1
|
Nedelcovych MT, Dash RP, Wu Y, Choi EY, Lapidus RS, Majer P, Jančařík A, Abou D, Penet MF, Nikolopoulou A, Amor-Coarasa A, Babich J, Thorek DL, Rais R, Kratochwil C, Slusher BS. JHU-2545 preferentially shields salivary glands and kidneys during PSMA-targeted imaging. Eur J Nucl Med Mol Imaging 2025; 52:1631-1641. [PMID: 39743616 PMCID: PMC11928385 DOI: 10.1007/s00259-024-07044-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 12/16/2024] [Indexed: 01/04/2025]
Abstract
PURPOSE Prostate-specific membrane antigen (PSMA) radioligand therapy is a promising treatment for metastatic castration-resistant prostate cancer (mCRPC). Several beta or alpha particle-emitting radionuclide-conjugated small molecules have shown efficacy in late-stage mCRPC and one, [[177Lu]Lu]Lu-PSMA-617, is FDA approved. In addition to tumor upregulation, PSMA is also expressed in kidneys and salivary glands where specific uptake can cause dose-limiting xerostomia and potential for nephrotoxicity. The PSMA inhibitor 2-(phosphonomethyl)pentanedioic acid (2-PMPA) can prevent kidney uptake in mice, but also blocks tumor uptake, precluding its clinical utility. Preferential delivery of 2-PMPA to non-malignant tissues could improve the therapeutic window of PSMA radioligand therapy. METHODS A tris(isopropoxycarbonyloxymethyl) (TrisPOC) prodrug of 2-PMPA, JHU-2545, was synthesized to enhance 2-PMPA delivery to non-malignant tissues. Mouse pharmacokinetic experiments were conducted to compare JHU-2545-mediated delivery of 2-PMPA to plasma, kidney, salivary glands, and C4-2 prostate tumor xenograft. Imaging studies were conducted in rats and mice to measure uptake of PSMA PET tracers in kidney, salivary glands, and prostate tumor xenografts with and without JHU-2545 pre-treatment. RESULTS JHU-2545 resulted in approximately 3- and 53-fold greater exposure of 2-PMPA in rodent salivary glands (18.0 ± 0.97 h*nmol/g) and kidneys (359 ± 4.16 h*nmol/g) versus prostate tumor xenograft (6.79 ± 0.19 h*nmol/g). JHU-2545 also blocked rodent kidneys and salivary glands uptake of the PSMA PET tracers [68Ga]Ga-PSMA-11 and [18 F]F-DCFPyL by up to 85% with little effect on tumor. CONCLUSIONS JHU-2545 pre-treatment may enable greater cumulative administered doses of PSMA radioligand therapy, possibly improving safety and efficacy.
Collapse
Affiliation(s)
- Michael T Nedelcovych
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- Johns Hopkins Drug Discovery, 855 North Wolfe Street, Baltimore, Maryland, 21205, USA.
| | - Ranjeet P Dash
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Ying Wu
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Eun Yong Choi
- Translational Laboratory Shared Service, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD, 21201, USA
| | - Rena S Lapidus
- Translational Laboratory Shared Service, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD, 21201, USA
| | - Pavel Majer
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic v.v.i, Prague, 166 10, Czech Republic
| | - Andrej Jančařík
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic v.v.i, Prague, 166 10, Czech Republic
| | - Diane Abou
- Depatment of Radiology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Marie-France Penet
- Departments of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Depatment of Radiology and Radiological Science, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Anastasia Nikolopoulou
- Division of Radiopharmaceutical Sciences and MI(3), Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Alex Amor-Coarasa
- Division of Radiopharmaceutical Sciences and MI(3), Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - John Babich
- Division of Radiopharmaceutical Sciences and MI(3), Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Daniel L Thorek
- Depatment of Radiology, Washington University School of Medicine, Saint Louis, MO, 63110, USA
| | - Rana Rais
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA
| | - Clemens Kratochwil
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Barbara S Slusher
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- Departments of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- Departments of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- Departments of Psychiatry, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- Departments of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, 21205, USA.
- Johns Hopkins Drug Discovery, 855 North Wolfe Street, Baltimore, Maryland, 21205, USA.
| |
Collapse
|
2
|
van der Gaag S, Vis AN, Bartelink IH, Koppes JCC, Hodolic M, Hendrikse H, Oprea-Lager DE. Exploring the Flare Phenomenon in Patients with Castration-Resistant Prostate Cancer: Enzalutamide-Induced PSMA Upregulation Observed on PSMA PET. J Nucl Med 2025; 66:373-376. [PMID: 39915123 DOI: 10.2967/jnumed.124.268340] [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: 07/02/2024] [Accepted: 12/05/2024] [Indexed: 03/05/2025] Open
Abstract
Androgen receptor-targeting agents, particularly enzalutamide, show promise in enhancing prostate cancer diagnostic and therapeutic strategies by modulating prostate-specific membrane antigen (PSMA). Methods: A retrospective clinical cohort study investigated 9 men with metastatic castration-resistant prostate cancer on enzalutamide. PSMA PET/CT scans were obtained before and after enzalutamide initiation to assess PSMA expression changes. Lesions and organs at risk were evaluated visually and semiquantitatively. The flare phenomenon was characterized by a significant increase (≥20%) in the SUVmax of existing lesions or the appearance of new PSMA-positive lesions. Results: Exposure to enzalutamide led to a significant PSMA expression increase in 56% of assessed lesions (n = 42), with new lesions detected in 1 patient (11%). PSMA expression in organs at risk remained largely unaffected, indicating a tumor-specific response. Conclusion: Enzalutamide induces PSMA upregulation in metastatic castration-resistant prostate cancer, potentially enhancing diagnostic and therapeutic strategies. Further exploration of the flare phenomenon's clinical implications is warranted.
Collapse
Affiliation(s)
- Suzanne van der Gaag
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - André N Vis
- Department of Urology, Prostate Cancer Network Amsterdam, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands;
| | - Imke H Bartelink
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Clinical Pharmacology and Pharmacy, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; and
| | - Josephina C C Koppes
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Marina Hodolic
- Nuclear Medicine Department, Faculty of Medicine and Dentistry, Palacký University Olomouc, Olomouc, Czech Republic
| | - Harry Hendrikse
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Daniela E Oprea-Lager
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Imaging and Biomarkers, Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
3
|
Laudicella R, Bauckneht M, Burger IA, Cacciola A, Fanti S, Farolfi A, Ficarra V, Iagaru A, Liberini V, Pergolizzi S, Santo G, Virgolini I, Minutoli F, Baldari S. The role of PSMA-based radioligand therapy in hormone-sensitive prostate cancer. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07083-8. [PMID: 39934300 DOI: 10.1007/s00259-025-07083-8] [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: 09/04/2024] [Accepted: 01/10/2025] [Indexed: 02/13/2025]
Abstract
PURPOSE Conventional systemic therapies are valuable options in prostate cancer (PCa); however, such treatments can determine adverse events and toxicity. The observed improvement in overall survival, coupled with PSA reduction and a favorable safety profile in the post-taxane castration-resistant PCa (CRPC) setting has prompted the consideration of PSMA-based radioligand therapy (RLT) earlier in the treatment sequence. In this review, we will describe the literature and ongoing clinical trials regarding the use of PSMA-based RLT in hormone-sensitive PCa (HSPC) including the neoadjuvant, de-novo/synchronous metastatic, adjuvant, and early BCR settings. METHODS We performed a systematic literature search on the PubMed/MEDLINE/EMBASE and clinicaltrials.gov databases for studies and protocols assessing the role of PSMA-based RLT in HSPC. RESULTS The literature search yielded 140 results. After screening titles and abstracts and applying inclusion and exclusion criteria, we selected 25 papers showing the potentialities of earlier RLT in HSPC, with several ongoing trials. CONCLUSION Early use of PSMA-based RLT holds significant potential in HSPC patients from the neoadjuvant to the BCR setting. In these stages, the lower tumor burden, more frequent exclusive nodal involvement, and higher organ reserve may improve treatment efficacy and allow for treatment combinations while maintaining a less toxic profile.
Collapse
Affiliation(s)
- Riccardo Laudicella
- Nuclear Medicine, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, Messina, Italy
| | - Matteo Bauckneht
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Nuclear Medicine, Department of Health Sciences (DISSAL), University of Genova, Genova, Italy
| | - Irene A Burger
- Department of Nuclear Medicine, University Hospital Zürich, University of Zurich, Zurich, Switzerland.
- Department of Nuclear Medicine, Cantonal Hospital Baden, affiliated Hospital for Research and Teaching, University of Zurich, Baden, Switzerland.
| | - Alberto Cacciola
- Brain Mapping Lab, Department of Biomedical, Dental Sciences and Morphological and Functional Imaging, University of Messina, Messina, Italy
| | - Stefano Fanti
- Nuclear Medicine Division, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Policlinico S. Orsola, Bologna, Italy
- Nuclear Medicine, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Andrea Farolfi
- Nuclear Medicine, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Vincenzo Ficarra
- Gaetano Barresi Department of Human and Paediatric Pathology, Urologic Section, University of Messina, Messina, Italy
| | - Andrei Iagaru
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, USA
| | | | - Stefano Pergolizzi
- Radiation Oncology Unit, Department of Biomedical, Dental and Morphological and Functional Imaging Sciences, University of Messina, Messina, Italy
| | - Giulia Santo
- Department of Nuclear Medicine, Medical University of Innsbruck, Innsbruck, Austria
- Department of Experimental and Clinical Medicine, ''Magna Graecia'' University of Catanzaro, Catanzaro, Italy
| | - Irene Virgolini
- Department of Nuclear Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Fabio Minutoli
- Nuclear Medicine, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, Messina, Italy
| | - Sergio Baldari
- Nuclear Medicine, Department of Biomedical and Dental Sciences and Morpho-Functional Imaging, University of Messina, Messina, Italy
| |
Collapse
|
4
|
Benitez CM, Sahlstedt H, Sonni I, Brynolfsson J, Berenji GR, Juarez JE, Kane N, Tsai S, Rettig M, Nickols NG, Duriseti S. Treatment Response Assessment According to Updated PROMISE Criteria in Patients with Metastatic Prostate Cancer Using an Automated Imaging Platform for Identification, Measurement, and Temporal Tracking of Disease. Eur Urol Oncol 2024:S2588-9311(24)00240-2. [PMID: 39521638 DOI: 10.1016/j.euo.2024.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/09/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND AND OBJECTIVE Prostate-specific membrane antigen (PSMA) molecular imaging is widely used for disease assessment in prostate cancer (PC). Artificial intelligence (AI) platforms such as automated Prostate Cancer Molecular Imaging Standardized Evaluation (aPROMISE) identify and quantify locoregional and distant disease, thereby expediting lesion identification and standardizing reporting. Our aim was to evaluate the ability of the updated aPROMISE platform to assess treatment responses based on integration of the RECIP (Response Evaluation Criteria in PSMA positron emission tomography-computed tomography [PET/CT]) 1.0 classification. METHODS The study included 33 patients with castration-sensitive PC (CSPC) and 34 with castration-resistant PC (CRPC) who underwent PSMA-targeted molecular imaging before and ≥2 mo after completion of treatment. Tracer-avid lesions were identified using aPROMISE for pretreatment and post-treatment PET/CT scans. Detected lesions were manually approved by an experienced nuclear medicine physician, and total tumor volume (TTV) was calculated. Response was assessed according to RECIP 1.0 as CR (complete response), PR (partial response), PD (progressive disease), or SD (stable disease). KEY FINDINGS AND LIMITATIONS: aPROMISE identified 1576 lesions on baseline scans and 1631 lesions on follow-up imaging, 618 (35%) of which were new. Of the 67 patients, aPROMISE classified four as CR, 16 as PR, 34 as SD, and 13 as PD; five cases were misclassified. The agreement between aPROMISE and clinician validation was 89.6% (κ = 0.79). CONCLUSIONS AND CLINICAL IMPLICATIONS aPROMISE may serve as a novel assessment tool for treatment response that integrates PSMA PET/CT results and RECIP imaging criteria. The precision and accuracy of this automated process should be validated in prospective clinical studies. PATIENT SUMMARY We used an artificial intelligence (AI) tool to analyze scans for prostate cancer before and after treatment to see if we could track how cancer spots respond to treatment. We found that the AI approach was successful in tracking individual tumor changes, showing which tumors disappeared, and identifying new tumors in response to prostate cancer treatment.
Collapse
Affiliation(s)
- Cecil M Benitez
- Department of Radiation Oncology, University of California-Los Angeles, Los Angeles, CA, USA
| | | | - Ida Sonni
- VA Greater Los Angeles Healthcare System, Department of Nuclear Medicine, Los Angeles, CA, USA; Department of Radiological Sciences, University of California-Los Angeles, Los Angeles, CA, USA
| | | | - Gholam Reza Berenji
- VA Greater Los Angeles Healthcare System, Department of Nuclear Medicine, Los Angeles, CA, USA
| | - Jesus Eduardo Juarez
- Department of Radiation Oncology, University of California-Los Angeles, Los Angeles, CA, USA
| | - Nathanael Kane
- Department of Radiation Oncology, University of California-Los Angeles, Los Angeles, CA, USA; Department of Radiation Oncology, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Sonny Tsai
- Department of Radiation Oncology, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Matthew Rettig
- Department of Hematology-Oncology, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA; Department of Medicine, University of California-Los Angeles, Los Angeles, CA, USA; Department of Urology, University of California-Los Angeles, Los Angeles, CA, USA
| | - Nicholas George Nickols
- Department of Radiation Oncology, University of California-Los Angeles, Los Angeles, CA, USA; Department of Radiation Oncology, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA; Department of Urology, University of California-Los Angeles, Los Angeles, CA, USA
| | - Sai Duriseti
- Department of Radiation Oncology, University of California-Los Angeles, Los Angeles, CA, USA; Department of Radiation Oncology, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA.
| |
Collapse
|
5
|
Nelson BJ, Krol V, Bansal A, Andersson JD, Wuest F, Pandey MK. Aspects and prospects of preclinical theranostic radiopharmaceutical development. Theranostics 2024; 14:6446-6470. [PMID: 39479448 PMCID: PMC11519794 DOI: 10.7150/thno.100339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 07/31/2024] [Indexed: 11/02/2024] Open
Abstract
This article provides an overview of preclinical theranostic radiopharmaceutical development, highlighting aspects of the preclinical development stages that can lead towards a clinical trial. The key stages of theranostic radiopharmaceutical development are outlined, including target selection, tracer development, radiopharmaceutical synthesis, automation and quality control, in vitro radiopharmaceutical analysis, selecting a suitable in vivo model, preclinical imaging and pharmacokinetic analysis, preclinical therapeutic analysis, dosimetry, toxicity, and preparing for clinical translation. Each stage is described and augmented with examples from the literature. Finally, an outlook on the prospects for the radiopharmaceutical theranostics field is provided.
Collapse
Affiliation(s)
- Bryce J.B. Nelson
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta, T6G 1Z2 Canada
| | - Viktoria Krol
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Aditya Bansal
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jan D. Andersson
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta, T6G 1Z2 Canada
- Edmonton Radiopharmaceutical Center, Alberta Health Services, Edmonton, Alberta, T6G 1Z2, Canada
| | - Frank Wuest
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta, T6G 1Z2 Canada
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, Alberta, T6G 2E1, Canada
| | - Mukesh K. Pandey
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
- Mayo Clinic Comprehensive Cancer Center, Rochester, MN 55905, USA
| |
Collapse
|
6
|
Maes J, Gesquière S, De Spiegeleer A, Maes A, Van de Wiele C. Prostate-Specific Membrane Antigen Biology and Pathophysiology in Prostate Carcinoma, an Update: Potential Implications for Targeted Imaging and Therapy. Int J Mol Sci 2024; 25:9755. [PMID: 39273701 PMCID: PMC11396261 DOI: 10.3390/ijms25179755] [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: 07/04/2024] [Revised: 08/28/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
Prostate-specific membrane antigen (PSMA), a transmembrane glycoprotein, was shown to be expressed 100-1000 fold higher in prostate adenocarcinoma as compared to normal prostate epithelium. Given the enzymatic function of PSMA with the presence of an internalization triggering motif, various Glu-urea-Lys-based inhibitors have been developed and, amongst others, radiolabeled with positron emitters for targeted positron emission tomography imaging such as 68Ga-PSMA-HBED-CC Glu-urea-Lys(Ahx) as well as with beta and alpha-emitting radioisotopes for targeted therapy, e.g., 177Lu-PSMA-617. In this paper, we review and discuss the potential implications for targeted imaging and therapy of altered PSMA-glycosylation, of PSMA-driven activation of the P13K/Akt/mTOR, of the evolution over time and the relationship with androgen signaling and changes in DNA methylation of PSMA, and of androgen deprivation therapy (ADT) in prostate carcinoma.
Collapse
Affiliation(s)
| | - Simon Gesquière
- Department of Diagnostic Sciences, University Ghent, De Pintelaan 185, 9000 Ghent, Belgium
| | | | - Alex Maes
- AZ Groeninge, 8500 Kortrijk, Belgium
- Department of Morphology and Functional Imaging, University Leuven, 3000 Leuven, Belgium
| | - Christophe Van de Wiele
- AZ Groeninge, 8500 Kortrijk, Belgium
- Department of Diagnostic Sciences, University Ghent, De Pintelaan 185, 9000 Ghent, Belgium
| |
Collapse
|
7
|
Cengiz TB, Kulkarni R, Novello M, Hafez A, Gavane S, Ghesani M, Ghesani N. Does enzalutamide related PSMA upregulation affect outcomes of lutetium-177 PSMA radioligand therapy? Urologia 2024; 91:525-530. [PMID: 38752520 DOI: 10.1177/03915603241249230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/07/2024]
Abstract
BACKGROUND Enzalutamide is an antiandrogen drug used prior to lutetium-177 prostate specific membrane antigen (Lu-PSMA) radioligand therapy and has shown promising results for upregulating the PSMA expression on prostate cancer cells. In this study, we aim to compare prostate specific antigen (PSA) level changes in prostate cancer patients who received enzalutamide to those who did not. METHODS Prostate cancer patients who underwent Lu-PSMA between 2021 and 2023 were retrospectively included. Patients were grouped based on prior enzalutamide therapy: those who received enzalutamide (EZ+) for at least 14 days and those who did not (EZ-). PSA changes and F-18 DCFPyL SUV (Standardized Uptake Values) were compared. RESULTS Thirty-seven patients were included, 18 EZ+ and 19 EZ-. The median age, Gleason score, and prior chemo/hormonal therapies were similar for EZ+ and EZ-, except for radium-223. Eleven patients (61%) in EZ+ and 13 patients (68%) in EZ- showed a decrease in PSA after the first cycle (p = 0.64). Four patients (22%) in EZ+ and seven patients (37%) in EZ- had more than 50% decrease in PSA after the first cycle (p = 0.33). The average percent decline at the end of the treatment was 23.3% in EZ+ and 50.4% in EZ- (p = 0.4). There was no difference in terms of lesion with highest SUVmax, mean SUV, total tumor volume or activity on pre-therapy PSMA imaging. CONCLUSION Enzalutamide treatment prior to Lu-PSMA does not improve patient outcomes when applied remotely. Larger studies evaluating the combination therapies and the timing of enzalutamide are needed to assess its correlation with Lu-PSMA outcomes.
Collapse
Affiliation(s)
- Turgut Bora Cengiz
- Section of Nuclear Medicine, Department of Radiology, Atrium Health Wake Forest Baptist Hospital, Winston-Salem, NC, USA
| | - Raksha Kulkarni
- Division of Nuclear Medicine, Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matteo Novello
- Division of Nuclear Medicine, Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Anthony Hafez
- Division of Nuclear Medicine, Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Somali Gavane
- Division of Nuclear Medicine, Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Munir Ghesani
- Division of Nuclear Medicine, Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nasrin Ghesani
- Division of Nuclear Medicine, Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
8
|
Georgiev T, Principi L, Galbiati A, Gilardoni E, Neri D, Cazzamalli S. Targeted interleukin-2 enhances the in vivo anti-cancer activity of Pluvicto™. Eur J Nucl Med Mol Imaging 2024; 51:2332-2337. [PMID: 38563883 DOI: 10.1007/s00259-024-06705-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 03/22/2024] [Indexed: 04/04/2024]
Abstract
PURPOSE Pluvicto™ ([177Lu]Lu-PSMA-617), a radioligand therapeutic targeting prostate-specific membrane antigen (PSMA), has been recently approved for the treatment of metastatic castration-resistant prostate cancer (mCRPR). The drug suffers from salivary gland and kidney uptake that prevents its dose escalation to potentially curative doses. In this work, we sought to potentiate the in vivo anti-cancer activity of Pluvicto™ by combining it with L19-IL2, a clinical-stage investigational medicinal product based on tumor-targeted interleukin-2. METHODS We established a new PSMA-expressing model (HT-1080.hPSMA) and validated it using a fluoresceine analogue of PSMA-617 (compound 1). The HT-1080.hPSMA model was used to study the saturation and tumor retention of Pluvicto™ (compound 2) and to run combination therapy studies with L19-IL2. To complement our understanding of the mechanism of action of this novel combination, we conducted proteomics experiments on tumor samples after therapy with Pluvicto™ alone or in combination with the immunocytokine. RESULTS High, selective, and long-lived tumor uptake was observed for Pluvicto™ (2) in the novel HT-1080.hPSMA model. Therapy studies in HT-1080.hPSMA tumor-bearing mice revealed that the combination of Pluvicto™ (2) plus L19-IL2 mediated curative and durable responses in all animals. Potent in vivo anti-cancer activity was observed solely for the combination modality, at doses that were well tolerated by treated animals. Proteomics studies indicated that L19-IL2 boosts the activation of the immune system in animals pre-treated with Pluvicto™. CONCLUSION The therapeutic efficacy of Pluvicto™ at low radioactive doses can be effectively enhanced by the combination with L19-IL2. Our findings warrant further clinical exploration of this novel combination modality.
Collapse
Affiliation(s)
- Tony Georgiev
- R&D Department, Philochem AG, Libernstrasse 3, CH-8112, Otelfingen, ZH, Switzerland
| | - Lucrezia Principi
- R&D Department, Philochem AG, Libernstrasse 3, CH-8112, Otelfingen, ZH, Switzerland
| | - Andrea Galbiati
- R&D Department, Philochem AG, Libernstrasse 3, CH-8112, Otelfingen, ZH, Switzerland
| | - Ettore Gilardoni
- R&D Department, Philochem AG, Libernstrasse 3, CH-8112, Otelfingen, ZH, Switzerland
| | - Dario Neri
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, CH-8093, Zurich, Switzerland.
- Philogen S.p.A., I-53100, Siena, Italy.
| | - Samuele Cazzamalli
- R&D Department, Philochem AG, Libernstrasse 3, CH-8112, Otelfingen, ZH, Switzerland.
| |
Collapse
|
9
|
Schatz CA, Zitzmann-Kolbe S, Moen I, Klotz M, Nair S, Stargard S, Bjerke RM, Wickstrøm Biseth K, Feng YZ, Indrevoll B, Cruciani V, Karlsson J, Haendler B, Nielsen CH, Alfsen MZ, Hammer S, Hennekes H, Cuthbertson A, Hagemann UB, Larsen Å. Preclinical Efficacy of a PSMA-Targeted Actinium-225 Conjugate (225Ac-Macropa-Pelgifatamab): A Targeted Alpha Therapy for Prostate Cancer. Clin Cancer Res 2024; 30:2531-2544. [PMID: 38593212 DOI: 10.1158/1078-0432.ccr-23-3746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 02/07/2024] [Accepted: 04/03/2024] [Indexed: 04/11/2024]
Abstract
PURPOSE Initially, prostate cancer responds to hormone therapy, but eventually resistance develops. Beta emitter-based prostate-specific membrane antigen (PSMA)-targeted radionuclide therapy is approved for the treatment of metastatic castration-resistant prostate cancer. Here we introduce a targeted alpha therapy (TAT) consisting of the PSMA antibody pelgifatamab covalently linked to a macropa chelator and labeled with actinium-225 and compare its efficacy and tolerability with other TATs. EXPERIMENTAL DESIGN The in vitro characteristics and in vivo biodistribution, antitumor efficacy, and tolerability of 225Ac-macropa-pelgifatamab (225Ac-pelgi) and other TATs were investigated in cell line- and patient-derived prostate cancer xenograft models. The antitumor efficacy of 225Ac-pelgi was also investigated in combination with the androgen receptor inhibitor darolutamide. RESULTS Actinium-225-labeling of 225Ac-pelgi was efficient already at room temperature. Potent in vitro cytotoxicity was seen in PSMA-expressing (LNCaP, MDA-PCa-2b, and C4-2) but not in PSMA-negative (PC-3 and DU-145) cell lines. High tumor accumulation was seen for both 225Ac-pelgi and 225Ac-DOTA-pelgi in the MDA-PCa-2b xenograft model. In the C4-2 xenograft model, 225Ac-pelgi showed enhanced antitumor efficacy with a T/Cvolume (treatment/control) ratio of 0.10 compared with 225Ac-DOTA-pelgi, 225Ac-DOTA-J591, and 227Th-HOPO-pelgifatamab (227Th-pelgi; all at 300 kBq/kg) with T/Cvolume ratios of 0.37, 0.39, and 0.33, respectively. 225Ac-pelgi was less myelosuppressive than 227Th-pelgi. 225Ac-pelgi showed dose-dependent treatment efficacy in the patient-derived KuCaP-1 model and strong combination potential with darolutamide in both cell line- (22Rv1) and patient-derived (ST1273) xenograft models. CONCLUSIONS These results provide a strong rationale to investigate 225Ac-pelgi in patients with prostate cancer. A clinical phase I study has been initiated (NCT06052306).
Collapse
|
10
|
Emmett L, Subramaniam S, Crumbaker M, Nguyen A, Joshua AM, Weickhardt A, Lee ST, Ng S, Francis RJ, Goh JC, Pattison DA, Tan TH, Kirkwood ID, Gedye C, Rutherford NK, Sandhu S, Kumar AR, Pook D, Ramdave S, Nadebaum DP, Voskoboynik M, Redfern AD, Macdonald W, Krieger L, Schembri G, Chua W, Lin P, Horvath L, Bastick P, Butler P, Zhang AY, Yip S, Thomas H, Langford A, Hofman MS, McJannett M, Martin AJ, Stockler MR, Davis ID. [ 177Lu]Lu-PSMA-617 plus enzalutamide in patients with metastatic castration-resistant prostate cancer (ENZA-p): an open-label, multicentre, randomised, phase 2 trial. Lancet Oncol 2024; 25:563-571. [PMID: 38621400 DOI: 10.1016/s1470-2045(24)00135-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 02/23/2024] [Accepted: 02/28/2024] [Indexed: 04/17/2024]
Abstract
BACKGROUND Enzalutamide and lutetium-177 [177Lu]Lu-prostate-specific membrane antigen (PSMA)-617 both improve overall survival in patients with metastatic castration-resistant prostate cancer. Androgen and PSMA receptors have a close intracellular relationship, with data suggesting complementary benefit if targeted concurrently. In this study, we assessed the activity and safety of enzalutamide plus adaptive-dosed [177Lu]Lu-PSMA-617 versus enzalutamide alone as first-line treatment for metastatic castration-resistant prostate cancer. METHODS ENZA-p was an open-label, randomised, controlled phase 2 trial done at 15 hospitals in Australia. Participants were men aged 18 years or older with metastatic castration-resistant prostate cancer not previously treated with docetaxel or androgen receptor pathway inhibitors for metastatic castration-resistant prostate cancer, gallium-68 [68Ga]Ga-PSMA-PET-CT (PSMA-PET-CT) positive disease, Eastern Cooperative Oncology Group performance status of 0-2, and at least two risk factors for early progression on enzalutamide. Participants were randomly assigned (1:1) by a centralised, web-based system using minimisation with a random component to stratify for study site, disease burden, use of early docetaxel, and previous treatment with abiraterone acetate. Patients were either given oral enzalutamide 160 mg daily alone or with adaptive-dosed (two or four doses) intravenous 7·5 GBq [177Lu]Lu-PSMA-617 every 6-8 weeks dependent on an interim PSMA-PET-CT (week 12). The primary endpoint was prostate-specific antigen (PSA) progression-free survival, defined as the interval from the date of randomisation to the date of first evidence of PSA progression, commencement of non-protocol anticancer therapy, or death. The analysis was done in the intention-to-treat population, using stratified Cox proportional hazards regression. This trial is registered with ClinicalTrials.gov, NCT04419402, and participant follow-up is ongoing. FINDINGS 162 participants were randomly assigned between Aug 17, 2020, and July 26, 2022. 83 men were assigned to the enzalutamide plus [177Lu]Lu-PSMA-617 group, and 79 were assigned to the enzalutamide group. Median follow-up in this interim analysis was 20 months (IQR 18-21), with 32 (39%) of 83 patients in the enzalutamide plus [177Lu]Lu-PSMA-617 group and 16 (20%) of 79 patients in the enzalutamide group remaining on treatment at the data cutoff date. Median age was 71 years (IQR 64-76). Median PSA progression-free survival was 13·0 months (95% CI 11·0-17·0) in the enzalutamide plus [177Lu]Lu-PSMA-617 group and 7·8 months (95% CI 4·3-11·0) in the enzalutamide group (hazard ratio 0·43, 95% CI 0·29-0·63, p<0·0001). The most common adverse events (all grades) were fatigue (61 [75%] of 81 patients), nausea (38 [47%]), and dry mouth (32 [40%]) in the enzalutamide plus [177Lu]Lu-PSMA-617 group and fatigue (55 [70%] of 79), nausea (21 [27%]), and constipation (18 [23%]) in the enzalutamide group. Grade 3-5 adverse events occurred in 32 (40%) of 81 patients in the enzalutamide plus [177Lu]Lu-PSMA-617 group and 32 (41%) of 79 patients in the enzalutamide group. Grade 3 events that occurred only in the enzalutamide plus [177Lu]Lu-PSMA-617 group included anaemia (three [4%] of 81 participants) and decreased platelet count (one [1%] participant). No grade 4 or 5 events were attributed to treatment on central review in either group. INTERPRETATION The addition of [177Lu]Lu-PSMA-617 to enzalutamide improved PSA progression-free survival providing evidence of enhanced anticancer activity in patients with metastatic castration-resistant prostate cancer with risk factors for early progression on enzalutamide and warrants further evaluation of the combination more broadly in metastatic prostate cancer. FUNDING Prostate Cancer Research Alliance (Movember and Australian Federal Government), St Vincent's Clinic Foundation, GenesisCare, Roy Morgan Research, and Endocyte (a Novartis company).
Collapse
Affiliation(s)
- Louise Emmett
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, NSW, Australia; St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia; Garvan Institute of Medical Research, Sydney, NSW, Australia.
| | - Shalini Subramaniam
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia; Department of Medical Oncology, Bankstown-Lidcombe Hospital, Sydney, NSW, Australia
| | - Megan Crumbaker
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, NSW, Australia; Department of Medical Oncology, Kinghorn Cancer Centre, St Vincent's Hospital, Sydney, NSW, Australia; St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia; Garvan Institute of Medical Research, Sydney, NSW, Australia; Macquarie University Hospital, Sydney, NSW, Australia
| | - Andrew Nguyen
- Department of Theranostics and Nuclear Medicine, St Vincent's Hospital, Sydney, NSW, Australia; St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Anthony M Joshua
- Department of Medical Oncology, Kinghorn Cancer Centre, St Vincent's Hospital, Sydney, NSW, Australia; Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Andrew Weickhardt
- Olivia Newton-John Cancer and Wellness Centre, Austin Health, Melbourne, VIC, Australia; School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
| | - Sze-Ting Lee
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, VIC, Australia; School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia; Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia; Department of Medicine and Department of Surgery, University of Melbourne, Melbourne, VIC, Australia
| | - Siobhan Ng
- Department of Oncology, Sir Charles Gairdner Hospital, Perth, WA, Australia; Department of Oncology, University of Western Australia, Perth, WA, Australia
| | - Roslyn J Francis
- Department of Nuclear Medicine, Sir Charles Gairdner Hospital, Perth, WA, Australia; Medical School, University of Western Australia, Perth, WA, Australia
| | - Jeffrey C Goh
- Department of Medical Oncology, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia; Queensland University of Technology, Brisbane, QLD, Australia
| | - David A Pattison
- Department of Nuclear Medicine and Specialised PET Services, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia; School of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Thean Hsiang Tan
- Department of Medical Oncology, Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Ian D Kirkwood
- Nuclear Medicine, PET and Bone Densitometry, Royal Adelaide Hospital, Adelaide, SA, Australia; Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Craig Gedye
- Department of Medical Oncology, Calvary Mater Newcastle, Waratah, NSW, Australia
| | - Natalie K Rutherford
- Department of Nuclear Medicine, Hunter New England Health, Newcastle, NSW, Australia
| | - Shahneen Sandhu
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia; Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Aravind Ravi Kumar
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - David Pook
- Department of Oncology, Monash Health, Melbourne, VIC, Australia
| | - Shakher Ramdave
- Monash Health Imaging, Monash Health, Melbourne, VIC, Australia
| | - David P Nadebaum
- Department of Oncology, Alfred Health, Melbourne, VIC, Australia
| | - Mark Voskoboynik
- Department of Oncology, Alfred Health, Melbourne, VIC, Australia; Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Andrew D Redfern
- Medical School, University of Western Australia, Perth, WA, Australia; Department of Medical Oncology, Fiona Stanley Hospital, Perth, WA, Australia
| | - William Macdonald
- Medical School, University of Western Australia, Perth, WA, Australia; Department of Nuclear Medicine, Fiona Stanley Hospital, Perth, WA, Australia
| | | | - Geoff Schembri
- Nuclear Medicine, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Wei Chua
- Department of Medical Oncology, Liverpool Hospital, Sydney, NSW, Australia; Western Sydney University, Sydney, NSW, Australia
| | - Peter Lin
- South Western Sydney Clinical School, University of New South Wales, Sydney, NSW, Australia; Department of Nuclear Medicine and PET, Liverpool Hospital, Sydney, NSW, Australia
| | - Lisa Horvath
- Department of Medical Oncology, Chris O'Brien Lifehouse, Sydney, NSW, Australia
| | - Patricia Bastick
- Department of Medical Oncology, St George Hospital, Sydney, NSW, Australia
| | - Patrick Butler
- Department of Nuclear Medicine, St George Hospital, Sydney, NSW, Australia
| | - Alison Yan Zhang
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia; Macquarie University Hospital, Sydney, NSW, Australia; Department of Medical Oncology, Chris O'Brien Lifehouse, Sydney, NSW, Australia
| | - Sonia Yip
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Hayley Thomas
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Ailsa Langford
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Michael S Hofman
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia; Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Margaret McJannett
- Australian and New Zealand Urogenital and Prostate Cancer Trials Group, Sydney, NSW, Australia
| | - Andrew James Martin
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia; Centre for Clinical Research, University of Queensland, Brisbane, QLD, Australia
| | - Martin R Stockler
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia; Department of Medical Oncology, Chris O'Brien Lifehouse, Sydney, NSW, Australia
| | - Ian D Davis
- Monash University Eastern Health Clinical School, Melbourne, VIC, Australia; Eastern Health, Melbourne, VIC, Australia
| |
Collapse
|
11
|
Corpetti M, Müller C, Beltran H, de Bono J, Theurillat JP. Prostate-Specific Membrane Antigen-Targeted Therapies for Prostate Cancer: Towards Improving Therapeutic Outcomes. Eur Urol 2024; 85:193-204. [PMID: 38104015 DOI: 10.1016/j.eururo.2023.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/08/2023] [Accepted: 11/20/2023] [Indexed: 12/19/2023]
Abstract
CONTEXT Prostate-specific membrane antigen (PSMA) is a transmembrane glycoprotein overexpressed in most prostate cancers and exploited as a target for PSMA-targeted therapies. Different approaches to target PSMA-expressing cancer cells have been developed, showing promising results in clinical trials. OBJECTIVE To discuss the regulation of PSMA expression and the main PSMA-targeted therapeutic concepts illustrating their clinical development and rationalizing combination approaches with examples. EVIDENCE ACQUISITION We performed a detailed literature search using PubMed and reviewed the American Society of Clinical Oncology and European Society of Medical Oncology annual meeting abstracts up to September 2023. EVIDENCE SYNTHESIS We present an overarching description of the different strategies to target PSMA. The outcomes of PSMA-targeted therapies strongly rely on surface-bound PSMA expression. However, PSMA heterogeneity at different levels (interpatient and inter/intratumoral) limits the efficacy of PSMA-targeted therapies. We highlight the molecular mechanisms governing PSMA regulation, the understanding of which is crucial to designing therapeutic strategies aimed at upregulating PSMA expression. Thus far, homeobox B13 (HOXB13) and androgen receptor (AR) have emerged as critical transcription factors positively and negatively regulating PSMA expression, respectively. Furthermore, epigenetic regulation of PSMA has been also reported recently. In addition, many established therapeutic approaches harbor the potential to upregulate PSMA levels as well as potentiate DNA damage mediated by current radioligands. CONCLUSIONS PSMA-targeted therapies are rapidly advancing, but their efficacy is strongly limited by the heterogeneous expression of the target. A thorough comprehension of how PSMA is regulated will help improve the outcomes through increasing PSMA expression and will provide the basis for synergistic combination therapies. PATIENT SUMMARY Prostate-specific membrane antigen (PSMA) is overexpressed in most prostate cancers. PSMA-targeted therapies have shown promising results, but the heterogeneous expression of PSMA limits their efficacy. We propose to better elucidate the regulation of PSMA expression to increase the levels of the target and improve the therapeutic outcomes.
Collapse
Affiliation(s)
- Matteo Corpetti
- Institute of Oncology Research, Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
| | - Cristina Müller
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland; Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, Switzerland
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Johann de Bono
- The Institute of Cancer Research, London, UK; The Royal Marsden Hospital, London, UK
| | - Jean-Philippe Theurillat
- Institute of Oncology Research, Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland.
| |
Collapse
|
12
|
Arbuznikova D, Eder M, Grosu AL, Meyer PT, Gratzke C, Zamboglou C, Eder AC. Towards Improving the Efficacy of PSMA-Targeting Radionuclide Therapy for Late-Stage Prostate Cancer-Combination Strategies. Curr Oncol Rep 2023; 25:1363-1374. [PMID: 37861915 PMCID: PMC10640479 DOI: 10.1007/s11912-023-01458-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2023] [Indexed: 10/21/2023]
Abstract
PURPOSE OF REVIEW [177Lu]Lu-PSMA-617 is a radiopharmaceutical that emits beta-minus radiation and targets prostate-specific membrane antigen (PSMA)-positive prostate cancer. Despite its clinical success, there are still patients not showing sufficient response rates. This review compiles latest studies aiming at therapy improvement in [177Lu]Lu-PSMA-617-naïve and -resistant patients by alternative or combination treatments. RECENT FINDINGS A variety of agents to combine with [177Lu]Lu-PSMA-617 are currently under investigation including alpha radiation-emitting pharmaceuticals, radiosensitizers, taxane chemotherapeutics, androgen receptor pathway inhibitors, immune checkpoint inhibitors, and external beam radiation. Actinium-225 (225Ac)-labeled PSMA-targeting inhibitors are the most studied pharmaceuticals for combination therapy or as an alternative for treatment after progression under [177Lu]Lu-PSMA-617 therapy. Alpha emitters seem to have a potential of achieving a response to PSMA-targeting radionuclide therapy in both initial non-responders or responders to [177Lu]Lu-PSMA-617 later developing treatment resistance. Emerging evidence for immunostimulatory effects of radiopharmaceuticals and first prospective studies support the combination of [177Lu]Lu-PSMA-617 and immune checkpoint inhibition for late-stage prostate cancer.
Collapse
Affiliation(s)
- Daria Arbuznikova
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
- Division of Radiopharmaceutical Development, German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany and German Cancer Research Center, Heidelberg, Germany
- Department of Radiation Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Matthias Eder
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany.
- Division of Radiopharmaceutical Development, German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany and German Cancer Research Center, Heidelberg, Germany.
| | - Anca-Ligia Grosu
- Department of Radiation Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Philipp T Meyer
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Christian Gratzke
- Department of Urology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Constantinos Zamboglou
- Department of Radiation Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ann-Christin Eder
- Department of Nuclear Medicine, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
- Division of Radiopharmaceutical Development, German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany and German Cancer Research Center, Heidelberg, Germany
| |
Collapse
|
13
|
Sonni I, Gafita A, Unterrainer LM, Alano RM, Lira S, Shen J, Drakaki A, Grogan T, Rettig MB, Czernin J, Calais J. Effects of novel androgen receptor signaling inhibitors on PSMA PET signal intensity in patients with castrate-resistant prostate cancer: a prospective exploratory serial imaging study. EJNMMI Res 2023; 13:95. [PMID: 37902861 PMCID: PMC10616012 DOI: 10.1186/s13550-023-01048-4] [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: 08/15/2023] [Accepted: 10/22/2023] [Indexed: 11/01/2023] Open
Abstract
BACKGROUND PSMA expression is influenced by hormonal status. We evaluated changes in PSA and whole-body 68Ga-PSMA-11 PET/CT (WB-PSMA PET) after initiation of androgen receptor signaling inhibitors (ARSi). METHODS Prospectively enrolled patients with metastatic castration-resistant prostate cancer (mCRPC) initiating ARSi underwent serial PSA measurements and WB-PSMA PET at baseline, 1-week, and 3-months post-ARSi. We correlated WB-PSMA PET metrics and PSA kinetics after ARSi to 1-year clinical outcome. RESULTS Due to low enrollment rate, the study was closed before reaching the recruitment goal of 30 patients. Nine patients were enrolled. At 1-year, unfavorable outcome was documented in 6/9 (66%) patients. Nine/9 patients completed PSMA PET at 1-week, 5/9 at 3-months. Changes in PSA, PSMA-VOL, SUVmean and SUVmax were - 12%, + 5%, + 3%, and + 10% at 1-week, - 42%, - 16%, - 15% and - 17% at 3-months, respectively. CONCLUSIONS Our prospective trial involving 9 mCRPC patients initiating ARSi did not show significant modulation of PSMA expression measured on WB-PSMA PET at 1-week. This study was registered on clinicaltrials.gov (NCT04279561).
Collapse
Affiliation(s)
- Ida Sonni
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Radiological Sciences, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095-7370, USA.
- Department of Experimental and Clinical Medicine, University Magna Graecia, Catanzaro, Italy.
| | - Andrei Gafita
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Lena M Unterrainer
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Nuclear Medicine, Ludwig Maximilian University of Munich LMU, Munich, Germany
| | - Rejah M Alano
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Stephanie Lira
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - John Shen
- Department of Medical Oncology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Alexandra Drakaki
- Department of Medical Oncology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Tristan Grogan
- Department of Medicine Statistics Core, University of California, Los Angeles, Los Angeles, CA, USA
| | - Matthew B Rettig
- Department of Medical Oncology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Johannes Czernin
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jeremie Calais
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
14
|
Alati S, Singh R, Pomper MG, Rowe SP, Banerjee SR. Preclinical Development in Radiopharmaceutical Therapy for Prostate Cancer. Semin Nucl Med 2023; 53:663-686. [PMID: 37468417 DOI: 10.1053/j.semnuclmed.2023.06.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/21/2023]
Abstract
Prostate cancer is a leading cause of cancer death in men worldwide. Among the various treatment options, radiopharmaceutical therapy has shown notable success in metastatic, castration-resistant disease. Radiopharmaceutical therapy is a systemic approach that delivers cytotoxic radiation doses precisely to the malignant tumors and/or tumor microenvironment. Therapeutic radiopharmaceuticals are composed of a therapeutic radionuclide and a high-affinity, tumor-targeting carrier molecule. Therapeutic radionuclides used in preclinical prostate cancer studies are primarily α-, β--, or Auger-electron-emitting radiometals or radiohalogens. Monoclonal antibodies, antibody-derived fragments, peptides, and small molecules are frequently used as tumor-targeting molecules. Over the years, several important membrane-associated proteases and receptors have been identified, validated, and subsequently used for preclinical radiotherapeutic development for prostate cancer. Prostate-specific membrane antigen (PSMA) is the most well-studied prostate cancer-associated protease in preclinical literature. PSMA-targeting radiotherapeutic agents are being investigated using high-affinity antibody- and small-molecule-based agents for safety and efficacy. Early generations of such agents were developed simply by replacing radionuclides of the imaging agents with therapeutic ones. Later, extensive structure-activity relationship studies were conducted to address the safety and efficacy issues obtained from initial patient data. Recent regulatory approval of the 177Lu-labeled low-molecular-weight agent, 177Lu-PSMA-617, is a significant accomplishment. Current preclinical experiments are focused on the structural modification of 177Lu-PSMA-617 and relevant investigational agents to increase tumor targeting and reduce off-target binding and toxicity in healthy organs. While lutetium-177 (177Lu) remains the most widely used radionuclide, radiolabeled analogs with iodine-131 (128I), yttrium-90 (89Y), copper-67 (67Cu), and terbium-161 (161Tb) have been evaluated as potential alternatives in recent years. In addition, agents carrying the α-particle-emitting radiohalogen, astatine-211 (211At), or radiometals, actinium-225 (225Ac), lead-212 (212Pb), radium-223 (223Ra), and thorium-227 (227Th), have been increasingly investigated in preclinical research. Besides PSMA-based radiotherapeutics, other prominent prostate cancer-related proteases, for example, human kallikrein peptidases (HK2 and HK3), have been explored using monoclonal-antibody-(mAb)-based targeting platforms. Several promising mAbs targeting receptors overexpressed on the different stages of prostate cancer have also been developed for radiopharmaceutical therapy, for example, Delta-like ligand 3 (DLL-3), CD46, and CUB domain-containing protein 1 (CDCP1). Progress is also being made using peptide-based targeting platforms for the gastrin-releasing peptide receptor (GRPR), a well-established membrane-associated receptor expressed in localized and metastatic prostate cancers. Furthermore, mechanism-driven combination therapies appear to be a burgeoning area in the context of preclinical prostate cancer radiotherapeutics. Here, we review the current developments related to the preclinical radiopharmaceutical therapy of prostate cancer. These are summarized in two major topics: (1) therapeutic radionuclides and (2) tumor-targeting approaches using monoclonal antibodies, small molecules, and peptides.
Collapse
Affiliation(s)
- Suresh Alati
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD; Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Rajan Singh
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD; Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Martin G Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD; Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Steven P Rowe
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD; Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD
| | - Sangeeta Ray Banerjee
- Russell H. Morgan Department of Radiology and Radiological Science, Baltimore, MD; Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD.
| |
Collapse
|
15
|
Kuzmanov A, Salemi S, Schmid FA, Burger IA, Eberli D, Kranzbühler B. Improved Prostate-Specific Membrane Antigen (PSMA) Stimulation Using a Super Additive Effect of Dutasteride and Lovastatin In Vitro. Int J Mol Sci 2023; 24:12338. [PMID: 37569712 PMCID: PMC10419009 DOI: 10.3390/ijms241512338] [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: 07/07/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023] Open
Abstract
Prostate-specific membrane antigen (PSMA)-based imaging improved the detection of primary, recurrent and metastatic prostate cancer. However, in certain patients, a low PSMA surface expression can be a limitation for this promising diagnostic tool. Pharmacological induction of PSMA might be useful to further improve the detection rate of PSMA-based imaging. To achieve this, we tested dutasteride (Duta)-generally used for treatment of benign prostatic enlargement-and lovastatin (Lova)-a compound used to reduce blood lipid concentrations. We aimed to compare the individual effects of Duta and Lova on cell proliferation as well as PSMA expression. In addition, we tested if a combination treatment using lower concentrations of Duta and Lova can further induce PSMA expression. Our results show that a treatment with ≤1 μM Duta and ≥1 μM Lova lead to a significant upregulation of whole and cell surface PSMA expression in LNCaP, C4-2 and VCaP cells. Lower concentrations of Duta and Lova in combination (0.5 μM Duta + 0.5 μM Lova or 0.5 μM Duta + 1 μM Lova) were further capable of enhancing PSMA protein expression compared to a single compound treatment using higher concentrations in all tested cell lines (LNCaP, C4-2 and VCaP).
Collapse
Affiliation(s)
- Aleksandar Kuzmanov
- Laboratory for Urologic Oncology and Stem Cell Therapy, Department of Urology, University Hospital Zürich, University of Zurich, 8091 Zurich, Switzerland
| | - Souzan Salemi
- Laboratory for Urologic Oncology and Stem Cell Therapy, Department of Urology, University Hospital Zürich, University of Zurich, 8091 Zurich, Switzerland
| | - Florian A. Schmid
- Laboratory for Urologic Oncology and Stem Cell Therapy, Department of Urology, University Hospital Zürich, University of Zurich, 8091 Zurich, Switzerland
| | - Irene A. Burger
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8091 Zurich, Switzerland
- Department of Nuclear Medicine, Baden Cantonal Hospital, 5404 Baden, Switzerland
| | - Daniel Eberli
- Laboratory for Urologic Oncology and Stem Cell Therapy, Department of Urology, University Hospital Zürich, University of Zurich, 8091 Zurich, Switzerland
| | - Benedikt Kranzbühler
- Laboratory for Urologic Oncology and Stem Cell Therapy, Department of Urology, University Hospital Zürich, University of Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
16
|
Nabavi N, Mahdavi SR, Ardalan MA, Chamanara M, Mosaed R, Lara A, Bastos D, Harsini S, Askari E, Velho PI, Bagheri H. Bipolar Androgen Therapy: When Excess Fuel Extinguishes the Fire. Biomedicines 2023; 11:2084. [PMID: 37509723 PMCID: PMC10377678 DOI: 10.3390/biomedicines11072084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/12/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Androgen deprivation therapy (ADT) remains the cornerstone of advanced prostate cancer treatment. However, the progression towards castration-resistant prostate cancer is inevitable, as the cancer cells reactivate androgen receptor signaling and adapt to the castrate state through autoregulation of the androgen receptor. Additionally, the upfront use of novel hormonal agents such as enzalutamide and abiraterone acetate may result in long-term toxicities and may trigger the selection of AR-independent cells through "Darwinian" treatment-induced pressure. Therefore, it is crucial to develop new strategies to overcome these challenges. Bipolar androgen therapy (BAT) is one such approach that has been devised based on studies demonstrating the paradoxical inhibitory effects of supraphysiologic testosterone on prostate cancer growth, achieved through a variety of mechanisms acting in concert. BAT involves rapidly alternating testosterone levels between supraphysiological and near-castrate levels over a period of a month, achieved through monthly intramuscular injections of testosterone plus concurrent ADT. BAT is effective and well-tolerated, improving quality of life and potentially re-sensitizing patients to previous hormonal therapies after progression. By exploring the mechanisms and clinical evidence for BAT, this review seeks to shed light on its potential as a promising new approach to prostate cancer treatment.
Collapse
Affiliation(s)
- Nima Nabavi
- Nuclear Medicine Research Center, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran
- Radiation Sciences Research Center, AJA University of Medical Sciences, Tehran 14117-18541, Iran
| | - Seied Rabi Mahdavi
- Department of Medical Physics, Radiation Biology Research Center, Iran University of Medical Sciences, Tehran 14117-18541, Iran
| | - Mohammad Afshar Ardalan
- Department of Internal Medicine, School of Medicine, AJA University of Medical Sciences, Tehran 14117-18541, Iran
| | - Mohsen Chamanara
- Department of Pharmacology, School of Medicine, AJA University of Medical Sciences, Tehran 14117-18541, Iran
| | - Reza Mosaed
- Department of Clinical Pharmacy, School of Medicine, AJA University of Medical Sciences, Tehran 14117-18541, Iran
| | - Aline Lara
- Hospital Sírio-Libanês, São Paulo 01308-050, Brazil
- Hospital do Câncer UOPECCAN, Cascavel 85806-300, Brazil
| | - Diogo Bastos
- Oncology Department, Hospital Sirio-Libanês, São Paulo 01308-050, Brazil
| | - Sara Harsini
- BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
| | - Emran Askari
- Nuclear Medicine Research Center, Mashhad University of Medical Sciences, Mashhad 13944-91388, Iran
| | - Pedro Isaacsson Velho
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD 21231, USA
- Hospital Moinhos de Vento, Porto Alegre 90035-000, Brazil
| | - Hamed Bagheri
- Radiation Sciences Research Center, AJA University of Medical Sciences, Tehran 14117-18541, Iran
- School of Medicine, AJA University of Medical Sciences, Tehran 14118-13389, Iran
| |
Collapse
|
17
|
Simon H, Henkel D, Chiron P, Helissey C. New perspectives on metabolic imaging in the management of prostate cancer in 2022: A focus on radiolabeled PSMA‑PET/CT (Review). Mol Clin Oncol 2023; 19:51. [PMID: 37323248 PMCID: PMC10265585 DOI: 10.3892/mco.2023.2647] [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: 11/07/2022] [Accepted: 01/04/2023] [Indexed: 06/17/2023] Open
Abstract
Nuclear medicine is an essential part of prostate cancer management concerning initial staging, patient follow-up and even therapy. Prostate-specific membrane antigen (PSMA) is a glutamate carboxypeptidase II transmembrane glycoprotein expressed by 80% of prostatic cells. The interest in this protein is due to its specificity for prostatic tissue. The use of 68GaPSMA PET/CT in the context of disease staging is thus well-established and recommended, especially for high-risk disease with metastases and lymph node involvement. However, the risk of false positives raises questions regarding its place in the management of patients with prostate cancer. The present study aimed to determine the use of PET-PSMA in the care of patients with prostate cancer but also to assess its limits of use.
Collapse
Affiliation(s)
- Hélène Simon
- Clinical Research Unit, Department of Oncology, Military Hospital Begin, 94160 Saint-Mandé, France
| | - Daniel Henkel
- Unité de Formation et de Recherche 5, University of Paris 8 Vincennes-St. Denis, 93200 Paris, France
| | - Paul Chiron
- Department of Urology, Military Hospital Begin, 94160 Saint-Mandé, France
| | - Carole Helissey
- Clinical Research Unit, Department of Oncology, Military Hospital Begin, 94160 Saint-Mandé, France
| |
Collapse
|
18
|
Derlin T, Riethdorf S, Schumacher U, Lafos M, Peine S, Coith C, Ross TL, Pantel K, Bengel FM. PSMA-heterogeneity in metastatic castration-resistant prostate cancer: Circulating tumor cells, metastatic tumor burden, and response to targeted radioligand therapy. Prostate 2023. [PMID: 37147881 DOI: 10.1002/pros.24549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/11/2023] [Accepted: 04/24/2023] [Indexed: 05/07/2023]
Abstract
BACKGROUND We explored the interrelation between prostate-specific membrane antigen (PSMA) expression on circulating tumor cells (CTCs) and that of solid metastatic lesions as determined by whole-body PSMA-targeted positron emission tomography (PET) to refine the prediction of response to subsequent PSMA-targeted radioligand therapy (RLT). METHODS A prospective study was performed in 20 patients with advanced mCRPC. Of these, 16 underwent subsequent RLT with [177 Lu]Lu-PSMA-617 at a dose of 7.4 GBq every 6-8 weeks. PSMA expression on CTCs using the CellSearch system was compared to clinical and serological results, and to marker expression in targeted imaging and available histological sections of prostatectomy specimens (19% of RLT patients). Clinical outcome was obtained after two cycles of RLT. RESULTS Marked heterogeneity of PSMA expression was observed already at first diagnosis in available histological specimens. Targeted whole-body imaging also showed heterogeneous inter- and intra-patient PSMA expression between metastases. Heterogeneity of CTC PSMA expression was partially paralleled by heterogeneity of whole-body tumor burden PSMA expression. Twenty percent of CTC samples showed no PSMA expression, despite unequivocal PSMA expression of solid metastases at PET. A high fraction of PSMA-negative CTCs emerged as the sole predictor of poor RLT response (odds ratio [OR]: 0.9379 [95% confidence interval, CI, 0.8558-0.9902]; p = 0.0160), and was prognostic for both shorter progression-free survival (OR: 1.236 [95% CI, 1.035-2.587]; p = 0.0043) and overall survival (OR: 1.056 [95% CI, 1.008-1.141]; p = 0.0182). CONCLUSION This proof-of-principle study suggests that liquid biopsy for CTC PSMA expression is complementary to PET for individual PSMA phenotyping of mCRPC.
Collapse
Affiliation(s)
- Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Sabine Riethdorf
- University Medical Center Hamburg-Eppendorf, Institute of Tumor Biology, Hamburg, Germany
| | - Udo Schumacher
- Department of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Medical School Berlin, Berlin, Germany
| | - Marcel Lafos
- Hannover Medical School, Institute of Pathology, Hannover, Germany
| | - Sven Peine
- University Medical Center Hamburg-Eppendorf, Institute of Transfusion Medicine, Hamburg, Germany
| | - Cornelia Coith
- University Medical Center Hamburg-Eppendorf, Institute of Tumor Biology, Hamburg, Germany
| | - Tobias L Ross
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Klaus Pantel
- University Medical Center Hamburg-Eppendorf, Institute of Tumor Biology, Hamburg, Germany
| | - Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| |
Collapse
|
19
|
Duriseti S, Berenji G, Tsai S, Rettig M, Nickols NG. Quantitative assessment of PSMA PET response to therapy in castration-sensitive prostate cancer using an automated imaging platform for disease identification and measurement. Eur J Hybrid Imaging 2023; 7:7. [PMID: 37009941 PMCID: PMC10068685 DOI: 10.1186/s41824-023-00165-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 02/14/2023] [Indexed: 04/04/2023] Open
Abstract
RATIONALE Prostate cancer treatment response may be automatically quantified using a molecular imaging analysis platform targeting prostate-specific membrane antigen (PSMA). METHODS A retrospective analysis of patients with castration-sensitive prostate cancer who underwent PSMA-targeted molecular imaging prior to and 3 months or more after treatment was conducted. Disease burden was analyzed with aPROMISE, an artificial intelligence imaging platform that automatically quantifies PSMA-positive lesions. The calculated PSMA scores for prostate/bed, nodal, and osseous disease sites were compared with prostate-specific antigen (PSA) values. RESULTS Of 30 eligible patients, the median decline in prostate/bed, nodal, and osseous disease PSMA scores were 100% (range 52-100%), 100% (range - 87-100%), and 100% (range - 21-100%), respectively. PSMA score decline was significantly associated with PSA decline. CONCLUSION Changes in aPROMISE PSMA scores are associated with changes in PSA and may quantify treatment response.
Collapse
Affiliation(s)
- Sai Duriseti
- VA Greater Los Angeles, Radiation Oncology Service, 11301 Wilshire Blvd, Building 500, Suite 0426, Los Angeles, CA, 90073, USA.
- Departments of Radiation Oncology, University of California, Los Angeles, 200 UCLA Medical Plaza, Suite B265, Los Angeles, CA, 90095, USA.
| | - Gholam Berenji
- VA Greater Los Angeles, Nuclear Medicine Service, 11301 Wilshire Blvd, Building 500, Suite 0090, Los Angeles, CA, 90073, USA
- Radiological Sciences, University of California, Los Angeles, 200 Medical Plaza, Suite B-114, Los Angeles, CA, 90095, USA
| | - Sonny Tsai
- Greater Los Angeles VA, Hematology and Oncology Section, 11301 Wilshire Blvd. Building 304, Suite E2-218, Los Angeles, CA, 90073, USA
| | - Matthew Rettig
- Greater Los Angeles VA, Hematology and Oncology Section, 11301 Wilshire Blvd. Building 304, Suite E2-218, Los Angeles, CA, 90073, USA
- Department of Urology, University of California, Los Angeles, 200 Medical Plaza, Suite 140, Los Angeles, CA, 90095, USA
- Department of Medicine, University of California, Los Angeles, 200 Medical Plaza, Suite 140, Los Angeles, CA, 90095, USA
| | - Nicholas G Nickols
- VA Greater Los Angeles, Radiation Oncology Service, 11301 Wilshire Blvd, Building 500, Suite 0426, Los Angeles, CA, 90073, USA
- Departments of Radiation Oncology, University of California, Los Angeles, 200 UCLA Medical Plaza, Suite B265, Los Angeles, CA, 90095, USA
- Department of Urology, University of California, Los Angeles, 200 Medical Plaza, Suite 140, Los Angeles, CA, 90095, USA
| |
Collapse
|
20
|
Fendler WP, Eiber M, Beheshti M, Bomanji J, Calais J, Ceci F, Cho SY, Fanti S, Giesel FL, Goffin K, Haberkorn U, Jacene H, Koo PJ, Kopka K, Krause BJ, Lindenberg L, Marcus C, Mottaghy FM, Oprea-Lager DE, Osborne JR, Piert M, Rowe SP, Schöder H, Wan S, Wester HJ, Hope TA, Herrmann K. PSMA PET/CT: joint EANM procedure guideline/SNMMI procedure standard for prostate cancer imaging 2.0. Eur J Nucl Med Mol Imaging 2023; 50:1466-1486. [PMID: 36604326 PMCID: PMC10027805 DOI: 10.1007/s00259-022-06089-w] [Citation(s) in RCA: 169] [Impact Index Per Article: 84.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Accepted: 12/18/2022] [Indexed: 01/07/2023]
Abstract
Here we aim to provide updated guidance and standards for the indication, acquisition, and interpretation of PSMA PET/CT for prostate cancer imaging. Procedures and characteristics are reported for a variety of available PSMA small radioligands. Different scenarios for the clinical use of PSMA-ligand PET/CT are discussed. This document provides clinicians and technicians with the best available evidence, to support the implementation of PSMA PET/CT imaging in research and routine practice.
Collapse
Affiliation(s)
- Wolfgang P Fendler
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
- PET Committee of the German Society of Nuclear Medicine, Marburg, Germany
| | - Matthias Eiber
- Department of Nuclear Medicine, Klinikum Rechts Der Isar, Technical University of Munich, Munich, Germany
| | - Mohsen Beheshti
- Division of Molecular Imaging & Theranostics, Department of Nuclear Medicine, University Hospital Salzburg, Paracelsus Medical University, Salzburg, Austria
| | - Jamshed Bomanji
- Institute of Nuclear Medicine, UCLH NHS Foundation Trust, London, UK
| | - Jeremie Calais
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA, USA
| | - Francesco Ceci
- Division of Nuclear Medicine and Theranostics, IEO European Institute of Oncology, IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Steve Y Cho
- Department of Radiology, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | | | - Frederik L Giesel
- Department of Nuclear Medicine, University Hospital Düsseldorf, Medical Faculty, Heinrich-Heine-University and Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Karolien Goffin
- Department of Nuclear Medicine, Division of Nuclear Medicine and Molecular Imaging, University Hospital Leuven, KU Leuven, Louvain, Belgium
| | - Uwe Haberkorn
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Heather Jacene
- Dana-Farber Cancer Institute/Brigham and Women's Hospital, Boston, USA
| | | | - Klaus Kopka
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- School of Science, Faculty of Chemistry and Food Chemistry, Technical University Dresden, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, Dresden, Germany
| | - Bernd J Krause
- Department of Nuclear Medicine, University Medical Center, University of Rostock, Rostock, Germany
| | - Liza Lindenberg
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Charles Marcus
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Emory University School of Medicine, Atlanta, GA, USA
| | - Felix M Mottaghy
- Department of Nuclear Medicine, University Hospital RWTH Aachen University, Aachen, Germany
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center (MUMC+), Maastricht, The Netherlands
| | - Daniela E Oprea-Lager
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical Centers, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | - Joseph R Osborne
- Department of Radiology, Division of Molecular Imaging and Therapeutics, Weill Cornell Medicine, New York, NY, USA
| | - Morand Piert
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, University of Michigan, Ann Arbor, MI, USA
| | - Steven P Rowe
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Heiko Schöder
- Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Simon Wan
- Institute of Nuclear Medicine, UCLH NHS Foundation Trust, London, UK
| | - Hans-Jürgen Wester
- Pharmaceutical Radiochemistry, Technische Universität München, Walther-Meißner-Str. 3, 85748, Garching, Germany
| | - Thomas A Hope
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany.
| |
Collapse
|
21
|
Storey CM, Altai M, Bicak M, Veach DR, Lückerath K, Adrian G, McDevitt MR, Kalidindi T, Park JE, Herrmann K, Abou D, Zedan W, Peekhaus N, Klein RJ, Damoiseaux R, Larson SM, Lilja H, Thorek D, Ulmert D. Quantitative In Vivo Imaging of the Androgen Receptor Axis Reveals Degree of Prostate Cancer Radiotherapy Response. Mol Cancer Res 2023; 21:307-315. [PMID: 36608299 PMCID: PMC10355285 DOI: 10.1158/1541-7786.mcr-22-0736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 12/13/2022] [Accepted: 01/03/2023] [Indexed: 01/09/2023]
Abstract
Noninvasive biomarkers for androgen receptor (AR) pathway activation are urgently needed to better monitor patient response to prostate cancer therapies. AR is a critical driver and mediator of resistance of prostate cancer but currently available noninvasive prostate cancer biomarkers to monitor AR activity are discordant with downstream AR pathway activity. External beam radiotherapy (EBRT) remains a common treatment for all stages of prostate cancer, and DNA damage induced by EBRT upregulates AR pathway activity to promote therapeutic resistance. [89Zr]11B6-PET is a novel modality targeting prostate-specific protein human kallikrein 2 (hK2), which is a surrogate biomarker for AR activity. Here, we studied whether [89Zr]11B6-PET can accurately assess EBRT-induced AR activity.Genetic and human prostate cancer mouse models received EBRT (2-50 Gy) and treatment response was monitored by [89Zr]11B6-PET/CT. Radiotracer uptake and expression of AR and AR target genes was quantified in resected tissue.EBRT increased AR pathway activity and [89Zr]11B6 uptake in LNCaP-AR and 22RV1 tumors. EBRT increased prostate-specific [89Zr]11B6 uptake in prostate cancer-bearing mice (Hi-Myc x Pb_KLK2) with no significant changes in uptake in healthy (Pb_KLK2) mice, and this correlated with hK2 protein levels. IMPLICATIONS hK2 expression in prostate cancer tissue is a proxy of EBRT-induced AR activity that can noninvasively be detected using [89Zr]11B6-PET; further clinical evaluation of hK2-PET for monitoring response and development of resistance to EBRT in real time is warranted.
Collapse
Affiliation(s)
- Claire M Storey
- Department of Molecular & Medical Pharmacology, University of California Los Angeles (UCLA), Los Angeles, USA
| | - Mohamed Altai
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Mesude Bicak
- Hasso Plattner Institute for Digital Health, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Darren R Veach
- Department of Radiology, Memorial Sloan Kettering Cancer Center (MSKCC), New York, USA
| | - Katharina Lückerath
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, DKTK, Essen, Germany
| | - Gabriel Adrian
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Michael R McDevitt
- Department of Radiology, Memorial Sloan Kettering Cancer Center (MSKCC), New York, USA
| | - Teja Kalidindi
- Department of Radiology, Memorial Sloan Kettering Cancer Center (MSKCC), New York, USA
| | - Julie E Park
- Department of Molecular & Medical Pharmacology, University of California Los Angeles (UCLA), Los Angeles, USA
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, DKTK, Essen, Germany
| | - Diane Abou
- Department of Radiology, Washington University School of Medicine, St. Louis, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, USA
| | - Wahed Zedan
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Norbert Peekhaus
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Robert J Klein
- Icahn Institute for Genomics and Multiscale Biology, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Robert Damoiseaux
- Department of Molecular & Medical Pharmacology, University of California Los Angeles (UCLA), Los Angeles, USA
- California NanoSystems Institute, UCLA, Los Angeles, USA
| | - Steven M Larson
- Department of Radiology, Memorial Sloan Kettering Cancer Center (MSKCC), New York, USA
- Department of Radiology, Weill Cornell Medical College, New York, USA
| | - Hans Lilja
- Genitourinary Oncology Service, Department of Medicine, MSKCC, New York, USA
- Urology Service, Department of Surgery, MSKCC, New York, USA
- Department of Laboratory Medicine, MSKCC, New York, USA
- Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Daniel Thorek
- Department of Radiology, Washington University School of Medicine, St. Louis, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, USA
- Department of Biomedical Engineering, Washington University School of Medicine, St. Louis, USA
| | - David Ulmert
- Department of Molecular & Medical Pharmacology, University of California Los Angeles (UCLA), Los Angeles, USA
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Lund, Sweden
- California NanoSystems Institute, UCLA, Los Angeles, USA
- Department of Urology, Institute of Urologic Oncology, UCLA, Los Angeles, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, UCLA, Los Angeles, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, USA
| |
Collapse
|
22
|
Esen B, Herrmann K, Bavbek S, Kordan Y, Tilki D, Esen T. Prostate-specific Membrane Antigen Positron Emission Tomography as a Biomarker to Assess Treatment Response in Patients with Advanced Prostate Cancer. Eur Urol Focus 2023:S2405-4569(23)00040-8. [PMID: 36842919 DOI: 10.1016/j.euf.2023.02.001] [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: 12/21/2022] [Revised: 01/11/2023] [Accepted: 02/08/2023] [Indexed: 02/28/2023]
Abstract
CONTEXT Prostate-specific membrane antigen (PSMA)-targeted positron emission tomography (PET) has superior accuracy for detection of metastatic lesions in patients with prostate cancer (PC). Although PSMA PET has a prominent role in primary and secondary imaging of PC, data on its role in assessing treatment response in advanced PC are limited. OBJECTIVE To review current data in the literature regarding the impact of antiandrogen therapy on PSMA expression of metastatic sites and the role of serial (baseline and at least 1 follow-up scan) PSMA PET to assess treatment response in patients with metastatic PC. EVIDENCE ACQUISITION A comprehensive literature search in the PubMed database was performed using the terms "PSMA expression prostate", "PSMA regulation", "PSMA PET response assessment", and "serial PSMA PET". EVIDENCE SYNTHESIS Serial PSMA PET studies (baseline and at least 1 follow-up scan) provide valuable data regarding PSMA expression changes after systemic treatment in patients with metastatic PC. PSMA PET-detected flare and upregulation of PSMA expression following hormonal intervention seem to be early events resolving after 3 mo of treatment. PSMA PET imaging is essential in selecting patients for 177Lu-PSMA radioligand therapy (RLT). Growing evidence favors its use in assessing treatment responses after RLT. Preliminary evidence indicates the value of PSMA PET for assessment of the treatment response in patients receiving systemic treatment other than RLT for metastatic PC. CONCLUSIONS PSMA flare following antiandrogen therapy seems to be an early event and thus PET scans should be performed no earlier than 3 mo after the start of treatment. PSMA PET has a promising role in tailoring treatment according to the specific needs of individual patients and assessing responses following systemic treatment in patients with advanced PC. PATIENT SUMMARY This review describes how a sensitive imaging method can be used to assess the tumor response to treatment for metastatic prostate cancer.
Collapse
Affiliation(s)
- Barış Esen
- Department of Urology, School of Medicine, Koç University, Istanbul, Turkey.
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium-University Hospital Essen, Essen, Germany
| | - Sevil Bavbek
- Department of Medical Oncology, VKF American Hospital, Istanbul, Turkey
| | - Yakup Kordan
- Department of Urology, School of Medicine, Koç University, Istanbul, Turkey
| | - Derya Tilki
- Department of Urology, School of Medicine, Koç University, Istanbul, Turkey; Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, Hamburg, Germany; Department of Urology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Tarık Esen
- Department of Urology, School of Medicine, Koç University, Istanbul, Turkey
| |
Collapse
|
23
|
van der Gaag S, Bartelink IH, Vis AN, Burchell GL, Oprea-Lager DE, Hendrikse H. Pharmacological Optimization of PSMA-Based Radioligand Therapy. Biomedicines 2022; 10:3020. [PMID: 36551776 PMCID: PMC9775864 DOI: 10.3390/biomedicines10123020] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/09/2022] [Accepted: 11/18/2022] [Indexed: 11/25/2022] Open
Abstract
Prostate cancer (PCa) is the most common malignancy in men of middle and older age. The standard treatment strategy for PCa ranges from active surveillance in low-grade, localized PCa to radical prostatectomy, external beam radiation therapy, hormonal treatment and chemotherapy. Recently, the use of prostate-specific membrane antigen (PSMA)-targeted radioligand therapy (RLT) for metastatic castration-resistant PCa has been approved. PSMA is predominantly, but not exclusively, expressed on PCa cells. Because of its high expression in PCa, PSMA is a promising target for diagnostics and therapy. To understand the currently used RLT, knowledge about pharmacokinetics (PK) and pharmacodynamics (PD) of the PSMA ligand and the PSMA protein itself is crucial. PK and PD properties of the ligand and its target determine the duration and extent of the effect. Knowledge on the concentration-time profile, the target affinity and target abundance may help to predict the effect of RLT. Increased specific binding of radioligands to PSMA on PCa cells may be associated with better treatment response, where nonspecific binding may increase the risk of toxicity in healthy organs. Optimization of the radioligand, as well as synergistic effects of concomitant agents and an improved dosing strategy, may lead to more individualized treatment and better overall survival.
Collapse
Affiliation(s)
- Suzanne van der Gaag
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
| | - Imke H. Bartelink
- Cancer Center Amsterdam, Imaging and Biomarkers, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
- Department of Clinical Pharmacology and Pharmacy, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - André N. Vis
- Department of Urology, Prostate Cancer Network Amsterdam, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - George L. Burchell
- Medical Library, VU University, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Daniela E. Oprea-Lager
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
| | - Harry Hendrikse
- Department of Radiology and Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
24
|
Sheehan B, Guo C, Neeb A, Paschalis A, Sandhu S, de Bono JS. Prostate-specific Membrane Antigen Biology in Lethal Prostate Cancer and its Therapeutic Implications. Eur Urol Focus 2022; 8:1157-1168. [PMID: 34167925 DOI: 10.1016/j.euf.2021.06.006] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/07/2021] [Accepted: 06/09/2021] [Indexed: 12/16/2022]
Abstract
CONTEXT Prostate-specific membrane antigen (PSMA) is a promising, novel theranostic target in advanced prostate cancer (PCa). Multiple PSMA-targeted therapies are currently in clinical development, with some agents showing impressive antitumour activity, although optimal patient selection and therapeutic resistance remain ongoing challenges. OBJECTIVE To review the biology of PSMA and recent advances in PSMA-targeted therapies in PCa, and to discuss potential strategies for patient selection and further therapeutic development. EVIDENCE ACQUISITION A comprehensive literature search was performed using PubMed and review of American Society of Clinical Oncology and European Society of Medical Oncology annual meeting abstracts up to April 2021. EVIDENCE SYNTHESIS PSMA is a largely extracellular protein that is frequently, but heterogeneously, expressed by PCa cells. PSMA expression is associated with disease progression, worse clinical outcomes and the presence of tumour defects in DNA damage repair (DDR). PSMA is also expressed by other cancer cell types and is implicated in glutamate and folate metabolism. It may confer a tumour survival advantage in conditions of cellular stress. PSMA regulation is complex, and recent studies have shed light on interactions with androgen receptor, PI3K/Akt, and DDR signalling. A phase 2 clinical trial has shown that 177Lu-PSMA-617 causes tumour shrinkage and delays disease progression in a significant subset of patients with metastatic castration-resistant PCa in comparison to second-line chemotherapy. Numerous novel PSMA-targeting immunotherapies, small molecules, and antibody therapies are currently in clinical development, including in earlier stages of PCa, with emerging evidence of antitumour activity. To date, the regulation and function of PSMA in PCa cells remain poorly understood. CONCLUSIONS There has been rapid recent progress in PSMA-targeted therapies for the management of advanced PCa. Dissection of PSMA biology will help to identify biomarkers for and resistance mechanisms to these therapies and facilitate further therapeutic development to improve PCa patient outcomes. PATIENT SUMMARY There have been major advances in the development of therapies targeting a molecule, PSMA, in PCa. Radioactive molecules targeting PSMA can cause tumour shrinkage and delay progression in some patients with lethal disease. Future studies are needed to determine which patients are most likely to respond, and how other treatments can be combined with therapies targeting PSMA so that more patients may benefit.
Collapse
Affiliation(s)
| | - Christina Guo
- The Institute of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, London, UK
| | - Antje Neeb
- The Institute of Cancer Research, London, UK
| | - Alec Paschalis
- The Institute of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, London, UK
| | - Shahneen Sandhu
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia; University of Melbourne, Melbourne, Australia
| | - Johann S de Bono
- The Institute of Cancer Research, London, UK; The Royal Marsden NHS Foundation Trust, London, UK.
| |
Collapse
|
25
|
Stenberg VY, Tornes AJK, Nilsen HR, Revheim ME, Bruland ØS, Larsen RH, Juzeniene A. Factors Influencing the Therapeutic Efficacy of the PSMA Targeting Radioligand 212Pb-NG001. Cancers (Basel) 2022; 14:cancers14112784. [PMID: 35681766 PMCID: PMC9179904 DOI: 10.3390/cancers14112784] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/18/2022] [Accepted: 06/01/2022] [Indexed: 12/10/2022] Open
Abstract
Simple Summary Prostate-specific membrane antigen (PSMA) is a protein overexpressed in metastatic castration-resistant prostate cancer and a promising target for targeted radionuclide therapy. PSMA-targeted alpha therapy is of growing interest due to the high-emission energy and short range of alpha particles, resulting in a prominent cytotoxic potency. This study assesses the influence of various factors on the in vitro and in vivo therapeutic efficacy of the alpha particle generating PSMA-targeting radioligand 212Pb-NG001. Abstract This study aimed to determine the influence of cellular PSMA expression, radioligand binding and internalization, and repeated administrations on the therapeutic effects of the PSMA-targeting radioligand 212Pb-NG001. Cellular binding and internalization, cytotoxicity, biodistribution, and the therapeutic efficacy of 212Pb-NG001 were investigated in two human prostate cancer cell lines with different PSMA levels: C4-2 (PSMA+) and PC-3 PIP (PSMA+++). Despite 10-fold higher PSMA expression on PC-3 PIP cells, cytotoxicity and therapeutic efficacy of the radioligand was only 1.8-fold better than for the C4-2 model, possibly explained by lower cellular internalization and less blood-rich stroma in PC-3 PIP xenografts. Mice bearing subcutaneous PC-3 PIP xenografts were treated with 0.2, 0.4, and 0.8 MBq of 212Pb-NG001 that resulted in therapeutic indexes of 2.7, 3.0, and 3.5, respectively. A significant increase in treatment response was observed in mice that received repeated injections compared to the corresponding single dose (therapeutic indexes of 3.6 for 2 × 0.2 MBq and 4.4 for 2 × 0.4 MBq). The results indicate that 212Pb-NG001 can induce therapeutic effects at clinically transferrable doses, both in the C4-2 model that resembles solid tumors and micrometastases with natural PSMA expression and in the PC-3 PIP model that mimics poorly vascularized metastases.
Collapse
Affiliation(s)
- Vilde Yuli Stenberg
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway; (A.J.K.T.); (A.J.)
- Nucligen AS, 0379 Oslo, Norway;
- Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway; (M.-E.R.); (Ø.S.B.)
- Correspondence: ; Tel.: +47-9012-8434
| | - Anna Julie Kjøl Tornes
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway; (A.J.K.T.); (A.J.)
- Nucligen AS, 0379 Oslo, Norway;
- Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway; (M.-E.R.); (Ø.S.B.)
| | - Hogne Røed Nilsen
- Department of Pathology, Rikshospitalet, Oslo University Hospital, 0372 Oslo, Norway;
| | - Mona-Elisabeth Revheim
- Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway; (M.-E.R.); (Ø.S.B.)
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, 0379 Oslo, Norway
| | - Øyvind Sverre Bruland
- Institute of Clinical Medicine, University of Oslo, 0318 Oslo, Norway; (M.-E.R.); (Ø.S.B.)
- Department of Oncology, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
| | | | - Asta Juzeniene
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway; (A.J.K.T.); (A.J.)
| |
Collapse
|
26
|
Jadvar H, Colletti PM. Molecular Imaging Assessment of Androgen Deprivation Therapy in Prostate Cancer. PET Clin 2022; 17:389-397. [PMID: 35662493 DOI: 10.1016/j.cpet.2022.02.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Hormonal therapy has long been recognized as a mainstay treatment for prostate cancer. New generation imaging agents have provided unprecedented opportunities at all phases along the natural history of prostate cancer. We review the literature on the effect of androgens and androgen deprivation therapy on prostate tumor at its various biological phases using the new generation molecular imaging agents in conjunction with positron emission tomography.
Collapse
Affiliation(s)
- Hossein Jadvar
- Division of Nuclear Medicine, Department of Radiology, USC Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Kenneth Norris Jr. Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA.
| | - Patrick M Colletti
- Division of Nuclear Medicine, Department of Radiology, USC Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
27
|
Addition of Standard Enzalutamide Medication Shows Synergistic Effects on Response to [ 177Lu]Lu-PSMA-617 Radioligand Therapy in mCRPC Patients with Imminent Treatment Failure-Preliminary Evidence of Pilot Experience. Cancers (Basel) 2022; 14:cancers14112691. [PMID: 35681671 PMCID: PMC9179420 DOI: 10.3390/cancers14112691] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/24/2022] [Accepted: 05/28/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary In this study, we investigated co-medication with enzalutamide, a well-established newer androgen axis drug, as a potential re-sensitizer for prostate-specific membrane antigen (PSMA)-targeted radioligand therapy (RLT) in n = 10 patients with imminent treatment failure on standard 177Lu-based PSMA-RLT. After the introduction of enzalutamide medication, all patients showed a PSA decrease (7/10 patients with partial remission). This pilot experience suggests the synergistic potential of adding enzalutamide to PSMA-RLT derived from the intra-individual comparison of 177Lu-based PSMA-RLT ± enzalutamide. Abstract Well-received strong efficacy of prostate-specific membrane antigen (PSMA)-targeted radioligand therapy (RLT) does not prevent patients from either early or eventual disease progression under this treatment. In this study, we investigated co-medication with enzalutamide as a potential re-sensitizer for PSMA-RLT in patients with imminent treatment failure on standard 177Lu-based PSMA-RLT. Ten mCRPC patients who exhibited an insufficient response to conventional [177Lu]Lu-PSMA-617 RLT received oral medication of enzalutamide 160 mg/d as an adjunct to continued PSMA-RLT. Prostate-specific antigen (PSA) and standard toxicity screening lab work-up were performed to assess the treatment efficacy and safety in these individuals. The mean PSA increase under PSMA-RLT before starting the re-sensitizing procedure was 22.4 ± 26.5%. After the introduction of enzalutamide medication, all patients experienced a PSA decrease, –43.4 ± 20.0% and –48.2 ± 39.0%, after one and two cycles of enzalutamide-augmented PSMA-RLT, respectively. A total of 70% of patients (7/10) experienced partial remission, with a median best PSA response of –62%. Moreover, 5/6 enzalutamide-naïve patients and 2/4 patients who had previously failed enzalutamide exhibited a partial remission. There was no relevant enzalutamide-induced toxicity observed in this small cohort. This pilot experience suggests the synergistic potential of adding enzalutamide to PSMA-RLT derived from the intra-individual comparison of 177Lu-based PSMA-RLT ± enzalutamide.
Collapse
|
28
|
Upregulation of PSMA Expression by Enzalutamide in Patients with Advanced mCRPC. Cancers (Basel) 2022; 14:cancers14071696. [PMID: 35406467 PMCID: PMC8997007 DOI: 10.3390/cancers14071696] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 02/27/2022] [Accepted: 03/24/2022] [Indexed: 12/24/2022] Open
Abstract
In this study, we investigated upregulation of prostate-specific membrane antigen (PSMA) by enzalutamide in a cohort (n = 30) of patients with advanced metastatic castration-resistant prostate cancer (mCRPC). Patients were examined by [68Ga]Ga-PSMA-11 PET/CT pre- and post-enzalutamide medication (mean 13 ± 7 days). Imaging results were compared based on quantification of whole-body PSMA tumor burden: total lesion PSMA (TLP) and normalized TLP values to liver (TLP-LR) and to parotid gland (TLP-PR). In addition, lesion-based analyses were performed. The median (mean) increases in TLP, TLP-LR and TLP-PR after enzalutamide medication were 10.1% (20.2%), 29.5% (34.8%) and 27.6% (24.4%), respectively. These increases were statistically significant (p = 0.002, p < 0.001, and p < 0.001), while prostate-specific antigen (PSA) serum values did not change significantly (p = 0.483). The increase was independent of prior patient exposure to enzalutamide. SUVmax increased substantially (>10%) in 49.6% of target lesions. The relative change was significantly higher in the subgroup of lesions with SUVmax < 10 (p < 0.001). In conclusion, short-term enzalutamide medication significantly increases PSMA expression in patients with mCRPC, irrespective of prior enzalutamide exposure. The relative PSMA upregulation effect seems to be more pronounced in lesions with only moderate baseline PSMA expression. Enzalutamide may provide a potential enhancer medication for PSMA-targeted radioligand therapy.
Collapse
|
29
|
Sommer U, Siciliano T, Ebersbach C, Beier AMK, Stope MB, Jöhrens K, Baretton GB, Borkowetz A, Thomas C, Erb HHH. Impact of Androgen Receptor Activity on Prostate-Specific Membrane Antigen Expression in Prostate Cancer Cells. Int J Mol Sci 2022; 23:1046. [PMID: 35162969 PMCID: PMC8835452 DOI: 10.3390/ijms23031046] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/02/2021] [Accepted: 01/17/2022] [Indexed: 12/15/2022] Open
Abstract
Prostate-specific membrane antigen (PSMA) is an essential molecular regulator of prostate cancer (PCa) progression coded by the FOLH1 gene. The PSMA protein has become an important factor in metastatic PCa diagnosis and radioligand therapy. However, low PSMA expression is suggested to be a resistance mechanism to PSMA-based imaging and therapy. Clinical studies revealed that androgen receptor (AR) inhibition increases PSMA expression. The mechanism has not yet been elucidated. Therefore, this study investigated the effect of activation and inhibition of androgen signaling on PSMA expression levels in vitro and compared these findings with PSMA levels in PCa patients receiving systemic therapy. To this end, LAPC4, LNCaP, and C4-2 PCa cells were treated with various concentrations of the synthetic androgen R1881 and antiandrogens. Changes in FOLH1 mRNA were determined using qPCR. Open access databases were used for ChIP-Seq and tissue expression analysis. Changes in PSMA protein were determined using western blot. For PSMA staining in patients' specimens, immunohistochemistry (IHC) was performed. Results revealed that treatment with the synthetic androgen R1881 led to decreased FOLH1 mRNA and PSMA protein. This effect was partially reversed by antiandrogen treatment. However, AR ChIP-Seq analysis revealed no canonical AR binding sites in the regulatory elements of the FOLH1 gene. IHC analysis indicated that androgen deprivation only resulted in increased PSMA expression in patients with low PSMA levels. The data demonstrate that AR activation and inhibition affects PSMA protein levels via a possible non-canonical mechanism. Moreover, analysis of PCa tissue reveals that low PSMA expression rates may be mandatory to increase PSMA by androgen deprivation.
Collapse
Affiliation(s)
- Ulrich Sommer
- Institute of Pathology, Universitätsklinikum Carl Gustav Carus Dresden, 01307 Dresden, Germany
| | - Tiziana Siciliano
- Department of Urology, Technische Universität Dresden, 01307 Dresden, Germany
| | - Celina Ebersbach
- Department of Urology, Technische Universität Dresden, 01307 Dresden, Germany
- Mildred Scheel Early Career Center, Department of Urology, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Alicia-Marie K Beier
- Department of Urology, Technische Universität Dresden, 01307 Dresden, Germany
- Mildred Scheel Early Career Center, Department of Urology, Medical Faculty and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Matthias B Stope
- Department of Gynecology and Gynecological Oncology, University Hospital Bonn, 53127 Bonn, Germany
- UroFors Consortium (Natural Scientists in Urological Research), German Society of Urology, 14163 Berlin, Germany
| | - Korinna Jöhrens
- Institute of Pathology, Universitätsklinikum Carl Gustav Carus Dresden, 01307 Dresden, Germany
| | - Gustavo B Baretton
- Institute of Pathology, Universitätsklinikum Carl Gustav Carus Dresden, 01307 Dresden, Germany
- National Center for Tumor Diseases Partner Site Dresden and German Cancer Center, 69120 Heidelberg, Germany
- Tumor and Normal Tissue Bank of the University Cancer Center (UCC), University Hospital and Faculty of Medicine, Technische Universität Dresden, 01069 Dresden, Germany
| | - Angelika Borkowetz
- Department of Urology, Technische Universität Dresden, 01307 Dresden, Germany
| | - Christian Thomas
- Department of Urology, Technische Universität Dresden, 01307 Dresden, Germany
| | - Holger H H Erb
- Department of Urology, Technische Universität Dresden, 01307 Dresden, Germany
- UroFors Consortium (Natural Scientists in Urological Research), German Society of Urology, 14163 Berlin, Germany
| |
Collapse
|
30
|
Ong JS, Hofman MS. PET imaging of prostate cancer. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00111-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
31
|
Suman S, Parghane RV, Joshi A, Prabhash K, Talole S, Basu S. Combined 177 Lu-PSMA-617 PRLT and abiraterone acetate versus 177 Lu-PSMA-617 PRLT monotherapy in metastatic castration-resistant prostate cancer: An observational study comparing the response and durability. Prostate 2021; 81:1225-1234. [PMID: 34469602 DOI: 10.1002/pros.24219] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/03/2021] [Accepted: 08/20/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVE The aim of present study was to determine and compare the overall response rates, progression-free survival (PFS), overall survival (OS), and clinical toxicity of the combination of 177Lu-PSMA-617 radioligand therapy (PRLT) and abiraterone acetate (AA) versus 177Lu-PSMA-617 PRLT as monotherapy in metastatic castration-resistant prostate cancer (mCRPC) patients. MATERIALS AND METHODS The mCRPC patients who received at least one cycle of 177 Lu-PSMA-617 PRLT with or without AA therapy, were included and analyzed in the present study. The patients were divided into two major groups. Group 1 received only 177 Lu-PSMA PRLT and Group 2 received combined 177 Lu-PSMA PRLT + AA therapy. Therapeutic dose of 177 Lu-PSMA-617 PRLT was 4.4-5.55 GBq per patient per cycle administered at intervals of 10-12 weeks in both groups. The Group 2 patients additionally received a dose of 1000 mg of AA once daily and 5 mg of prednisone twice daily. Treatment response in two groups was evaluated under four broad categories (a) symptomatic, (b) biochemical (serum prostate-specific antigen level), (c) objective molecular imaging (68 Ga-PSMA-11 and 18 F-FDG PET/CT), and (d) objective anatomical imaging (computed tomography). For assessing treatment response, patients in two groups were categorized into responders (complete response [CR], partial response [PR], and stable disease [SD]) and nonresponders (progressive disease [PD]). The Kaplan-Meier product-limit method was used to calculate PFS and OS following first 177 Lu-PSMA PRLT in the two groups. Univariate analysis was used to compare the patients' characteristics in two groups using a χ2 or Fisher exact test. The Kaplan-Meier curves of PFS and OS between two groups were compared by using the log-rank test (p < 0.05 significant). RESULTS A total of 58 mCRPC patients (Group 1, 38 patients and Group 2, 20 patients) were included in this study analysis. The clinical and demographic characteristics of these patients (age, Gleason score, FDG avid disease, metastatic disease burden, and average number of 177 Lu-PSMA PRLT cycles) in two groups were compared and found to be similar (p > 0.05). Post-treatment, symptomatic, biochemical, molecular, and anatomic imaging responders were found in 22 patients (58%) and 17 patients (85%), 22 patients (58%) and 16 patients (80%), 19 patients (54%) and 14 patients (78%), and 19 patients (54%) and 14 patients (78%) in Group 1 and Group 2, respectively. The median PFS of 7 months and median OS of 8 months were documented in Group 1, whereas median PFS was not reached and median OS of 16 months registered in Group 2. Transient hematological toxicity of Grades 1 and 2 was found in total seven patients (five patients in Group 1 and two patients in Group 2). On comparison of the treatment outcome between two groups, significant p value was found for symptomatic responders (58% in Group 1 vs. 85% in Group 2), median PFS (7 months in Group 1 vs. not reached in Group 2), and median OS (8 months in Group 1 vs. 16 months in Group 2), with better outcome in Group 2 patients for these variables. CONCLUSION In the present study, the combination of 177 Lu-PSMA-617 PRLT and AA therapy showed significant improvement in mCRPC patients' symptomatic response, PFS, and OS as compared to 177 Lu-PSMA-617 PRLT monotherapy.
Collapse
Affiliation(s)
- Sonam Suman
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Centre Annexe, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Rahul V Parghane
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Centre Annexe, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Amit Joshi
- Homi Bhabha National Institute, Mumbai, India
- Department of Medical Oncology, Tata Memorial Centre, Mumbai, India
| | - Kumar Prabhash
- Homi Bhabha National Institute, Mumbai, India
- Department of Medical Oncology, Tata Memorial Centre, Mumbai, India
| | - Sanjay Talole
- Homi Bhabha National Institute, Mumbai, India
- Department of Biostatistics, Tata Memorial Centre, Mumbai, India
| | - Sandip Basu
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Centre Annexe, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
32
|
Jacob A, Raj R, Allison DB, Myint ZW. Androgen Receptor Signaling in Prostate Cancer and Therapeutic Strategies. Cancers (Basel) 2021; 13:5417. [PMID: 34771580 PMCID: PMC8582395 DOI: 10.3390/cancers13215417] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/21/2021] [Accepted: 10/27/2021] [Indexed: 11/17/2022] Open
Abstract
Understanding of the molecular mechanisms of prostate cancer has led to development of therapeutic strategies targeting androgen receptor (AR). These androgen-receptor signaling inhibitors (ARSI) include androgen synthesis inhibitor-abiraterone and androgen receptor antagonists-enzalutamide, apalutamide, and darolutamide. Although these medications provide significant improvement in survival among men with prostate cancer, drug resistance develops in nearly all patients with time. This could be through androgen-dependent or androgen-independent mechanisms. Even weaker signals and non-canonical steroid ligands can activate AR in the presence of truncated AR-splice variants, AR overexpression, or activating mutations in AR. AR splice variant, AR-V7 is the most studied among these and is not targeted by available ARSIs. Non-androgen receptor dependent resistance mechanisms are mediated by activation of an alternative signaling pathway when AR is inhibited. DNA repair pathway, PI3K/AKT/mTOR pathway, BRAF-MAPK and Wnt signaling pathway and activation by glucocorticoid receptors can restore downstream signaling in prostate cancer by alternative proteins. Multiple clinical trials are underway exploring therapeutic strategies to overcome these resistance mechanisms.
Collapse
Affiliation(s)
- Aasems Jacob
- Department of Medicine, Division of Hematology & Oncology, Pikeville Medical Center, Pikeville, KY 41501, USA;
| | - Rishi Raj
- Department of Medicine, Division of Endocrinology, Diabetes & Metabolism, Pikeville Medical Center, Pikeville, KY 41501, USA;
| | - Derek B. Allison
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA;
- Department of Pathology and Laboratory Medicine, University of Kentucky, Lexington, KY 40536, USA
- Department of Urology, University of Kentucky, Lexington, KY 40536, USA
| | - Zin W. Myint
- Markey Cancer Center, University of Kentucky, Lexington, KY 40536, USA;
- Department of Medicine, Division of Medical Oncology, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
33
|
Comparing the Effect of Multiple Histone Deacetylase Inhibitors on SSTR2 Expression and [ 111In]In-DOTATATE Uptake in NET Cells. Cancers (Basel) 2021; 13:cancers13194905. [PMID: 34638389 PMCID: PMC8508045 DOI: 10.3390/cancers13194905] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/21/2021] [Accepted: 09/26/2021] [Indexed: 01/23/2023] Open
Abstract
Simple Summary Patients diagnosed with neuroendocrine tumors (NETs) are often treated with peptide receptor radionuclide therapy (PRRT). This therapy targets the somatostatin type-2 receptors (SSTR2) frequently overexpressed on these types of tumors. Although this therapy has proven to be effective, complete responses are rare and therapy improvement is desirable. We aimed to increase SSTR2 expression on NET cells, potentially increasing the number of patients eligible for SSTR2-targeted PRRT and improving clinical outcomes. We used histone deacetylase inhibitors (HDACis) to manipulate the epigenetic machinery and hereby aimed to increase SSTR2 gene transcription. Our results showed that the HDACis increased SSTR2 expression in several NET cell lines. Moreover, the uptake of radiolabeled DOTATATE, the tracer used for PRRT, was enhanced. The observed reversibility profile after HDACi withdrawal of the induced effects suggests that proper timing of HDACi treatment is likely essential. Abstract The aim of this study was to increase somatostatin type-2 receptor (SSTR2) expression on neuroendocrine tumor (NET) cells using histone deacetylase inhibitors (HDACis), potentially increasing the uptake of SSTR2-targeted radiopharmaceuticals and subsequently improving treatment efficacy of peptide receptor radionuclide therapy (PRRT). Human NET cell lines BON-1, NCI-H727, and GOT1 were treated with HDACis (i.e., CI-994, entinostat, LMK-235, mocetinostat, panobinostat, or valproic acid (VPA); entinostat and VPA were the HDACis tested in GOT1 cells) to examine SSTR2 mRNA expression levels and uptake of SSTR2-targeting radiotracer [111In]In-DOTATATE. Reversibility of the induced effects was examined after drug-withdrawal. Finally, the effect of VPA on radiosensitivity was investigated. A strong stimulatory effect in BON-1, NCI-H727, and GOT1 cells was observed after HDACi treatment, both on SSTR2 mRNA expression levels and [111In]In-DOTATATE uptake. The effects of the HDACis were largely reversible over a period of seven days, demonstrating largest reductions within the first day. The reversibility profile of the induced effects suggests that proper timing of HDACi treatment is most likely essential for a beneficial outcome. In addition to increasing SSTR2 expression levels, VPA enhanced the radiosensitivity of all cell lines. In conclusion, HDACi treatment increased SSTR2 expression, and radiosensitivity was also enhanced upon VPA treatment.
Collapse
|
34
|
Zhang H, Koumna S, Pouliot F, Beauregard JM, Kolinsky M. PSMA Theranostics: Current Landscape and Future Outlook. Cancers (Basel) 2021; 13:4023. [PMID: 34439177 PMCID: PMC8391520 DOI: 10.3390/cancers13164023] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 08/04/2021] [Accepted: 08/06/2021] [Indexed: 12/14/2022] Open
Abstract
INTRODUCTION Prostate-specific membrane antigen (PSMA) is a promising novel molecular target for imaging diagnostics and therapeutics (theranostics). There has been a growing body of evidence supporting PSMA theranostics approaches in optimizing the management of prostate cancer and potentially altering its natural history. METHODS We utilized PubMed and Google Scholar for published studies, and clinicaltrials.gov for planned, ongoing, and completed clinical trials in PSMA theranostics as of June 2021. We presented evolving evidence for various PSMA-targeted radiopharmaceutical agents in the treatment paradigm for prostate cancer, as well as combination treatment strategies with other targeted therapy and immunotherapy. We highlighted the emerging evidence of PSMA and fluorodeoxyglucose (FDG) PET/CT as a predictive biomarker for PSMA radioligand therapy. We identified seven ongoing clinical trials in oligometastatic-directed therapy using PSMA PET imaging. We also presented a schematic overview of 17 key PSMA theranostic clinical trials throughout the various stages of prostate cancer. CONCLUSIONS In this review, we presented the contemporary and future landscape of theranostic applications in prostate cancer with a focus on PSMA ligands. As PSMA theranostics will soon become the standard of care for the management of prostate cancer, we underscore the importance of integrating nuclear medicine physicians into the multidisciplinary team.
Collapse
Affiliation(s)
- Hanbo Zhang
- Department of Medical Oncology and Hematology, University of Manitoba, Winnipeg, MB R3E 0V9, Canada;
| | - Stella Koumna
- Department of Diagnostic Imaging, Cross Cancer Institute, Edmonton, AB T6G 1Z2, Canada;
| | - Frédéric Pouliot
- Department of Surgery, Université Laval, Québec City, QC G1R 3S1, Canada;
| | - Jean-Mathieu Beauregard
- Department of Radiology and Nuclear Medicine, Université Laval, Québec City, QC G1R 3S1, Canada;
| | - Michael Kolinsky
- Department of Medical Oncology, University of Alberta, Edmonton, AB T6G 1Z2, Canada
| |
Collapse
|
35
|
Hammer S, Schlicker A, Zitzmann-Kolbe S, Baumgart S, Hagemann UB, Scholz A, Haendler B, Lejeune P, Karlsson J, Ellingsen C, Hennekes H, Nielsen CH, Juul MU, Mumberg D, Schatz CA. Darolutamide Potentiates the Antitumor Efficacy of a PSMA-targeted Thorium-227 Conjugate by a Dual Mode of Action in Prostate Cancer Models. Clin Cancer Res 2021; 27:4367-4378. [PMID: 34035067 PMCID: PMC9401501 DOI: 10.1158/1078-0432.ccr-21-0342] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/23/2021] [Accepted: 05/21/2021] [Indexed: 01/07/2023]
Abstract
PURPOSE Androgen receptor (AR) inhibitors are well established in the treatment of castration-resistant prostate cancer and have recently shown efficacy also in castration-sensitive prostate cancer. Although most patients respond well to initial therapy, resistance eventually develops, and thus, more effective therapeutic approaches are needed. Prostate-specific membrane antigen (PSMA) is highly expressed in prostate cancer and presents an attractive target for radionuclide therapy. Here, we evaluated the efficacy and explored the mode of action of the PSMA-targeted thorium-227 conjugate (PSMA-TTC) BAY 2315497, an antibody-based targeted alpha-therapy, in combination with the AR inhibitor darolutamide. EXPERIMENTAL DESIGN The in vitro and in vivo antitumor efficacy and mode of action of the combination treatment were investigated in preclinical cell line-derived and patient-derived prostate cancer xenograft models with different levels of PSMA expression. RESULTS Darolutamide induced the expression of PSMA in androgen-sensitive VCaP and LNCaP cells in vitro, and the efficacy of darolutamide in combination with PSMA-TTC was synergistic in these cells. In vivo, the combination treatment showed synergistic antitumor efficacy in the low PSMA-expressing VCaP and in the high PSMA-expressing ST1273 prostate cancer models, and enhanced efficacy in the enzalutamide-resistant KUCaP-1 model. The treatments were well tolerated. Mode-of-action studies revealed that darolutamide induced PSMA expression, resulting in higher tumor uptake of PSMA-TTC, and consequently, higher antitumor efficacy, and impaired PSMA-TTC-mediated induction of DNA damage repair genes, potentially contributing to increased DNA damage. CONCLUSIONS These results provide a strong rationale to investigate PSMA-TTC in combination with AR inhibitors in patients with prostate cancer.
Collapse
Affiliation(s)
| | | | | | - Simon Baumgart
- Bayer U.S. LLC, Pharmaceuticals, Cambridge, Massachusetts
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Target Heterogeneity in Oncology: The Best Predictor for Differential Response to Radioligand Therapy in Neuroendocrine Tumors and Prostate Cancer. Cancers (Basel) 2021; 13:cancers13143607. [PMID: 34298822 PMCID: PMC8304541 DOI: 10.3390/cancers13143607] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/04/2021] [Accepted: 07/07/2021] [Indexed: 12/27/2022] Open
Abstract
Simple Summary In the era of precision medicine, novel targets have emerged on the surface of cancer cells, which have been exploited for the purpose of radioligand therapy. However, there have been variations in the way these receptors are expressed, especially in prostate cancers and neuroendocrine tumors. This variable expression of receptors across the grades of cancers led to the concept of ‘target heterogeneity’, which has not just impacted therapeutic decisions but also their outcomes. Radiopharmaceuticals targeting receptors need to be used when there are specific indicators—either clinical, radiological, or at molecular level—warranting their use. In addition, response to these radioligands can be assessed using different techniques, whereby we can prognosticate further outcomes. We shall also discuss, in this review, the conventional as well as novel approaches of detecting heterogeneity in prostate cancers and neuroendocrine tumors. Abstract Tumor or target heterogeneity (TH) implies presence of variable cellular populations having different genomic characteristics within the same tumor, or in different tumor sites of the same patient. The challenge is to identify this heterogeneity, as it has emerged as the most common cause of ‘treatment resistance’, to current therapeutic agents. We have focused our discussion on ‘Prostate Cancer’ and ‘Neuroendocrine Tumors’, and looked at the established methods for demonstrating heterogeneity, each with its advantages and drawbacks. Also, the available theranostic radiotracers targeting PSMA and somatostatin receptors combined with targeted systemic agents, have been described. Lu-177 labeled PSMA and DOTATATE are the ‘standard of care’ radionuclide therapeutic tracers for management of progressive treatment-resistant prostate cancer and NET. These approved therapies have shown reasonable benefit in treatment outcome, with improvement in quality of life parameters. Various biomarkers and predictors of response to radionuclide therapies targeting TH which are currently available and those which can be explored have been elaborated in details. Imaging-based features using artificial intelligence (AI) need to be developed to further predict the presence of TH. Also, novel theranostic tools binding to newer targets on surface of cancer cell should be explored to overcome the treatment resistance to current treatment regimens.
Collapse
|
37
|
Evolving Castration Resistance and Prostate Specific Membrane Antigen Expression: Implications for Patient Management. Cancers (Basel) 2021; 13:cancers13143556. [PMID: 34298770 PMCID: PMC8307676 DOI: 10.3390/cancers13143556] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 12/11/2022] Open
Abstract
Metastatic castration-resistant prostate cancer (mCRPC) remains an incurable disease, despite multiple novel treatment options. The role of prostate-specific membrane antigen (PSMA) in the process of mCRPC development has long been underestimated. During the last years, a new understanding of the underlying molecular mechanisms of rising PSMA expression and its association with disease progression has emerged. Accurate understanding of these complex interactions is indispensable for a precise diagnostic process and ultimately successful treatment of advanced prostate cancer. The combination of different novel therapeutics such as androgen deprivation agents, 177LU-PSMA radioligand therapy and PARP inhibitors promises a new kind of efficacy. In this review, we summarize the current knowledge about the most relevant molecular mechanisms around PSMA in mCRPC development and how they can be implemented in mCRPC management.
Collapse
|
38
|
Mei R, Farolfi A, Castellucci P, Nanni C, Zanoni L, Fanti S. PET/CT variants and pitfalls in prostate cancer: What you might see on PET and should never forget. Semin Nucl Med 2021; 51:621-632. [PMID: 34266631 DOI: 10.1053/j.semnuclmed.2021.06.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
2-deoxy-2-[18F]fluorodeoxyglucose (FDG) positron emission tomography (PET) gained an impressive role in the diagnostic management of many oncological diseases, even though its use in imaging prostate cancer (PC) is limited to selected cases, mostly advanced stage of PC and selection for prostate specific antigen membrane (PSMA) radioligand therapy (RLT). In the past years, several PET tracers have been developed for both staging and restaging PC. The three most employed PET molecules in daily practice are [11C] or [18F]F-Choline, [18F]F-Fluciclovine (Anti-1- amino-3-[18F]Fluorocyclobutane-1-Carboxylic Acid, also known as (Anti-[18F]FACBC), [68Ga]Ga-PSMA and recently FDA approved the first Fluorinated PSMA-based named [18F]F-DCFPyl. Each one has its own physiological and peculiarity which are worth exploring. Moreover, an increasing number of case reports and studies have reported tracers' variants, pitfalls, or even non-prostatic diseases (benign and malignant) incidentally detected. In prostate oncology, PET can be performed with several indications in different stages of disease, as highlighted in the EAU Guidelines on PC. A correct scan interpretation depends on the knowledge of both the physiological distribution of the tracers and the uptake of possible variants and pitfalls. The aim of this critical review is to provide a comprehensive knowledge of physiological distribution of these three tracers, as well as an updated overview of variants and pitfalls.
Collapse
Affiliation(s)
- Riccardo Mei
- Nuclear Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum, University of Bologna, Italy.
| | - Andrea Farolfi
- Nuclear Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Paolo Castellucci
- Nuclear Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Cristina Nanni
- Nuclear Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Lucia Zanoni
- Nuclear Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Stefano Fanti
- Nuclear Medicine Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Department of Experimental, Diagnostic and Specialty Medicine, Alma Mater Studiorum, University of Bologna, Italy
| |
Collapse
|
39
|
Enzalutamide Enhances PSMA Expression of PSMA-Low Prostate Cancer. Int J Mol Sci 2021; 22:ijms22147431. [PMID: 34299051 PMCID: PMC8304389 DOI: 10.3390/ijms22147431] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/02/2021] [Accepted: 07/08/2021] [Indexed: 12/11/2022] Open
Abstract
Prostate-specific membrane antigen (PSMA)-directed radioligand therapy (RLT) prolongs overall survival in men with metastatic castration-resistant prostate cancer (mCRPC). However, men with low PSMA expression are excluded from RLT. We explored the effect of androgen receptor blockade with enzalutamide on PSMA expression. Assessment of PSMA and androgen receptor (AR) expression on the human PC cell lines 22Rv1, C4-2, and LNCaP by immunohistochemistry and flow cytometry revealed low (22Rv1) and high (C4-2 and LNCaP) PSMA expression, and high, comparable AR positivity. Treatment with enzalutamide increased PSMA levels in 22Rv1, C4-2, and LNCaP (2.2/2.3/2.6-fold, p = 0.0005/0.03/0.046) after one week compared to DMSO-treated controls as assessed by flow cytometry. NOD/Scid mice bearing 22Rv1 tumors were treated with enzalutamide for two weeks. Positron emission tomography/computed tomography (PET/CT) demonstrated higher tumor uptake of 68Ga-PSMA after enzalutamide treatment (p = 0.004). Similarly, a clinical case with low baseline PSMA avidity demonstrated increased uptake of 68Ga-PSMA after enzalutamide on PET/CT and post-therapeutic 177Lu-PSMA scintigraphy in a patient with mCRPC. Enzalutamide induced PSMA expression in the 22Rv1 xenograft model and in an mCRPC patient, both with low baseline tumoral PSMA levels. Therefore, enzalutamide pre-treatment might render patients with low PSMA expression eligible for 177Lu-PSMA RLT.
Collapse
|
40
|
Van Simaeys G, Doumont G, De Maeseneire C, Passon N, Lacroix S, Lentz C, Horion A, Warnier C, Torres D, Martens C, Vierasu I, Egrise D, Goldman S. [ 18F]-JK-PSMA-7 and [ 18F]-FDG tumour PET uptake in treated xenograft human prostate cancer model in mice. Eur J Nucl Med Mol Imaging 2021; 48:1773-1784. [PMID: 33398412 DOI: 10.1007/s00259-020-05169-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 12/15/2020] [Indexed: 10/22/2022]
Abstract
PURPOSE This preclinical study aims to evaluate the extent to which a change in prostate-specific membrane antigen (PSMA) expression of castration-resistant prostate cancer (CRPC) following standard treatment is reflected in [18F]JK-PSMA-7 PET/CT. METHODS Castrated mice supplemented with testosterone implant were xenografted with human LNCaP CRPC. After appropriate tumour growth, androgen deprivation therapy (ADT) was carried out by the removal of the implant followed by a single injection of docetaxel (400 μg/20-g mouse) 2 weeks later. [18F]JK-PSMA-7 PET/CT were performed before ADT, then before and at days 12, 26, 47 and 69 after docetaxel administration. The [18F]JK-PSMA-7 PET data were compared to corresponding unspecific metabolic [18F]FDG PET/CT and ex vivo quantification of PSMA expression estimated by flow cytometry on repeated tumour biopsies. RESULTS ADT alone had no early effect on LNCaP tumours that pursued their progression. Until day 12 post-docetaxel, the [18F]JK-PSMA7 uptake was significantly higher than that of [18F]FDG, indicating the persistence of PSMA expression at those time points. From day 26 onwards when the tumours were rapidly expanding, both [18F]JK-PSMA7 and [18F]FDG uptake continuously decreased although the decrease in [18F]JK-PSMA uptake was markedly faster. The fraction of PSMA-positive cells in tumour biopsies decreased similarly over time to reach a non-specific level after the same time period. CONCLUSION Applying PSMA-based imaging for therapy monitoring in patients with CRPC should be considered with caution since a reduction in [18F]JK-PSMA-7 PET uptake after successive ADT and chemotherapy may be related to downregulation of PSMA expression in dedifferentiated and rapidly proliferating tumour cells.
Collapse
Affiliation(s)
- Gaetan Van Simaeys
- Center for Microscopy and Molecular Imaging, Université libre de Bruxelles, Charleroi, Belgium.
- Service de médecine nucléaire, Hôpital Érasme, Université libre de Bruxelles, Brussels, Belgium.
| | - Gilles Doumont
- Center for Microscopy and Molecular Imaging, Université libre de Bruxelles, Charleroi, Belgium
| | - Coraline De Maeseneire
- Center for Microscopy and Molecular Imaging, Université libre de Bruxelles, Charleroi, Belgium
| | - Nicolas Passon
- Center for Microscopy and Molecular Imaging, Université libre de Bruxelles, Charleroi, Belgium
| | - Simon Lacroix
- Center for Microscopy and Molecular Imaging, Université libre de Bruxelles, Charleroi, Belgium
- Service de médecine nucléaire, Hôpital Érasme, Université libre de Bruxelles, Brussels, Belgium
| | | | | | | | - David Torres
- Institute for Medical Immunology, Université libre de Bruxelles, Charleroi, Belgium
| | - Corentin Martens
- Center for Microscopy and Molecular Imaging, Université libre de Bruxelles, Charleroi, Belgium
- Service de médecine nucléaire, Hôpital Érasme, Université libre de Bruxelles, Brussels, Belgium
| | - Irina Vierasu
- Service de médecine nucléaire, Hôpital Érasme, Université libre de Bruxelles, Brussels, Belgium
| | - Dominique Egrise
- Center for Microscopy and Molecular Imaging, Université libre de Bruxelles, Charleroi, Belgium
- Service de médecine nucléaire, Hôpital Érasme, Université libre de Bruxelles, Brussels, Belgium
| | - Serge Goldman
- Center for Microscopy and Molecular Imaging, Université libre de Bruxelles, Charleroi, Belgium
- Service de médecine nucléaire, Hôpital Érasme, Université libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
41
|
Mathy CS, Mayr T, Kürpig S, Meisenheimer M, Dolscheid-Pommerich RC, Stoffel-Wagner B, Kristiansen G, Essler M, Muders MH, Bundschuh RA. Antihormone treatment differentially regulates PSA secretion, PSMA expression and 68Ga-PSMA uptake in LNCaP cells. J Cancer Res Clin Oncol 2021; 147:1733-1743. [PMID: 33760944 PMCID: PMC8076114 DOI: 10.1007/s00432-021-03583-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 02/28/2021] [Indexed: 01/12/2023]
Abstract
BACKGROUND In recent years, a variety of innovative therapeutics for castration-resistant prostate cancer have been developed, including novel anti-androgenic drugs, such as abiraterone or VPC-13566. Therapeutic monitoring of these pharmaceuticals is performed either by measuring PSA levels in serum or by imaging. PET using PSMA ligands labeled with Fluor-18 or Gallium-68 is the most sensitive and specific imaging modality for detection of metastases in advanced prostate cancer. To date, it remains unclear how PSMA expression is modulated by anti-hormonal treatment and how it correlates with PSA secretion. METHODS We analyzed modulation of PSMA-mRNA and protein expression, 68Ga-PSMA uptake and regulation of PSA secretion by abiraterone or VPC-13566 in LNCaP cells in vitro. RESULTS We found that abiraterone and VPC-13566 upregulate PSMA protein and mRNA expression but block PSA secretion in LNCaP cells. Both anti-androgens also enhanced 68Ga-PSMA uptake normalized by the number of cells, whereas abiraterone and VPC-13566 reduced 68Ga-PSMA uptake in total LNCaP monolayers treated due to cell death. CONCLUSION Our data indicate that PSA secretion and PSMA expression are differentially regulated upon anti-androgen treatment. This finding might be important for the interpretation of 68Ga-PSMA PET images in monitoring therapies with abiraterone and VPC-13566 in prostate cancer patients, but needs to be validated in vivo.
Collapse
Affiliation(s)
- C S Mathy
- Department of Nuclear Medicine, University Hospital Bonn, Venusberg Campus 1, 53127, Bonn, Germany
- Department of Pathology, University Hospital Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - T Mayr
- Department of Pathology, University Hospital Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - S Kürpig
- Department of Nuclear Medicine, University Hospital Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - M Meisenheimer
- Department of Nuclear Medicine, University Hospital Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - R C Dolscheid-Pommerich
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - B Stoffel-Wagner
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - G Kristiansen
- Department of Pathology, University Hospital Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - M Essler
- Department of Nuclear Medicine, University Hospital Bonn, Venusberg Campus 1, 53127, Bonn, Germany
| | - M H Muders
- Department of Pathology, University Hospital Bonn, Venusberg Campus 1, 53127, Bonn, Germany.
| | - R A Bundschuh
- Department of Nuclear Medicine, University Hospital Bonn, Venusberg Campus 1, 53127, Bonn, Germany.
| |
Collapse
|
42
|
Kranzbühler B, Sousa R, Prause L, Burger IA, Rupp NJ, Sulser T, Salemi S, Eberli D. Impact of short-term Dutasteride treatment on prostate-specific membrane antigen expression in a mouse xenograft model. Cancer Rep (Hoboken) 2021; 4:e1418. [PMID: 34008909 PMCID: PMC8714546 DOI: 10.1002/cnr2.1418] [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: 11/22/2020] [Revised: 03/29/2021] [Accepted: 04/19/2021] [Indexed: 11/30/2022] Open
Abstract
Background Dutasteride has been shown to increase expression of the prostate‐specific membrane antigen (PSMA) in prostate cancer cells in previous in vitro studies. This 5‐alpha‐reductase inhibitor is commonly used for the treatment of symptomatic benign prostatic enlargement. The modulation of PSMA expression might affect PSMA‐based prostate cancer imaging and therapy. Aim The purpose of this work was to further analyze concentration‐dependent effects of Dutasteride on PSMA expression in a mouse xenograft model. Methods and results Four groups of mice bearing LNCaP xenografts were treated for 14 days with daily intraperitoneal injections of either vehicle control or different concentrations of Dutasteride (0.1, 1, 10 mg/kg). Total expression of PSMA, androgen receptor (AR), and caspase‐3 protein was analyzed using immunoblotting (WES). In addition, PSMA, cleaved caspase‐3 and Ki‐67 expression was assessed and quantified by immunohistochemistry. Tumor size was measured by caliper on day 7 and 14, tumor weight was assessed following tissue harvesting. The mean PSMA protein expression in mice increased significantly after treatment with 1 mg/kg (10‐fold) or 10 mg/kg (sixfold) of Dutasteride compared to vehicle control. The mean fluorescence intensity significantly increased by daily injections of 0.1 mg/kg Dutasteride (1.6‐fold) as well as 1 and 10 mg/kg Dutasteride (twofold). While the reduction in tumor volume following treatment with high concentrations of 10 mg/kg Dutasteride was nonsignificant, no changes in AR, caspase‐3, cleaved caspase‐3, and Ki‐67 expression were observed. Conclusion Short‐term Dutasteride treatments with concentrations of 1 and 10 mg/kg significantly increase the total PSMA protein expression in a mouse LNCaP xenograft model. PSMA fluorescence intensity increases significantly even using lower daily concentrations of 0.1 mg/kg Dutasteride. Further investigations are needed to elucidate the impact of Dutasteride treatment on PSMA expression in patients.
Collapse
Affiliation(s)
- Benedikt Kranzbühler
- Department of Urology, University Hospital Zürich, University of Zürich, Laboratory for Urologic Oncology and Stem Cell Therapy, Zürich, Switzerland
| | - Rosa Sousa
- Department of Urology, University Hospital Zürich, University of Zürich, Laboratory for Urologic Oncology and Stem Cell Therapy, Zürich, Switzerland
| | - Lukas Prause
- Department of Urology, University Hospital Zürich, University of Zürich, Laboratory for Urologic Oncology and Stem Cell Therapy, Zürich, Switzerland
| | - Irene A Burger
- Department of Nuclear Medicine, University Hospital of Zürich, University of Zürich, Zürich, Switzerland.,Department of Nuclear Medicine, Kantonsspital Baden, Baden, Switzerland
| | - Niels J Rupp
- Department of Pathology and Molecular Pathology, University Hospital of Zürich, University of Zürich, Zürich, Switzerland
| | - Tullio Sulser
- Department of Urology, University Hospital Zürich, University of Zürich, Laboratory for Urologic Oncology and Stem Cell Therapy, Zürich, Switzerland
| | - Souzan Salemi
- Department of Urology, University Hospital Zürich, University of Zürich, Laboratory for Urologic Oncology and Stem Cell Therapy, Zürich, Switzerland
| | - Daniel Eberli
- Department of Urology, University Hospital Zürich, University of Zürich, Laboratory for Urologic Oncology and Stem Cell Therapy, Zürich, Switzerland
| |
Collapse
|
43
|
Stenberg VY, Larsen RH, Ma LW, Peng Q, Juzenas P, Bruland ØS, Juzeniene A. Evaluation of the PSMA-Binding Ligand 212Pb-NG001 in Multicellular Tumour Spheroid and Mouse Models of Prostate Cancer. Int J Mol Sci 2021; 22:ijms22094815. [PMID: 34062920 PMCID: PMC8124365 DOI: 10.3390/ijms22094815] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/27/2021] [Accepted: 04/29/2021] [Indexed: 01/12/2023] Open
Abstract
Radioligand therapy targeting the prostate-specific membrane antigen (PSMA) is rapidly evolving as a promising treatment for metastatic castration-resistant prostate cancer. The PSMA-targeting ligand p-SCN-Bn-TCMC-PSMA (NG001) labelled with 212Pb efficiently targets PSMA-positive cells in vitro and in vivo. The aim of this preclinical study was to evaluate the therapeutic potential of 212Pb-NG001 in multicellular tumour spheroid and mouse models of prostate cancer. The cytotoxic effect of 212Pb-NG001 was tested in human prostate C4-2 spheroids. Biodistribution at various time points and therapeutic effects of different activities of the radioligand were investigated in male athymic nude mice bearing C4-2 tumours, while long-term toxicity was studied in immunocompetent BALB/c mice. The radioligand induced a selective cytotoxic effect in spheroids at activity concentrations of 3–10 kBq/mL. In mice, the radioligand accumulated rapidly in tumours and was retained over 24 h, while it rapidly cleared from nontargeted tissues. Treatment with 0.25, 0.30 or 0.40 MBq of 212Pb-NG001 significantly inhibited tumour growth and improved median survival with therapeutic indexes of 1.5, 2.3 and 2.7, respectively. In BALB/c mice, no signs of long-term radiation toxicity were observed at activities of 0.05 and 0.33 MBq. The obtained results warrant clinical studies to evaluate the biodistribution, therapeutic efficacy and toxicity of 212Pb-NG001.
Collapse
Affiliation(s)
- Vilde Yuli Stenberg
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway; (L.-W.M.); (A.J.)
- Department of Research and Development, Nucligen AS, 0379 Oslo, Norway;
- Institute for Clinical Medicine, University of Oslo, 0318 Oslo, Norway;
- Correspondence: ; Tel.: +47-9012-8434
| | | | - Li-Wei Ma
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway; (L.-W.M.); (A.J.)
| | - Qian Peng
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway; (Q.P.); (P.J.)
| | - Petras Juzenas
- Department of Pathology, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway; (Q.P.); (P.J.)
| | - Øyvind Sverre Bruland
- Institute for Clinical Medicine, University of Oslo, 0318 Oslo, Norway;
- Department of Oncology, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
| | - Asta Juzeniene
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway; (L.-W.M.); (A.J.)
| |
Collapse
|
44
|
Ceci F, Oprea-Lager DE, Emmett L, Adam JA, Bomanji J, Czernin J, Eiber M, Haberkorn U, Hofman MS, Hope TA, Kumar R, Rowe SP, Schwarzenboeck SM, Fanti S, Herrmann K. E-PSMA: the EANM standardized reporting guidelines v1.0 for PSMA-PET. Eur J Nucl Med Mol Imaging 2021; 48:1626-1638. [PMID: 33604691 PMCID: PMC8113168 DOI: 10.1007/s00259-021-05245-y] [Citation(s) in RCA: 229] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 02/07/2021] [Indexed: 12/13/2022]
Abstract
RATIONALE The development of consensus guidelines for interpretation of Prostate-Specific Membrane Antigen (PSMA)-Positron Emission Tomography (PET) is needed to provide more consistent reports in clinical practice. The standardization of PSMA-PET interpretation may also contribute to increasing the data reproducibility within clinical trials. Finally, guidelines in PSMA-PET interpretation are needed to communicate the exact location of findings to referring physicians, to support clinician therapeutic management decisions. METHODS A panel of worldwide experts in PSMA-PET was established. Panelists were selected based on their expertise and publication record in the diagnosis or treatment of PCa, in their involvement in clinical guidelines and according to their expertise in the clinical application of radiolabeled PSMA inhibitors. Panelists were actively involved in all stages of a modified, nonanonymous, Delphi consensus process. RESULTS According to the findings obtained by modified Delphi consensus process, panelist recommendations were implemented in a structured report for PSMA-PET. CONCLUSIONS The E-PSMA standardized reporting guidelines, a document supported by the European Association of Nuclear Medicine (EANM), provide consensus statements among a panel of experts in PSMA-PET imaging, to develop a structured report for PSMA-PET in prostate cancer and to harmonize diagnostic interpretation criteria.
Collapse
Affiliation(s)
- Francesco Ceci
- Nuclear Medicine, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Daniela E Oprea-Lager
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical Centers, VU University Medical Center, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands.
| | - Louise Emmett
- St. Vincent's Clinical School, University of New South Wales, Kensington, NSW, Australia
- Department of Theranostics and Nuclear Medicine, St. Vincent's Hospital Sydney, Darlinghurst, NSW, Australia
| | - Judit A Adam
- Department of Radiology & Nuclear Medicine, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Jamshed Bomanji
- Department of Nuclear Medicine, University College London Hospitals NHS Foundation Trust, London, UK
| | - Johannes Czernin
- Ahmanson Translational Theranostics Division, Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Matthias Eiber
- School of Medicine, Department of Nuclear Medicine, Technische Universität München, Munich, Germany
| | - Uwe Haberkorn
- Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael S Hofman
- Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Thomas A Hope
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Rakesh Kumar
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Steven P Rowe
- Division of Nuclear Medicine, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Stefano Fanti
- Nuclear Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, University of Bologna, Bologna, Italy
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
| |
Collapse
|
45
|
Vaz S, Hadaschik B, Gabriel M, Herrmann K, Eiber M, Costa D. Influence of androgen deprivation therapy on PSMA expression and PSMA-ligand PET imaging of prostate cancer patients. Eur J Nucl Med Mol Imaging 2021; 47:9-15. [PMID: 31654093 DOI: 10.1007/s00259-019-04529-8] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Sofia Vaz
- Nuclear Medicine Radiopharmacology, Champalimaud Centre for the Unknown, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal.
| | - Boris Hadaschik
- Department of Urology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Michael Gabriel
- Institute of Nuclear Medicine and Endocrinology, Kepler University Hospital, Linz, Austria
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Matthias Eiber
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
| | - Durval Costa
- Nuclear Medicine Radiopharmacology, Champalimaud Centre for the Unknown, Champalimaud Foundation, Av. Brasília, 1400-038, Lisbon, Portugal
| |
Collapse
|
46
|
Weber M, Hadaschik B, Ferdinandus J, Rahbar K, Bögemann M, Herrmann K, Fendler WP, Kesch C. Prostate-specific Membrane Antigen-based Imaging of Castration-resistant Prostate Cancer. Eur Urol Focus 2021; 7:279-287. [PMID: 33483289 DOI: 10.1016/j.euf.2021.01.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 12/10/2020] [Accepted: 01/06/2021] [Indexed: 12/13/2022]
Abstract
CONTEXT Positron emission tomography (PET) targeting prostate-specific membrane antigen (PSMA) has unprecedented accuracy for localization of initial or recurrent prostate cancer (PC). There is now growing evidence regarding the value of PSMA-PET in patients with advanced PC. OBJECTIVE To review the value of PSMA-PET/computed tomography (CT) in the context of castration-resistant PC (CRPC). EVIDENCE ACQUISITION A search of the PubMed database using the terms "PSMA PET castration resistant prostate cancer" (years 2011-2020) was performed. Reviews, case reports/series, non-English articles, preclinical studies, access-restricted studies, and studies on PSMA radioligand therapy without further analysis of PSMA-PET parameters were subsequently excluded. EVIDENCE SYNTHESIS Compared to conventional imaging, PSMA-PET better identifies the true extent of CRPC, especially nonmetastatic CRPC. The clinical benefit of this stage migration is still unclear and needs to be evaluated in further studies. High accuracy of PSMA-PET holds promise for better, PET-guided metastasis-directed treatment in patients with oligometastatic CRPC. PSMA-PET is an essential eligibility criterion for [177Lu]-PSMA theranostic applications. Preliminary evidence indicates the value of PSMA-PET for the assessment of treatment responses. CONCLUSIONS Among other applications, PSMA-PET offers more precise staging for nonmetastatic CRPC. In particular, target localization for metastasis-directed therapy and target expression assessment for PSMA radioligand therapy also hold promise. Potential translation of this diagnostic tool into an oncologic benefit needs to be defined in future trials. PATIENT SUMMARY This review describes how prostate-specific membrane antigen positron emission tomography (PSMA-PET), a new sensitive imaging tool for prostate cancer, might help to guide clinicians in making treatment decisions for advanced prostate cancer.
Collapse
Affiliation(s)
- Manuel Weber
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany; West German Cancer Center.
| | - Boris Hadaschik
- West German Cancer Center; Department of Urology, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| | - Justin Ferdinandus
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany; West German Cancer Center
| | - Kambiz Rahbar
- West German Cancer Center; Department of Nuclear Medicine, University Hospital Münster, Münster, Germany
| | - Martin Bögemann
- West German Cancer Center; Department of Urology, University Hospital Münster, Münster, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany; West German Cancer Center
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany; West German Cancer Center
| | - Claudia Kesch
- West German Cancer Center; Department of Urology, University of Duisburg-Essen and German Cancer Consortium (DKTK)-University Hospital Essen, Essen, Germany
| |
Collapse
|
47
|
Veach DR, Storey CM, Lückerath K, Braun K, von Bodman C, Lamminmäki U, Kalidindi T, Strand SE, Strand J, Altai M, Damoiseaux R, Zanzonico P, Benabdallah N, Pankov D, Scher HI, Scardino P, Larson SM, Lilja H, McDevitt MR, Thorek DLJ, Ulmert D. PSA-Targeted Alpha-, Beta-, and Positron-Emitting Immunotheranostics in Murine Prostate Cancer Models and Nonhuman Primates. Clin Cancer Res 2021; 27:2050-2060. [PMID: 33441295 DOI: 10.1158/1078-0432.ccr-20-3614] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 11/13/2020] [Accepted: 01/07/2021] [Indexed: 12/22/2022]
Abstract
PURPOSE Most patients with prostate cancer treated with androgen receptor (AR) signaling inhibitors develop therapeutic resistance due to restoration of AR functionality. Thus, there is a critical need for novel treatment approaches. Here we investigate the theranostic potential of hu5A10, a humanized mAb specifically targeting free PSA (KLK3). EXPERIMENTAL DESIGN LNCaP-AR (LNCaP with overexpression of wildtype AR) xenografts (NSG mice) and KLK3_Hi-Myc transgenic mice were imaged with 89Zr- or treated with 90Y- or 225Ac-labeled hu5A10; biodistribution and subcellular localization were analyzed by gamma counting, PET, autoradiography, and microscopy. Therapeutic efficacy of [225Ac]hu5A10 and [90Y]hu5A10 in LNCaP-AR tumors was assessed by tumor volume measurements, time to nadir (TTN), time to progression (TTP), and survival. Pharmacokinetics of [89Zr]hu5A10 in nonhuman primates (NHP) were determined using PET. RESULTS Biodistribution of radiolabeled hu5A10 constructs was comparable in different mouse models. Specific tumor uptake increased over time and correlated with PSA expression. Treatment with [90Y]/[225Ac]hu5A10 effectively reduced tumor burden and prolonged survival (P ≤ 0.0054). Effects of [90Y]hu5A10 were more immediate than [225Ac]hu5A10 (TTN, P < 0.0001) but less sustained (TTP, P < 0.0001). Complete responses were observed in 7 of 18 [225Ac]hu5A10 and 1 of 9 mice [90Y]hu5A10. Pharmacokinetics of [89Zr]hu5A10 were consistent between NHPs and comparable with those in mice. [89Zr]hu5A10-PET visualized the NHP-prostate over the 2-week observation period. CONCLUSIONS We present a complete preclinical evaluation of radiolabeled hu5A10 in mouse prostate cancer models and NHPs, and establish hu5A10 as a new theranostic agent that allows highly specific and effective downstream targeting of AR in PSA-expressing tissue. Our data support the clinical translation of radiolabeled hu5A10 for treating prostate cancer.
Collapse
Affiliation(s)
- Darren R Veach
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Claire M Storey
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California
| | - Katharina Lückerath
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California.,Ahmanson Translational Imaging Division, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.,Department of Urology, David Geffen School of Medicine, Institute of Urologic Oncology, University of California, Los Angeles, Los Angeles, California.,Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Katharina Braun
- Department of Urology, Marien Hospital Herne, Ruhr-University Bochum, Herne, Germany
| | | | - Urpo Lamminmäki
- Department of Biochemistry, University of Turku, Turku, Finland
| | - Teja Kalidindi
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sven-Erik Strand
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University, Lund, Sweden
| | - Joanna Strand
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University, Lund, Sweden
| | - Mohamed Altai
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University, Lund, Sweden
| | - Robert Damoiseaux
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California
| | - Pat Zanzonico
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nadia Benabdallah
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Dmitry Pankov
- Immunology Program, Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Howard I Scher
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Peter Scardino
- Department of Medicine, Weill Cornell Medical College, New York, New York.,Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Steven M Larson
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Hans Lilja
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Translational Medicine, Lund University, Malmö, Sweden
| | - Michael R McDevitt
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Radiology, Weill Cornell Medical College, New York, New York
| | - Daniel L J Thorek
- Department of Radiology, Washington University School of Medicine, St. Louis, Missouri.,Department of Biomedical Engineering, Washington University School of Medicine, St. Louis, Missouri
| | - David Ulmert
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, Los Angeles, California. .,Ahmanson Translational Imaging Division, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.,Department of Urology, David Geffen School of Medicine, Institute of Urologic Oncology, University of California, Los Angeles, Los Angeles, California.,Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California.,Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|
48
|
Brumberg J, Beckl M, Dierks A, Schirbel A, Krebs M, Buck A, Kübler H, Lapa C, Seitz AK. Detection Rate of 68Ga-PSMA Ligand PET/CT in Patients with Recurrent Prostate Cancer and Androgen Deprivation Therapy. Biomedicines 2020; 8:biomedicines8110511. [PMID: 33217931 PMCID: PMC7698713 DOI: 10.3390/biomedicines8110511] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 11/08/2020] [Accepted: 11/16/2020] [Indexed: 01/10/2023] Open
Abstract
Prostate-specific membrane antigen (PSMA) ligand PET/CT enables the localization of tumor lesions in patients with recurrent prostate cancer, but it is unclear whether androgen deprivation therapy (ADT) influences diagnostic accuracy. The aim of this study was to evaluate the effect of ADT on the detection rate of 68Ga-PSMA ligand PET/CT. Thus, 399 patients with initial radical prostatectomy and 68Ga-PSMA ligand PET/CT during PSA relapse were retrospectively evaluated. Propensity score matching was used to create two balanced groups of 62 subjects who either did or did not receive ADT within six months before imaging. All 68Ga-PSMA ligand PET/CT were evaluated visually and with semiquantitative measures. The detection rate of tumor recurrence was significantly higher in the group with ADT (88.7% vs. 72.6%, p = 0.02) and improved with increasing PSA-levels in both groups. In subjects with pathological PET/CT and ADT, whole-body total lesion PSMA (p < 0.01) and PSMA-derived tumor volume (p < 0.01) were significantly higher than in those without ADT. More PSMA-positive lesions and higher PSMA-derived volumetric parameters in patients with ADT suggest that a better detection rate is related to a (biologically) more advanced disease stage. Due to high detection rates in patients with PSA-levels < 2 ng/mL, the withdrawal of ADT before PSMA ligand PET/CT cannot be recommended.
Collapse
Affiliation(s)
- Joachim Brumberg
- Department of Nuclear Medicine, University Hospital Würzburg and Julius-Maximilian University Würzburg, 97080 Würzburg, Germany; (M.B.); (A.D.); (A.S.); (A.B.); (C.L.)
- Correspondence: ; Tel.: +49-931-201-35000
| | - Melanie Beckl
- Department of Nuclear Medicine, University Hospital Würzburg and Julius-Maximilian University Würzburg, 97080 Würzburg, Germany; (M.B.); (A.D.); (A.S.); (A.B.); (C.L.)
| | - Alexander Dierks
- Department of Nuclear Medicine, University Hospital Würzburg and Julius-Maximilian University Würzburg, 97080 Würzburg, Germany; (M.B.); (A.D.); (A.S.); (A.B.); (C.L.)
- Department of Nuclear Medicine, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany
| | - Andreas Schirbel
- Department of Nuclear Medicine, University Hospital Würzburg and Julius-Maximilian University Würzburg, 97080 Würzburg, Germany; (M.B.); (A.D.); (A.S.); (A.B.); (C.L.)
| | - Markus Krebs
- Department of Urology, University Hospital Würzburg and Julius-Maximilian University Würzburg, 97080 Würzburg, Germany; (M.K.); (H.K.); (A.K.S.)
- Comprehensive Cancer Center Mainfranken, University Hospital Würzburg and Julius-Maximilian University Würzburg, 97080 Würzburg, Germany
| | - Andreas Buck
- Department of Nuclear Medicine, University Hospital Würzburg and Julius-Maximilian University Würzburg, 97080 Würzburg, Germany; (M.B.); (A.D.); (A.S.); (A.B.); (C.L.)
| | - Hubert Kübler
- Department of Urology, University Hospital Würzburg and Julius-Maximilian University Würzburg, 97080 Würzburg, Germany; (M.K.); (H.K.); (A.K.S.)
| | - Constantin Lapa
- Department of Nuclear Medicine, University Hospital Würzburg and Julius-Maximilian University Würzburg, 97080 Würzburg, Germany; (M.B.); (A.D.); (A.S.); (A.B.); (C.L.)
- Department of Nuclear Medicine, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany
| | - Anna Katharina Seitz
- Department of Urology, University Hospital Würzburg and Julius-Maximilian University Würzburg, 97080 Würzburg, Germany; (M.K.); (H.K.); (A.K.S.)
| |
Collapse
|
49
|
Miyahira AK, Pienta KJ, Babich JW, Bander NH, Calais J, Choyke P, Hofman MS, Larson SM, Lin FI, Morris MJ, Pomper MG, Sandhu S, Scher HI, Tagawa ST, Williams S, Soule HR. Meeting report from the Prostate Cancer Foundation PSMA theranostics state of the science meeting. Prostate 2020; 80:1273-1296. [PMID: 32865839 PMCID: PMC8442561 DOI: 10.1002/pros.24056] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 12/12/2022]
Abstract
INTRODUCTION The Prostate Cancer Foundation (PCF) convened a PCF prostate-specific membrane antigen (PSMA) Theranostics State of the Science Meeting on 18 November 2019, at Weill Cornell Medicine, New York, NY. METHODS The meeting was attended by 22 basic, translational, and clinical researchers from around the globe, with expertise in PSMA biology, development and use of PSMA theranostics agents, and clinical trials. The goal of this meeting was to discuss the current state of knowledge, the most important biological and clinical questions, and critical next steps for the clinical development of PSMA positron emission tomography (PET) imaging agents and PSMA-targeted radionuclide agents for patients with prostate cancer. RESULTS Several major topic areas were discussed including the biology of PSMA, the role of PSMA-targeted PET imaging in prostate cancer, the physics and performance of different PSMA-targeted PET imaging agents, the current state of clinical development of PSMA-targeted radionuclide therapy (RNT) agents, the role of dosimetry in PSMA RNT treatment planning, barriers and challenges in PSMA RNT clinical development, optimization of patient selection for PSMA RNT trials, and promising combination treatment approaches with PSMA RNT. DISCUSSION This article summarizes the presentations from the meeting for the purpose of globally disseminating this knowledge to advance the use of PSMA-targeted theranostic agents for imaging and treatment of patients with prostate cancer.
Collapse
Affiliation(s)
- Andrea K. Miyahira
- Science Department, Prostate Cancer Foundation, Santa Monica, California
| | - Kenneth J. Pienta
- Department of Urology, The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - John W. Babich
- Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Neil H. Bander
- Laboratory of Urologic Oncology, Department of Urology and Meyer Cancer Center, Weill Cornell Medicine, New York, New York
| | - Jeremie Calais
- Ahmanson Translational Theranostics Division, Department of Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California
| | - Peter Choyke
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Michael S. Hofman
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Peter MacCallum Cancer Centre, The University of Melbourne, Melbourne, Australia
- Department of Molecular Imaging and Therapeutic Nuclear Medicine, Peter MacCallum Cancer Centre, The University of Melbourne, Melbourne, Australia
| | - Steven M. Larson
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Frank I. Lin
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Michael J. Morris
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Martin G. Pomper
- Department of Urology, The Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Shahneen Sandhu
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Peter MacCallum Cancer Centre, The University of Melbourne, Melbourne, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, The University of Melbourne, Melbourne, Australia
| | - Howard I. Scher
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Scott T. Tagawa
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Scott Williams
- Prostate Cancer Theranostics and Imaging Centre of Excellence (ProsTIC), Peter MacCallum Cancer Centre, The University of Melbourne, Melbourne, Australia
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, The University of Melbourne, Melbourne, Australia
| | - Howard R. Soule
- Science Department, Prostate Cancer Foundation, Santa Monica, California
| |
Collapse
|
50
|
Kessel K, Seifert R, Weckesser M, Roll W, Humberg V, Schlack K, Bögemann M, Bernemann C, Rahbar K. Molecular analysis of circulating tumor cells of metastatic castration-resistant Prostate Cancer Patients receiving 177Lu-PSMA-617 Radioligand Therapy. Theranostics 2020; 10:7645-7655. [PMID: 32685010 PMCID: PMC7359074 DOI: 10.7150/thno.44556] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 05/20/2020] [Indexed: 12/21/2022] Open
Abstract
Rationale: Lu-177-PSMA-617 radioligand therapy (RLT) is currently under approval for treatment of metastatic castration resistant prostate cancer (mCRPC) patients with late stage disease. However, previous studies demonstrated both heterogeneity of prostate specific membrane antigen (PSMA) expression, as well as response to PSMA treatment among mCRPC patients. Thus, there is an unmet need for identifying predictive parametres prior or under PSMA-RLT treatment. We therefore aimed to correlate several clinical and molecular parameters with response to PSMA treatment in a cohort of mCRPC patients undergoing PSMA RLT followed by a detailed analysis of promising candidates. Methods: Nineteen patients, median age 68.8 years (range: 56.9 - 83.3) with mCRPC were included in this study. We performed baseline analysis of clinical parameters based on PSMA PET/CT, (metabolic tumor volume (MTV), total tumor volume (TTV)), serum PSA, ALP, LDH and gene expression analysis of circulating tumor cells (expression of AR full length (AR-FL), AR splice variant 7 (AR-V7), PSA and PSMA) as well as common markers for neuroendocrine differentiation (NED). Results: Patients presented with bone, lymph node, and visceral metastases (89%, 68%, and 21%, respectively). All patients were pretreated with docetaxel, either abiraterone or enzalutamide, or both. Biochemical response in terms of PSA decline ≥50 or ≥30% was observed in 42% and 63%, respectively. There were significant correlations between PSA and PSMA mRNA expression, as well as tumor volumes (both MTV and TTV), AR-FL and AR-V7 mRNA expression. However, there was no correlation with response to PSMA treatment. Furthermore, none of these parameters was significantly correlated with baseline serum PSA values. Common NED markers were shown to be specifically high expressed and revealed impact on OS independent from AR-V7 gene expression. Conclusion: We demonstrate that AR-FL and its splice variant AR-V7 might serve as prognostic biomarkers displaying high tumor burden in mCRPC patient prior to PSMA-RLT. Contrary, PSMA, which has been discussed as a biomarker for PSMA targeted treatment, does not display strong prognostic ability - at least on the mRNA level. Surprisingly, none of these parameters correlates to response to PSMA treatment. In contrast, commom NED markers such as SYP and ENO2 as well as FOXA1 expression level seem to predict OS, but not PFS, more reliably. We admit that a limitation of our study is the focus on mRNA expression of potential biomarkers only. Further investigations analyzing the potential role of protein expression of these markers are therefore warranted.
Collapse
|