collapse
Affiliation(s)
- Longxiyu Meng
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, 321 Zhongshan Rd, Nanjing, Jiangsu, China
| | - Shun Zhang
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, 321 Zhongshan Rd, Nanjing, Jiangsu, China
- Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, 321 Zhongshan Rd, Nanjing, 210008, Jiangsu, China
| | - Jie Gao
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, 321 Zhongshan Rd, Nanjing, Jiangsu, China
| | - Qinfeng Xu
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, 68 Changle Rd, Nanjing, Jiangsu, China
| | - Yao Fu
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Rd, Nanjing, Jiangsu, China
| | - Yi-Hua Zhou
- Departments of Laboratory Medicine and Infectious Diseases, Nanjing Drum Tower Hospital, Nanjing University Medical School, 321 Zhongshan Rd, Nanjing, Jiangsu, China.
| | - Feng Wang
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, 68 Changle Rd, Nanjing, Jiangsu, China.
| | - Hongqian Guo
- Department of Urology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Institute of Urology, Nanjing University, 321 Zhongshan Rd, Nanjing, Jiangsu, China.
- Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, 321 Zhongshan Rd, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
27
|
Leder T, Drescher R, Gühne F, Theis B, Freesmeyer M. De Quervain Subacute Thyroiditis With Moderate PSMA Uptake Mimicking Thyroid Metastasis of Renal Cell Carcinoma. Clin Nucl Med 2022; 47:221-222. [PMID: 34690294 DOI: 10.1097/rlu.0000000000003952] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT PSMA-targeted PET/CT can be used for diagnosis and staging of clear cell renal cell carcinoma. A 68-year-old woman with suspected hepatic metastases from clear cell renal cell carcinoma underwent PET/CT with 68Ga-PSMA, which showed a focus of moderately increased tracer uptake in the right thyroid lobe. Sonography displayed a correlating hypoechogenic, ill-defined node (TIRADS 5). Right hemithyroidectomy was performed in the assumption of a solitary metastasis of renal cell carcinoma. Histologic examination revealed subacute granulomatous thyroiditis (De Quervain thyroiditis) without evidence of malignancy.
Collapse
Affiliation(s)
- Theresa Leder
- From the Clinic of Nuclear Medicine, Jena University Hospital, Jena, Germany
| | | | | | | | | |
Collapse
|
28
|
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: 11.5] [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
|
29
|
Application of 18F Prostate-Specific Membrane Antigen Positron Emission Tomography/Computed Tomography in Monitoring Gastric Metastasis and Cancer Thrombi from Renal Cell Carcinoma. JOURNAL OF ONCOLOGY 2022; 2022:5681463. [PMID: 35154318 PMCID: PMC8837453 DOI: 10.1155/2022/5681463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/11/2022] [Indexed: 11/17/2022]
Abstract
Background. Renal cell carcinoma (RCC) with gastric metastasis is rare, particularly accompanied by multiple cancer thrombi. Methods. We reported a 66-year-old man with a history of a right radical nephrectomy because of RCC. The patient underwent 18F prostate-specific membrane antigen (PSMA) positron emission tomography/computed tomography (PET/CT) scanning after 6 months of targeted therapy because of gastric metastasis and cancer thrombi. We conducted a systematic review of the literature and identified 73 cases of RCC with gastric metastasis. We analyzed the clinicopathological characteristics, therapies, and outcomes of patients. Results. 18F-PSMA PET/CT showed a large mass in the gastric fundus and cancer thrombi in the right atrium, inferior vena cava, and splenic vein with intense tracer uptake. Other metastases with increased tracer uptake included multiple bones and abdominal lymph nodes. The majority of gastric metastasis of RCC were men (53/73, 72.6%), with a median age at presentation of 67 (from 48 to 87) years. Gastric metastasis of RCC was mainly metachronous, and presented with small polyps or mass appearance and often accompanied by multiple-site metastases and gastrointestinal symptoms. An overall median interval between nephrectomy and diagnosis of gastric metastasis was 6 (from 0.1 to 23) years, and an overall median survival time was 14 (from 0.25 to 72) months. The median interval time of solitary gastric metastasis was longer than gastric metastasis with multiple-site metastases (7 vs.5 years;
). Patients with gastric and multiple-site metastases had higher mortality than patients with solitary metastasis (17 vs.1;
). The patients with synchronous gastric metastasis had a shorter survival time than metachronous gastric metastasis (6 vs.17 months;
). Conclusions. Postoperative follow-up of multiple imaging modalities to monitor recurrence and metastasis is necessary and important. PSMA PET/CT can improve the detection sensitivity of RCC, especially in metastatic clear cell renal cell carcinoma (ccRCC), and could provide a basis for disease staging, restaging, and therapeutic efficacy evaluation.
Collapse
|
30
|
Seront E, Lhommel R, Tombal B. Case Report: Early 68Ga-PSMA-PET Metabolic Assessment and Response to Systemic Treatment for First-Line Metastatic Clear Cell Renal Cell Carcinoma; About Two Clinical Cases. Front Oncol 2021; 11:782166. [PMID: 34950588 PMCID: PMC8689125 DOI: 10.3389/fonc.2021.782166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 11/16/2021] [Indexed: 01/03/2023] Open
Abstract
Early evaluation of response to anticancer treatment in metastatic renal cell carcinoma (RCC) is challenging as responses are sometimes delayed, as mixed responses can occur, and as conventional imaging have some limitations. As PSMA has been previously identified in neovasculature of clear cell RCC (ccRCC), 68Ga-PSMA-Positron Emitted Tomography (PET) could appear as an interesting tool to evaluate therapeutic response. We describe the association of an early decrease in 68Ga metabolism (at 8 weeks after treatment onset) and further radiological response (at 12 weeks after treatment onset) to treatment in two patients with different sensitivity to axitinib–pembrolizumab combination. Interestingly, one of these patients presented an initial progressive disease on pembrolizumab alone and a subsequent response to axitinib alone in the disease course; these response profiles were associated with absence of decrease and subsequent decrease in the 68Ga metabolism, respectively. Even if further prospective trials are needed, 68Ga-PSMA-PET may appear as a promising way for early prediction of response to ccRCC systemic treatment.
Collapse
Affiliation(s)
- Emmanuel Seront
- Division of Medical Oncology, Institut de Recherche Clinique (IREC), Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Renaud Lhommel
- Division of Nuclear Medicine, Institut de Recherche Clinique (IREC), Cliniques Universitaires Saint Luc, Brussels, Belgium
| | - Bertrand Tombal
- Division of Urology, Institut de Recherche Clinique (IREC), Cliniques Universitaires Saint Luc, Brussels, Belgium
| |
Collapse
|
31
|
Tariq A, Kwok M, Pearce A, Rhee H, Kyle S, Marsh P, Raveenthiran S, Wong D, McBean R, Westera J, Dunglison N, Esler R, Navaratnam A, Yaxley JW, Thomas P, Pattison DA, Roberts MJ. The role of dual tracer PSMA and FDG PET/CT in renal cell carcinoma (RCC) compared to conventional imaging: A multi-institutional case series with intra-individual comparison. Urol Oncol 2021; 40:66.e1-66.e9. [PMID: 34895817 DOI: 10.1016/j.urolonc.2021.11.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 11/05/2021] [Indexed: 01/03/2023]
Abstract
PURPOSE The objective of this study was to perform an intra-individual dual tracer comparison of Fluorodeoxyglucose (FDG) and Prostate Specific Membrane Antigen (PSMA) computed tomography (CT)/Positron Emission Tomography (PET) against standard of care (SOC) imaging for the characterisation, staging and restaging of renal cell carcinoma (RCC). METHODS A multicentre retrospective cohort study was performed at 3 major tertiary referral institutions in Brisbane, Australia between 2015 and 2020. All patients who underwent both PSMA and FDG PET/CT following SOC imaging for investigation of RCC were identified. Clinical details, imaging characteristics and histopathology were collected prior to univariate statistical analysis. RESULTS Eleven patients who underwent dual tracer PET/CT were included. Mean age was 65.5 years (SD 8.8). Most patients were male (64%) with clear cell morphology (91%). The indication for dual tracer PET was staging (36%) and restaging after radical/partial nephrectomy (64%). Primary tumour assessment showed mixed avidity patterns (concordant 40%, discordant favouring PSMA 20%, and FDG 40%). Metastatic disease assessment showed concordant avidity in 6 patients (55%), concordant negative in 3 (27%), and discordant uptake favouring PSMA. PET outperformed SOC imaging for assessment of metastatic disease in 5 patients (45%) and equivalent for the remainder. A change in management was noted in three cases (27%). CONCLUSION Dual tracer FDG and PSMA PET/CT for assessment of primary and metastatic RCC were mostly concordant. PET imaging outperformed conventional imaging and led to a change in management for 1 in 4 patients. Further studies with larger samples sizes are required to validate these findings and identify characteristics to guide patient selection for selective or dual tracer use.
Collapse
Affiliation(s)
- Arsalan Tariq
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia; Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Michael Kwok
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia; Department of Urology, Redcliffe Hospital, Queensland, Australia
| | - Adam Pearce
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia; Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia; Wesley Urology Clinic, The Wesley Hospital, Brisbane, Queensland, Australia
| | - Handoo Rhee
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia; Department of Urology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Samuel Kyle
- Department of Diagnostic Radiology, Princess Alexandra Hospital, Brisbane, Queensland, Australia; Department of Nuclear Medicine, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Phillip Marsh
- Department of Diagnostic Radiology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Sheliyan Raveenthiran
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia; Department of Urology, Redcliffe Hospital, Queensland, Australia
| | - David Wong
- I-MED Radiology, The Wesley Hospital, Brisbane, Queensland, Australia
| | - Rhiannon McBean
- I-MED Radiology, The Wesley Hospital, Brisbane, Queensland, Australia
| | - Jurjen Westera
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia; Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Nigel Dunglison
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia; Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Rachel Esler
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia; Wesley Urology Clinic, The Wesley Hospital, Brisbane, Queensland, Australia
| | - Anojan Navaratnam
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia; Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - John W Yaxley
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia; Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia; Wesley Urology Clinic, The Wesley Hospital, Brisbane, Queensland, Australia
| | - Paul Thomas
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia; Department of Nuclear Medicine, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - David A Pattison
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia; Department of Nuclear Medicine, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Matthew J Roberts
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia; Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia; Department of Urology, Redcliffe Hospital, Queensland, Australia; Centre for Clinical Research, The University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
32
|
Tariq A, Westera J, Navaratnam A, Dunglison N, Esler R, Roberts MJ. Extra-peritoneal oligometastatic renal cell carcinoma: atypical pattern of metastasis and novel localization techniques. ANZ J Surg 2021; 92:1553-1555. [PMID: 34719834 DOI: 10.1111/ans.17335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 10/10/2021] [Accepted: 10/12/2021] [Indexed: 11/28/2022]
Affiliation(s)
- Arsalan Tariq
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Jurjen Westera
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Anojan Navaratnam
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Nigel Dunglison
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Rachel Esler
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Matthew J Roberts
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia.,Prostate Theranostics and Urological Diseases Group, Centre for Clinical Research, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
33
|
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: 1.7] [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
|
34
|
68Ga-EMP-100 PET/CT-a novel ligand for visualizing c-MET expression in metastatic renal cell carcinoma-first in-human biodistribution and imaging results. Eur J Nucl Med Mol Imaging 2021; 49:1711-1720. [PMID: 34708249 PMCID: PMC8940803 DOI: 10.1007/s00259-021-05596-6] [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: 08/03/2021] [Accepted: 10/13/2021] [Indexed: 01/29/2023]
Abstract
Background 68Ga-EMP-100 is a novel positron emission tomography (PET) ligand that directly targets tumoral c-MET expression. Upregulation of the receptor tyrosin kinase c-MET in renal cell carcinoma (RCC) is correlated with overall survival in metastatic disease (mRCC). Clinicopathological staging of c-MET expression could improve patient management prior to systemic therapy with for instance inhibitors targeting c-MET such as cabozantinib. We present the first in-human data of 68Ga-EMP-100 in mRCC patients evaluating uptake characteristics in metastases and primary RCC. Methods Twelve patients with mRCC prior to anticipated cabozantinib therapy underwent 68Ga-EMP-100 PET/CT imaging. We compared the biodistribution in normal organs and tumor uptake of mRCC lesions by standard uptake value (SUVmean) and SUVmax measurements. Additionally, metastatic sites on PET were compared to contrast-enhanced computed tomography (CT) and the respective, quantitative PET parameters were assessed and then compared inter- and intra-individually. Results Overall, 87 tumor lesions were analyzed. Of these, 68/87 (79.3%) were visually rated c-MET-positive comprising a median SUVmax of 4.35 and SUVmean of 2.52. Comparing different tumor sites, the highest uptake intensity was found in tumor burden at the primary site (SUVmax 9.05 (4.86–29.16)), followed by bone metastases (SUVmax 5.56 (0.97–15.85)), and lymph node metastases (SUVmax 3.90 (2.13–6.28)) and visceral metastases (SUVmax 3.82 (0.11–16.18)). The occurrence of visually PET-negative lesions (20.7%) was distributed heterogeneously on an intra- and inter-individual level; the largest proportion of PET-negative metastatic lesions were lung and liver metastases. The highest physiological 68Ga-EMP-100 accumulation besides the urinary bladder content was seen in the kidneys, followed by moderate uptake in the liver and the spleen, whereas significantly lower uptake intensity was observed in the pancreas and the intestines. Conclusion Targeting c-MET expression, 68Ga-EMP-100 shows distinctly elevated uptake in mRCC patients with partially high inter- and intra-individual differences comprising both c-MET-positive and c-MET-negative lesions. Our first clinical results warrant further systemic studies investigating the clinical use of 68Ga-EMP-100 as a biomarker in mRCC patients.
Collapse
|
35
|
Shahrokhi P, Masteri Farahani A, Tamaddondar M, Rezazadeh F. The utility of radiolabeled PSMA ligands for tumor imaging. Chem Biol Drug Des 2021; 99:136-161. [PMID: 34472217 DOI: 10.1111/cbdd.13946] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/06/2021] [Accepted: 08/16/2021] [Indexed: 01/19/2023]
Abstract
Prostate-specific membrane antigen (PSMA) is a glycosylated type-II transmembrane protein expressed in prostatic tissue and significantly overexpressed in several prostate cancer cells. Despite its name, PSMA has also been reported to be overexpressed in endothelial cells of benign and malignant non-prostate disease. So its clinical use was extended to detection, staging, and therapy of various tumor types. Recently small molecules targeting PSMA have been developed as imaging probes for diagnosis of several malignancies. Preliminary studies are emerging improved diagnostic sensitivity and specificity of PSMA imaging, leading to a change in patient management. In this review, we evaluated the first preclinical and clinical studies on PSMA ligands resulting future perspectives radiolabeled PSMA in staging and molecular characterization, based on histopathologic examinations of PSMA expression.
Collapse
Affiliation(s)
- Pejman Shahrokhi
- Nuclear Medicine Center, Payambar Azam Hospital, Hormozgan University of Medical Sciences, Bandar Abbas, Hormozgan, Iran
| | - Arezou Masteri Farahani
- Nuclear Medicine Center, Payambar Azam Hospital, Hormozgan University of Medical Sciences, Bandar Abbas, Hormozgan, Iran
| | - Mohammad Tamaddondar
- Nephrology Department, Payambar Azam Hospital, Hormozgan University of Medical Sciences, Bandar Abbas, Hormozgan, Iran
| | - Farzaneh Rezazadeh
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
36
|
Immunohistochemical Reactivity of Prostate-Specific Membrane Antigen in Salivary Gland Tumors. Head Neck Pathol 2021; 16:427-433. [PMID: 34420181 PMCID: PMC9187811 DOI: 10.1007/s12105-021-01376-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 08/16/2021] [Indexed: 10/20/2022]
Abstract
Prostate-specific membrane antigen (PSMA) is a transmembrane glycoprotein that is overexpressed in the prostate gland and prostate cancer. PSMA has been recently used in positron emission tomography/computed tomography (PET/CT) imaging and targeted alpha-radiation therapy (TAT) for prostate cancer. Recently, the tubarial gland, a type of minor salivary gland that is described as a new organ situated in the pharynx, is reported to express PMSA. Here, we studied the expression of PSMA in common benign and malignant salivary gland tumors. We performed immunohistochemistry for PSMA in 55 salivary gland tumors comprising 10 pleomorphic adenomas, 10 Warthin tumors, 9 basal cell adenomas, 9 adenoid cystic carcinomas, 9 mucoepidermoid carcinomas, and 8 salivary duct carcinomas. PSMA was expressed in 97% of benign tumors and 77% of malignant tumors. Moreover, PSMA was expressed in 59% of normal salivary glands adjacent to the tumor. PSMA is relatively expressed in salivary gland tumors and salivary glands. Therefore, salivary gland neoplasm, and normal salivary gland, possibly demonstrate the accumulation of PSMA in PET/CT. Thus, we need to monitor the side effects in the salivary glands during TAT for prostate cancer.
Collapse
|
37
|
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: 3.7] [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
|
38
|
Zeng Y, Luo J, Liao H, Chen P. Utility of 18F-Prostate-Specific Membrane Antigen 1007 in Imaging of Tumor Thrombus of Renal Cell Carcinoma. Clin Nucl Med 2021; 46:697-699. [PMID: 33883493 DOI: 10.1097/rlu.0000000000003664] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT A 64-year-old man was referred for 18F-prostate-specific membrane antigen 1007 PET/CT with a suspicion of recurrence and metastases of renal cell carcinoma after the resection of right renal cell carcinoma. The scan showed intense tracer concentration in the inferior vena cava and the right atrium, which was later proven on histopathologic examination as tumor thrombus of renal cell carcinoma. Adding on to previous studies with 18F-prostate-specific membrane antigen 1007 in the primary renal tumor, the case proved that the tumor thrombus of renal cell carcinoma could also show intense tracer concentration and further highlights its utility in renal cell carcinoma.
Collapse
Affiliation(s)
- Yuping Zeng
- From the Department of Nuclear Medicine, Guangzhou Universal Medical Imaging Diagnostic Center, Guangzhou, China
| | | | | | | |
Collapse
|
39
|
Diagnostic potential of serum extracellular vesicles expressing prostate-specific membrane antigen in urologic malignancies. Sci Rep 2021; 11:15000. [PMID: 34294841 PMCID: PMC8298409 DOI: 10.1038/s41598-021-94603-9] [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: 02/06/2021] [Accepted: 07/09/2021] [Indexed: 12/12/2022] Open
Abstract
We aimed to develop a sandwich ELISA to detect prostate-specific membrane antigen (PSMA) on small extracellular vesicles (EVs) using T-cell immunoglobulin domain and mucin domain-containing protein 4 (Tim4) as a capture molecule for EVs and to evaluate its diagnostic potential in urologic malignancies. First, we optimized the conditions for sandwich ELISA measuring the PSMA level on EVs captured from serum by Tim4 and found that the use of highly-purified EVs released from Tim4 that had captured EVs in serum reduced the background. Second, we confirmed its validity by studying mouse xenograft model for prostate cancer (PC). Lastly, we measured PSMA-EVs in serum of patients with urologic malignancies. The PSMA-EV levels were significantly higher in metastatic PC and castration-resistant PC (CRPC) patients than in therapy-naïve PC patients. In renal cell carcinoma (RCC) patients, PSMA-EVs were elevated in those with metastasis compared with those without metastasis, which may reflect the development of the neovasculature positive for PSMA in tumors. In conclusion, we developed a sandwich ELISA for detection of PSMA-EVs using highly-purified EVs isolated from serum by Tim4. Our results suggest that PSMA-EVs may be useful to diagnose and monitor not only PC but also RCC and possibly other hypervascular solid tumors.
Collapse
|
40
|
Abstract
Hybrid FDG PET/CT plays a vital role in oncologic imaging and has been widely adopted for the staging and restaging of a variety of malignancies. Its diagnostic value in urogenital malignancies is less well-known, not at least because of the variable FDG avidity of these tumor entities, the sites of these tumors, and technical challenges associated with sequential imaging of CT and PET. PET/CT interpretation thus can be especially challenging and is associated with many pitfalls, which can lead to both false-positive and false-negative diagnoses as well as incorrect assessment of metabolic change following therapy. Currently, FDG PET/CT is not the standard of care for the initial diagnosis or staging of early-stage or low-risk urogenital cancers; however, it can help evaluate distant metastatic disease, response to therapy, and disease recurrence in high-risk patients. Knowledge of imaging features of tumor metabolic avidity and pitfalls is essential for accurate interpretation.
Collapse
Affiliation(s)
- Anil Vasireddi
- Department of Radiology, University of Pittsburgh Medical Center, UPMC Presbyterian Hospital, Pittsburgh, PA
| | - Nghi C Nguyen
- Department of Radiology, University of Pittsburgh Medical Center, UPMC Presbyterian Hospital, Pittsburgh, PA.
| |
Collapse
|
41
|
Siow A, Kowalczyk R, Brimble MA, Harris PWR. Evolution of Peptide-Based Prostate-Specific Membrane Antigen (PSMA) Inhibitors: An Approach to Novel Prostate Cancer Therapeutics. Curr Med Chem 2021; 28:3713-3752. [PMID: 33023429 DOI: 10.2174/0929867327666201006153847] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/24/2020] [Accepted: 08/30/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Prostate cancer is one of the most common cancers worldwide, with approximately 1.1 million cases diagnosed annually. The rapid development of molecular imaging has facilitated greater structural understanding, which can help formulate novel combinations of therapeutic regimens and more accurate diagnosis, avoiding unnecessary prostate biopsies. This accumulated knowledge also provides a greater understanding of the aggressive stages of the disease and tumor recurrence. Recently, much progress has been made on developing peptidomimetic-based inhibitors as promising candidates to effectively bind to the prostate- specific membrane antigen (PSMA), which is expressed by prostate cancer cells. OBJECTIVE In this review, recent advances covering small-molecule and peptide-based PSMA inhibitors will be extensively reviewed, providing a base for the rational design of future PSMA inhibitors. METHOD Herein, the literature on selected PSMA inhibitors that have been developed from 1996 to 2020 were reviewed, emphasizing recent synthetic advances and chemical strategies whilst highlighting therapeutic potential and drawbacks of each inhibitor. RESULTS Synthesized inhibitors presented in this review demonstrate the clinical application of certain PSMA inhibitors, exhibited in vitro and in vivo. CONCLUSION This review highlights the clinical potential of PSMA inhibitors, analyzing the advantages and setbacks of the chemical synthetic methodologies utilized, setting precedence for the discovery of novel PSMA inhibitors for future clinical applications.
Collapse
Affiliation(s)
- Andrew Siow
- School of Chemical Sciences, The University of Auckland, 23 Symonds Street, Private Bag: 92019, Auckland 1010, New Zealand
| | - Renata Kowalczyk
- School of Chemical Sciences, The University of Auckland, 23 Symonds Street, Private Bag: 92019, Auckland 1010, New Zealand
| | - Margaret A Brimble
- School of Chemical Sciences, The University of Auckland, 23 Symonds Street, Private Bag: 92019, Auckland 1010, New Zealand
| | - Paul W R Harris
- School of Chemical Sciences, The University of Auckland, 23 Symonds Street, Private Bag: 92019, Auckland 1010, New Zealand
| |
Collapse
|
42
|
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: 8] [Impact Index Per Article: 2.7] [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
|
43
|
Jeevanandam J, Sabbih G, Tan KX, Danquah MK. Oncological Ligand-Target Binding Systems and Developmental Approaches for Cancer Theranostics. Mol Biotechnol 2021; 63:167-183. [PMID: 33423212 DOI: 10.1007/s12033-020-00296-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2020] [Indexed: 02/07/2023]
Abstract
Targeted treatment of cancer hinges on the identification of specific intracellular molecular receptors on cancer cells to stimulate apoptosis for eventually inhibiting growth; the development of novel ligands to target biomarkers expressed by the cancer cells; and the creation of novel multifunctional carrier systems for targeted delivery of anticancer drugs to specific malignant sites. There are numerous receptors, antigens, and biomarkers that have been discovered as oncological targets (oncotargets) for cancer diagnosis and treatment applications. Oncotargets are critically important to navigate active anticancer drug ingredients to specific disease sites with no/minimal effect on surrounding normal cells. In silico techniques relating to genomics, proteomics, and bioinformatics have catalyzed the discovery of oncotargets for various cancer types. Effective oncotargeting requires high-affinity probes engineered for specific binding of receptors associated with the malignancy. Computational methods such as structural modeling and molecular dynamic (MD) simulations offer opportunities to structurally design novel ligands and optimize binding affinity for specific oncotargets. This article proposes a streamlined approach for the development of ligand-oncotarget bioaffinity systems via integrated structural modeling and MD simulations, making use of proteomics, genomic, and X-ray crystallographic resources, to support targeted diagnosis and treatment of cancers and tumors.
Collapse
Affiliation(s)
- Jaison Jeevanandam
- CQM-Centro de Química da Madeira, MMRG, Universidade da Madeira, Campus da Penteada, 9020-105, Funchal, Portugal
| | - Godfred Sabbih
- Chemical Engineering Department, University of Tennessee, Chattanooga, TN, 37403, USA
| | - Kei X Tan
- School of Materials Science and Engineering, Nanyang Technological University, Singapore, 639798, Singapore
| | - Michael K Danquah
- Chemical Engineering Department, University of Tennessee, Chattanooga, TN, 37403, USA.
| |
Collapse
|
44
|
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: 19] [Impact Index Per Article: 4.8] [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
|
45
|
Abstract
The major applications for molecular imaging with PET in clinical practice concern cancer imaging. Undoubtedly, 18F-FDG represents the backbone of nuclear oncology as it remains so far the most widely employed positron emitter compound. The acquired knowledge on cancer features, however, allowed the recognition in the last decades of multiple metabolic or pathogenic pathways within the cancer cells, which stimulated the development of novel radiopharmaceuticals. An endless list of PET tracers, substantially covering all hallmarks of cancer, has entered clinical routine or is being investigated in diagnostic trials. Some of them guard significant clinical applications, whereas others mostly bear a huge potential. This chapter summarizes a selected list of non-FDG PET tracers, described based on their introduction into and impact on clinical practice.
Collapse
|
46
|
Eich C, Giessing M. [Radical nephrectomy and partial nephrectomy]. Aktuelle Urol 2020; 51:441-449. [PMID: 32722827 DOI: 10.1055/a-1190-3102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Renal cell carcinoma is the 2nd most frequent urological malignancy in women and the third most frequent in men, with an age peak in the seventh decade of life. If detected early in a local non-metastatic stage, options for complete recovery are excellent. While two decades ago, even locally limited cancers of the kidney were cured by radical nephrectomy, treatment today mostly consists of local treatment for locally confined cancers. Guidelines today recommend local surgical excision (open or minimally-invasive) or - in selected cases - topical energy application (radio-frequency ablation, cryoablation). The surgeon's expertise is most important in the selection of the appropriate kind of surgery and different guidelines have slightly different recommendations.Treatment decisions should be made on an individual basis in due consideration of an individual's age and co-morbidities. This may lead to the recommendation that, due to low perioperative morbidity, even localised carcinomas should be treated by (minimally-invasive) radical nephrectomy instead of nephron-sparing surgery and, in other cases, a non-interventional, active surveillance strategy may be pursued without compromising the patient's life expectancy. For higher-grade renal cell carcinomas, there is usually an indication for radical nephrectomy, as long as no metastases are detected. This also applies to carcinomas with venous thrombi extending into the atrium of the heart. Complications in the treatment of renal carcinomas are usually rare and easily treatable in most cases.
Collapse
Affiliation(s)
| | - Markus Giessing
- Universitätsklinikum Düsseldorf, Klinik für Urologie, Düsseldorf
| |
Collapse
|
47
|
Gao J, Xu Q, Fu Y, He K, Zhang C, Zhang Q, Shi J, Zhao X, Wang F, Guo H. Comprehensive evaluation of 68Ga-PSMA-11 PET/CT parameters for discriminating pathological characteristics in primary clear-cell renal cell carcinoma. Eur J Nucl Med Mol Imaging 2020; 48:561-569. [PMID: 32623502 DOI: 10.1007/s00259-020-04916-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 06/07/2020] [Indexed: 01/25/2023]
Abstract
PURPOSE To evaluate parameters derived from 68Ga-PSMA-11 PET/CT images for discriminating pathological characteristics in primary clear-cell renal cell carcinoma (ccRCC). METHODS The study retrospectively examined data for 36 ccRCC patients with preoperative 68Ga-PSMA-11 PET/CT scan and surgical specimens. Radiological parameters including maximal tumor diameter, mean CT value, and maximal standard uptake value (SUVmax) were derived from PET/CT images. Pathological characteristics included WHO/ISUP grade and adverse pathology (tumor necrosis or sarcomatoid or rhabdoid feature). Values of radiological parameters were compared within subgroups of pathological characteristics. Receiver operating characteristic (ROC) curve analysis was used for the effectiveness of radiological parameters in differentiating pathological characteristics, estimating area under the ROC curve (AUC) and 95% confidence intervals (CIs). RESULTS The WHO/ISUP grade distribution for 36 tumors was grade 1, 9 (25.0%); grade 2, 12 (33.3%); grade 3, 9 (25.0%); and grade 4, 6 (16.7%). Adverse pathology was positive for 15 (41.7%). Radiological tumor diameter and SUVmax significantly differed by WHO/ISUP grade, pT stage, and adverse pathology (all P < 0.05), with no difference by CT value. Tumor diameter demonstrated sensitivity 86% and specificity 88% for pT stage, with cutoff 6.70 and AUC 0.91 (95% CI, 0.79-1.00, P < 0.001). SUVmax could effectively differentiate WHO/ISUP grade (3-4 vs. 1-2) and adverse pathology (positive vs. negative), with AUC 0.89 (95% CI, 0.81-0.98, P < 0.001), cutoff 16.4, sensitivity 100%, and specificity 71% and AUC 0.92 (95% CI, 0.85-0.99, P < 0.001), cutoff 18.5, sensitivity 94%, and specificity 87%, respectively. CONCLUSION 68Ga-PSMA-11 PET/CT could effectively identify aggressive pathological features of ccRCC.
Collapse
Affiliation(s)
- Jie Gao
- Department of Urology, Nanjing Drum Tower Hospital, Institute of Urology Nanjing University, The Affiliated Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu Province, China
| | - Qinfeng Xu
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yao Fu
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Kuiqiang He
- Department of Urology, Nanjing Drum Tower Hospital, Institute of Urology Nanjing University, The Affiliated Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu Province, China
| | - Chengwei Zhang
- Department of Urology, Nanjing Drum Tower Hospital, Institute of Urology Nanjing University, The Affiliated Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu Province, China
| | - Qing Zhang
- Department of Urology, Nanjing Drum Tower Hospital, Institute of Urology Nanjing University, The Affiliated Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu Province, China
| | - Jiong Shi
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Xiaozhi Zhao
- Department of Urology, Nanjing Drum Tower Hospital, Institute of Urology Nanjing University, The Affiliated Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu Province, China.
| | - Feng Wang
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Hongqian Guo
- Department of Urology, Nanjing Drum Tower Hospital, Institute of Urology Nanjing University, The Affiliated Hospital of Nanjing University Medical School, No. 321 Zhongshan Road, Nanjing, 210008, Jiangsu Province, China.
| |
Collapse
|
48
|
Yao X, Zha Z, Ploessl K, Choi SR, Zhao R, Alexoff D, Zhu L, Kung HF. Synthesis and evaluation of novel radioiodinated PSMA targeting ligands for potential radiotherapy of prostate cancer. Bioorg Med Chem 2020; 28:115319. [PMID: 32001090 DOI: 10.1016/j.bmc.2020.115319] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/03/2020] [Accepted: 01/08/2020] [Indexed: 12/19/2022]
Abstract
Radioligand therapy (RLT) using prostate-specific membrane antigen (PSMA) targeting ligands is an attractive option for the treatment of Prostate cancer (PCa) and its metastases. We report herein a series of radioiodinated glutamate-urea-lysine-phenylalanine derivatives as new PSMA ligands in which l-tyrosine and l-glutamic acid moieties were added to increase hydrophilicity concomitant with improvement of in vivo targeting properties. Compounds 8, 15, 19a/19b and 23a/23b were synthesized and radiolabeled with 125I by iododestannylation. All iodinated compounds displayed high binding affinities toward PSMA (IC50 = 1-13 nM). In vitro cell uptake studies demonstrated that compounds containing an l-tyrosine linker moiety (8, 15 and 19a/19b) showed higher internalization than MIP-1095 and 23a/23b, both without the l-tyrosine linker moiety. Biodistribution studies in mice bearing PC3-PIP and PC3 xenografts showed that [125I]8 and [125I]15 with higher lipophilicity exhibited higher nonspecific accumulations in the liver and intestinal tract, whereas [125I]19a/19b and [125I]23a/23b containing additional glutamic acid moieties showed higher accumulations in the kidney and implanted PC3-PIP (PSMA+) tumors. [125I]23b displayed a promising biodistribution profile with favorable tumor retention, fast clearance from the kidney, and 2-3-fold lower uptake in the liver and blood than that observed for [125I]MIP-1095. [125/131I]23b may serve as an optimal PSMA ligand for radiotherapy treatment of prostate cancer over-expressing PSMA.
Collapse
Affiliation(s)
- Xinyue Yao
- College of Chemistry, Beijing Normal University, Beijing 100875, PR China; Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Zhihao Zha
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Karl Ploessl
- Five Eleven Pharma Inc., Philadelphia, PA 19104, USA
| | - Seok Rye Choi
- Five Eleven Pharma Inc., Philadelphia, PA 19104, USA
| | - Ruiyue Zhao
- College of Chemistry, Beijing Normal University, Beijing 100875, PR China; Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David Alexoff
- Five Eleven Pharma Inc., Philadelphia, PA 19104, USA
| | - Lin Zhu
- College of Chemistry, Beijing Normal University, Beijing 100875, PR China.
| | - Hank F Kung
- Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA; Five Eleven Pharma Inc., Philadelphia, PA 19104, USA.
| |
Collapse
|
49
|
Toriihara A, Nobashi T, Baratto L, Duan H, Moradi F, Park S, Hatami N, Aparici CM, Davidzon G, Iagaru A. Comparison of 3 Interpretation Criteria for 68Ga-PSMA11 PET Based on Inter- and Intrareader Agreement. J Nucl Med 2019; 61:533-539. [PMID: 31562226 DOI: 10.2967/jnumed.119.232504] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 08/27/2019] [Indexed: 11/16/2022] Open
Abstract
PET using radiolabeled prostate-specific membrane antigen (PSMA) is now being more widely adopted as a valuable tool to evaluate patients with prostate cancer (PC). Recently, 3 different criteria for interpretation of PSMA PET were published: the European Association of Nuclear Medicine (EANM) criteria, the Prostate Cancer Molecular Imaging Standardized Evaluation criteria, and the PSMA Reporting and Data System. We compared these 3 criteria in terms of interreader, intrareader, and intercriteria agreement. Methods: Data from 104 patients prospectively enrolled in research protocols at our institution were retrospectively reviewed. The cohort consisted of 2 groups: 47 patients (mean age, 64.2 y old) who underwent Glu-NH-CO-NH-Lys-(Ahx)-[68Ga(HBED-CC)] (68Ga-PSMA11) PET/MRI for initial staging of biopsy-proven intermediate- or high-risk PC, and 57 patients (mean age, 70.5 y old) who underwent 68Ga-PSMA11 PET/CT because of biochemically recurrent PC. Three nuclear medicine physicians independently evaluated all 68Ga-PSMA11 PET/MRI and PET/CT studies according to the 3 interpretation criteria. Two of them reevaluated all studies 6 mo later in the same manner and masked to the initial reading. The Gwet agreement coefficient was calculated to evaluate interreader, intrareader, and intercriteria agreement based on the following sites: local lesion (primary tumor or prostate bed after radical prostatectomy), lymph node metastases, and other metastases. Results: In the PET/MRI group, interreader, intrareader, and intercriteria agreement ranged from substantial to almost perfect for any site according to all 3 criteria. In the PET/CT group, interreader agreement ranged from substantial to almost perfect except for judgment of distant metastases based on the PSMA Reporting and Data System (Gwet agreement coefficient, 0.57; moderate agreement), in which the most frequent cause of disagreement was lung nodules. Intrareader agreement ranged from substantial to almost perfect for any site according to all 3 criteria. Intercriteria agreement for each site was also substantial to almost perfect. Conclusion: Although the 3 published criteria have good interreader and intrareader reproducibility in evaluating 68Ga-PSMA11 PET, there are some factors causing interreader disagreement. Further work is needed to address this issue.
Collapse
Affiliation(s)
- Akira Toriihara
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Tomomi Nobashi
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Lucia Baratto
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Heying Duan
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Farshad Moradi
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Sonya Park
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Negin Hatami
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Carina Mari Aparici
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Guido Davidzon
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| | - Andrei Iagaru
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, California
| |
Collapse
|
50
|
Ali S, Ahn T, Papa N, Perera M, Teloken P, Coughlin G, Wood ST, Roberts MJ. Changing trends in surgical management of renal tumours from 2000 to 2016: a nationwide study of Medicare claims data. ANZ J Surg 2019; 90:48-52. [PMID: 31478323 DOI: 10.1111/ans.15385] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 06/29/2019] [Accepted: 07/02/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Guidelines recommend nephron sparing surgery where possible for patients with T1 renal tumours. The trends of nephron sparing surgery outside the USA are limited, particularly since the introduction of robotic-assisted partial nephrectomy (RAPN). The aim of this study was to describe contemporary surgical management patterns of renal tumours in Australia according to Medicare claims data. METHODS Claims data according to the Medicare Benefits Schedule on surgical management of renal tumours in adult Australians between January 2000 and December 2016 was collated. Analysis of absolute number, population-adjusted rate and renal cancer-adjusted rate of interventions according to age and gender were performed, as well as proportion of RAPN. RESULTS Between 2000 and 2016, the rate of partial nephrectomy (PN) increased while radical nephrectomy (RN) remained stable (PN: 0.87-4.16, RN: 6.52-6.70 per 100 000 population). Since 2015, PN has become more common than RN in patients aged 25 to 44 years (0.98 versus 0.95 procedures per 100 000 population). Renal cancer-adjusted rate exhibited a trend towards increasing utilization of PN and reduced RN across all age groups. An increase in overall surgical treatment was observed (25%-41%), mainly due to increased treatment of patients older than 75 years. The proportion of RAPN was seen to rapidly increase (4.7% in 2010 to 58% in 2016). CONCLUSIONS Treatment utilization for renal masses has markedly changed in Australia according to Medicare claims. PN is increasingly replacing RN in younger patients, and older patients are receiving more surgical treatment. The impact of increased RAPN utilization is yet to be determined.
Collapse
Affiliation(s)
- Stephen Ali
- Department of Surgery, St Vincent's Hospital, Sydney, New South Wales, Australia.,Faculty of Medicine, The University of New South Wales, Sydney, New South Wales, Australia
| | - Thomas Ahn
- Department of Urology, Princess Alexandra Hospital, Brisbane, Queensland, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Nathan Papa
- Department of Surgery, Austin Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Marlon Perera
- Department of Urology, Princess Alexandra Hospital, Brisbane, Queensland, Australia.,Department of Surgery, Austin Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Patrick Teloken
- Department of Urology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Geoffrey Coughlin
- Department of Urology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Simon T Wood
- Department of Urology, Princess Alexandra Hospital, Brisbane, Queensland, Australia.,Centre for Kidney Disease Research, Translational Research Institute, Brisbane, Queensland, Australia
| | - Matthew J Roberts
- Department of Urology, Princess Alexandra Hospital, Brisbane, Queensland, Australia.,Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.,Nepean Urology Research Group, Nepean Hospital, Sydney, New South Wales, Australia
| |
Collapse
|