1
|
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
|
2
|
Li W, Hua B, Song S, Pan W, Yang Q, Xu B. From sutureless to standard: a comprehensive analysis of conversion rates in laparoscopic partial nephrectomy. BMC Urol 2024; 24:183. [PMID: 39198764 PMCID: PMC11351731 DOI: 10.1186/s12894-024-01578-6] [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: 04/02/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024] Open
Abstract
OBJECTIVE To assess the rate at which sutureless partial nephrectomy (SLPN) transitions to standard partial nephrectomy (SPN), focusing on preoperative factors that might prompt such conversions. PATIENTS AND METHODS In this retrospective study, we analyzed the efficacy of SLPN performed on adults at our institution from 2016 to 2023. The subjects were patients diagnosed with localized solid renal tumors. The primary technique employed was resection with scissors and argon beam coagulation for hemostasis, with suturing techniques used only when necessary. Predictive factors necessitating conversion to SPN were identified, and the associations among multiple variables were explored using various statistical analysis methods, including logistic regression, to identify key preoperative predictive factors. RESULTS Our institution performed 353 SLPN, with 21 cases (5.9%) necessitating conversion to SPN. The conversion rates for the Laparoscopic Partial Nephrectomy (LPN) subgroup and the Robotic-assist Partial Nephrectomy (RPN) subgroup were 7.9% (17/215) and 2.9% (4/138), respectively, nearing statistical significance (P = .066). Significant differences were observed between the conversion group and the no conversion group in terms of preoperative estimated Glomerular Filtration Rate (eGFR), age at surgery, tumor size, and exophytic/endophytic characteristics. Multivariate analysis identified age at surgery, preoperative eGFR, radiological tumor size, and tumor exophytic/endophytic nature as significant predictors for conversion to SPN. CONCLUSION This investigation highlights the efficacy and feasibility of SLPN while identifying critical factors influencing the necessity for conversion to SPN. The identified predictors, including younger surgical age, superior preoperative eGFR, and specific tumor characteristics, provide valuable insights for refining surgical strategies.
Collapse
Affiliation(s)
- Wenfeng Li
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Bao Hua
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Sangqing Song
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Weixin Pan
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Qing Yang
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Bin Xu
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
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
|
Rowe SP, Krueger S, Gorin MA, Fishman EK. Cinematic rendering of 18F-DCFPyL PET/CT fusion data in a patient with metastatic clear cell renal cell carcinoma. BJUI COMPASS 2024; 5:548-550. [PMID: 38873347 PMCID: PMC11168768 DOI: 10.1002/bco2.324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 12/12/2023] [Indexed: 06/15/2024] Open
Affiliation(s)
- Steven P. Rowe
- Department of RadiologyUniversity of North CarolinaChapel HillNorth CarolinaUSA
| | | | - Michael A. Gorin
- Milton and Carroll Petrie Department of UrologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Elliot K. Fishman
- The Russell H. Morgan Department of Radiology and Radiological ScienceJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
5
|
Bellin MF, Valente C, Bekdache O, Maxwell F, Balasa C, Savignac A, Meyrignac O. Update on Renal Cell Carcinoma Diagnosis with Novel Imaging Approaches. Cancers (Basel) 2024; 16:1926. [PMID: 38792005 PMCID: PMC11120239 DOI: 10.3390/cancers16101926] [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: 03/21/2024] [Revised: 05/06/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
This review highlights recent advances in renal cell carcinoma (RCC) imaging. It begins with dual-energy computed tomography (DECT), which has demonstrated a high diagnostic accuracy in the evaluation of renal masses. Several studies have suggested the potential benefits of iodine quantification, particularly for distinguishing low-attenuation, true enhancing solid masses from hyperdense cysts. By determining whether or not a renal mass is present, DECT could avoid the need for additional imaging studies, thereby reducing healthcare costs. DECT can also provide virtual unenhanced images, helping to reduce radiation exposure. The review then provides an update focusing on the advantages of multiparametric magnetic resonance (MR) imaging performance in the histological subtyping of RCC and in the differentiation of benign from malignant renal masses. A proposed standardized stepwise reading of images helps to identify clear cell RCC and papillary RCC with a high accuracy. Contrast-enhanced ultrasound may represent a promising diagnostic tool for the characterization of solid and cystic renal masses. Several combined pharmaceutical imaging strategies using both sestamibi and PSMA offer new opportunities in the diagnosis and staging of RCC, but their role in risk stratification needs to be evaluated. Although radiomics and tumor texture analysis are hampered by poor reproducibility and need standardization, they show promise in identifying new biomarkers for predicting tumor histology, clinical outcomes, overall survival, and the response to therapy. They have a wide range of potential applications but are still in the research phase. Artificial intelligence (AI) has shown encouraging results in tumor classification, grade, and prognosis. It is expected to play an important role in assessing the treatment response and advancing personalized medicine. The review then focuses on recently updated algorithms and guidelines. The Bosniak classification version 2019 incorporates MRI, precisely defines previously vague imaging terms, and allows a greater proportion of masses to be placed in lower-risk classes. Recent studies have reported an improved specificity of the higher-risk categories and better inter-reader agreement. The clear cell likelihood score, which adds standardization to the characterization of solid renal masses on MRI, has been validated in recent studies with high interobserver agreement. Finally, the review discusses the key imaging implications of the 2017 AUA guidelines for renal masses and localized renal cancer.
Collapse
Affiliation(s)
- Marie-France Bellin
- Service de Radiologie Diagnostique et Interventionnelle, Hôpital de Bicêtre AP-HP, 78 Rue du Général Leclerc, 94275 Le Kremlin-Bicêtre, France; (C.V.); (O.B.); (F.M.); (A.S.); (O.M.)
- Faculté de Médecine, University of Paris-Saclay, 63 Rue Gabriel Péri, 94276 Le Kremlin-Bicêtre, France
- BioMaps, UMR1281 INSERM, CEA, CNRS, University of Paris-Saclay, 94805 Villejuif, France
| | - Catarina Valente
- Service de Radiologie Diagnostique et Interventionnelle, Hôpital de Bicêtre AP-HP, 78 Rue du Général Leclerc, 94275 Le Kremlin-Bicêtre, France; (C.V.); (O.B.); (F.M.); (A.S.); (O.M.)
| | - Omar Bekdache
- Service de Radiologie Diagnostique et Interventionnelle, Hôpital de Bicêtre AP-HP, 78 Rue du Général Leclerc, 94275 Le Kremlin-Bicêtre, France; (C.V.); (O.B.); (F.M.); (A.S.); (O.M.)
| | - Florian Maxwell
- Service de Radiologie Diagnostique et Interventionnelle, Hôpital de Bicêtre AP-HP, 78 Rue du Général Leclerc, 94275 Le Kremlin-Bicêtre, France; (C.V.); (O.B.); (F.M.); (A.S.); (O.M.)
| | - Cristina Balasa
- Service de Radiologie Diagnostique et Interventionnelle, Hôpital de Bicêtre AP-HP, 78 Rue du Général Leclerc, 94275 Le Kremlin-Bicêtre, France; (C.V.); (O.B.); (F.M.); (A.S.); (O.M.)
| | - Alexia Savignac
- Service de Radiologie Diagnostique et Interventionnelle, Hôpital de Bicêtre AP-HP, 78 Rue du Général Leclerc, 94275 Le Kremlin-Bicêtre, France; (C.V.); (O.B.); (F.M.); (A.S.); (O.M.)
| | - Olivier Meyrignac
- Service de Radiologie Diagnostique et Interventionnelle, Hôpital de Bicêtre AP-HP, 78 Rue du Général Leclerc, 94275 Le Kremlin-Bicêtre, France; (C.V.); (O.B.); (F.M.); (A.S.); (O.M.)
- Faculté de Médecine, University of Paris-Saclay, 63 Rue Gabriel Péri, 94276 Le Kremlin-Bicêtre, France
- BioMaps, UMR1281 INSERM, CEA, CNRS, University of Paris-Saclay, 94805 Villejuif, France
| |
Collapse
|
6
|
Marvaso G, Jereczek-Fossa BA, Zaffaroni M, Vincini MG, Corrao G, Andratschke N, Balagamwala EH, Bedke J, Blanck O, Capitanio U, Correa RJM, De Meerleer G, Franzese C, Gaeta A, Gandini S, Garibaldi C, Gerszten PC, Gillessen S, Grubb WR, Guckenberger M, Hannan R, Jhaveri PM, Josipovic M, Kerkmeijer LGW, Lehrer EJ, Lindskog M, Louie AV, Nguyen QN, Ost P, Palma DA, Procopio G, Rossi M, Staehler M, Tree AC, Tsang YM, Van As N, Zaorsky NG, Zilli T, Pasquier D, Siva S. Delphi consensus on stereotactic ablative radiotherapy for oligometastatic and oligoprogressive renal cell carcinoma-a European Society for Radiotherapy and Oncology study endorsed by the European Association of Urology. Lancet Oncol 2024; 25:e193-e204. [PMID: 38697165 DOI: 10.1016/s1470-2045(24)00023-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/09/2024] [Accepted: 01/11/2024] [Indexed: 05/04/2024]
Abstract
The purpose of this European Society for Radiotherapy and Oncology (ESTRO) project, endorsed by the European Association of Urology, is to explore expert opinion on the management of patients with oligometastatic and oligoprogressive renal cell carcinoma by means of stereotactic ablative radiotherapy (SABR) on extracranial metastases, with the aim of developing consensus recommendations for patient selection, treatment doses, and concurrent systemic therapy. A questionnaire on SABR in oligometastatic renal cell carcinoma was prepared by a core group and reviewed by a panel of ten prominent experts in the field. The Delphi consensus methodology was applied, sending three rounds of questionnaires to clinicians identified as key opinion leaders in the field. At the end of the third round, participants were able to find consensus on eight of the 37 questions. Specifically, panellists agreed to apply no restrictions regarding age (25 [100%) of 25) and primary renal cell carcinoma histology (23 [92%] of 25) for SABR candidates, on the upper threshold of three lesions to offer ablative treatment in patients with oligoprogression, and on the concomitant administration of immune checkpoint inhibitor. SABR was indicated as the treatment modality of choice for renal cell carcinoma bone oligometatasis (20 [80%] of 25) and for adrenal oligometastases 22 (88%). No consensus or major agreement was reached regarding the appropriate schedule, but the majority of the poll (54%-58%) retained the every-other-day schedule as the optimal choice for all the investigated sites. The current ESTRO Delphi consensus might provide useful direction for the application of SABR in oligometastatic renal cell carcinoma and highlight the key areas of ongoing debate, perhaps directing future research efforts to close knowledge gaps.
Collapse
Affiliation(s)
- Giulia Marvaso
- Division of Radiation Oncology, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Barbara Alicja Jereczek-Fossa
- Division of Radiation Oncology, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Mattia Zaffaroni
- Division of Radiation Oncology, European Institute of Oncology, IRCCS, Milan, Italy.
| | - Maria Giulia Vincini
- Division of Radiation Oncology, European Institute of Oncology, IRCCS, Milan, Italy
| | - Giulia Corrao
- Division of Radiation Oncology, European Institute of Oncology, IRCCS, Milan, Italy
| | - Nicolaus Andratschke
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ehsan H Balagamwala
- Department of Radiation Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jens Bedke
- Department of Urology and Transplantation surgery, Klinikum Stuttgart, Stuttgart, Germany
| | - Oliver Blanck
- Department of Radiation Oncology, University Medical Center Schleswig Holstein, Kiel, Germany
| | - Umberto Capitanio
- IRCCS San Raffaele Scientific Institute, Milan, Italy; University Vita-Salute San Raffaele, Milan, Italy
| | - Rohann J M Correa
- Department of Radiation Oncology, London Health Sciences Centre, London, ON, Canada
| | - Gert De Meerleer
- Department of Radiation Oncology, Leuven University Hospitals, Leuven, Belgium
| | - Ciro Franzese
- Department of Radiotherapy and Radiosurgery, IRCCS Humanitas Research Hospital, Milan, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Aurora Gaeta
- Department of Experimental Oncology, European Institute of Oncology, IRCCS, Milan, Italy
| | - Sara Gandini
- Department of Experimental Oncology, European Institute of Oncology, IRCCS, Milan, Italy
| | - Cristina Garibaldi
- Unit of Radiation Research, European Institute of Oncology, IRCCS, Milan, Italy
| | - Peter C Gerszten
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Silke Gillessen
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Università della Svizzera Italiana, Lugano, Switzerland
| | - William R Grubb
- Department of Radiation Oncology, Augusta University Medical Center, Augusta, GA, USA
| | - Matthias Guckenberger
- Department of Radiation Oncology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Raquibul Hannan
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Pavan M Jhaveri
- Department of Radiation Oncology, Baylor College of Medicine, Houston, TX, USA
| | - Mirjana Josipovic
- Section of Radiotherapy, Department of Oncology, Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, Copenhagen University Hospital, Copenhagen, Denmark
| | - Linda G W Kerkmeijer
- Department of Radiation Oncology, Radboud University Medical Centre, Nijmegen, Netherlands
| | - Eric J Lehrer
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, USA
| | - Magnus Lindskog
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden; Department of Pelvic Cancer, Section of Genitourinary Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Alexander V Louie
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON, Canada
| | - Quynh-Nhu Nguyen
- Departments of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Piet Ost
- Department of Human Structure and Repair, Ghent University, Ghent, Belgium and Department of Radiation Oncology, Iridium Network, Antwerp, Belgium
| | - David A Palma
- Department of Radiation Oncology, London Health Sciences Centre, London, ON, Canada
| | - Giuseppe Procopio
- Dipartimento Di Oncologia Medica, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Maddalena Rossi
- Department of Radiation Oncology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Michael Staehler
- Interdisciplinary Centre on Renal Tumours, University of Munich, Munich, Germany
| | - Alison C Tree
- Department of Urology, The Institute of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, Sutton, Surrey, UK
| | - Yat Man Tsang
- Radiation Medicine Program, Princess Margaret Cancer Centre, Toronto, ON, Canada
| | - Nicholas Van As
- Department of Urology, The Institute of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, Sutton, Surrey, UK; The Institute of Cancer Research, London, UK
| | - Nicholas G Zaorsky
- University Hospitals Seidman Cancer Center and Case Western Reserve University, Cleveland, OH, USA
| | - Thomas Zilli
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Università della Svizzera Italiana, Lugano, Switzerland
| | - David Pasquier
- Academic Department of Radiation Oncology, Centre O Lambret, Lille, France; University of Lille, Centrale Lille, CNRS, UMR 9189-CRIStAL, Lille, France
| | - Shankar Siva
- Peter MacCallum Cancer Centre, Department of Radiation Oncology, University of Melbourne, Parkville, VIC, Australia; Faculty of Medicine, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
7
|
Oldan JD, Almaguel F, Voter AF, Duran A, Gafita A, Pomper MG, Hope TA, Rowe SP. PSMA-Targeted Radiopharmaceuticals for Prostate Cancer Diagnosis and Therapy. Cancer J 2024; 30:176-184. [PMID: 38753752 DOI: 10.1097/ppo.0000000000000718] [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: 05/18/2024]
Abstract
ABSTRACT Prostate cancer (PCa) is the most common noncutaneous malignancy in men. Until recent years, accurate imaging of men with newly diagnosed PCa, or recurrent or low-volume metastatic disease, was limited. Further, therapeutic options for men with advanced, metastatic, castration-resistant disease were increasingly limited as a result of increasing numbers of systemic therapies being combined in the upfront metastatic setting. The advent of urea-based, small-molecule inhibitors of prostate-specific membrane antigen (PSMA) has partially addressed those shortcomings in diagnosis and therapy of PCa. On the diagnostic side, there are multiple pivotal phase III trials with several different agents having demonstrated utility in the initial staging setting, with generally modest sensitivity but very high specificity for determining otherwise-occult pelvic nodal involvement. That latter statistic drives the utility of the scan by allowing imaging interpreters to read with very high sensitivity while maintaining a robust specificity. Other pivotal phase III trials have demonstrated high detection efficiency in patients with biochemical failure, with high positive predictive value at the lesion level, opening up possible new avenues of therapy such as metastasis-directed therapy. Beyond the diagnostic aspects of PSMA-targeted radiotracers, the same urea-based chemical scaffolds can be altered to deliver therapeutic isotopes to PCa cells that express PSMA. To date, one such agent, when combined with best standard-of-care therapy, has demonstrated an ability to improve overall survival, progression-free survival, and freedom from skeletal events relative to best standard-of-care therapy alone in men with metastatic, castration-resistant PCa who are post chemotherapy. Within the current milieu, there are a number of important future directions including the use of artificial intelligence to better leverage diagnostic findings, further medicinal chemistry refinements to the urea-based structure that may allow improved tumor targeting and decreased toxicities, and the incorporation of new radionuclides that may better balance efficacy with toxicities than those nuclides that are available.
Collapse
Affiliation(s)
- Jorge D Oldan
- From the Department of Radiology, University of North Carolina, Chapel Hill, NC
| | - Frankis Almaguel
- Department of Radiology, Loma Linda University School of Medicine, Loma Linda, CA
| | - Andrew F Voter
- The Russell H. Morgan Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Alfonso Duran
- Department of Radiology, Loma Linda University School of Medicine, Loma Linda, CA
| | - Andrei Gafita
- The Russell H. Morgan Department of Radiology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Martin G Pomper
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Thomas A Hope
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA
| | - Steven P Rowe
- From the Department of Radiology, University of North Carolina, Chapel Hill, NC
| |
Collapse
|
8
|
Olmstead T, Emmerling M, Bantumilli S, Raynor M, Nielsen ME, Bjurlin MA, Rose TL. Detection of Testicular Metastasis from Renal Cell Carcinoma on PSMA-PET Scan. J Kidney Cancer VHL 2024; 11:49-53. [PMID: 38464887 PMCID: PMC10923653 DOI: 10.15586/jkcvhl.v11i1.268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 01/15/2024] [Indexed: 03/12/2024] Open
Abstract
The use of prostate-specific membrane antigen-positron emission tomography (PSMA-PET) is becoming more widespread for the diagnosis and management of prostate cancer. Here we report a case of oligometastatic renal cell carcinoma (RCC) to the testes diagnosed incidentally on PSMA-PET imaging. This case demonstrates the potential for diagnosis of nonprostate disease with PSMA-PET imaging, as well as the promising nature of PSMA-PET for the diagnosis and surveillance of RCC. In addition, this case report discusses the rare occurrence of oligometastatic RCC to the testis.
Collapse
Affiliation(s)
| | - Michael Emmerling
- Department of Urology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Surekha Bantumilli
- Department of Pathology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Mathew Raynor
- Department of Urology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Matthew E. Nielsen
- Department of Urology, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Marc A. Bjurlin
- Department of Urology, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Tracy L. Rose
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Division of Oncology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
9
|
David C, Muhammad A, Cristian U, Ben T, Arun A, Lewis A, Lavinia S, Marlon P, Shankar S. SABR for oligometastatic renal cell carcinoma. Clin Transl Radiat Oncol 2024; 45:100739. [PMID: 38380117 PMCID: PMC10877104 DOI: 10.1016/j.ctro.2024.100739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/25/2024] [Accepted: 01/27/2024] [Indexed: 02/22/2024] Open
Abstract
Stereotactic ablative body radiotherapy (SABR) aims to accurately deliver a higher than conventional dose of radiotherapy to a well-defined target tumour incorporating advanced immobilisation and imaging techniques. SABR is an emerging treatment option for primary kidney cancer especially when surgery is contraindicated. Increasingly, SABR is being incorporated into the management of low-volume stage IV kidney cancers to delay the need for systemic therapy or to prolong the duration of ongoing systemic treatment. This review will evaluate the evidence and limitations of SABR for oligometastatic renal cell carcinoma.
Collapse
Affiliation(s)
- Chang David
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Ali Muhammad
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Udovicich Cristian
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Tran Ben
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Azad Arun
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Au Lewis
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Spain Lavinia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Perera Marlon
- Division of Surgical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Siva Shankar
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
10
|
Singhal T, Singh P, Parida GK, Agrawal K. Role of PSMA-targeted PET-CT in renal cell carcinoma: a systematic review and meta-analysis. Ann Nucl Med 2024; 38:176-187. [PMID: 38340144 DOI: 10.1007/s12149-024-01904-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/08/2024] [Indexed: 02/12/2024]
Abstract
Fluoro-deoxy glucose positron emission tomography/computed tomography (PET/CT), the workhorse of nuclear medicine, has limited utility for renal cell carcinoma (RCC), particularly clear cell variant. Thus, various other tracers have been tried for evaluation of RCC. One of the most promising targets for radiotracers is prostate-specific membrane antigen (PSMA) expressed in abundance in carcinoma-associated neo-vasculature. Thus, we tried to review and analyse the role of PSMA-targeted PET/CT in evaluation of RCC. Databases like PubMed, EMBASE and SCOPUS were searched for original studies published on PSMA-targeted PET/CT in RCC till 30 September 2023. Revised Tool for the Quality Assessment of Diagnostic Accuracy Studies-2 (QUADAS-2) checklist was used to assess the included studies. Pooled sensitivity and specificity were calculated and represented with 95% confidence intervals (95%CI). Heterogeneity in the studies was assessed by I-square index. Pooled sensitivity and specificity of PSMA-targeted PET/CT for detection of local disease estimates were 87.2% (95%CI: 77-94%) and 100% (95%CI: 92.9-100%), respectively. Pooled sensitivity and specificity for detection of local recurrent disease are 100% (95%CI: 71.5-100%) and 100% (95%CI: 89.4-100%), respectively. Pooled sensitivity and specificity for detection of metastatic disease are 92% (95%CI: 86.2-96%) and 96.9% (95%CI: 83.8-99.9%), respectively. Pooled sensitivity of PSMA-targeted PET/CT for detection of clear cell renal cell carcinoma (ccRCC) and non-ccRCC are 94.7% (95%CI: 88-98.3%) and 75% (95%CI: 35-96.8%), respectively. PSMA-targeted PET-CT demonstrated better diagnostic efficacy for the detection of recurrent RCC. Whilst for staging RCC, it had higher specificity but lower sensitivity. Thus, it can serve as a non-invasive adjuvant tool to conventional imaging in the evaluation of staging of RCC, particularly clear cell variant.
Collapse
Affiliation(s)
- Tejasvini Singhal
- Department of Nuclear Medicine, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Parneet Singh
- Department of Nuclear Medicine, All India Institute of Medical Sciences, Bhubaneswar, India
| | - Girish Kumar Parida
- Department of Nuclear Medicine, All India Institute of Medical Sciences, Bhubaneswar, India.
| | - Kanhaiyalal Agrawal
- Department of Nuclear Medicine, All India Institute of Medical Sciences, Bhubaneswar, India
| |
Collapse
|
11
|
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
|
12
|
Qi W, Guan W. A Comprehensive Review on the Importance of MiRNA-206 in the Animal Model and Human Diseases. Curr Neuropharmacol 2024; 22:1064-1079. [PMID: 37032500 PMCID: PMC10964108 DOI: 10.2174/1570159x21666230407124146] [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: 01/18/2023] [Revised: 02/08/2023] [Accepted: 02/15/2023] [Indexed: 04/11/2023] Open
Abstract
MicroRNA-206 (miR-206) is a microRNA that is involved in many human diseases, such as myasthenia gravis, osteoarthritis, depression, cancers, etc. Both inhibition effects and progression roles of miR-206 have been reported for the past few years. High expression of miR-206 was observed in patients with osteoarthritis, gastric cancer and epithelial ovarian cancer compared to normal people. The study also showed that miR-206 promotes cancer progression in breast cancer patients and avascular necrosis of the femoral head. Meanwhile, several studies have shown that expression levels of miR-206 were down-regulated in laryngeal carcinoma cell multiplication, as well as in hepatocellular carcinoma, non-small lung cancer and infantile hemangioma. Moreover, miR-206 was up-regulated in the mild stage of amyotrophic lateral sclerosis patients and then down-regulated in the moderate and severe stages, indicating that miR-206 has the double effects of starting and aggravating the disease. In neuropsychiatric disorders, such as depression, miR-206 also plays an important role in the progression of the disease; the level of miR-206 is most highly expressed in the brains of patients with depression. In the current review, we summarize the role of miR-206 in various diseases, and miR-206 may be developed as a new biomarker for diagnosing diseases in the near future.
Collapse
Affiliation(s)
- Wang Qi
- Department of Pharmacology, The First People's Hospital of Yancheng, Yancheng, 224000, Jiangsu, China
| | - Wei Guan
- Department of Pharmacology, Pharmacy College, Nantong University, Nantong, 226001, Jiangsu, China
- School of Medicine, Nantong University, Nantong, China
| |
Collapse
|
13
|
Urso L, Bauckneht M, Albano D, Chondrogiannis S, Grassetto G, Lanfranchi F, Dondi F, Fornarini G, Lazzeri M, Evangelista L. The evolution of PET imaging in renal, bladder, upper urinary tract urothelial, testicular and penile carcinoma - Today's impact, tomorrow's potential. Expert Rev Med Devices 2024; 21:55-72. [PMID: 38072680 DOI: 10.1080/17434440.2023.2293919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Accepted: 12/07/2023] [Indexed: 02/05/2024]
Abstract
INTRODUCTION The advancement of hybrid PET/CT or PET/MRI imaging for non-prostate genitourinary cancers has not experienced the rapid progress of prostate cancer. Nevertheless, these neoplasms are aggressive and reliable imaging plays a pivotal role in enhancing patients' quality of life and prognosis. AREAS COVERED the main evidence regarding [18F]FDG and non-[18F]FDG PET/CT or PET/MRI in non-prostate uro-oncological malignancies are summarized and discussed. Moreover, potential future directions concerning PET imaging in these neoplasms are debated, with the aim to stimulate future research projects covering these fields. EXPERT OPINION In Renal Cell Carcinoma (RCC), [18F]FDG PET/CT demonstrates varying efficacy in staging, restaging, and prognostic stratification, but PSMA PET/CT is emerging as a potential game-changer, particularly in advanced, high-grade aggressive clear cell RCC. [18F]FDG PET/CT may see an increased use in N and M-staging of bladder cancer, as well as for detecting recurrence and response to neoadjuvant chemotherapy. Preliminary data regarding [68Ga]-FAPI also looks promising in this context. [18F]FDG PET/MRI could be useful for the T-staging of bladder cancer, while upper tract urothelial carcinoma still lacks of molecular imaging literature reports. In testicular and penile cancer [18F]FDG PET/CT has demonstrated its usefulness in several clinical settings, although experiences with non-[18F]FDG radiotracers are lacking.
Collapse
Affiliation(s)
- Luca Urso
- Department of Nuclear Medicine - PET/CT Center, S. Maria Della Misericordia Hospital, Rovigo, Italy
| | - Matteo Bauckneht
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico, San Martino, Genova, Italy
- Department of Health Sciences (DISSAL), University of Genoa, Genova, Italy
| | - Domenico Albano
- Nuclear Medicine Department, ASST Spedali Civili of Brescia, University of Brescia, Brescia, Italy
| | - Sotirios Chondrogiannis
- Department of Nuclear Medicine - PET/CT Center, S. Maria Della Misericordia Hospital, Rovigo, Italy
| | - Gaia Grassetto
- Department of Nuclear Medicine - PET/CT Center, S. Maria Della Misericordia Hospital, Rovigo, Italy
| | - Francesco Lanfranchi
- Nuclear Medicine Unit, IRCCS Ospedale Policlinico, San Martino, Genova, Italy
- Department of Health Sciences (DISSAL), University of Genoa, Genova, Italy
| | - Francesco Dondi
- Nuclear Medicine Department, ASST Spedali Civili of Brescia, University of Brescia, Brescia, Italy
| | - Giuseppe Fornarini
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico, San Martino, Genova, Italy
| | - Massimo Lazzeri
- Department of Urology, IRCCS Humanitas Clinical and Research Hospital, Rozzano, Italy
| | - Laura Evangelista
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Italy
- Nuclear Medicine Unit, IRCCS Humanitas Research Hospital, Rozzano, Italy
| |
Collapse
|
14
|
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
|
15
|
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
|
16
|
Bui VN, Unterrainer LM, Brendel M, Kunte SC, Holzgreve A, Allmendinger F, Bartenstein P, Klauschen F, Unterrainer M, Staehler M, Ledderose S. PSMA-Expression Is Highly Associated with Histological Subtypes of Renal Cell Carcinoma: Potential Implications for Theranostic Approaches. Biomedicines 2023; 11:3095. [PMID: 38002095 PMCID: PMC10668989 DOI: 10.3390/biomedicines11113095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 10/13/2023] [Accepted: 10/19/2023] [Indexed: 11/26/2023] Open
Abstract
In renal cell carcinoma (RCC), accurate imaging methods are required for treatment planning and response assessment to therapy. In addition, there is an urgent need for new therapeutic options, especially in metastatic RCC. One way to combine diagnostics and therapy in a so-called theranostic approach is the use of radioligands directed against surface antigens. For instance, radioligands against prostate-specific membrane antigen (PSMA) have already been successfully used for diagnosis and radionuclide therapy of metastatic prostate cancer. Recent studies have demonstrated that PSMA is expressed not only in prostate cancer but also in the neovasculature of several solid tumors, which has raised hopes to use PSMA-guided theranostic approaches in other tumor entities, too. However, data on PSMA expression in different histopathological subtypes of RCC are sparse. Because a better understanding of PSMA expression in RCC is critical to assess which patients would benefit most from theranostic approaches using PSMA-targeted ligands, we investigated the expression pattern of PSMA in different subtypes of RCC on protein level. Immunohistochemical staining for PSMA was performed on formalin-fixed, paraffin-embedded archival material of major different histological subtypes of RCC (clear cell RCC (ccRCC)), papillary RCC (pRCC) and chromophobe RCC (cpRCC). The extent and intensity of PSMA staining were scored semi-quantitatively and correlated with the histological RCC subtypes. Group comparisons were calculated with the Kruskal-Wallis test. In all cases, immunoreactivity was detected only in the tumor-associated vessels and not in tumor cells. Staining intensity was the strongest in ccRCC, followed by cpRCC and pRCC. ccRCC showed the most diffuse staining pattern, followed by cpRCC and pRCC. Our results provide a rationale for PSMA-targeted theranostic approaches in ccRCC and cpRCC.
Collapse
Affiliation(s)
- Vinh Ngoc Bui
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (M.B.); (S.C.K.); (F.A.); (P.B.); (M.U.)
| | - Lena M. Unterrainer
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (M.B.); (S.C.K.); (F.A.); (P.B.); (M.U.)
| | - Matthias Brendel
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (M.B.); (S.C.K.); (F.A.); (P.B.); (M.U.)
- Munich Cluster for Systems Neurology (SyNergy), 81377 Munich, Germany
| | - Sophie C. Kunte
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (M.B.); (S.C.K.); (F.A.); (P.B.); (M.U.)
| | - Adrien Holzgreve
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (M.B.); (S.C.K.); (F.A.); (P.B.); (M.U.)
| | - Fabian Allmendinger
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (M.B.); (S.C.K.); (F.A.); (P.B.); (M.U.)
| | - Peter Bartenstein
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (M.B.); (S.C.K.); (F.A.); (P.B.); (M.U.)
| | | | - Marcus Unterrainer
- Department of Nuclear Medicine, LMU University Hospital, LMU Munich, 81377 Munich, Germany; (M.B.); (S.C.K.); (F.A.); (P.B.); (M.U.)
- Die RADIOLOGIE, 80331 Munich, Germany
| | - Michael Staehler
- Department of Urology, LMU University Hospital, LMU Munich, 81377 Munich, Germany;
| | - Stephan Ledderose
- Institute of Pathology, LMU Munich, 81377 Munich, Germany; (F.K.); (S.L.)
| |
Collapse
|
17
|
Chen SH, Lin BH, Chen SM, Qiu QRS, Ruan ZT, Chen ZJ, Wei Y, Zheng QS, Xue XY, Miao WB, Xu N. Head-to-head comparisons of enhanced CT, 68Ga-PSMA-11 PET/CT and 18F-FDG PET/CT in identifying adverse pathology of clear-cell renal cell carcinoma: a prospective study. Int Braz J Urol 2023; 49:716-731. [PMID: 37624658 PMCID: PMC10947621 DOI: 10.1590/s1677-5538.ibju.2023.0312] [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: 07/04/2023] [Accepted: 07/31/2023] [Indexed: 08/27/2023] Open
Abstract
OBJECTIVES Accurate preoperative prediction of adverse pathology is crucial for treatment planning of renal cell carcinoma (RCC). Previous studies have emphasized the potential of prostate-specific membrane antigen positron emission tomography / computed tomography (PSMA PET/CT) in differentiating between benign and malignant localized renal tumors. However, there is a scarcity of case reports elucidating the identification of aggressive pathological features using PET/CT. Our study was designed to prospectively compare the diagnostic value of enhanced CT, 68Ga-PSMA-11 and 18F-fluorodeoxyglucose (18F-FDG) PET/CT in clear-cell renal cell carcinoma (ccRCC) with necrosis or sarcomatoid or rhabdoid differentiation. MATERIALS AND METHODS A prospective case series of patients with a newly diagnosed renal mass who underwent enhanced CT, 68Ga-PSMA-11 and 18F-FDG PET/CT within 30 days prior to nephrectomy was included. Complete preoperative and postoperative clinicopathological data were recorded. Patients who received neoadjuvant targeted therapy, declined enhanced CT or PET/CT scanning, refused surgical treatment or had non-ccRCC pathological indications were excluded. Radiological parameters were compared within subgroups of pathological characteristics. Bonferroni corrections were used to adjust for multiple testing and statistical significance was set at a p-value less than 0.017. RESULTS Seventy-two patients were available for the final analysis. Enhanced CT demonstrated poor performance in identifying necrosis, sarcomatoid or rhabdoid differentiation and adverse pathology (all P > 0.05). The maximum standardized uptake value (SUVmax) of 68Ga-PSMA-11 PET/CT was more effective than 18F-FDG PET/CT in identifying tumor necrosis and adverse pathology, with an area under the curve (AUC) of 0.85 (cutoff value=25.26, p<0.001; Delong test z=2.709, p=0.007) for tumor necrosis and AUC of 0.90 (cutoff value=25.26, p<0.001; Delong test z=3.433, p<0.001) for adverse pathology. However, no significant statistical difference was found between 68Ga-PSMA-11 and 18F-FDG PET/CT in predicting sarcomatoid or rhabdoid feature (AUC of 0.91 vs.0.75, Delong test z=1.998, p=0.046). Subgroup analyses based on age, sex, tumor location, maximal diameter, stage and WHO/ISUP grade demonstrated that 68Ga-PSMA-11 PET/CT SUVmax had a significant predictive value for adverse pathology. Enhanced CT value and SUVmax demonstrated strong reliability [intraclass correlation coefficient (ICC) > 0.80], indicating a robust correlation. CONCLUSIONS 68Ga-PSMA-11 PET/CT demonstrates distinct advantages in identifying aggressive pathological features of primary ccRCC when compared to enhanced CT and 18F-FDG PET/CT. Further research and assessment are warranted to fully establish the clinical utility of 68Ga-PSMA-11 PET/CT in ccRCC.
Collapse
Affiliation(s)
- Shao-Hao Chen
- The First Affiliated Hospital of Fujian Medical UniversityDepartment of UrologyUrology Research InstituteFuzhouChinaDepartment of Urology, Urology Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Medical UniversityDepartment of UrologyNational Region Medical centerFuzhouChinaDepartment of Urology, National Region Medical center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Bo-Han Lin
- The First Affiliated Hospital of Fujian Medical UniversityDepartment of UrologyUrology Research InstituteFuzhouChinaDepartment of Urology, Urology Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Medical UniversityDepartment of UrologyNational Region Medical centerFuzhouChinaDepartment of Urology, National Region Medical center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Shao-Ming Chen
- The First Affiliated Hospital of Fujian Medical UniversityDepartment of Nuclear MedicineFuzhouChinaDepartment of Nuclear Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Qian-Ren-Shun Qiu
- The First Affiliated Hospital of Fujian Medical UniversityDepartment of UrologyUrology Research InstituteFuzhouChinaDepartment of Urology, Urology Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Medical UniversityDepartment of UrologyNational Region Medical centerFuzhouChinaDepartment of Urology, National Region Medical center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Zhong-Tian Ruan
- The First Affiliated Hospital of Fujian Medical UniversityDepartment of UrologyUrology Research InstituteFuzhouChinaDepartment of Urology, Urology Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Medical UniversityDepartment of UrologyNational Region Medical centerFuzhouChinaDepartment of Urology, National Region Medical center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Ze-Jia Chen
- The First Affiliated Hospital of Fujian Medical UniversityDepartment of UrologyUrology Research InstituteFuzhouChinaDepartment of Urology, Urology Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Medical UniversityDepartment of UrologyNational Region Medical centerFuzhouChinaDepartment of Urology, National Region Medical center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Yong Wei
- The First Affiliated Hospital of Fujian Medical UniversityDepartment of UrologyUrology Research InstituteFuzhouChinaDepartment of Urology, Urology Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Medical UniversityDepartment of UrologyNational Region Medical centerFuzhouChinaDepartment of Urology, National Region Medical center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Qing-Shui Zheng
- The First Affiliated Hospital of Fujian Medical UniversityDepartment of UrologyUrology Research InstituteFuzhouChinaDepartment of Urology, Urology Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Medical UniversityDepartment of UrologyNational Region Medical centerFuzhouChinaDepartment of Urology, National Region Medical center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xue-Yi Xue
- The First Affiliated Hospital of Fujian Medical UniversityDepartment of UrologyUrology Research InstituteFuzhouChinaDepartment of Urology, Urology Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Medical UniversityDepartment of UrologyNational Region Medical centerFuzhouChinaDepartment of Urology, National Region Medical center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- The First Affiliated Hospital of Fujian Medical UniversityFujian Key Laboratory of Precision Medicine for CancerFuzhouChinaFujian Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Wei-Bing Miao
- The First Affiliated Hospital of Fujian Medical UniversityDepartment of Nuclear MedicineFuzhouChinaDepartment of Nuclear Medicine, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- The First Affiliated Hospital of Fujian Medical UniversityFujian Key Laboratory of Precision Medicine for CancerFuzhouChinaFujian Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Ning Xu
- The First Affiliated Hospital of Fujian Medical UniversityDepartment of UrologyUrology Research InstituteFuzhouChinaDepartment of Urology, Urology Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Medical UniversityDepartment of UrologyNational Region Medical centerFuzhouChinaDepartment of Urology, National Region Medical center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
- The First Affiliated Hospital of Fujian Medical UniversityFujian Key Laboratory of Precision Medicine for CancerFuzhouChinaFujian Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
18
|
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
|
19
|
Giraudet AL, Vinceneux A, Pretet V, Paquet E, Lajusticia AS, Khayi F, Badel JN, Boyle H, Flechon A, Kryza D. Rationale for Prostate-Specific-Membrane-Antigen-Targeted Radionuclide Theranostic Applied to Metastatic Clear Cell Renal Carcinoma. Pharmaceuticals (Basel) 2023; 16:995. [PMID: 37513907 PMCID: PMC10383345 DOI: 10.3390/ph16070995] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/04/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Prostate-specific membrane antigen (PSMA), whose high expression has been demonstrated in metastatic aggressive prostate adenocarcinoma, is also highly expressed in the neovessels of various solid tumors, including clear cell renal cell carcinoma (ccRCC). In the VISION phase III clinical trial, PSMA-targeted radioligand therapy (PRLT) with lutetium 177 demonstrated a 4-month overall survival OS benefit compared to the best standard of care in heavily pretreated metastatic prostate cancer. Despite the improvement in the management of metastatic clear cell renal cell carcinoma (mccRCC) with antiangiogenic tyrosine kinase inhibitor (TKI) and immunotherapy, there is still a need for new treatments for patients who progress despite these drugs. In this study, we discuss the rationale of PRLT applied to the treavtment of mccRCC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - David Kryza
- Lumen Nuclear Medicine Department, Hospices Civils de Lyon, 69437 Lyon, France
- UNIV Lyon-Université Claude Bernard Lyon 1, LAGEPP UMR 5007 CNRS Villeurbanne, 69100 Villeurbanne, France
| |
Collapse
|
20
|
Gaudreault M, Chang D, Hardcastle N, McIntosh L, Jackson P, Kron T, Udovicich C, Hofman MS, Siva S. Feasibility of biology-guided radiotherapy for metastatic renal cell carcinoma driven by PSMA PET imaging. Clin Transl Radiat Oncol 2023; 40:100608. [PMID: 36942088 PMCID: PMC10023908 DOI: 10.1016/j.ctro.2023.100608] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/15/2023] [Accepted: 02/23/2023] [Indexed: 03/02/2023] Open
Abstract
Background Biology-guided radiotherapy (BgRT) is a novel treatment where the detection of positron emission originating from a volume called the biological tracking zone (BTZ) initiates dose delivery. Prostate-specific membrane antigen (PSMA) positron emission tomography (PET) is a novel imaging technique that may improve patient selection for metastasis-directed therapy in renal cell carcinoma (RCC). This study aims to determine the feasibility of BgRT treatment for RCC. Material and methods All consecutive patients that underwent PSMA PET/CT scan for RCC staging at our institution between 2014 and 2020 were retrospectively considered for inclusion. GTVs were contoured on the CT component of the PET/CT scan. The tumor-to-background ratio was quantified from the normalized standardized uptake value (nSUV), defined as the ratio between SUVmax inside the GTV and SUVmean inside the margin expansion. Tumors were classified suitable for BgRT if (1) nSUV was greater or equal to an nSUV threshold and (2) if the BTZ was free of any PET-avid region other than the tumor. Results Out of this cohort of 83 patients, 47 had metastatic RCC and were included in this study. In total, 136 tumors were delineated, 1 to 22 tumors per patient, mostly in lung (40%). Using a margin expansion of 5 mm/10 mm/20 mm and nSUV threshold = 3, 66%/63%/41% of tumors were suitable for BgRT treatment. Uptake originating from another tumor, the kidney, or the liver was typically inside the BTZ in tumors judged unsuitable for BgRT. Conclusions More than 60% of tumors were found to be suitable for BgRT in this cohort of patients with RCC. However, the proximity of PET-avid organs such as the liver or the kidney may affect BgRT delivery.
Collapse
Key Words
- Abdo, abdomen
- BTZ
- BTZ, biological tracking zone
- BgRT
- BgRT, biology-guided radiotherapy
- GTV, gross tumor volume
- LN&ST, lymph node and soft tissue
- MIP, maximum intensity projection
- PET, positron emission tomography
- PSMA
- PSMA, prostate specific membrane antigen
- RCC
- RCC, renal cell carcinoma
- SUV, standardized uptake value
- TPS, treatment planning system
- nSUV, normalized SUV
Collapse
Affiliation(s)
- Mathieu Gaudreault
- Department of Physical Sciences, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria 3010, Australia
- Corresponding author at: Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, 3000, Australia.
| | - David Chang
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria 3010, Australia
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Nicholas Hardcastle
- Department of Physical Sciences, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria 3010, Australia
- Centre for Medical Radiation Physics, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Lachlan McIntosh
- Department of Physical Sciences, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Price Jackson
- Department of Physical Sciences, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria 3010, Australia
| | - Tomas Kron
- Department of Physical Sciences, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria 3010, Australia
- Centre for Medical Radiation Physics, University of Wollongong, Wollongong, NSW 2522, Australia
| | - Cristian Udovicich
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria 3010, Australia
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Michael S. Hofman
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria 3010, Australia
- Molecular Imaging and Therapeutic Nuclear Medicine, Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| | - Shankar Siva
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Victoria 3010, Australia
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria 3000, Australia
| |
Collapse
|
21
|
Wang G, Li L, Wang J, Zang J, Chen J, Xiao Y, Fan X, Zhu L, Kung HF, Zhu Z. Head-to-head comparison of [ 68Ga]Ga-P16-093 and 2-[ 18F]FDG PET/CT in patients with clear cell renal cell carcinoma: a pilot study. Eur J Nucl Med Mol Imaging 2023; 50:1499-1509. [PMID: 36600099 DOI: 10.1007/s00259-022-06101-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/24/2022] [Indexed: 01/06/2023]
Abstract
PURPOSE This pilot study was prospectively designed to evaluate and compare the diagnostic value of PET/CT using a PSMA-specific tracer [68Ga]Ga-P16-093 and a glucose metabolism probe 2-[18F]FDG in clear cell renal cell carcinoma (ccRCC) patients. METHODS Forty-two pathologically confirmed ccRCC patients were included. Within 1 week, each patient underwent [68Ga]Ga-P16-093 and 2-[18F]FDG PET/CT. In addition to visual analysis of tumor number, the standardized uptake value (SUV) was measured for semiquantitative comparison and correlation analysis. RESULTS For primary ccRCC patients, [68Ga]Ga-P16-093 PET/CT demonstrated a significantly higher detection rate (19/22 vs. 13/22, P = 0.031) and higher tumor uptake (15.7 ± 9.0 vs. 5.1 ± 3.4, P < 0.001) than 2-[18F]FDG PET/CT. In addition, the SUVmax of the primary tumor on [68Ga]Ga-P16-093 and 2-[18F]FDG PET/CT was significantly correlated with pT stage (for [68Ga]Ga-P16-093, r = 0.550, P = 0.008; for 2-[18F]FDG, r = 0.514, P = 0.014) and WHO/ISUP grade (for [68Ga]Ga-P16-093, r = 0.566, P = 0.006; for 2-[18F]FDG, r = 0.492, P = 0.020), respectively. For metastatic ccRCC patients, [68Ga]Ga-P16-093 PET/CT also demonstrated a better detection rate (21/22 vs. 14/22, P = 0.008) and higher tumor uptake (11.0 ± 6.4 vs. 4.4 ± 2.7, P < 0.001) than 2-[18F]FDG PET/CT. The SUVmax on [68Ga]Ga-P16-093 PET/CT had a significant association with PSMA expression in primary ccRCC (r = 0.776, P < 0.001) and metastatic ccRCC (r = 0.626, P = 0.029). CONCLUSIONS [68Ga]Ga-P16-093 PET/CT demonstrates significantly better tumor detectability than 2-[18F]FDG PET/CT for ccRCC patients. TRIAL REGISTRATION 68Ga-P16-093 and 18F-FDG PET/CT Imaging in the Same Group of Clear Cell Renal Cell Carcinoma Patients (NCT05432947, Registered 27 June 2021, retrospectively registered) URL OF REGISTRY: https://clinicaltrials.gov/ct2/show/NCT05432947 .
Collapse
Affiliation(s)
- Guochang Wang
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100730, China
| | - Linlin Li
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100730, China
| | - Jiarou Wang
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100730, China
| | - Jie Zang
- Department of Nuclear Medicine, Fujian Provincial Key Laboratory of Precision Medicine for Cancer, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350005, China
| | - Jingci Chen
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yu Xiao
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xinrong Fan
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100730, China.
| | - Lin Zhu
- College of Chemistry, Key Laboratory of Radiopharmaceuticals, Ministry of Education, Beijing Normal University, Beijing, 100875, China.
| | - Hank F Kung
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Zhaohui Zhu
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
22
|
Posada Calderon L, Eismann L, Reese SW, Reznik E, Hakimi AA. Advances in Imaging-Based Biomarkers in Renal Cell Carcinoma: A Critical Analysis of the Current Literature. Cancers (Basel) 2023; 15:cancers15020354. [PMID: 36672304 PMCID: PMC9856305 DOI: 10.3390/cancers15020354] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/31/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Cross-sectional imaging is the standard diagnostic tool to determine underlying biology in renal masses, which is crucial for subsequent treatment. Currently, standard CT imaging is limited in its ability to differentiate benign from malignant disease. Therefore, various modalities have been investigated to identify imaging-based parameters to improve the noninvasive diagnosis of renal masses and renal cell carcinoma (RCC) subtypes. MRI was reported to predict grading of RCC and to identify RCC subtypes, and has been shown in a small cohort to predict the response to targeted therapy. Dynamic imaging is promising for the staging and diagnosis of RCC. PET/CT radiotracers, such as 18F-fluorodeoxyglucose (FDG), 124I-cG250, radiolabeled prostate-specific membrane antigen (PSMA), and 11C-acetate, have been reported to improve the identification of histology, grading, detection of metastasis, and assessment of response to systemic therapy, and to predict oncological outcomes. Moreover, 99Tc-sestamibi and SPECT scans have shown promising results in distinguishing low-grade RCC from benign lesions. Radiomics has been used to further characterize renal masses based on semantic and textural analyses. In preliminary studies, integrated machine learning algorithms using radiomics proved to be more accurate in distinguishing benign from malignant renal masses compared to radiologists' interpretations. Radiomics and radiogenomics are used to complement risk classification models to predict oncological outcomes. Imaging-based biomarkers hold strong potential in RCC, but require standardization and external validation before integration into clinical routines.
Collapse
Affiliation(s)
- Lina Posada Calderon
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Lennert Eismann
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Stephen W. Reese
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ed Reznik
- Computational Oncology, Department of Epidemiology & Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Abraham Ari Hakimi
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Correspondence:
| |
Collapse
|
23
|
Rizzo A, Racca M, Dall’Armellina S, Rescigno P, Banna GL, Albano D, Dondi F, Bertagna F, Annunziata S, Treglia G. The Emerging Role of PET/CT with PSMA-Targeting Radiopharmaceuticals in Clear Cell Renal Cancer: An Updated Systematic Review. Cancers (Basel) 2023; 15:355. [PMID: 36672305 PMCID: PMC9857064 DOI: 10.3390/cancers15020355] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 12/30/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Recent articles proposed the employment of positron emission tomography/computed tomography (PET/CT) with prostate-specific membrane antigen (PSMA)-targeting radiopharmaceuticals in clear cell renal cell carcinoma (ccRCC). METHODS The authors performed a comprehensive literature search of studies on the performance of PET/CT with PSMA-targeting radiopharmaceuticals in ccRCC. Original articles concerning this imaging examination were included in newly diagnosed ccRCC patients and ccRCC patients with disease recurrence. RESULTS A total of sixteen papers concerning the diagnostic performance of PSMA-targeted PET/CT in ccRCC (331 patients) were included in this systematic review. The included articles demonstrated an excellent detection rate of PSMA-targeting PET/CT in ccRCC. CONCLUSIONS PSMA-targeted PET/CT seems promising in detecting ccRCC lesions as well as in discriminating the presence of aggressive phenotypes. Prospective multicentric studies are warranted to strengthen the role of PSMA-targeting PET/CT in ccRCC.
Collapse
Affiliation(s)
- Alessio Rizzo
- Department of Nuclear Medicine, Candiolo Cancer Institute, FPO–IRCCS, 10060 Turin, Italy
| | - Manuela Racca
- Department of Nuclear Medicine, Candiolo Cancer Institute, FPO–IRCCS, 10060 Turin, Italy
| | - Sara Dall’Armellina
- Nuclear Medicine Unit, Department of Medical Sciences, AOU Città della Salute e della Scienza, University of Turin, 10126 Turin, Italy
| | - Pasquale Rescigno
- Department of Oncology, Candiolo Cancer Institute, FPO-IRCCS, 10060 Turin, Italy
| | | | - Domenico Albano
- Division of Nuclear Medicine, Università degli Studi di Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Francesco Dondi
- Division of Nuclear Medicine, Università degli Studi di Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Francesco Bertagna
- Division of Nuclear Medicine, Università degli Studi di Brescia and ASST Spedali Civili di Brescia, 25123 Brescia, Italy
| | - Salvatore Annunziata
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Giorgio Treglia
- Clinic of Nuclear Medicine, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, 6501 Bellinzona, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| |
Collapse
|
24
|
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
|
25
|
Rizzo A, Dall’Armellina S, Pizzuto DA, Perotti G, Zagaria L, Lanni V, Treglia G, Racca M, Annunziata S. PSMA Radioligand Uptake as a Biomarker of Neoangiogenesis in Solid Tumours: Diagnostic or Theragnostic Factor? Cancers (Basel) 2022; 14:4039. [PMID: 36011032 PMCID: PMC9406909 DOI: 10.3390/cancers14164039] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/10/2022] [Accepted: 08/17/2022] [Indexed: 01/10/2023] Open
Abstract
Due to its overexpression on the surface of prostate cancer cells, prostate-specific membrane antigen (PSMA) is a relatively novel effective target for molecular imaging and radioligand therapy (RLT) in prostate cancer. Recent studies reported that PSMA is expressed in the neovasculature of various types of cancer and regulates tumour cell invasion as well as tumour angiogenesis. Several authors explored the role of diagnostic and therapeutic PSMA radioligands in various malignancies. In this narrative review, we describe the current status of the literature on PSMA radioligands' application in solid tumours other than prostate cancer to explore their potential role as diagnostic or therapeutic agents, with particular regard to the relevance of PSMA radioligand uptake as neoangiogenetic biomarker. Hence, a comprehensive review of the literature was performed to find relevant articles on the applications of PSMA radioligands in non-prostate solid tumours. Data on the general, methodological and clinical aspects of all included studies were collected. Forty full-text papers were selected for final review, 8 of which explored PSMA radioligand PET/CT performances in gliomas, 3 in salivary gland malignancies, 6 in thyroid cancer, 2 in breast cancer, 16 in renal cell carcinoma and 5 in hepatocellular carcinoma. In the included studies, PSMA radioligand PET showed promising performance in patients with non-prostate solid tumours. Further studies are needed to better define its potential role in oncological patients management, especially in those undergoing antineoangiogenic therapies, and to assess the efficacy of PSMA-RLT in this clinical context.
Collapse
Affiliation(s)
- Alessio Rizzo
- Department of Nuclear Medicine, Candiolo Cancer Institute, FPO—IRCCS, 10060 Turin, Italy
| | - Sara Dall’Armellina
- Nuclear Medicine Unit, Department of Medical Sciences, AOU Città della Salute e della Scienza, University of Turin, 10134 Turin, Italy
| | - Daniele Antonio Pizzuto
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Germano Perotti
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Luca Zagaria
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Valerio Lanni
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Giorgio Treglia
- Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, 6501 Bellinzona, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, 1011 Lausanne, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, 6900 Lugano, Switzerland
| | - Manuela Racca
- Department of Nuclear Medicine, Candiolo Cancer Institute, FPO—IRCCS, 10060 Turin, Italy
| | - Salvatore Annunziata
- Unità di Medicina Nucleare, TracerGLab, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| |
Collapse
|
26
|
Boinapally S, Lisok A, Lofland G, Minn I, Yan Y, Jiang Z, Shin MJ, Merino VF, Zheng L, Brayton C, Pomper MG, Banerjee SR. Hetero-bivalent agents targeting FAP and PSMA. Eur J Nucl Med Mol Imaging 2022; 49:4369-4381. [PMID: 35965291 DOI: 10.1007/s00259-022-05933-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 08/01/2022] [Indexed: 11/29/2022]
Abstract
PURPOSE We developed a theranostic radiopharmaceutical that engages two key cell surface proteases, fibroblast activation protein alpha (FAP) and prostate-specific membrane antigen (PSMA), each frequently overexpressed within the tumor microenvironment (TME). The latter is also expressed in most prostate tumor epithelium. To engage a broader spectrum of cancers for imaging and therapy, we conjugated small-molecule FAP and PSMA-targeting moieties using an optimized linker to provide 64Cu-labeled compounds. METHODS We synthesized FP-L1 and FP-L2 using two linker constructs attaching the FAP and PSMA-binding pharmacophores. We determined in vitro inhibition constants (Ki) for FAP and PSMA. Cell uptake assays and flow cytometry were conducted in human glioma (U87), melanoma (SK-MEL-24), prostate cancer (PSMA + PC3 PIP and PSMA - PC3 flu), and clear cell renal cell carcinoma lines (PSMA + /PSMA - 786-O). Quantitative positron emission tomography/computed tomography (PET/CT) and tissue biodistribution studies were performed using U87, SK-MEL-24, PSMA + PC3 PIP, and PSMA + 786-O experimental xenograft models and the KPC genetically engineered mouse model of pancreatic cancer. RESULTS 64Cu-FP-L1 and 64Cu-FP-L2 were produced in high radiochemical yields (> 98%) and molar activities (> 19 MBq/nmol). Ki values were in the nanomolar range for both FAP and PSMA. PET imaging and biodistribution studies revealed high and specific targeting of 64Cu-FP-L1 and 64Cu-FP-L2 for FAP and PSMA. 64Cu-FP-L1 displayed more favorable pharmacokinetics than 64Cu-FP-L2. In the U87 tumor model at 2 h post-injection, tumor uptake of 64Cu-FP-L1 (10.83 ± 1.02%ID/g) was comparable to 64Cu-FAPI-04 (9.53 ± 2.55%ID/g). 64Cu-FP-L1 demonstrated high retention 5.34 ± 0.29%ID/g at 48 h in U87 tumor. Additionally, 64Cu-FP-L1 showed high retention in PSMA + PC3 PIP tumor (12.06 ± 0.78%ID/g at 2 h and 10.51 ± 1.82%ID/g at 24 h). CONCLUSIONS 64Cu-FP-L1 demonstrated high and specific tumor targeting of FAP and PSMA. This compound should enable imaging of lesions expressing FAP, PSMA, or both on the tumor cell surface or within the TME. FP-L1 can readily be converted into a theranostic for the management of heterogeneous tumors.
Collapse
Affiliation(s)
- Srikanth Boinapally
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, USA
| | - Alla Lisok
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, USA
| | - Gabriela Lofland
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, USA
| | - Il Minn
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, USA
| | - Yu Yan
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, USA
| | - Zirui Jiang
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, USA
| | - Min Jay Shin
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, USA
| | - Vanessa F Merino
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, USA
| | - Lei Zheng
- Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Cory Brayton
- Department of Molecular and Comparative Pathobiology, Baltimore, MD, USA
| | - Martin G Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, USA. .,Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| | - Sangeeta Ray Banerjee
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD, USA. .,Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA.
| |
Collapse
|
27
|
Rowe SP, Buck A, Bundschuh RA, Lapa C, Serfling SE, Derlin T, Higuchi T, Gorin MA, Pomper MG, Werner RA. [18F]DCFPyL PET/CT for Imaging of Prostate Cancer. Nuklearmedizin 2022; 61:240-246. [PMID: 35030637 DOI: 10.1055/a-1659-0010] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Prostate-specific membrane antigen (PSMA)-directed positron emission tomography (PET) has gained increasing interest for imaging of men affected by prostate cancer (PC). In recent years, 68Ga-labeled PSMA compounds have been widely utilized, although there is a trend towards increased utilization of 18F-labeled agents. Among others, [18F]DCFPyL (piflufolastat F 18, PYLARIFY) has been tested in multiple major trials, such as OSPREY and CONDOR, which provided robust evidence on the clinical utility of this compound for staging, restaging, and change in management. Recent explorative prospective trials have also utilized [18F]DCFPyL PET/CT for response assessment, e.g., in patients under abiraterone or enzalutamide, rendering this 18F-labeled PSMA radiotracer as an attractive biomarker for image-guided strategies in men with PC. After recent approval by the U.S. Food and Drug Administration, one may expect more widespread use, not only in the U.S., but also in Europe in the long term. In the present review, we will provide an overview of the current clinical utility of [18F]DCFPyL in various clinical settings for men with PC.
Collapse
Affiliation(s)
- Steven P Rowe
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, United States.,Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, United States.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Andreas Buck
- Nuclear Medicine, Würzburg University Medical Center Clinic for Nuclear Medicine, Würzburg, Germany
| | - Ralph A Bundschuh
- Department of Nuclear Medicine, University Hospital Bonn, Bonn, Germany
| | | | - Sebastian E Serfling
- Nuclear Medicine, Würzburg University Medical Center Clinic for Nuclear Medicine, Würzburg, Germany
| | - Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Takahiro Higuchi
- Nuclear Medicine, Würzburg University Medical Center Clinic for Nuclear Medicine, Würzburg, Germany.,Dentistry and Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Okayama, Japan
| | - Michael A Gorin
- Urology Associates and UPMC Western Maryland, Cumberland, United States.,Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, United States
| | - Martin G Pomper
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, United States.,Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, United States.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, United States
| | - Rudolf A Werner
- Nuclear Medicine, Würzburg University Medical Center Clinic for Nuclear Medicine, Würzburg, Germany
| |
Collapse
|
28
|
Voter AF, Werner RA, Pienta KJ, Gorin MA, Pomper MG, Solnes LB, Rowe SP. Piflufolastat F-18 ( 18F-DCFPyL) for PSMA PET imaging in prostate cancer. Expert Rev Anticancer Ther 2022; 22:681-694. [DOI: 10.1080/14737140.2022.2081155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Andrew F. Voter
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Transitional Year Residency Program, Aurora St. Luke’s Medical Center, Advocate Aurora Health, Milwaukee, WI, USA
| | - Rudolf A. Werner
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Kenneth J. Pienta
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael A. Gorin
- Urology Associates and UPMC Western Maryland, Cumberland, MD, USA
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Martin G. Pomper
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lilja B. Solnes
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Steven P. Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
29
|
Roussel E, Capitanio U, Kutikov A, Oosterwijk E, Pedrosa I, Rowe SP, Gorin MA. Novel Imaging Methods for Renal Mass Characterization: A Collaborative Review. Eur Urol 2022; 81:476-488. [PMID: 35216855 PMCID: PMC9844544 DOI: 10.1016/j.eururo.2022.01.040] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 01/08/2022] [Accepted: 01/21/2022] [Indexed: 01/19/2023]
Abstract
CONTEXT The incidental detection of localized renal masses has been rising steadily, but a significant proportion of these tumors are benign or indolent and, in most cases, do not require treatment. At the present time, a majority of patients with an incidentally detected renal tumor undergo treatment for the presumption of cancer, leading to a significant number of unnecessary surgical interventions that can result in complications including loss of renal function. Thus, there exists a clinical need for improved tools to aid in the pretreatment characterization of renal tumors to inform patient management. OBJECTIVE To systematically review the evidence on noninvasive, imaging-based tools for solid renal mass characterization. EVIDENCE ACQUISITION The MEDLINE database was systematically searched for relevant studies on novel imaging techniques and interpretative tools for the characterization of solid renal masses, published in the past 10 yr. EVIDENCE SYNTHESIS Over the past decade, several novel imaging tools have offered promise for the improved characterization of indeterminate renal masses. Technologies of particular note include multiparametric magnetic resonance imaging of the kidney, molecular imaging with targeted radiopharmaceutical agents, and use of radiomics as well as artificial intelligence to enhance the interpretation of imaging studies. Among these, 99mTc-sestamibi single photon emission computed tomography/computed tomography (CT) for the identification of benign renal oncocytomas and hybrid oncocytic chromophobe tumors, and positron emission tomography/CT imaging with radiolabeled girentuximab for the identification of clear cell renal cell carcinoma, are likely to be closest to implementation in clinical practice. CONCLUSIONS A number of novel imaging tools stand poised to aid in the noninvasive characterization of indeterminate renal masses. In the future, these tools may aid in patient management by providing a comprehensive virtual biopsy, complete with information on tumor histology, underlying molecular abnormalities, and ultimately disease prognosis. PATIENT SUMMARY Not all renal tumors require treatment, as a significant proportion are either benign or have limited metastatic potential. Several innovative imaging tools have shown promise for their ability to improve the characterization of renal tumors and provide guidance in terms of patient management.
Collapse
Affiliation(s)
- Eduard Roussel
- Department of Urology, University Hospitals Leuven, Leuven, Belgium
| | - Umberto Capitanio
- Department of Urology, University Vita-Salute, San Raffaele Scientific Institute, Milan, Italy; Division of Experimental Oncology, URI, Urological Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alexander Kutikov
- Division of Urology, Department of Surgery, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA, USA
| | - Egbert Oosterwijk
- Department of Urology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, The Netherlands
| | - Ivan Pedrosa
- Department of Radiology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Advanced Imaging Research Center. University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Urology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Steven P Rowe
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA; The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Michael A Gorin
- Urology Associates and UPMC Western Maryland, Cumberland, MD, USA; Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
30
|
Muselaers S, Erdem S, Bertolo R, Ingels A, Kara Ö, Pavan N, Roussel E, Pecoraro A, Marchioni M, Carbonara U, Marandino L, Amparore D, Campi R. PSMA PET/CT in Renal Cell Carcinoma: An Overview of Current Literature. J Clin Med 2022; 11:jcm11071829. [PMID: 35407435 PMCID: PMC8999609 DOI: 10.3390/jcm11071829] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 03/23/2022] [Indexed: 11/23/2022] Open
Abstract
Although the vast majority of prostate-specific membrane antigen (PSMA) positron emission tomography (PET) imaging occurs in the field of prostate cancer, PSMA is also highly expressed on the cell surface of the microvasculature of several other solid tumors, including renal cell carcinoma (RCC). This makes it a potentially interesting imaging target for the staging and monitoring of RCC. The objective of this review is to provide an overview of the current evidence regarding the use of PSMA PET/Computed Tomography in RCC patients.
Collapse
Affiliation(s)
- Stijn Muselaers
- Department of Urology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
- Correspondence: ; Tel.: +31-243-613-735
| | - Selcuk Erdem
- Division of Urologic Oncology, Department of Urology, Faculty of Medicine, Istanbul University, Istanbul 34452, Turkey;
| | - Riccardo Bertolo
- Department of Urology, San Carlo Di Nancy Hospital, 00165 Rome, Italy;
| | - Alexandre Ingels
- Department of Urology, University Hospital Henri Mondor, APHP, 94000 Créteil, France;
| | - Önder Kara
- Department of Urology, School of Medicine, Kocaeli University, İzmit 41001, Turkey;
| | - Nicola Pavan
- Urology Clinic, Department of Medical, Surgical and Health Science, University of Trieste, 34127 Trieste, Italy;
| | - Eduard Roussel
- Department of Urology, University Hospitals Leuven, 3000 Leuven, Belgium;
| | - Angela Pecoraro
- Department of Urology, San Luigi Gonzaga Hospital, University of Turin, Orbassano, 10124 Turin, Italy; (A.P.); (D.A.)
| | - Michele Marchioni
- Department of Medical, Oral and Biotechnological Sciences, G. d’Annunzio University of Chieti, 66100 Chieti, Italy;
| | - Umberto Carbonara
- Department of Emergency and Organ Transplantation-Urology, Andrology and Kidney Transplantation, University of Bari, 70121 Bari, Italy;
| | - Laura Marandino
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, 20132 Milan, Italy;
| | - Daniele Amparore
- Department of Urology, San Luigi Gonzaga Hospital, University of Turin, Orbassano, 10124 Turin, Italy; (A.P.); (D.A.)
| | - Riccardo Campi
- Unit of Urological Robotic Surgery and Renal Transplantation, Careggi Hospital, University of Florence, 50121 Florence, Italy;
| | | |
Collapse
|
31
|
Chen Y, Minn I, Rowe SP, Lisok A, Chatterjee S, Brummet M, Banerjee SR, Mease RC, Pomper MG. A Series of PSMA-Targeted Near-Infrared Fluorescent Imaging Agents. Biomolecules 2022; 12:biom12030405. [PMID: 35327597 PMCID: PMC8946146 DOI: 10.3390/biom12030405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/18/2022] [Accepted: 02/23/2022] [Indexed: 02/01/2023] Open
Abstract
We have synthesized a series of 10 new, PSMA-targeted, near-infrared imaging agents intended for use in vivo for fluorescence-guided surgery (FGS). Compounds were synthesized from the commercially available amine-reactive active NHS ester of DyLight800. We altered the linker between the PSMA-targeting urea moiety and the fluorophore with a view to improve the pharmacokinetics. Chemical yields for the conjugates ranged from 51% to 86%. The Ki values ranged from 0.10 to 2.19 nM. Inclusion of an N-bromobenzyl substituent at the ε-amino group of lysine enhanced PSMA+ PIP tumor uptake, as did hydrophilic substituents within the linker. The presence of a polyethylene glycol chain within the linker markedly decreased renal uptake. In particular, DyLight800-10 demonstrated high specific uptake relative to background signal within kidney, confirmed by immunohistochemistry. These compounds may be useful for FGS in prostate, renal or other PSMA-expressing cancers.
Collapse
|
32
|
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
|
33
|
DePalatis L, Martiniova L, de Almeida Graff T, Ravizzini G. Applications of PSMA-PET in tumors other than prostate cancer. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00116-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
34
|
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
|
35
|
Klinkhammer BM, Lammers T, Mottaghy FM, Kiessling F, Floege J, Boor P. Non-invasive molecular imaging of kidney diseases. Nat Rev Nephrol 2021; 17:688-703. [PMID: 34188207 PMCID: PMC7612034 DOI: 10.1038/s41581-021-00440-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2021] [Indexed: 02/05/2023]
Abstract
In nephrology, differential diagnosis or assessment of disease activity largely relies on the analysis of glomerular filtration rate, urinary sediment, proteinuria and tissue obtained through invasive kidney biopsies. However, currently available non-invasive functional parameters, and most serum and urine biomarkers, cannot capture intrarenal molecular disease processes specifically. Moreover, although histopathological analyses of kidney biopsy samples enable the visualization of pathological morphological and molecular alterations, they only provide information about a small part of the kidney and do not allow longitudinal monitoring. These limitations not only hinder understanding of the dynamics of specific disease processes in the kidney, but also limit the targeting of treatments to active phases of disease and the development of novel targeted therapies. Molecular imaging enables non-invasive and quantitative assessment of physiological or pathological processes by combining imaging technologies with specific molecular probes. Here, we discuss current preclinical and clinical molecular imaging approaches in nephrology. Non-invasive visualization of the kidneys through molecular imaging can be used to detect and longitudinally monitor disease activity and can therefore provide companion diagnostics to guide clinical trials, as well as the safe and effective use of drugs.
Collapse
Affiliation(s)
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany
- Department of Pharmaceutics, Utrecht University, Utrecht, Netherlands
- Department of Targeted Therapeutics, University of Twente, Enschede, Netherlands
| | - Felix M Mottaghy
- Department of Nuclear Medicine, University Hospital RWTH, Aachen, Germany
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, Netherlands
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany
- Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany
| | - Jürgen Floege
- Department of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany
| | - Peter Boor
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany.
- Department of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany.
- Electron Microscopy Facility, RWTH Aachen University Hospital, Aachen, Germany.
| |
Collapse
|
36
|
Abstract
Piflufolastat F 18 (PYLARIFY®) is an 18F-labelled diagnostic imaging agent that has been developed by Progenics Pharmaceuticals Inc., a Lantheus company, for positron emission tomography (PET) that targets prostate-specific membrane antigen (PSMA). Piflufolastat F 18 was approved in the USA on 27 May 2021 for PET of PSMA positive lesions in men with prostate cancer with suspected metastasis who are candidates for initial definitive therapy or with suspected recurrence based on elevated serum prostate specific antigen (PSA) level. This article summarizes the milestones in the development of piflufolastat F 18 leading to this approval as a radioactive diagnostic agent in prostate cancer.
Collapse
Affiliation(s)
- Susan J Keam
- Springer Nature, Private Bag 65901, Mairangi Bay, Auckland, 0754, New Zealand.
| |
Collapse
|
37
|
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
|
38
|
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
|
39
|
PSMA radioligand therapy for solid tumors other than prostate cancer: background, opportunities, challenges, and first clinical reports. Eur J Nucl Med Mol Imaging 2021; 48:4350-4368. [PMID: 34120192 PMCID: PMC8566635 DOI: 10.1007/s00259-021-05433-w] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 05/24/2021] [Indexed: 12/11/2022]
Abstract
In the past decade, a growing body of literature has reported promising results for prostate-specific membrane antigen (PSMA)-targeted radionuclide imaging and therapy in prostate cancer. First clinical studies evaluating the efficacy of [177Lu]Lu-PSMA radioligand therapy (PSMA-RLT) demonstrated favorable results in prostate cancer patients. [177Lu]Lu-PSMA is generally well tolerated due to its limited side effects. While PSMA is highly overexpressed in prostate cancer cells, varying degrees of PSMA expression have been reported in other malignancies as well, particularly in the tumor-associated neovasculature. Hence, it is anticipated that PSMA-RLT could be explored for other solid cancers. Here, we describe the current knowledge of PSMA expression in other solid cancers and define a perspective towards broader clinical implementation of PSMA-RLT. This review focuses specifically on salivary gland cancer, glioblastoma, thyroid cancer, renal cell carcinoma, hepatocellular carcinoma, lung cancer, and breast cancer. An overview of the (pre)clinical data on PSMA immunohistochemistry and PSMA PET/CT imaging is provided and summarized. Furthermore, the first clinical reports of non-prostate cancer patients treated with PSMA-RLT are described.
Collapse
|
40
|
Mittlmeier LM, Ledderose ST, Schott M, Brendel M, Beyer L, Theurich S, Mayr D, Walz C, Kunz WG, Ricke J, Bartenstein P, Ilhan H, Staehler M, Unterrainer M. Immature Plasma Cell Myeloma Mimics Metastatic Renal Cell Carcinoma on 18F-PSMA-1007 PET/CT Due to Endothelial PSMA-Expression. Diagnostics (Basel) 2021; 11:423. [PMID: 33802288 PMCID: PMC8000301 DOI: 10.3390/diagnostics11030423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 02/28/2021] [Accepted: 03/01/2021] [Indexed: 11/23/2022] Open
Abstract
We present a 71-year-old female patient who underwent 18F-PSMA-1007 PET/CT for suspected metastatic renal cell carcinoma (RCC), as RCC also shows high PSMA-expression in tumor neovascularization. 18F-PSMA-1007 PET/CT showed a high PSMA-avidity in the renal tumor, enlarged intra-abdominal and mediastinal lymph nodes. Moreover, PSMA-positive pleural, pulmonal and osseous lesions were found. However, histopathology revealed an immature plasma cell myeloma with an endothelial PSMA-expression of the neovasculature. This case illustrates the increased PSMA-avidity in multiple myeloma and highlights PSMA as a potential theragnostic target in multiple myeloma. For clinical routine, lymphatic diseases such as extramedullary myeloma should be considered as differential diagnosis in PSMA-avid renal masses on PET/CT.
Collapse
Affiliation(s)
- Lena M. Mittlmeier
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany; (L.M.M.); (M.B.); (L.B.); (P.B.); (H.I.)
| | - Stephan T. Ledderose
- Institute of Pathology, LMU Munich, 81377 Munich, Germany; (S.T.L.); (D.M.); (C.W.)
| | - Melanie Schott
- Department of Urology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.S.); (M.S.)
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany; (L.M.M.); (M.B.); (L.B.); (P.B.); (H.I.)
| | - Leonie Beyer
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany; (L.M.M.); (M.B.); (L.B.); (P.B.); (H.I.)
| | - Sebastian Theurich
- Department of Medicine III, University Hospital, LMU Munich, 81377 Munich, Germany;
| | - Doris Mayr
- Institute of Pathology, LMU Munich, 81377 Munich, Germany; (S.T.L.); (D.M.); (C.W.)
| | - Christoph Walz
- Institute of Pathology, LMU Munich, 81377 Munich, Germany; (S.T.L.); (D.M.); (C.W.)
| | - Wolfgang G. Kunz
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (W.G.K.); (J.R.)
| | - Jens Ricke
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (W.G.K.); (J.R.)
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany; (L.M.M.); (M.B.); (L.B.); (P.B.); (H.I.)
| | - Harun Ilhan
- Department of Nuclear Medicine, University Hospital, LMU Munich, 81377 Munich, Germany; (L.M.M.); (M.B.); (L.B.); (P.B.); (H.I.)
| | - Michael Staehler
- Department of Urology, University Hospital, LMU Munich, 81377 Munich, Germany; (M.S.); (M.S.)
| | - Marcus Unterrainer
- Department of Radiology, University Hospital, LMU Munich, 81377 Munich, Germany; (W.G.K.); (J.R.)
| |
Collapse
|
41
|
Mittlmeier LM, Unterrainer M, Rodler S, Todica A, Albert NL, Burgard C, Cyran CC, Kunz WG, Ricke J, Bartenstein P, Stief CG, Ilhan H, Staehler M. 18F-PSMA-1007 PET/CT for response assessment in patients with metastatic renal cell carcinoma undergoing tyrosine kinase or checkpoint inhibitor therapy: preliminary results. Eur J Nucl Med Mol Imaging 2020; 48:2031-2037. [PMID: 33369689 PMCID: PMC8113284 DOI: 10.1007/s00259-020-05165-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 12/13/2020] [Indexed: 11/26/2022]
Abstract
Introduction Tyrosine kinase (TKI) and checkpoint inhibitors (CI) prolonged overall survival in metastatic renal cell carcinoma (mRCC). Early prediction of treatment response is highly desirable for the individualization of patient management and improvement of therapeutic outcome; however, serum biochemistry is unable to predict therapeutic efficacy. Therefore, we compared 18F-PSMA-1007 PET imaging for response assessment in mRCC patients undergoing TKI or CI therapy compared to CT-based response assessment as the current imaging reference standard. Methods 18F-PSMA-1007 PET/CT was performed in mRCC patients prior to initiation of systemic treatment and 8 weeks after therapy initiation. Treatment response was evaluated separately on 18F-PSMA-PET and CT. Changes on PSMA-PET (SUVmean) were assessed on a per patient basis using a modified PERCIST scoring system. Complete response (CRPET) was defined as absence of any uptake in all target lesions on posttreatment PET. Partial response (PRPET) was defined as decrease in summed SUVmean of > 30%. The appearance of new, PET-positive lesions or an increase in summed SUVmean of > 30% was defined as progressive disease (PDPET). A change in summed SUVmean of ± 30% defined stable disease (SDPET). RECIST 1.1 criteria were used for response assessment on CT. Results of radiographic response assessment on PSMA-PET and CT were compared. Results Overall, 11 mRCC patients undergoing systemic treatment were included. At baseline PSMA-PET1, all mRCC patients showed at least one PSMA-avid lesion. On follow-up PET2, 3 patients showed CRPET, 3 PRPET, 4 SDPET, and 1 PDPET. According to RECIST 1.1, 1 patient showed PRCT, 9 SDCT, and 1 PDCT. Overall, concordant classifications were found in only 2 cases (2 SDCT + PET). Patients with CRPET on PET were classified as 3 SDCT on CT using RECIST 1.1. By contrast, the patient classified as PRCT on CT showed PSMA uptake without major changes during therapy (SDPET). However, among 9 patients with SDCT on CT, 3 were classified as CRPET, 3 as PRPET, 1 as PDPET, and only 2 as SDPET on PSMA-PET. Conclusion On PSMA-PET, heterogeneous courses were observed during systemic treatment in mRCC patients with highly diverging results compared to RECIST 1.1. In the light of missing biomarkers for early response assessment, PSMA-PET might allow more precise response assessment to systemic treatment, especially in patients classified as SD on CT.
Collapse
Affiliation(s)
- L M Mittlmeier
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - M Unterrainer
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - S Rodler
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - A Todica
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - N L Albert
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - C Burgard
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - C C Cyran
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - W G Kunz
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - J Ricke
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - P Bartenstein
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - C G Stief
- Department of Urology, University Hospital, LMU Munich, Munich, Germany
| | - H Ilhan
- Department of Nuclear Medicine, University Hospital, LMU Munich, Munich, Germany
| | - M Staehler
- Department of Urology, University Hospital, LMU Munich, Munich, Germany.
- Head Interdisciplinary Center on Renal Tumors, Department of Urology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany.
| |
Collapse
|
42
|
|
43
|
MiR-206 suppresses proliferation and epithelial-mesenchymal transition of renal cell carcinoma by inhibiting CDK6 expression. Hum Cell 2020; 33:750-758. [PMID: 32277426 DOI: 10.1007/s13577-020-00355-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 03/25/2020] [Indexed: 12/11/2022]
Abstract
Increasing evidence indicates that miRNAs are involved in tumorigenesis of human renal cell carcinoma (RCC). However, the role of miR-206 is still unknown. This study aimed to investigate the possible mechanism of miR-206 in progression of RCC. Here, compared with adjacent normal renal tissues and HK-2 cells, miR-206 level was markedly decreased, whereas CDK6 level was obviously increased in RCC tissues and cell lines. MiR-206 was inversely associated with lymph node metastasis and TNM stage, and acted as an independent prognostic factor in RCC. MiR-206 effectively caused apoptosis and cell cycle arrest at G0/G1 phase, and affected the growth of xenograft tumor of nude mice. MiR-206 also inhibited migration and invasion of RCC cells by modulating the expressions of EMT-related genes. Dual-luciferase assay demonstrated CDK6 was a direct target of miR-206. CDK6 silencing aggravated the inhibition effects of miR-206. In conclusion, miR-206 suppresses proliferation and EMT of RCC by inhibiting CDK6 expression. The miR-206/CDK6 axis may provide a novel insight into tumorigenesis of RCC.
Collapse
|
44
|
Van de Wiele C, Sathekge M, de Spiegeleer B, de Jonghe PJ, Beels L, Maes A. PSMA-Targeting Positron Emission Agents for Imaging Solid Tumors Other Than Non-Prostate Carcinoma: A Systematic Review. Int J Mol Sci 2019; 20:E4886. [PMID: 31581638 PMCID: PMC6801742 DOI: 10.3390/ijms20194886] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/30/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022] Open
Abstract
Despite its name, prostate-specific membrane antigen (PSMA) has been shown using immunohistochemistry (IHC) to also be over-expressed in the tumor neovasculature of a wide variety of solid tumors other than prostate carcinoma. Accordingly, positron-emitting radiolabeled small molecules targeting PSMA, initially developed for positron emission tomography in prostate carcinomas, are currently being explored for their staging and restaging potential as an alternative imaging modality in other solid tumor types where 18-F-fluorodeoxyglucose (FDG)-PET imaging has low diagnostic accuracy. In this paper, the currently available literature in this field is reviewed. Preliminary, mainly retrospective studies are encouraging, with evidence of improved diagnostic sensitivity and specificity in clear cell renal carcinoma, glioma, and hepatocellular carcinoma, leading to a change in patient management in several patients. However, the results published thus far warrant confirmation by larger prospective studies additionally assessing the longitudinal impact on patient outcomes.
Collapse
Affiliation(s)
- Christophe Van de Wiele
- Department of Nuclear Medicine, AZ Groeninge, 8500 Kortrijk, Belgium.
- Department of Nuclear Medicine and Radiology, University Ghent, 9000 GHent, Belgium.
| | - Mike Sathekge
- Department of Nuclear Medicine, University of Pretoria, 0001 Pretoria, South-Africa.
| | - Bart de Spiegeleer
- Laboratory of Drug Quality and Registration, University Ghent, 9000 Ghent, Belgium.
| | | | - Laurence Beels
- Department of Nuclear Medicine, AZ Groeninge, 8500 Kortrijk, Belgium.
| | - Alex Maes
- Department of Nuclear Medicine, AZ Groeninge, 8500 Kortrijk, Belgium.
- Department of Imaging and Pathology, KULAK, University of Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
45
|
Derks YH, Löwik DWPM, Sedelaar JPM, Gotthardt M, Boerman OC, Rijpkema M, Lütje S, Heskamp S. PSMA-targeting agents for radio- and fluorescence-guided prostate cancer surgery. Am J Cancer Res 2019; 9:6824-6839. [PMID: 31660071 PMCID: PMC6815946 DOI: 10.7150/thno.36739] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/16/2019] [Indexed: 11/15/2022] Open
Abstract
Despite recent improvements in imaging and therapy, prostate cancer (PCa) still causes substantial morbidity and mortality. In surgical treatment, incomplete resection of PCa and understaging of possible undetected metastases may lead to disease recurrence and consequently poor patient outcome. To increase the chance of accurate staging and subsequently complete removal of all cancerous tissue, prostate specific membrane antigen (PSMA) targeting agents may provide the surgeon an aid for the intraoperative detection and resection of PCa lesions. Two modalities suitable for this purpose are radionuclide detection, which allows sensitive intraoperative localization of tumor lesions with a gamma probe, and fluorescence imaging, allowing tumor visualization and delineation. Next to fluorescence, use of photosensitizers may enable intraoperative targeted photodynamic therapy to eradicate remaining tumor lesions. Since radiodetection and optical imaging techniques each have their own strengths and weaknesses, a combination of both modalities could be of additional value. Here, we provide an overview of recent preclinical and clinical advances in PSMA-targeted radio- and fluorescence-guided surgery of PCa.
Collapse
|