1
|
Ling SW, van der Veldt A, Segbers M, Luiting H, Brabander T, Verburg F. Tubarial salivary glands show a low relative contribution to functional salivary gland tissue mass. Ann Nucl Med 2024; 38:913-918. [PMID: 39060832 PMCID: PMC11489203 DOI: 10.1007/s12149-024-01965-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: 06/11/2024] [Accepted: 07/22/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND In 2021, the tubarial salivary glands (TSGs) were newly identified on prostate-specific membrane antigen (PSMA) positron emission tomography/computed tomography (PET/CT) as macroscopic glands in the nasopharyngeal wall. However, the relative contribution of the TSGs to the total salivary gland function, and consequently on the development of xerostomia after external beam radiotherapy (EBRT) or PSMA-targeted radionuclide therapy (RNT) is not known. Therefore, we aimed to determine the presence of the TSGs and to quantify uptake in the TSGs on PSMA PET. METHODS Qualitative and quantitative analyses were performed on 68Ga-PSMA-11 PET/CT scans of 100 patients with prostate cancer. The mean and maximum standardized uptake value (SUVmean and SUVmax) in the TSGs were measured and compared to the parotid, submandibular and sublingual salivary glands (PSGs, SMSGs and SLSGs, respectively). Furthermore, proportional function of the TSGs was compared to the PSGs, SMSGs and SLSGs based on the total organ PSMA (TO-PSMA). RESULTS The TSGs were visible on 95% of the 68Ga-PSMA-11 PET/CT scans. The normalized median SUVmean and SUVmax was significantly higher for the PSGs (p < 0.001) and SMSGs (p < 0.001) compared to the TSGs, but not for the SLSGs (p = 0.242 and p = 0.300, respectively). The normalized median TO-PSMA was significantly higher for the PSGs (p < 0.001) and SMSGs (p < 0.001), and significant lower for the SLSGs (p < 0.001) compared the TSGs. CONCLUSIONS The SUVmean, SUVmax and TO-PSMA of the TSGs were most comparable to the SLSGs. However, the measured PSMA uptake may be disproportional towards the saliva production. Therefore, future studies should focus on the relation between PSMA uptake and salivary function before and after PSMA therapy.
Collapse
Affiliation(s)
- Sui Wai Ling
- Department of Radiology and Nuclear Medicine, Erasmus MC, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands.
| | - Astrid van der Veldt
- Department of Radiology and Nuclear Medicine, Erasmus MC, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
- Department of Medical Oncology, Erasmus MC, Rotterdam, the Netherlands
| | - Marcel Segbers
- Department of Radiology and Nuclear Medicine, Erasmus MC, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Henk Luiting
- Department of Urology, Erasmus MC, Rotterdam, the Netherlands
| | - Tessa Brabander
- Department of Radiology and Nuclear Medicine, Erasmus MC, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Frederik Verburg
- Department of Radiology and Nuclear Medicine, Erasmus MC, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| |
Collapse
|
2
|
Lindland K, Malenge MM, Li RG, Wouters R, Bønsdorff TB, Juzeniene A, Dragovic SM. Antigen targeting and anti-tumor activity of a novel anti-CD146 212Pb internalizing alpha-radioimmunoconjugate against malignant peritoneal mesothelioma. Sci Rep 2024; 14:25941. [PMID: 39472474 PMCID: PMC11522520 DOI: 10.1038/s41598-024-76778-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 10/16/2024] [Indexed: 11/02/2024] Open
Abstract
Malignant mesothelioma, a highly aggressive cancer that primarily affects the serosal membranes, has limited therapeutic options, particularly for cavitary tumors, such as peritoneal and pleural malignant mesothelioma. Intracavitary administration of a radioimmunoconjugate to locally target mesothelioma cancer cells has been proposed as a treatment. CD146, upregulated in mesothelioma but not in healthy tissues, is a promising therapeutic target. This study characterized CD146 expression and binding/internalization kinetics of the CD146-targeting antibody OI-3 coupled with 212Pb (212Pb-TCMC-OI-3) in human mesothelioma cells. Flow cytometry showed that both chimeric (chOI-3) and murine (mOI-3) antibodies rapidly bound and internalized within 1-6 h in MSTO-211H cells. 212Pb-TCMC-chOI-3 exhibited 3.1- to 13.7-fold and 3.1- to 8.5-fold increased internalized 212Pb and 212Bi atoms per cell at 2 and 24 h, respectively, compared to isotype control, underscoring enhanced internalization efficiency. Intraperitoneal administration of 212Pb-TCMC-mOI-3 to mice with intraperitoneal MSTO-211H xenografts improved median survival by a ratio of 1.3 compared to non-binding 212Pb-TCMC-mIgG1. The ability of 212Pb-TCMC-mOI-3 to target and inhibit the growth of intraperitoneal mesothelioma xenografts supports targeted radionuclide therapy's efficacy for metastatic peritoneal mesothelioma. This study highlights the potential of localized CD146-targeted radioimmunotherapy for malignant mesothelioma, offering a new avenue for improving patient outcomes.
Collapse
Affiliation(s)
- Kim Lindland
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0316, Oslo, Norway.
- Department of Radiation Biology, Institute of Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379, Oslo, Norway.
- Oncoinvent ASA, 0484, Oslo, Norway.
| | | | | | - Roxanne Wouters
- Oncoinvent ASA, 0484, Oslo, Norway
- Laboratory of Tumour Immunology and Immunotherapy, Department of Oncology, Leuven Cancer Institute, KU Leuven, 3000, Leuven, Belgium
| | | | - Asta Juzeniene
- Department of Radiation Biology, Institute of Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, 0379, Oslo, Norway
- Department of Physics, University of Oslo, Oslo, Norway
| | | |
Collapse
|
3
|
Muniz M, Loprinzi CL, Orme JJ, Koch RM, Mahmoud AM, Kase AM, Riaz IB, Andrews JR, Thorpe MP, Johnson GB, Kendi AT, Kwon ED, Nauseef JT, Morgans AK, Sartor O, Childs DS. Salivary toxicity from PSMA-targeted radiopharmaceuticals: What we have learned and where we are going. Cancer Treat Rev 2024; 127:102748. [PMID: 38703593 PMCID: PMC11160931 DOI: 10.1016/j.ctrv.2024.102748] [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: 03/20/2024] [Revised: 04/18/2024] [Accepted: 04/29/2024] [Indexed: 05/06/2024]
Abstract
Clinical trials of prostate-specific membrane antigen (PSMA) targeted radiopharmaceuticals have shown encouraging results. Some agents, like lutetium-177 [177Lu]Lu-PSMA-617 ([177Lu]Lu-PSMA-617), are already approved for late line treatment of metastatic castration-resistant prostate cancer (mCRPC). Projections are for continued growth of this treatment modality; [177Lu]Lu-PSMA-617 is being studied both in earlier stages of disease and in combination with other anti-cancer therapies. Further, the drug development pipeline is deep with variations of PSMA-targeting radionuclides, including higher energy alpha particles conjugated to PSMA-honing vectors. It is safe to assume that an increasing number of patients will be exposed to PSMA-targeted radiopharmaceuticals during the course of their cancer treatment. In this setting, it is important to better understand and mitigate the most commonly encountered toxicities. One particularly vexing side effect is xerostomia. In this review, we discuss the scope of the problem, inventories to better characterize and monitor this troublesome side effect, and approaches to preserve salivary function and effectively palliate symptoms. This article aims to serve as a useful reference for prescribers of PSMA-targeted radiopharmaceuticals, while also commenting on areas of missing data and opportunities for future research.
Collapse
Affiliation(s)
- Miguel Muniz
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, US.
| | | | - Jacob J Orme
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, US.
| | - Regina M Koch
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, US.
| | | | - Adam M Kase
- Department of Medical Oncology, Mayo Clinic, Jacksonville FL, US.
| | - Irbaz B Riaz
- Division of Hematology and Medical Oncology, Mayo Clinic, Scottsdale, AZ, US.
| | - Jack R Andrews
- Department of Urology, Mayo Clinic Arizona, Phoenix, AZ, US.
| | - Matthew P Thorpe
- Department of Radiology, Division of Nuclear Medicine, Mayo Clinic, Rochester, MN, US.
| | - Geoffrey B Johnson
- Department of Radiology, Division of Nuclear Medicine, Mayo Clinic, Rochester, MN, US; Department of Immunology, Mayo Clinic, Rochester, MN, US.
| | - Ayse T Kendi
- Department of Radiology, Division of Nuclear Medicine, Mayo Clinic, Rochester, MN, US.
| | - Eugene D Kwon
- Department of Urology, Mayo Clinic, Rochester, MN, US.
| | - Jones T Nauseef
- Division of Hematology & Medical Oncology, Weill Cornell Medicine, New York, NY, US.
| | - Alicia K Morgans
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, US.
| | - Oliver Sartor
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, US; Department of Radiology, Division of Nuclear Medicine, Mayo Clinic, Rochester, MN, US.
| | - Daniel S Childs
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, US.
| |
Collapse
|
4
|
Mattana F, Muraglia L, Barone A, Colandrea M, Saker Diffalah Y, Provera S, Cascio AS, Omodeo Salè E, Ceci F. Prostate-Specific Membrane Antigen-Targeted Therapy in Prostate Cancer: History, Combination Therapies, Trials, and Future Perspective. Cancers (Basel) 2024; 16:1643. [PMID: 38730595 PMCID: PMC11083597 DOI: 10.3390/cancers16091643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/22/2024] [Accepted: 04/23/2024] [Indexed: 05/13/2024] Open
Abstract
In the last decades, the development of PET/CT radiopharmaceuticals, targeting the Prostate-Specific Membrane Antigen (PSMA), changed the management of prostate cancer (PCa) patients thanks to its higher diagnostic accuracy in comparison with conventional imaging both in staging and in recurrence. Alongside molecular imaging, PSMA was studied as a therapeutic agent targeted with various isotopes. In 2021, results from the VISION trial led to the Food and Drug Administration (FDA) approval of [177Lu]Lu-PSMA-617 as a novel therapy for metastatic castration-resistant prostate cancer (mCRPC) and set the basis for a radical change in the future perspectives of PCa treatment and the history of Nuclear Medicine. Despite these promising results, primary resistance in patients treated with single-agent [177Lu]Lu-PSMA-617 remains a real issue. Emerging trials are investigating the use of [177Lu]Lu-PSMA-617 in combination with other PCa therapies in order to cover the multiple oncologic resistance pathways and to overcome tumor heterogeneity. In this review, our aim is to retrace the history of PSMA-targeted therapy from the first preclinical studies to its future applications in PCa.
Collapse
Affiliation(s)
- Francesco Mattana
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy; (A.B.); (M.C.); (A.S.C.); (F.C.)
| | - Lorenzo Muraglia
- Division of Nuclear Medicine, Humanitas IRCCS, 20141 Milan, Italy;
| | - Antonio Barone
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy; (A.B.); (M.C.); (A.S.C.); (F.C.)
| | - Marzia Colandrea
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy; (A.B.); (M.C.); (A.S.C.); (F.C.)
| | - Yasmina Saker Diffalah
- Division of Nuclear Medicine, Hospital Clínico Universitario Lozano Blesa, 50009 Zaragoza, Spain;
| | - Silvia Provera
- Division of Pharmacy, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy; (S.P.); (E.O.S.)
| | - Alfio Severino Cascio
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy; (A.B.); (M.C.); (A.S.C.); (F.C.)
| | - Emanuela Omodeo Salè
- Division of Pharmacy, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy; (S.P.); (E.O.S.)
| | - Francesco Ceci
- Division of Nuclear Medicine, IEO European Institute of Oncology IRCCS, 20141 Milan, Italy; (A.B.); (M.C.); (A.S.C.); (F.C.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| |
Collapse
|
5
|
Raitanen J, Barta B, Fuchs H, Hacker M, Balber T, Georg D, Mitterhauser M. Radiobiological Assessment of Targeted Radionuclide Therapy with [ 177Lu]Lu-PSMA-I&T in 2D vs. 3D Cell Culture Models. Int J Mol Sci 2023; 24:17015. [PMID: 38069337 PMCID: PMC10706939 DOI: 10.3390/ijms242317015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
In vitro therapeutic efficacy studies are commonly conducted in cell monolayers. However, three-dimensional (3D) tumor spheroids are known to better represent in vivo tumors. This study used [177Lu]Lu-PSMA-I&T, an already clinically applied radiopharmaceutical for targeted radionuclide therapy against metastatic castrate-resistant prostate cancer, to demonstrate the differences in the radiobiological response between 2D and 3D cell culture models of the prostate cancer cell lines PC-3 (PSMA negative) and LNCaP (PSMA positive). After assessing the target expression in both models via Western Blot, cell viability, reproductive ability, and growth inhibition were assessed. To investigate the geometric effects on dosimetry for the 2D vs. 3D models, Monte Carlo simulations were performed. Our results showed that PSMA expression in LNCaP spheroids was highly preserved, and target specificity was shown in both models. In monolayers of LNCaP, no short-term (48 h after treatment), but only long-term (14 days after treatment) radiobiological effects were evident, showing decreased viability and reproductive ability with the increasing activity. Further, LNCaP spheroid growth was inhibited with the increasing activity. Overall, treatment efficacy was higher in LNCaP spheroids compared to monolayers, which can be explained by the difference in the resulting dose, among others.
Collapse
Affiliation(s)
- Julia Raitanen
- Ludwig Boltzmann Institute Applied Diagnostics, 1090 Vienna, Austria; (J.R.)
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, 1090 Vienna, Austria
- Vienna Doctoral School of Chemistry (DoSChem), University of Vienna, 1090 Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
| | - Bernadette Barta
- Ludwig Boltzmann Institute Applied Diagnostics, 1090 Vienna, Austria; (J.R.)
| | - Hermann Fuchs
- Department of Radiation Oncology, Division of Medical Radiation Physics, Medical University of Vienna, 1090 Vienna, Austria
| | - Marcus Hacker
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, 1090 Vienna, Austria
| | - Theresa Balber
- Ludwig Boltzmann Institute Applied Diagnostics, 1090 Vienna, Austria; (J.R.)
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, 1090 Vienna, Austria
- Joint Applied Medicinal Radiochemistry Facility, Medical University of Vienna, University of Vienna, 1090 Vienna, Austria
| | - Dietmar Georg
- Department of Radiation Oncology, Division of Medical Radiation Physics, Medical University of Vienna, 1090 Vienna, Austria
| | - Markus Mitterhauser
- Ludwig Boltzmann Institute Applied Diagnostics, 1090 Vienna, Austria; (J.R.)
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, 1090 Vienna, Austria
- Institute of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria
- Joint Applied Medicinal Radiochemistry Facility, Medical University of Vienna, University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
6
|
Morgan KA, Rudd SE, Noor A, Donnelly PS. Theranostic Nuclear Medicine with Gallium-68, Lutetium-177, Copper-64/67, Actinium-225, and Lead-212/203 Radionuclides. Chem Rev 2023; 123:12004-12035. [PMID: 37796539 DOI: 10.1021/acs.chemrev.3c00456] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/06/2023]
Abstract
Molecular changes in malignant tissue can lead to an increase in the expression levels of various proteins or receptors that can be used to target the disease. In oncology, diagnostic imaging and radiotherapy of tumors is possible by attaching an appropriate radionuclide to molecules that selectively bind to these target proteins. The term "theranostics" describes the use of a diagnostic tool to predict the efficacy of a therapeutic option. Molecules radiolabeled with γ-emitting or β+-emitting radionuclides can be used for diagnostic imaging using single photon emission computed tomography or positron emission tomography. Radionuclide therapy of disease sites is possible with either α-, β-, or Auger-emitting radionuclides that induce irreversible damage to DNA. This Focus Review centers on the chemistry of theranostic approaches using metal radionuclides for imaging and therapy. The use of tracers that contain β+-emitting gallium-68 and β-emitting lutetium-177 will be discussed in the context of agents in clinical use for the diagnostic imaging and therapy of neuroendocrine tumors and prostate cancer. A particular emphasis is then placed on the chemistry involved in the development of theranostic approaches that use copper-64 for imaging and copper-67 for therapy with functionalized sarcophagine cage amine ligands. Targeted therapy with radionuclides that emit α particles has potential to be of particular use in late-stage disease where there are limited options, and the role of actinium-225 and lead-212 in this area is also discussed. Finally, we highlight the challenges that impede further adoption of radiotheranostic concepts while highlighting exciting opportunities and prospects.
Collapse
Affiliation(s)
- Katherine A Morgan
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Melbourne 3010, Australia
| | - Stacey E Rudd
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Melbourne 3010, Australia
| | - Asif Noor
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Melbourne 3010, Australia
| | - Paul S Donnelly
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Melbourne 3010, Australia
| |
Collapse
|
7
|
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
|
8
|
Kvassheim M, Tornes AJK, Juzeniene A, Stokke C, Revheim MER. Imaging of 212Pb in mice with a clinical SPECT/CT. EJNMMI Phys 2023; 10:47. [PMID: 37603123 PMCID: PMC10442031 DOI: 10.1186/s40658-023-00571-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 08/07/2023] [Indexed: 08/22/2023] Open
Abstract
INTRODUCTION 212Pb is a promising radionuclide for targeted alpha therapy. Here, the feasibility of visualising the tumour uptake and biodistribution of 212Pb-NG001 in mice with a clinical SPECT/CT scanner was investigated. METHODS A mouse phantom with 212Pb was imaged with a clinical- and a preclinical SPECT/CT scanner. Different acquisition and reconstruction settings were investigated on the clinical system (Siemens Symbia Intevo Bold). Two athymic nude mice carrying PC-3 PIP prostate cancer tumours of 235-830 μl received 1.44 MBq of 212Pb-NG001 and were imaged 2, 6, and 24 h post-injection on the clinical SPECT/CT with a Medium Energy collimator and a 40% energy window centred on 79 keV. All acquisition times were 30 min, except the mouse imaging 24 h post-injection which was 60 min. After the final imaging, the organs were harvested and measured on a gamma counter to give an indication of how much activity was present in organs of interest at the last imaging time point. RESULTS Four volumes in the mouse phantom of ~ 300 μl with 246-303 kBq/ml of 212Pb were distinguishable on images acquired with the clinical SPECT/CT with a high number of reconstruction updates. With the preclinical SPECT, the same volumes were easily distinguished with 49 kBq/ml of 212Pb. Clinical SPECT/CT images of the mice revealed uptake in tumours and bladders 2 h after injection and in tumours containing down to approximately 15 kBq/ml at 6 and 24 h after injection. CONCLUSION Although the preclinical scanner should be used preferentially in biodistribution studies in mice, the clinical SPECT/CT confirmed uptake in small volumes (e.g. ~ 300 μl volume with ~ 250 kBq/ml). Regardless of system, the resolution and sensitivity limits should be carefully determined, otherwise false negative or too low uptakes can be wrongly interpreted.
Collapse
Affiliation(s)
- Monika Kvassheim
- Division of Radiology and Nuclear Medicine, Department of Physics and Computational Radiology, Oslo University Hospital, Oslo, Norway.
- Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Anna Julie Kjøl Tornes
- Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- ARTBIO AS, Oslo, Norway
| | - Asta Juzeniene
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Physics, University of Oslo, Oslo, Norway
| | - Caroline Stokke
- Division of Radiology and Nuclear Medicine, Department of Physics and Computational Radiology, Oslo University Hospital, Oslo, Norway
- Department of Physics, University of Oslo, Oslo, Norway
| | - Mona-Elisabeth R Revheim
- Faculty of Medicine, University of Oslo, Oslo, Norway
- Division of Radiology and Nuclear Medicine, Department of Nuclear Medicine, Oslo University Hospital, Oslo, Norway
- The Intervention Centre, Division of Technology and Innovation, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
9
|
Deshayes E, Fersing C, Thibault C, Roumiguie M, Pourquier P, Houédé N. Innovation in Radionuclide Therapy for the Treatment of Prostate Cancers: Radiochemical Perspective and Recent Therapeutic Practices. Cancers (Basel) 2023; 15:3133. [PMID: 37370743 DOI: 10.3390/cancers15123133] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/31/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
Prostate cancer represents the second cause of death by cancer in males in western countries. While early-stage diseases are accessible to surgery and/or external radiotherapy, advanced metastatic prostate cancers are primarily treated with androgen deprivation therapy, to which new generation androgen receptor antagonists or taxane-based chemotherapies are added in the case of tumor relapse. Nevertheless, patients become invariably resistant to castration with a median survival that rarely exceeds 3 years. This fostered the search for alternative strategies, independent of the androgen receptor signaling pathway. In this line, radionuclide therapies may represent an interesting option as they could target either the microenvironment of sclerotic bone metastases with the use of radiopharmaceuticals containing samarium-153, strontium-89 or radium-223 or tumor cells expressing the prostate-specific membrane antigen (PSMA), a protein found at the surface of prostate cancer cells. This review gives highlights the chemical properties of radioligands targeting prostate cancer cells and recapitulates the clinical trials evaluating the efficacy of radionuclide therapies, alone or in combination with other approved treatments, in patients with castration-resistant prostate tumors. It discusses some of the encouraging results obtained, especially the benefit on overall survival that was reported with [177Lu]-PSMA-617. It also addresses the specific requirements for the use of this particular class of drugs, both in terms of medical staff coordination and adapted infrastructures for efficient radioprotection.
Collapse
Affiliation(s)
- Emmanuel Deshayes
- INSERM U1194, Montpellier Cancer Research Institute, University of Montpellier, 34298 Montpellier, France
- Department of Nuclear Medicine, Institute du Cancer de Montpellier (ICM), 34298 Montpellier, France
| | - Cyril Fersing
- Department of Nuclear Medicine, Institute du Cancer de Montpellier (ICM), 34298 Montpellier, France
- IBMM, University Montpellier, CNRS, ENSCM, 34293 Montpellier, France
| | - Constance Thibault
- Department of Medical Oncology, Hôpital Européen Georges Pompidou, Institut du Cancer Paris CARPEM, AP-HP Centre, 75015 Paris, France
| | - Mathieu Roumiguie
- Urology Department, Andrology and Renal Transplantation, CHU Rangueil, 31059 Toulouse, France
| | - Philippe Pourquier
- INSERM U1194, Montpellier Cancer Research Institute, University of Montpellier, 34298 Montpellier, France
| | - Nadine Houédé
- INSERM U1194, Montpellier Cancer Research Institute, University of Montpellier, 34298 Montpellier, France
- Medical Oncology Department, Institute de Cancérologie du Gard-CHU Caremeau, 30009 Nîmes, France
| |
Collapse
|
10
|
Let’s Go 3D! New Generation of Models for Evaluating Drug Response and Resistance in Prostate Cancer. Int J Mol Sci 2023; 24:ijms24065293. [PMID: 36982368 PMCID: PMC10049142 DOI: 10.3390/ijms24065293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/06/2023] [Accepted: 03/08/2023] [Indexed: 03/12/2023] Open
Abstract
Prostate cancer (PC) is the third most frequently diagnosed cancer worldwide and the second most frequent in men. Several risk factors can contribute to the development of PC, and those include age, family history, and specific genetic mutations. So far, drug testing in PC, as well as in cancer research in general, has been performed on 2D cell cultures. This is mainly because of the vast benefits these models provide, including simplicity and cost effectiveness. However, it is now known that these models are exposed to much higher stiffness; lose physiological extracellular matrix on artificial plastic surfaces; and show changes in differentiation, polarization, and cell–cell communication. This leads to the loss of crucial cellular signaling pathways and changes in cell responses to stimuli when compared to in vivo conditions. Here, we emphasize the importance of a diverse collection of 3D PC models and their benefits over 2D models in drug discovery and screening from the studies done so far, outlining their benefits and limitations. We highlight the differences between the diverse types of 3D models, with the focus on tumor–stroma interactions, cell populations, and extracellular matrix composition, and we summarize various standard and novel therapies tested on 3D models of PC for the purpose of raising awareness of the possibilities for a personalized approach in PC therapy.
Collapse
|
11
|
Juzeniene A, Stenberg VY, Bruland ØS, Revheim ME, Larsen RH. Dual targeting with 224Ra/ 212Pb-conjugates for targeted alpha therapy of disseminated cancers: A conceptual approach. Front Med (Lausanne) 2023; 9:1051825. [PMID: 36733936 PMCID: PMC9887039 DOI: 10.3389/fmed.2022.1051825] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 12/05/2022] [Indexed: 01/18/2023] Open
Abstract
Metastases are the primary cause of death among cancer patients and efficacious new treatments are sorely needed. Targeted alpha-emitting radiopharmaceuticals that are highly cytotoxic may fulfill this critical need. The focus of this paper is to describe and explore a novel technology that may improve the therapeutic effect of targeted alpha therapy by combining two radionuclides from the same decay chain in the same solution. We hypothesize that the dual targeting solution containing bone-seeking 224Ra and cell-directed complexes of progeny 212Pb is a promising approach to treat metastatic cancers with bone and soft tissue lesions as well as skeletal metastases of mixed lytic/osteoblastic nature. A novel liquid 224Ra/212Pb-generator for rapid preparation of a dual targeting solution is described. Cancer cell targeting monoclonal antibodies, their fragments, synthetic proteins or peptides can all be radiolabeled with 212Pb in the 224Ra-solution in transient equilibrium with daughter nuclides. Thus, 224Ra targets stromal elements in sclerotic bone metastases and 212Pb-chelated-conjugate targets tumor cells of metastatic prostate cancer or osteosarcoma. The dual targeting solution may also be explored to treat metastatic breast cancer or multiple myeloma after manipulation of bone metastases to a more osteoblastic phenotype by the use of bisphosphonates, denosumab, bortezomib or hormone therapy prior to treatment. This may improve targeting of bone-seeking 224Ra and render an augmented radiation dose deposited within metastases. Our preliminary preclinical studies provide conceptual evidence that the dual 224Ra-solution with bone or tumor-targeted delivery of 212Pb has potential to inhibit cancer metastases without significant toxicity. In some settings, the use of a booster dose of purified 212Pb-conjugate alone could be required to elevate the effect of this tumor cell directed component, if needed, e.g., in a fractionated treatment regimen, where the dual targeting solution will act as maintenance treatment.
Collapse
Affiliation(s)
- Asta Juzeniene
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Department of Physics, University of Oslo, Oslo, Norway
| | - Vilde Yuli Stenberg
- Department of Radiation Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Institute for Clinical Medicine, University of Oslo, Oslo, Norway
- ARTBIO AS, Oslo, Norway
| | - Øyvind Sverre Bruland
- Institute for Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Oncology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
| | - Mona-Elisabeth Revheim
- Institute for Clinical Medicine, University of Oslo, Oslo, Norway
- Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | | |
Collapse
|
12
|
Tornes AJK, Stenberg VY, Larsen RH, Bruland ØS, Revheim ME, Juzeniene A. Targeted alpha therapy with the 224Ra/ 212Pb-TCMC-TP-3 dual alpha solution in a multicellular tumor spheroid model of osteosarcoma. Front Med (Lausanne) 2022; 9:1058863. [PMID: 36507500 PMCID: PMC9727293 DOI: 10.3389/fmed.2022.1058863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/04/2022] [Indexed: 11/24/2022] Open
Abstract
Osteosarcoma patients with overt metastases at primary diagnosis have a 5-year survival rate of less than 20%. TP-3 is a murine IgG2b monoclonal antibody with high affinity for an epitope residing on the p80 osteosarcoma cell surface membrane antigen. The tumor-associated antigen p80 is overexpressed in osteosarcomas, and has very low normal tissue expression. We propose a novel dual alpha targeting solution containing two radionuclides from the same decay chain, including the bone-seeking 224Ra, and cancer cell-surface seeking 212Pb-TCMC-TP-3 for the treatment of osteoblastic bone cancers, circulating cancer cells and micrometastases. In this in vitro study, the cytotoxic effects of 212Pb-TCMC-TP-3 (single alpha solution) and 224Ra/212Pb-TCMC-TP-3 (dual alpha solution) were investigated in a multicellular spheroid model mimicking micrometastatic disease in osteosarcoma. OHS spheroids with diameters of 253 ± 98 μm treated with 4.5, 2.7, and 3.3 kBq/ml of 212Pb-TCMC-TP-3 for 1, 4, and 24 h, respectively, were disintegrated within 3 weeks. The 212Pb-TCMC-TP-3 induced a 7-fold delay in spheroid doubling time compared to a 28-times higher dose with the non-specific 212Pb-TCMC-rituximab. The 224Ra/212Pb-TCMC-TP-3 completely disintegrated spheroids with diameters of 218-476 μm within 3 and 2 weeks after 4 and 24 h incubation with 5 kBq/ml, respectively. Treatment with 1 kBq/ml of 224Ra/212Pb-TCMC-TP-3 for 24 h caused an 11.4-fold reduction in spheroid viability compared with unconjugated 224Ra/212Pb. The single and dual alpha solutions with TP-3 showed cytotoxicity in spheroids of clinically relevant size, which warrant further testing of the dual alpha solution using in vivo osteosarcoma models.
Collapse
Affiliation(s)
- Anna Julie Kjøl Tornes
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway,ArtBio AS, Oslo, Norway,Institute of Clinical Medicine, University of Oslo, Oslo, Norway,*Correspondence: Anna Julie Kjøl Tornes,
| | - Vilde Yuli Stenberg
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway,ArtBio AS, Oslo, Norway,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Øyvind Sverre Bruland
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Mona-Elisabeth Revheim
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway,Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Asta Juzeniene
- Department of Radiation Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway,Department of Physics, University of Oslo, Oslo, Norway
| |
Collapse
|
13
|
Kvassheim M, Revheim MER, Stokke C. Quantitative SPECT/CT imaging of lead-212: a phantom study. EJNMMI Phys 2022; 9:52. [PMID: 35925521 PMCID: PMC9352840 DOI: 10.1186/s40658-022-00481-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 07/20/2022] [Indexed: 12/23/2022] Open
Abstract
Background Lead-212 (212Pb) is a promising radionuclide for targeted therapy, as it decays to α-particle emitter bismuth-212 (212Bi) via β-particle emission. This extends the problematic short half-life of 212Bi. In preparation for upcoming clinical trials with 212Pb, the feasibility of quantitative single photon-emission computed tomography/computed tomography (SPECT/CT) imaging of 212Pb was studied, with the purpose to explore the possibility of individualised patient dosimetric estimation. Results Both acquisition parameters (combining two different energy windows and two different collimators) and iterative reconstruction parameters (varying the iterations x subsets between 10 × 1, 15 × 1, 30 × 1, 30 × 2, 30 × 3, 30 × 4, and 30 × 30) were investigated to evaluate visual quality and quantitative uncertainties based on phantom images. Calibration factors were determined using a homogeneous phantom and were stable when the total activity imaged exceeded 1 MBq for all the imaging protocols studied, but they increased sharply as the activity decayed below 1 MBq. Both a 20% window centred on 239 keV and a 40% window on 79 keV, with dual scatter windows of 5% and 20%, respectively, could be used. Visual quality at the lowest activity concentrations was improved with the High Energy collimator and the 79 keV energy window. Fractional uncertainty in the activity quantitation, including uncertainties from calibration factors and small volume effects, in spheres of 2.6 ml in the NEMA phantom was 16–21% for all protocols with the 30 × 4 filtered reconstruction except the High Energy collimator with the 239 keV energy window. Quantitative analysis was possible both with and without filters, but the visual quality of the images improved with a filter. Conclusions Only minor differences were observed between the imaging protocols which were all determined suitable for quantitative imaging of 212Pb. As uncertainties generally decreased with increasing iterative updates in the reconstruction and recovery curves did not converge with few iterations, a high number of reconstruction updates are recommended for quantitative imaging. Supplementary Information The online version contains supplementary material available at 10.1186/s40658-022-00481-z.
Collapse
Affiliation(s)
- Monika Kvassheim
- Department of Physics and Computational Radiology, Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway. .,Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Mona-Elisabeth R Revheim
- Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Nuclear Medicine, Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway
| | - Caroline Stokke
- Department of Physics and Computational Radiology, Division of Radiology and Nuclear Medicine, Oslo University Hospital, Oslo, Norway.,Department of Physics, University of Oslo, Oslo, Norway
| |
Collapse
|
14
|
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
|
15
|
Jeitner TM, Babich JW, Kelly JM. Advances in PSMA theranostics. Transl Oncol 2022; 22:101450. [PMID: 35597190 PMCID: PMC9123266 DOI: 10.1016/j.tranon.2022.101450] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/04/2022] [Accepted: 05/08/2022] [Indexed: 12/15/2022] Open
Abstract
PSMA is an appealing target for theranostic because it is a transmembrane protein with a known substrate that is overexpessed on prostate cancer cells and internalizes upon ligand binding. There are a number of PSMA theranostic ligands in clinical evaluation, clinical trial, or clinically approved. PSMA theranostic ligands increase progression-free survival, overall survival, and pain in patients with metastatic castration resistant prostate cancer. A major obstacle to PSMA-targeted radioligand therapy is off-target toxicity in salivary glands.
The validation of prostate specific membrane antigen (PSMA) as a molecular target in metastatic castration-resistant prostate cancer has stimulated the development of multiple classes of theranostic ligands that specifically target PSMA. Theranostic ligands are used to image disease or selectively deliver cytotoxic radioactivity to cells expressing PSMA according to the radioisotope conjugated to the ligand. PSMA theranostics is a rapidly advancing field that is now integrating into clinical management of prostate cancer patients. In this review we summarize published research describing the biological role(s) and activity of PSMA, highlight the most clinically advanced PSMA targeting molecules and biomacromolecules, and identify next generation PSMA ligands that aim to further improve treatment efficacy. The goal of this review is to provide a comprehensive assessment of the current state-of-play and a roadmap to achieving further advances in PSMA theranostics.
Collapse
Affiliation(s)
- Thomas M Jeitner
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, Belfer Research Building, 413 East 69th Street, Room BB-1604, New York, NY 10021, USA
| | - John W Babich
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, Belfer Research Building, 413 East 69th Street, Room BB-1604, New York, NY 10021, USA; Weill Cornell Medicine, Sandra and Edward Meyer Cancer Center, New York, NY 10021, USA; Weill Cornell Medicine, Citigroup Biomedical Imaging Center, New York, NY 10021, USA
| | - James M Kelly
- Molecular Imaging Innovations Institute, Department of Radiology, Weill Cornell Medicine, Belfer Research Building, 413 East 69th Street, Room BB-1604, New York, NY 10021, USA; Weill Cornell Medicine, Citigroup Biomedical Imaging Center, New York, NY 10021, USA.
| |
Collapse
|
16
|
On the Way for Patients with Prostate Cancer to the Best Use of PSMA. Int J Mol Sci 2022; 23:ijms23052478. [PMID: 35269620 PMCID: PMC8909989 DOI: 10.3390/ijms23052478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 02/17/2022] [Indexed: 02/01/2023] Open
|
17
|
Kokov KV, Egorova BV, German MN, Klabukov ID, Krasheninnikov ME, Larkin-Kondrov AA, Makoveeva KA, Ovchinnikov MV, Sidorova MV, Chuvilin DY. 212Pb: Production Approaches and Targeted Therapy Applications. Pharmaceutics 2022; 14:pharmaceutics14010189. [PMID: 35057083 PMCID: PMC8777968 DOI: 10.3390/pharmaceutics14010189] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/30/2021] [Accepted: 12/30/2021] [Indexed: 01/18/2023] Open
Abstract
Over the last decade, targeted alpha therapy has demonstrated its high effectiveness in treating various oncological diseases. Lead-212, with a convenient half-life of 10.64 h, and daughter alpha-emitter short-lived 212Bi (T1/2 = 1 h), provides the possibility for the synthesis and purification of complex radiopharmaceuticals with minimum loss of radioactivity during preparation. As a benefit for clinical implementation, it can be milked from a radionuclide generator in different ways. The main approaches applied for these purposes are considered and described in this review, including chromatographic, solution, and other techniques to isolate 212Pb from its parent radionuclide. Furthermore, molecules used for lead’s binding and radiochemical features of preparation and stability of compounds labeled with 212Pb are discussed. The results of preclinical studies with an estimation of therapeutic and tolerant doses as well as recently initiated clinical trials of targeted radiopharmaceuticals are presented.
Collapse
Affiliation(s)
- Konstantin V. Kokov
- Physical and Chemical Technology Center, National Research Center Kurchatov Institute, 123182 Moscow, Russia; (K.V.K.); (M.N.G.); (A.A.L.-K.); (K.A.M.); (D.Y.C.)
| | - Bayirta V. Egorova
- Department of Chemistry, Lomonosov Moscow State University, 119991 Moscow, Russia
- Correspondence: or
| | - Marina N. German
- Physical and Chemical Technology Center, National Research Center Kurchatov Institute, 123182 Moscow, Russia; (K.V.K.); (M.N.G.); (A.A.L.-K.); (K.A.M.); (D.Y.C.)
| | - Ilya D. Klabukov
- Department of Regenerative Medicine, National Medical Research Radiological Center, 249036 Obninsk, Russia;
| | - Michael E. Krasheninnikov
- Research and Educational Resource Center for Cellular Technologies, Peoples’ Friendship University of Russia, 117198 Moscow, Russia;
| | - Antonius A. Larkin-Kondrov
- Physical and Chemical Technology Center, National Research Center Kurchatov Institute, 123182 Moscow, Russia; (K.V.K.); (M.N.G.); (A.A.L.-K.); (K.A.M.); (D.Y.C.)
| | - Kseniya A. Makoveeva
- Physical and Chemical Technology Center, National Research Center Kurchatov Institute, 123182 Moscow, Russia; (K.V.K.); (M.N.G.); (A.A.L.-K.); (K.A.M.); (D.Y.C.)
| | - Michael V. Ovchinnikov
- Laboratory of Peptide Synthesis, National Medical Research Center of Cardiology, 121552 Moscow, Russia; (M.V.O.); (M.V.S.)
| | - Maria V. Sidorova
- Laboratory of Peptide Synthesis, National Medical Research Center of Cardiology, 121552 Moscow, Russia; (M.V.O.); (M.V.S.)
| | - Dmitry Y. Chuvilin
- Physical and Chemical Technology Center, National Research Center Kurchatov Institute, 123182 Moscow, Russia; (K.V.K.); (M.N.G.); (A.A.L.-K.); (K.A.M.); (D.Y.C.)
| |
Collapse
|
18
|
Yang H, Wilson JJ, Orvig C, Li Y, Wilbur DS, Ramogida CF, Radchenko V, Schaffer P. Harnessing α-Emitting Radionuclides for Therapy: Radiolabeling Method Review. J Nucl Med 2022; 63:5-13. [PMID: 34503958 PMCID: PMC8717181 DOI: 10.2967/jnumed.121.262687] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/05/2021] [Indexed: 11/16/2022] Open
Abstract
Targeted α-therapy (TAT) is an emerging powerful tool treating late-stage cancers for which therapeutic options are limited. At the core of TAT are targeted radiopharmaceuticals, where isotopes are paired with targeting vectors to enable tissue- or cell-specific delivery of α-emitters. DOTA (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid) and DTPA (diethylenetriamine pentaacetic acid) are commonly used to chelate metallic radionuclides but have limitations. Significant efforts are underway to develop effective stable chelators for α-emitters and are at various stages of development and community adoption. Isotopes such as 149Tb, 212/213Bi, 212Pb (for 212Bi), 225Ac, and 226/227Th have found suitable chelators, although further studies, especially in vivo studies, are required. For others, including 223Ra, 230U, and, arguably 211At, the ideal chemistry remains elusive. This review summarizes the methods reported to date for the incorporation of 149Tb, 211At, 212/213Bi, 212Pb (for 212Bi), 223Ra, 225Ac, 226/227Th, and 230U into radiopharmaceuticals, with a focus on new discoveries and remaining challenges.
Collapse
Affiliation(s)
- Hua Yang
- Life Sciences Division, TRIUMF, Vancouver, British Columbia, Canada;
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Justin J Wilson
- Chemistry and Chemical Biology, Cornell University, Ithaca, New York
| | - Chris Orvig
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada
| | - Yawen Li
- Department of Radiation Oncology, University of Washington, Seattle, Washington
| | - D Scott Wilbur
- Department of Radiation Oncology, University of Washington, Seattle, Washington
| | - Caterina F Ramogida
- Life Sciences Division, TRIUMF, Vancouver, British Columbia, Canada
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Valery Radchenko
- Life Sciences Division, TRIUMF, Vancouver, British Columbia, Canada
- Department of Chemistry, University of British Columbia, Vancouver, British Columbia, Canada; and
| | - Paul Schaffer
- Life Sciences Division, TRIUMF, Vancouver, British Columbia, Canada
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia, Canada
- Department of Radiology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
19
|
Neels OC, Kopka K, Liolios C, Afshar-Oromieh A. Radiolabeled PSMA Inhibitors. Cancers (Basel) 2021; 13:6255. [PMID: 34944875 PMCID: PMC8699044 DOI: 10.3390/cancers13246255] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/10/2021] [Accepted: 12/11/2021] [Indexed: 12/16/2022] Open
Abstract
PSMA has shown to be a promising target for diagnosis and therapy (theranostics) of prostate cancer. We have reviewed developments in the field of radio- and fluorescence-guided surgery and targeted photodynamic therapy as well as multitargeting PSMA inhibitors also addressing albumin, GRPr and integrin αvβ3. An overview of the regulatory status of PSMA-targeting radiopharmaceuticals in the USA and Europe is also provided. Technical and quality aspects of PSMA-targeting radiopharmaceuticals are described and new emerging radiolabeling strategies are discussed. Furthermore, insights are given into the production, application and potential of alternatives beyond the commonly used radionuclides for radiolabeling PSMA inhibitors. An additional refinement of radiopharmaceuticals is required in order to further improve dose-limiting factors, such as nephrotoxicity and salivary gland uptake during endoradiotherapy. The improvement of patient treatment achieved by the advantageous combination of radionuclide therapy with alternative therapies is also a special focus of this review.
Collapse
Affiliation(s)
- Oliver C. Neels
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Bautzner Landstrasse 400, 01328 Dresden, Germany;
| | - Klaus Kopka
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Bautzner Landstrasse 400, 01328 Dresden, Germany;
- Faculty of Chemistry and Food Chemistry, School of Science, Technical University Dresden, Mommsenstrasse 4, 01062 Dresden, Germany
| | - Christos Liolios
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, National & Kapodistrian University of Athens, Zografou, 15771 Athens, Greece;
- INRASTES, Radiochemistry Laboratory, NCSR “Demokritos”, Ag. Paraskevi Attikis, 15310 Athens, Greece
| | - Ali Afshar-Oromieh
- Department of Nuclear Medicine, Bern University Hospital (Inselspital), Freiburgstrasse 18, 3010 Bern, Switzerland;
| |
Collapse
|
20
|
A Novel Single-Step-Labeled 212Pb-CaCO 3 Microparticle for Internal Alpha Therapy: Preparation, Stability, and Preclinical Data from Mice. MATERIALS 2021; 14:ma14237130. [PMID: 34885283 PMCID: PMC8658347 DOI: 10.3390/ma14237130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/19/2021] [Accepted: 11/19/2021] [Indexed: 02/06/2023]
Abstract
Lead-212 is recognized as a promising radionuclide for targeted alpha therapy for tumors. Many studies of 212Pb-labeling of various biomolecules through bifunctional chelators have been conducted. Another approach to exploiting the cytotoxic effect is coupling the radionuclide to a microparticle acting as a carrier vehicle, which could be used for treating disseminated cancers in body cavities. Calcium carbonate may represent a suitable material, as it is biocompatible, biodegradable, and easy to synthesize. In this work, we explored 212Pb-labeling of various CaCO3 microparticles and developed a protocol that can be straightforwardly implemented by clinicians. Vaterite microparticles stabilized by pamidronate were effective as 212Pb carriers; labeling yields of ≥98% were achieved, and 212Pb was strongly retained by the particles in an in vitro stability assessment. Moreover, the amounts of 212Pb reaching the kidneys, liver, spleen, and skeleton of mice following intraperitoneal (i.p.) administration were very low compared to i.p. injection of unbound 212Pb2+, indicating that CaCO3-bound 212Pb exhibited stability when administered intraperitoneally. Therapeutic efficacy was observed in a model of i.p. ovarian cancer for all the tested doses, ranging from 63 to 430 kBq per mouse. Lead-212-labeled CaCO3 microparticles represent a promising candidate for treating intracavitary cancers.
Collapse
|