1
|
Sancho L, Roteta A, Torres I, de Arcocha M, Ramos R, Domínguez ML, Rosales JJ, Prieto E, Quincoces G. State of the art and future perspectives of new radionuclides in Nuclear Medicine. Part II. Rev Esp Med Nucl Imagen Mol 2025:500128. [PMID: 40147757 DOI: 10.1016/j.remnie.2025.500128] [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: 02/04/2025] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/29/2025]
Abstract
The state of the art and future perspectives of new radionuclides in Nuclear Medicine continue to evolve, driven by the development of isotopes with innovative applications in theragnostics. In this second part of the continuing education series, the clinical and therapeutic applications of terbium, actinium, and bismuth are analyzed in depth. The use of the four terbium isotopes (terbium-149, terbium-152, terbium-155, and terbium-161) is described, offering a versatile system for both diagnosis and treatment due to their chemical similarity to lutetium-177, along with the challenges related to their production and availability. Additionally, actinium-225, a powerful alpha-emitting radionuclide, is reviewed for its growing role in Targeted Alpha Therapy (TAT), particularly in prostate cancer and neuroendocrine tumors. Finally, bismuth-213, derived from actinium-225, is analyzed for its short half-life, making it a viable option for localized and selective therapies. Despite technical and production challenges, these radionuclides are driving the evolution of precision medicine, expanding therapeutic and diagnostic possibilities in Nuclear Medicine.
Collapse
Affiliation(s)
- L Sancho
- Servicio de Medicina Nuclear, Clínica Universidad de Navarra, Madrid, Spain
| | - A Roteta
- Servicio de Medicina Nuclear, Hospital Universitario de Donostia, San Sebastián, Spain
| | - I Torres
- Servicio de Medicina Nuclear, Hospital La Fe, Valencia, Spain
| | - M de Arcocha
- Unidad de Radiofarmacia, Servicio de Medicina Nuclear, Hospital Universitario Marqués de Valdecilla, Santander, Spain; Grupo de Imagen Molecular IDIVAL, Santander, Spain
| | - R Ramos
- Unidad de Radiofarmacia, Servicio de Medicina Nuclear, Clínica Universidad de Navarra, Madrid, Spain
| | - M L Domínguez
- Servicio de Medicina Nuclear, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - J J Rosales
- Servicio de Medicina Nuclear, Clínica Universidad de Navarra, Pamplona, Spain
| | - E Prieto
- Servicio de Radiofísica y Protección Radiológica, Clínica Universidad de Navarra, Pamplona, Spain.
| | - G Quincoces
- Unidad de Radiofarmacia, Servicio de Medicina Nuclear, Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
2
|
Lacerda S, de Kruijff RM, Djanashvili K. The Advancement of Targeted Alpha Therapy and the Role of Click Chemistry Therein. Molecules 2025; 30:1296. [PMID: 40142070 PMCID: PMC11944744 DOI: 10.3390/molecules30061296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/21/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Recent years have seen a swift rise in the use of α-emitting radionuclides such as 225Ac and 223Ra as various radiopharmaceuticals to treat (micro)metastasized tumors. They have shown remarkable effectiveness in clinical practice owing to the highly cytotoxic α-particles that are emitted, which have a very short range in tissue, causing mainly double-stranded DNA breaks. However, it is essential that both chelation and targeting strategies are optimized for their successful translation to clinical application, as α-emitting radionuclides have distinctly different features compared to β--emitters, including their much larger atomic radius. Furthermore, upon α-decay, any daughter nuclide irrevocably breaks free from the targeting molecule, known as the recoil effect, dictating the need for faster targeting to prevent healthy tissue toxicity. In this review we provide a brief overview of the current status of targeted α-therapy and highlight innovations in α-emitter-based chelator design, focusing on the role of click chemistry to allow for fast complexation to biomolecules at mild labeling conditions. Finally, an outlook is provided on different targeting strategies and the role that pre-targeting can play in targeted alpha therapy.
Collapse
Affiliation(s)
- Sara Lacerda
- Centre de Biophysique Moléculaire, CNRS UPR 4301, Université d’Orléans, Rue Charles Sadron, 45071 Orléans, France;
| | - Robin M. de Kruijff
- Department of Radiation Science and Technology, Delft University of Technology, Mekelweg 15, 2629 JB Delft, The Netherlands;
| | - Kristina Djanashvili
- Department of Radiation Science and Technology, Delft University of Technology, Mekelweg 15, 2629 JB Delft, The Netherlands;
- Department of Biotechnology, Delft University of Technology, Van der Maasweg 9, 2629 HZ Delft, The Netherlands
| |
Collapse
|
3
|
Wenker STM, van Lith SAM, Tamborino G, Konijnenberg MW, Bussink J, Heskamp S. The potential of targeted radionuclide therapy to treat hypoxic tumor cells. Nucl Med Biol 2025; 140-141:108971. [PMID: 39579561 DOI: 10.1016/j.nucmedbio.2024.108971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 11/06/2024] [Accepted: 11/11/2024] [Indexed: 11/25/2024]
Abstract
Tumor hypoxia contributes to cancer progression and therapy resistance. Several strategies have been investigated to relieve tumor hypoxia, of which some were successful. However, their clinical application remains challenging and therefore they are not used in daily clinical practice. Here, we review the potential of targeted radionuclide therapy (TRT) to eradicate hypoxic cancer cells. We present an overview of the published TRT strategies using β--particles, α-particles, and Auger electrons. Altogether, we conclude that α-particle emitting radionuclides are most promising since they can cause DNA double strand breaks independent of oxygen levels. Future directions for research are addressed, including more adequate in vitro and in vivo models to proof the potential of TRT to eliminate hypoxic cancer cells. Furthermore, dosimetry and radiobiology are identified as key to better understand the mechanism of action and dose-response relationships in hypoxic tumor areas. Finally, we can conclude that in order to achieve long-term anti-tumor efficacy, TRT combination treatment strategies may be necessary.
Collapse
Affiliation(s)
- S T M Wenker
- Department of Medical Imaging, Nuclear Medicine, Radboudumc, Nijmegen, the Netherlands; Department of Radiation Oncology, Radiotherapy & Oncoimmunology laboratory, Radboudumc, Nijmegen, the Netherlands
| | - S A M van Lith
- Department of Medical Imaging, Nuclear Medicine, Radboudumc, Nijmegen, the Netherlands
| | - G Tamborino
- Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - M W Konijnenberg
- Department of Medical Imaging, Nuclear Medicine, Radboudumc, Nijmegen, the Netherlands; Department of Radiology & Nuclear Medicine, Erasmus MC, Rotterdam, the Netherlands
| | - J Bussink
- Department of Radiation Oncology, Radiotherapy & Oncoimmunology laboratory, Radboudumc, Nijmegen, the Netherlands
| | - S Heskamp
- Department of Medical Imaging, Nuclear Medicine, Radboudumc, Nijmegen, the Netherlands.
| |
Collapse
|
4
|
Sakmár M, Kozempel J, Kučka J, Janská T, Štíbr M, Ondrák L, Ondrák Fialová K, Vlk M, Šefc L, Bruchertseifer F, Morgenstern A. In vitro and in vivo study of 221Fr and 213Bi progeny release from the 225Ac-labelled TiO 2 nanoparticles. Nucl Med Biol 2025; 140-141:108973. [PMID: 39579560 DOI: 10.1016/j.nucmedbio.2024.108973] [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: 06/10/2024] [Revised: 10/03/2024] [Accepted: 11/11/2024] [Indexed: 11/25/2024]
Abstract
BACKGROUND Targeted alpha therapy (TAT) is an effective option for cancer treatment. To maximize its efficacy and minimize side effects, carriers must deliver radionuclides to target tissues. Most of the nuclides used in TAT decay via the alpha cascade, producing several radioactive daughter nuclei with sufficient energy to escape from the original carrier. Therefore, studying these daughter atoms is crucial in the search for new carriers. Nanoparticles have potential as carriers due to their structure, which can prevent the escape of daughter atoms and reduce radiation exposure to non-target tissues. This work focuses on determining the released activity of 221Fr and 213Bi resulting from the decay of 225Ac labelled TiO2 nanoparticles. RESULTS Labelling of TiO2 nanoparticles has shown high sorption rates of 225Ac and its progeny, 221Fr and 213Bi, with over 92 % of activities sorbed on the nanoparticle surface for all measured radionuclides. However, in the quasi-dynamic in vitro system, the released activity of 221Fr and 213Bi is strongly dependent on the nanoparticles concentration, ranging from 15 % for a concentration of 1 mg/mL to approximately 50 % for a nanoparticle concentration of 10 μg/mL in saline solution. The released activities of 213Bi were lower, with a maximum value of around 20 % for concentrations of 0.05, 0.025, and 0.01 mg/mL. The leakage of 225Ac and its progeny was tested in various biological matrices. Minimal released activity was measured in saline at around 10 % after 48 h, while the maximum activity was measured in blood serum and plasma at 20 %. The amount of 225Ac released into the media was minimal (<3 %). The in vitro results were confirmed in a healthy mouse model. The difference in %ID/g was clearly visible immediately after dissection and again after 6 h when 213Bi reached equilibrium with 225Ac. CONCLUSION The study verified the potential release of 225Ac progeny from the labelled TiO2 nanoparticles. Experiments were performed to determine the dependence of released activity on nanoparticle concentration and the biological environment. The results demonstrated the high stability of the prepared 225Ac@TiO2 NPs and the potential release of progeny over time. In vivo studies confirmed our hypothesis. The data obtained suggest that the daughter atoms can escape from the original carrier and follow their own biological pathways in the organism.
Collapse
Affiliation(s)
- Michal Sakmár
- Department of Nuclear Chemistry, Faculty of Nuclear Sciences and Physical Engineering, Czech Technical University in Prague, Břehová 7, 11519 Prague 1, Czech Republic; Institute of Nuclear Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, U Nemocnice 5, 12808 Prague, Czech Republic
| | - Ján Kozempel
- Department of Nuclear Chemistry, Faculty of Nuclear Sciences and Physical Engineering, Czech Technical University in Prague, Břehová 7, 11519 Prague 1, Czech Republic.
| | - Jan Kučka
- Czech Academy of Sciences, Institute of Macromolecular Chemistry, Heyrovského náměstí 1888-2, 16000 Prague 6, Czech Republic
| | - Tereza Janská
- Department of Nuclear Chemistry, Faculty of Nuclear Sciences and Physical Engineering, Czech Technical University in Prague, Břehová 7, 11519 Prague 1, Czech Republic
| | - Matěj Štíbr
- Department of Nuclear Chemistry, Faculty of Nuclear Sciences and Physical Engineering, Czech Technical University in Prague, Břehová 7, 11519 Prague 1, Czech Republic
| | - Lukáš Ondrák
- Department of Nuclear Chemistry, Faculty of Nuclear Sciences and Physical Engineering, Czech Technical University in Prague, Břehová 7, 11519 Prague 1, Czech Republic
| | - Kateřina Ondrák Fialová
- Department of Nuclear Chemistry, Faculty of Nuclear Sciences and Physical Engineering, Czech Technical University in Prague, Břehová 7, 11519 Prague 1, Czech Republic
| | - Martin Vlk
- Department of Nuclear Chemistry, Faculty of Nuclear Sciences and Physical Engineering, Czech Technical University in Prague, Břehová 7, 11519 Prague 1, Czech Republic
| | - Luděk Šefc
- Charles University in Prague, 1(st) Faculty of Medicine, Center for Advanced Preclinical Imaging (CAPI), Salmovská 3, 12000 Prague 2, Czech Republic
| | | | | |
Collapse
|
5
|
Ballisat L, De Sio C, Beck L, Chambers AL, Dillingham MS, Guatelli S, Sakata D, Shi Y, Duan J, Velthuis J, Rosenfeld A. Simulation of cell cycle effects on DNA strand break induction due to α-particles. Phys Med 2025; 129:104871. [PMID: 39667143 DOI: 10.1016/j.ejmp.2024.104871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/04/2024] [Accepted: 11/30/2024] [Indexed: 12/14/2024] Open
Abstract
PURPOSE Understanding cell cycle variations in radiosensitivity is important for α-particle therapies. Differences are due to both repair response mechanisms and the quantity of initial radiation-induced DNA strand breaks. Genome compaction within the nucleus has been shown to impact the yield of strand breaks. Compaction changes during the cell cycle are therefore likely to contribute to radiosensitivity differences. Simulation allows the strand break yield to be calculated independently of repair mechanisms which would be challenging experimentally. METHODS Using Geant4 the impact of genome compaction changes on strand break induction due to α-particles was simulated. Genome compaction is considered to be described by three metrics: global base pair density, chromatin fibre packing fraction and chromosome condensation. Nuclei in the G1, S, G2 and M phases from two cancer cell lines and one normal cell line are simulated. Repair mechanisms are not considered to study only the impact of genome compaction changes. RESULTS The three compaction metrics have differing effects on the strand break yield. For all cell lines the strand break yield is greatest in G2 cells and least in G1 cells. More strand breaks are induced in the two cancer cell lines than in the normal cell line. CONCLUSIONS Compaction of the genome affects the initial yield of strand breaks. Some radiosensitivity differences between cell lines can be attributed to genome compaction changes between the phases of the cell cycle. This study provides a basis for further analysis of how repair deficiencies impact radiation-induced lethality in normal and malignant cells.
Collapse
Affiliation(s)
| | - Chiara De Sio
- School of Physics, University of Bristol, Bristol, UK
| | - Lana Beck
- School of Physics, University of Bristol, Bristol, UK
| | - Anna L Chambers
- DNA-Protein Interactions Unit, School of Biochemistry, University of Bristol, Bristol, UK
| | - Mark S Dillingham
- DNA-Protein Interactions Unit, School of Biochemistry, University of Bristol, Bristol, UK
| | - Susanna Guatelli
- Centre for Medical Radiation Physics (CMRP), University of Wollongong, NSW, Australia
| | - Dousatsu Sakata
- School of Physics, University of Bristol, Bristol, UK; Centre for Medical Radiation Physics (CMRP), University of Wollongong, NSW, Australia; Division of Health Sciences, Osaka University, Osaka 565-0871, Japan
| | - Yuyao Shi
- School of Physics, University of Bristol, Bristol, UK
| | - Jinyan Duan
- School of Physics, University of Bristol, Bristol, UK
| | - Jaap Velthuis
- School of Physics, University of Bristol, Bristol, UK
| | - Anatoly Rosenfeld
- Centre for Medical Radiation Physics (CMRP), University of Wollongong, NSW, Australia
| |
Collapse
|
6
|
Tosato M, Favaretto C, Kleynhans J, Burgoyne AR, Gestin JF, van der Meulen NP, Jalilian A, Köster U, Asti M, Radchenko V. Alpha Atlas: Mapping global production of α-emitting radionuclides for targeted alpha therapy. Nucl Med Biol 2024; 142-143:108990. [PMID: 39809026 DOI: 10.1016/j.nucmedbio.2024.108990] [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: 11/09/2024] [Revised: 12/06/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025]
Abstract
Targeted Alpha Therapy has shown great promise in cancer treatment, sparking significant interest over recent decades. However, its broad adoption has been impeded by the scarcity of alpha-emitters and the complexities related to their use. The availability of these radionuclides is often constrained by the intricate production processes and purification, as well as regulatory and logistical challenges. Moreover, the high cost and technical difficulties associated with handling and applying alpha-emitting radionuclides pose additional barriers to their clinical implementation. This Alpha Atlas provides an in-depth overview of the leading alpha-particle emitting radionuclide candidates for clinical use, focusing on their production processes and supply chains. By mapping the current facilities that produce and supply these radionuclides, this atlas aims to assist researchers, clinicians, and industries in initiating or scaling up the applications of alpha-emitters. The Alpha Atlas aspires to act as a strategic guide, facilitating collaboration and driving forward the integration of these potent therapeutic agents into cancer treatment practices.
Collapse
Affiliation(s)
- Marianna Tosato
- Radiopharmaceutical Chemistry Laboratory (RACHEL), Nuclear Medicine Unit, AUSL-IRCCS Reggio Emilia, 42123 Reggio Emilia, Italy.
| | - Chiara Favaretto
- Radiopharmacy and Cyclotron Department, IRCCS Sacro Cuore Don Calabria, Negrar 37024, Verona, Italy
| | - Janke Kleynhans
- Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Andrew R Burgoyne
- Oak Ridge National Laboratory, 1 Bethel Valley Road, Oak Ridge, TN 37830, United States
| | - Jean-François Gestin
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 44000 Nantes, France
| | - Nicholas P van der Meulen
- PSI Center for Life Sciences, 5232 Villigen-PSI, Switzerland; PSI Center for Nuclear Engineering and Sciences, 5232 Villigen-PSI, Switzerland
| | - Amirreza Jalilian
- Department of Nuclear Safety and Security, International Atomic Energy Agency, 1220 Vienna, Austria
| | - Ulli Köster
- Institut Laue-Langevin, 38042 Grenoble, France
| | - Mattia Asti
- Radiopharmaceutical Chemistry Laboratory (RACHEL), Nuclear Medicine Unit, AUSL-IRCCS Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Valery Radchenko
- Life Sciences Division, TRIUMF, BC V6T 2A3 Vancouver, British Columbia, Canada; Department of Chemistry, University of British Columbia, V6T 1Z1 Vancouver, British Columbia, Canada
| |
Collapse
|
7
|
Chan HW, Kuo DY, Shueng PW, Chuang HY. Visualizing the Tumor Microenvironment: Molecular Imaging Probes Target Extracellular Matrix, Vascular Networks, and Immunosuppressive Cells. Pharmaceuticals (Basel) 2024; 17:1663. [PMID: 39770505 PMCID: PMC11676442 DOI: 10.3390/ph17121663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
The tumor microenvironment (TME) is a critical factor in cancer progression, driving tumor growth, immune evasion, therapeutic resistance, and metastasis. Understanding the dynamic interactions within the TME is essential for advancing cancer management. Molecular imaging provides a non-invasive, real-time, and longitudinal approach to studying the TME, with techniques such as positron emission tomography (PET), magnetic resonance imaging (MRI), and fluorescence imaging offering complementary strengths, including high sensitivity, spatial resolution, and intraoperative precision. Recent advances in imaging probe development have enhanced the ability to target and monitor specific components of the TME, facilitating early cancer diagnosis, therapeutic monitoring, and deeper insights into tumor biology. By integrating these innovations, molecular imaging offers transformative potential for precision oncology, improving diagnostic accuracy and treatment outcomes through a comprehensive assessment of TME dynamics.
Collapse
Affiliation(s)
- Hui-Wen Chan
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., Beitou Dist., Taipei City 112, Taiwan;
| | - Deng-Yu Kuo
- Division of Radiation Oncology, Department of Radiology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
| | - Pei-Wei Shueng
- Division of Radiation Oncology, Department of Radiology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei City 112, Taiwan
| | - Hui-Yen Chuang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., Beitou Dist., Taipei City 112, Taiwan;
| |
Collapse
|
8
|
Hou R, Liu N, Li F. Nanoradiopharmaceuticals: An Attractive Concept in Oncotherapy. ChemMedChem 2024; 19:e202400423. [PMID: 39140435 DOI: 10.1002/cmdc.202400423] [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: 06/03/2024] [Revised: 08/10/2024] [Accepted: 08/12/2024] [Indexed: 08/15/2024]
Abstract
Radiopharmaceuticals are of significant importance in the fields of tumor imaging and therapy. In recent decades, the increasing role of nanotechnology has led to the attractive concept of nanoradiopharmaceuticals. Consequently, it is imperative to provide a concise summary of the necessary guidelines to facilitate the translation of nanoradiopharmaceuticals. In this work, we have presented the contents of radiolabeling strategies and some applications of nanoradiopharmaceuticals. Such a framework can assist researchers in identifying more pertinent insights or making more informed decisions in the study of nanoradiopharmaceuticals.
Collapse
Affiliation(s)
- Ruitong Hou
- Key Laboratory of Radiation Physics and Technology of the Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu, 610064, PR China
| | - Ning Liu
- Key Laboratory of Radiation Physics and Technology of the Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu, 610064, PR China
| | - Feize Li
- Key Laboratory of Radiation Physics and Technology of the Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu, 610064, PR China
| |
Collapse
|
9
|
Higuchi T, Klimek K, Groener D, Chen X, Werner RA. Norepinephrine Transporter-Targeted Cancer Theranostics-New Horizons. Clin Nucl Med 2024:00003072-990000000-01400. [PMID: 39570057 DOI: 10.1097/rlu.0000000000005567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
ABSTRACT In the evolving landscape of precision oncology, this review delineates the role of radiopharmaceuticals targeting the norepinephrine transporter (NET), with a particular focus on the current clinical application of 123I-MIBG diagnostic imaging and 131I-MIBG therapeutics, in particular for pheochromocytoma, neuroblastoma, or paraganglioma. We will also highlight recently introduced 18F-labeled NET targeting imaging radiotracers, which would offer unparalleled resolution, enhanced tumor localization, and staging properties. Complementing these novel second-generation PET agents in a theranostic approach, astatine-211 meta-astatobenzylguanidine (211At-MABG) would leverage the advantages of alpha-particles to selectively target and eradicate NET-expressing tumor cells with minimal off-target effects.
Collapse
Affiliation(s)
| | - Konrad Klimek
- Clinic for Radiology and Nuclear Medicine, Department of Nuclear Medicine, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
| | - Daniel Groener
- Clinic for Radiology and Nuclear Medicine, Department of Nuclear Medicine, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
| | - Xinyu Chen
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | | |
Collapse
|
10
|
Tomiyoshi K, Wilson LJ, Mourtada F, Mourtada JS, Namiki Y, Kamata W, Yang DJ, Inoue T. Optimization Processes of Clinical Chelation-Based Radiopharmaceuticals for Pathway-Directed Targeted Radionuclide Therapy in Oncology. Pharmaceutics 2024; 16:1458. [PMID: 39598580 PMCID: PMC11597032 DOI: 10.3390/pharmaceutics16111458] [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: 10/23/2024] [Revised: 11/09/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
Targeted radionuclide therapy (TRT) for internal pathway-directed treatment is a game changer for precision medicine. TRT improves tumor control while minimizing damage to healthy tissue and extends the survival for patients with cancer. The application of theranostic-paired TRT along with cellular phenotype and genotype correlative analysis has the potential for malignant disease management. Chelation chemistry is essential for the development of theranostic-paired radiopharmaceuticals for TRT. Among image-guided TRT, 68Ga and 99mTc are the current standards for diagnostic radionuclides, while 177Lu and 225Ac have shown great promise for β- and α-TRT, respectively. Their long half-lives, potent radiobiology, favorable decay schemes, and ability to form stable chelation conjugates make them ideal for both manufacturing and clinical use. The current challenges include optimizing radionuclide production processes, coordinating chelation chemistry stability of theranostic-paired isotopes to reduce free daughters [this pertains to 225Ac daughters 221Fr and 213Bi]-induced tissue toxicity, and improving the modeling of micro dosimetry to refine dose-response evaluation. The empirical approach to TRT delivery is based on standard radionuclide administered activity levels, although clinical trials have revealed inconsistent outcomes and normal-tissue toxicities despite equivalent administered activities. This review presents the latest optimization methods for chelation-based theranostic radiopharmaceuticals, advancements in micro-dosimetry, and SPECT/CT technologies for quantifying whole-body uptake and monitoring therapeutic response as well as cytogenetic correlative analyses.
Collapse
Affiliation(s)
- Katsumi Tomiyoshi
- Shonan Research Institute of Innovative Medicine, Shonan Kamakura General Hospital, Kamakura 247-8533, Japan
| | - Lydia J. Wilson
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA; (L.J.W.); (F.M.)
| | - Firas Mourtada
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA; (L.J.W.); (F.M.)
| | | | - Yuta Namiki
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura 247-8533, Japan; (Y.N.); (W.K.); (D.J.Y.)
| | - Wataru Kamata
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura 247-8533, Japan; (Y.N.); (W.K.); (D.J.Y.)
| | - David J. Yang
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura 247-8533, Japan; (Y.N.); (W.K.); (D.J.Y.)
| | - Tomio Inoue
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura 247-8533, Japan; (Y.N.); (W.K.); (D.J.Y.)
| |
Collapse
|
11
|
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
|
12
|
Zhang H, Zhu W, Pan W, Wan X, Li N, Tang B. Recent advances in spatio-temporally controllable systems for management of glioma. Asian J Pharm Sci 2024; 19:100954. [PMID: 39483717 PMCID: PMC11525460 DOI: 10.1016/j.ajps.2024.100954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 05/29/2024] [Accepted: 06/17/2024] [Indexed: 11/03/2024] Open
Abstract
Malignant glioma remains one of the most aggressive intracranial tumors with devastating clinical outcomes despite the great advances in conventional treatment approaches, including surgery and chemotherapy. Spatio-temporally controllable approaches to glioma are now being actively investigated due to the preponderance, including spatio-temporal adjustability, minimally invasive, repetitive properties, etc. External stimuli can be readily controlled by adjusting the site and density of stimuli to exert the cytotoxic on glioma tissue and avoid undesired injury to normal tissues. It is worth noting that the removability of external stimuli allows for on-demand treatment, which effectively reduces the occurrence of side effects. In this review, we highlight recent advancements in drug delivery systems for spatio-temporally controllable treatments of glioma, focusing on the mechanisms and design principles of sensitizers utilized in these controllable therapies. Moreover, the potential challenges regarding spatio-temporally controllable therapy for glioma are also described, aiming to provide insights into future advancements in this field and their potential clinical applications.
Collapse
Affiliation(s)
- Huiwen Zhang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, China
| | - Wanqi Zhu
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, China
| | - Wei Pan
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, China
| | - Xiuyan Wan
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, China
| | - Na Li
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong, Institute of Molecular and Nano Science, Shandong Normal University, Jinan 250014, China
- Laoshan Laboratory, Qingdao 266237, China
| |
Collapse
|
13
|
Bender AA, Kirkeby EK, Cross DJ, Minoshima S, Roberts AG, Mastren TE. Development of a 213Bi-Labeled Pyridyl Benzofuran for Targeted α-Therapy of Amyloid-β Aggregates. J Nucl Med 2024; 65:1467-1472. [PMID: 39054283 DOI: 10.2967/jnumed.124.267482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Alzheimer disease is a neurodegenerative disorder with limited treatment options. It is characterized by the presence of several biomarkers, including amyloid-β aggregates, which lead to oxidative stress and neuronal decay. Targeted α-therapy (TAT) has been shown to be efficacious against metastatic cancer. TAT takes advantage of tumor-localized α-particle emission to break disease-associated covalent bonds while minimizing radiation dose to healthy tissues due to the short, micrometer-level, distances traveled. We hypothesized that TAT could be used to break covalent bonds within amyloid-β aggregates and facilitate natural plaque clearance mechanisms. Methods: We synthesized a 213Bi-chelate-linked benzofuran pyridyl derivative (BiBPy) and generated [213Bi]BiBPy, with a specific activity of 120.6 GBq/μg, dissociation constant of 11 ± 1.5 nM, and logP of 0.14 ± 0.03. Results: As the first step toward the validation of [213Bi]BiBPy as a TAT agent for the reduction of Alzheimer disease-associated amyloid-β, we showed that brain homogenates from APP/PS1 double-transgenic male mice (6-9 mo old) incubated with [213Bi]BiBPy exhibited a marked reduction in amyloid-β plaque concentration as measured using both enzyme-linked immunosorbent and Western blotting assays, with a half-maximal effective concentration of 3.72 kBq/pg. Conclusion: This [213Bi]BiBPy-concentration-dependent activity shows that TAT can reduce amyloid plaque concentration in vitro and supports the development of targeting systems for in vivo validations.
Collapse
Affiliation(s)
- Aidan A Bender
- Nuclear Engineering Program, University of Utah, Salt Lake City, Utah
| | - Emily K Kirkeby
- Department of Chemistry, University of Utah, Salt Lake City, Utah; and
| | - Donna J Cross
- Department of Radiology, University of Utah, School of Medicine, Salt Lake City, Utah
| | - Satoshi Minoshima
- Department of Radiology, University of Utah, School of Medicine, Salt Lake City, Utah
| | - Andrew G Roberts
- Department of Chemistry, University of Utah, Salt Lake City, Utah; and
| | - Tara E Mastren
- Nuclear Engineering Program, University of Utah, Salt Lake City, Utah;
| |
Collapse
|
14
|
Winter RC, Amghar M, Wacker AS, Bakos G, Taş H, Roscher M, Kelly JM, Benešová-Schäfer M. Future Treatment Strategies for Cancer Patients Combining Targeted Alpha Therapy with Pillars of Cancer Treatment: External Beam Radiation Therapy, Checkpoint Inhibition Immunotherapy, Cytostatic Chemotherapy, and Brachytherapy. Pharmaceuticals (Basel) 2024; 17:1031. [PMID: 39204136 PMCID: PMC11359268 DOI: 10.3390/ph17081031] [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: 06/12/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 09/03/2024] Open
Abstract
Cancer is one of the most complex and challenging human diseases, with rising incidences and cancer-related deaths despite improved diagnosis and personalized treatment options. Targeted alpha therapy (TαT) offers an exciting strategy emerging for cancer treatment which has proven effective even in patients with advanced metastatic disease that has become resistant to other treatments. Yet, in many cases, more sophisticated strategies are needed to stall disease progression and overcome resistance to TαT. The combination of two or more therapies which have historically been used as stand-alone treatments is an approach that has been pursued in recent years. This review aims to provide an overview on TαT and the four main pillars of therapeutic strategies in cancer management, namely external beam radiation therapy (EBRT), immunotherapy with checkpoint inhibitors (ICI), cytostatic chemotherapy (CCT), and brachytherapy (BT), and to discuss their potential use in combination with TαT. A brief description of each therapy is followed by a review of known biological aspects and state-of-the-art treatment practices. The emphasis, however, is given to the motivation for combination with TαT as well as the pre-clinical and clinical studies conducted to date.
Collapse
Affiliation(s)
- Ruth Christine Winter
- Research Group Molecular Biology of Systemic Radiotherapy/Translational Radiotheranostics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; (R.C.W.); (M.A.); (G.B.); (H.T.)
| | - Mariam Amghar
- Research Group Molecular Biology of Systemic Radiotherapy/Translational Radiotheranostics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; (R.C.W.); (M.A.); (G.B.); (H.T.)
| | - Anja S. Wacker
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; (A.S.W.); (J.M.K.)
| | - Gábor Bakos
- Research Group Molecular Biology of Systemic Radiotherapy/Translational Radiotheranostics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; (R.C.W.); (M.A.); (G.B.); (H.T.)
| | - Harun Taş
- Research Group Molecular Biology of Systemic Radiotherapy/Translational Radiotheranostics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; (R.C.W.); (M.A.); (G.B.); (H.T.)
| | - Mareike Roscher
- Service Unit for Radiopharmaceuticals and Preclinical Studies, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany;
| | - James M. Kelly
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; (A.S.W.); (J.M.K.)
| | - Martina Benešová-Schäfer
- Research Group Molecular Biology of Systemic Radiotherapy/Translational Radiotheranostics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; (R.C.W.); (M.A.); (G.B.); (H.T.)
| |
Collapse
|
15
|
Simms ME, Li Z, Sibley MM, Ivanov AS, Lara CM, Johnstone TC, Kertesz V, Fears A, White FD, Thorek DLJ, Thiele NA. PYTA: a universal chelator for advancing the theranostic palette of nuclear medicine. Chem Sci 2024; 15:11279-11286. [PMID: 39055008 PMCID: PMC11268510 DOI: 10.1039/d3sc06854d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 06/05/2024] [Indexed: 07/27/2024] Open
Abstract
To clinically advance the growing arsenal of radiometals available to image and treat cancer, chelators with versatile binding properties are needed. Herein, we evaluated the ability of the py2[18]dieneN6 macrocycle PYTA to interchangeably bind and stabilize 225Ac3+, [177Lu]Lu3+, [111In]In3+ and [44Sc]Sc3+, a chemically diverse set of radionuclides that can be used complementarily for targeted alpha therapy, beta therapy, single-photon emission computed tomography (SPECT) imaging, and positron emission tomography (PET) imaging, respectively. Through NMR spectroscopy and X-ray diffraction, we show that PYTA possesses an unusual degree of flexibility for a macrocyclic chelator, undergoing dramatic conformational changes that enable it to optimally satisfy the disparate coordination properties of each metal ion. Subsequent radiolabeling studies revealed that PYTA quantitatively binds all 4 radiometals at room temperature in just minutes at pH 6. Furthermore, these complexes were found to be stable in human serum over 2 half-lives. These results surpass those obtained for 2 state-of-the-art chelators for nuclear medicine, DOTA and macropa. The stability of 225Ac-PYTA and [44Sc]Sc-PYTA, the complexes having the most disparity with respect to metal-ion size, was further probed in mice. The resulting PET images (44Sc) and ex vivo biodistribution profiles (44Sc and 225Ac) of the PYTA complexes differed dramatically from those of unchelated [44Sc]Sc3+ and 225Ac3+. These differences provide evidence that PYTA retains this size-divergent pair of radionuclides in vivo. Collectively, these studies establish PYTA as a new workhorse chelator for nuclear medicine and warrant its further investigation in targeted constructs.
Collapse
Affiliation(s)
- Megan E Simms
- Chemical Sciences Division, Oak Ridge National Laboratory Oak Ridge TN 37831 USA
| | - Zhiyao Li
- Department of Radiology, Washington University in St. Louis School of Medicine St. Louis MO 63110 USA
- Program in Quantitative Molecular Therapeutics, Washington University in St. Louis School of Medicine St. Louis MO 63110 USA
| | - Megan M Sibley
- Chemical Sciences Division, Oak Ridge National Laboratory Oak Ridge TN 37831 USA
| | - Alexander S Ivanov
- Chemical Sciences Division, Oak Ridge National Laboratory Oak Ridge TN 37831 USA
| | - Caroline M Lara
- Department of Biological Sciences, University of Notre Dame Notre Dame IN 46556 USA
| | - Timothy C Johnstone
- Department of Chemistry and Biochemistry, University of California Santa Cruz Santa Cruz CA 95064 USA
| | - Vilmos Kertesz
- Biosciences Division, Oak Ridge National Laboratory Oak Ridge TN 37831 USA
| | - Amanda Fears
- Department of Radiology, Washington University in St. Louis School of Medicine St. Louis MO 63110 USA
- Program in Quantitative Molecular Therapeutics, Washington University in St. Louis School of Medicine St. Louis MO 63110 USA
| | - Frankie D White
- Radioisotope Science and Technology Division, Oak Ridge National Laboratory Oak Ridge TN 37831 USA
| | - Daniel L J Thorek
- Department of Radiology, Washington University in St. Louis School of Medicine St. Louis MO 63110 USA
- Program in Quantitative Molecular Therapeutics, Washington University in St. Louis School of Medicine St. Louis MO 63110 USA
- Department of Biomedical Engineering, Washington University in St. Louis St. Louis MO 63110 USA
- Oncologic Imaging Program, Siteman Cancer Center, Washington University in St. Louis School of Medicine St. Louis MO 63110 USA
| | - Nikki A Thiele
- Chemical Sciences Division, Oak Ridge National Laboratory Oak Ridge TN 37831 USA
| |
Collapse
|
16
|
Suzuki H, Kannaka K, Hirayama M, Yamashita T, Kaizuka Y, Kobayashi R, Yasuda T, Takahashi K, Uehara T. In vivo stable 211At-labeled prostate-specific membrane antigen-targeted tracer using a neopentyl glycol structure. EJNMMI Radiopharm Chem 2024; 9:48. [PMID: 38884866 PMCID: PMC11183015 DOI: 10.1186/s41181-024-00278-8] [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: 03/25/2024] [Accepted: 05/31/2024] [Indexed: 06/18/2024] Open
Abstract
BACKGROUND Prostate cancer is a common cancer among men worldwide that has a very poor prognosis, especially when it progresses to metastatic castration-resistant prostate cancer (mCRPC). Therefore, novel therapeutic agents for mCRPC are urgently required. Because prostate-specific membrane antigen (PSMA) is overexpressed in mCRPC, targeted alpha therapy (TAT) for PSMA is a promising treatment for mCRPC. Astatine-211 (211At) is a versatile α-emitting radionuclide that can be produced using a cyclotron. Therefore, 211At-labeled PSMA compounds could be useful for TAT; however, 211At-labeled compounds are unstable against deastatination in vivo. In this study, to develop in vivo stable 211At-labeled PSMA derivatives, we designed and synthesized 211At-labeled PSMA derivatives using a neopentyl glycol (NpG) structure that can stably retain 211At in vivo. We also evaluated their biodistribution in normal and tumor-bearing mice. RESULTS We designed and synthesized 211At-labeled PSMA derivatives containing two glutamic acid (Glu) linkers between the NpG structure and asymmetric urea (NpG-L-PSMA ((L-Glu)2 linker used) and NpG-D-PSMA ((D-Glu)2 linker used)). First, we evaluated the characteristics of 125I-labeled NpG derivatives because 125I was readily available. [125I]I-NpG-L-PSMA and [125I]I-NpG-D-PSMA showed low accumulation in the stomach and thyroid, indicating their high in vivo stability against deiodination. [125I]I-NpG-L-PSMA was excreted in urine as hydrophilic radiometabolites in addition to the intact form. Meanwhile, [125I]I-NpG-D-PSMA was excreted in urine in an intact form. In both cases, no radioactivity was observed in the free iodine fraction. [125I]I-NpG-D-PSMA showed higher tumor accumulation than [125I]I-NpG-L-PSMA. We then developed 211At-labeled PSMA using the NpG-D-PSMA structure. [211At]At-NpG-D-PSMA showed low accumulation in the stomach and thyroid in normal mice, indicating its high stability against deastatination in vivo. Moreover, [211At]At-NpG-D-PSMA showed high accumulation in tumor similar to that of [125I]I-NpG-D-PSMA. CONCLUSIONS [211At]At-NpG-D-PSMA showed high in vivo stability against deastatination and high tumor accumulation. [211At]At-NpG-D-PSMA should be considered as a potential new TAT for mCRPC.
Collapse
Affiliation(s)
- Hiroyuki Suzuki
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Chuo-Ku, Inohana, Chiba, 260-8675, Japan
| | - Kento Kannaka
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Chuo-Ku, Inohana, Chiba, 260-8675, Japan
| | - Mizuki Hirayama
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Chuo-Ku, Inohana, Chiba, 260-8675, Japan
| | - Tomoki Yamashita
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Chuo-Ku, Inohana, Chiba, 260-8675, Japan
| | - Yuta Kaizuka
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Chuo-Ku, Inohana, Chiba, 260-8675, Japan
| | - Ryota Kobayashi
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Chuo-Ku, Inohana, Chiba, 260-8675, Japan
| | - Takahiro Yasuda
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Chuo-Ku, Inohana, Chiba, 260-8675, Japan
| | - Kazuhiro Takahashi
- Advanced Clinical Research Center, Fukushima Medical University, 1 Hikariga-Oka, Fukushima, 960-12195, Japan
| | - Tomoya Uehara
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Chuo-Ku, Inohana, Chiba, 260-8675, Japan.
| |
Collapse
|
17
|
Zannoni EM, Sankar P, Jin Y, Liu C, Sinusas AJ, Metzler SD, Meng LJ. Design and development of the DE-SPECT system: a clinical SPECT system for broadband multi-isotope imaging of peripheral vascular disease. Phys Med Biol 2024; 69:125016. [PMID: 38815617 PMCID: PMC11167601 DOI: 10.1088/1361-6560/ad5266] [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: 10/11/2023] [Revised: 05/05/2024] [Accepted: 05/30/2024] [Indexed: 06/01/2024]
Abstract
Objective. Peripheral Vascular Disease (PVD) affects more than 230 million people worldwide and is one of the leading causes of disability among people over age 60. Nowadays, PVD remains largely underdiagnosed and undertreated, and requires the development of tailored diagnostic approaches. We present the full design of the Dynamic Extremity SPECT (DE-SPECT) system, the first organ-dedicated SPECT system for lower extremity imaging, based on 1 cm thick Cadmium Zinc Telluride (CZT) spectrometers and a dynamic dual field-of-view (FOV) synthetic compound-eye (SCE) collimator.Approach. The proposed DE-SPECT detection system consists of 48 1 cm thick 3D-position-sensitive CZT spectrometers arranged in a partial ring of 59 cm in diameter in a checkerboard pattern. The detection system is coupled with a compact dynamic SCE collimator that allows the user to select between two different FOVs at any time during an imaging study: a wide-FOV (28 cm diameter) configuration for dual-leg or scout imaging or a high-resolution and high-sensitivity (HR-HS) FOV (16 cm diameter) for single-leg or focused imaging.Main results.The preliminary experimental data show that the CZT spectrometer achieves a 3D intrinsic spatial resolution of <0.75 mm FWHM and an excellent energy resolution over a broad energy range (2.6 keV FWHM at 218, 3.3 keV at 440 keV). From simulations, the wide-FOV configuration offers a 0.034% averaged sensitivity at 140 keV and <8 mm spatial resolution, whereas the HR-HS configuration presents a peak central sensitivity of 0.07% at 140 keV and a ∼5 mm spatial resolution. The dynamic SCE collimator enables the capability to perform joint reconstructions that would ensure an overall improvement in imaging performance.Significance. The DE-SPECT system is a stationary and high-performance SPECT system that offers an excellent spectroscopic performance with a unique computer-controlled dual-FOV imaging capability, and a relatively high sensitivity for multi-tracer and multi-functional SPECT imaging of the extremities.
Collapse
Affiliation(s)
- E M Zannoni
- Department of Nuclear, Plasma, and Radiological Engineering, University of Illinois at Urbana Champaign, Urbana, IL, United States of America
| | - P Sankar
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Y Jin
- Department of Nuclear, Plasma, and Radiological Engineering, University of Illinois at Urbana Champaign, Urbana, IL, United States of America
| | - C Liu
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, United States of America
| | - A J Sinusas
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, United States of America
- Department of Medicine, Yale University School of Medicine, New Haven, CT, United States of America
| | - S D Metzler
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States of America
| | - L J Meng
- Department of Nuclear, Plasma, and Radiological Engineering, University of Illinois at Urbana Champaign, Urbana, IL, United States of America
- Beckman Institute for Advance Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, United States of America
| |
Collapse
|
18
|
Ree SM, Greenwood H, Young JD, Roberts R, Livens FR, Heath SL, Sosabowski JK. Selection of radionuclide(s) for targeted alpha therapy based on their nuclear decay properties. Nucl Med Commun 2024; 45:465-473. [PMID: 38465420 DOI: 10.1097/mnm.0000000000001832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Targeted alpha therapy (TAT) is a promising form of oncology treatment utilising alpha-emitting radionuclides that can specifically accumulate at disease sites. The high energy and high linear energy transfer associated with alpha emissions causes localised damage at target sites whilst minimising that to surrounding healthy tissue. The lack of appropriate radionuclides has inhibited research in TAT. The identification of appropriate radionuclides should be primarily a function of the radionuclide's nuclear decay properties, and not their biochemistry or economic factors since these last two factors can change; however, the nuclear decay properties are fixed to that nuclide. This study has defined and applied a criterion based on nuclear decay properties useful for TAT. This down-selection exercise concluded that the most appropriate radionuclides are: 149 Tb, 211 At/ 211 Po, 212 Pb/ 212 Bi/ 212 Po, 213 Bi/ 213 Po, 224 Ra, 225 Ra/ 225 Ac/ 221 Fr, 226 Ac/ 226 Th, 227 Th/ 223 Ra/ 219 Rn, 229 U, 230 U/ 226 Th, and 253 Fm, the majority of which have previously been considered for TAT. 229 U and 253 Fm have been newly identified and could become new radionuclides of interest for TAT, depending on their decay chain progeny.
Collapse
Affiliation(s)
- Samantha M Ree
- National Nuclear Laboratory, Springfields, Salwick, Preston, Lancashire
| | - Howard Greenwood
- National Nuclear Laboratory, Springfields, Salwick, Preston, Lancashire
| | - Jennifer D Young
- School of Biomedical Engineering and Imaging Sciences, King's College London, London
| | - Rachel Roberts
- National Nuclear Laboratory, Springfields, Salwick, Preston, Lancashire
| | | | - Scott L Heath
- Department of Earth and Environmental Sciences, The University of Manchester, Manchester and
| | - Jane K Sosabowski
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London
| |
Collapse
|
19
|
Lapi SE, Scott PJH, Scott AM, Windhorst AD, Zeglis BM, Abdel-Wahab M, Baum RP, Buatti JM, Giammarile F, Kiess AP, Jalilian A, Knoll P, Korde A, Kunikowska J, Lee ST, Paez D, Urbain JL, Zhang J, Lewis JS. Recent advances and impending challenges for the radiopharmaceutical sciences in oncology. Lancet Oncol 2024; 25:e236-e249. [PMID: 38821098 PMCID: PMC11340123 DOI: 10.1016/s1470-2045(24)00030-5] [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: 02/14/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 06/02/2024]
Abstract
This paper is the first of a Series on theranostics that summarises the current landscape of the radiopharmaceutical sciences as they pertain to oncology. In this Series paper, we describe exciting developments in radiochemistry and the production of radionuclides, the development and translation of theranostics, and the application of artificial intelligence to our field. These developments are catalysing growth in the use of radiopharmaceuticals to the benefit of patients worldwide. We also highlight some of the key issues to be addressed in the coming years to realise the full potential of radiopharmaceuticals to treat cancer.
Collapse
Affiliation(s)
- Suzanne E Lapi
- Departments of Radiology and Chemistry, O'Neal Comprehensive Cancer Center, University of Alabama, Birmingham, AL, USA
| | - Peter J H Scott
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Andrew M Scott
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, VIC, Australia; Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia; School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia; Department of Surgery, Faculty of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - Albert D Windhorst
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, Amsterdam, Netherlands; Cancer Center Amsterdam, Vrije Universiteit, Amsterdam, Netherlands
| | - Brian M Zeglis
- Department of Chemistry, Hunter College, City University of New York, New York City, NY, USA; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA; Department of Radiology, Weill Cornell Medical College, New York City, NY, USA
| | - May Abdel-Wahab
- Division of Human Health, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| | - Richard P Baum
- Deutsche Klinik für Diagnostik (DKD Helios Klinik) Wiesbaden, Curanosticum MVZ Wiesbaden-Frankfurt, Center for Advanced Radiomolecular Precision Oncology, Germany
| | - John M Buatti
- Department of Radiation Oncology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Francesco Giammarile
- Nuclear Medicine and Diagnostic Imaging Section, Division of Human Health, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria; Centre Leon Bérard, Lyon, France
| | - Ana P Kiess
- Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Amirreza Jalilian
- Radiochemistry and Radiotechnology Section, Division of Physical and Chemical Sciences, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| | - Peter Knoll
- Dosimetry and Medical Radiation Physics Section, Division of Human Health, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| | - Aruna Korde
- Radiochemistry and Radiotechnology Section, Division of Physical and Chemical Sciences, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| | - Jolanta Kunikowska
- Nuclear Medicine Department, Medical University of Warsaw, Warsaw, Poland
| | - Sze Ting Lee
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, VIC, Australia; Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia; School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia; Department of Surgery, Faculty of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - Diana Paez
- Nuclear Medicine and Diagnostic Imaging Section, Division of Human Health, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| | - Jean-Luc Urbain
- Department of Radiology-Nuclear Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Jingjing Zhang
- Department of Diagnostic Radiology, National University of Singapore, Singapore; Clinical Imaging Research Centre, Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA; Department of Radiology, Weill Cornell Medical College, New York City, NY, USA; Department of Pharmacology, Weill Cornell Medical College, New York City, NY, USA.
| |
Collapse
|
20
|
Kleynhans J, Ebenhan T, Cleeren F, Sathekge MM. Can current preclinical strategies for radiopharmaceutical development meet the needs of targeted alpha therapy? Eur J Nucl Med Mol Imaging 2024; 51:1965-1980. [PMID: 38676735 PMCID: PMC11139742 DOI: 10.1007/s00259-024-06719-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/15/2024] [Indexed: 04/29/2024]
Abstract
Preclinical studies are essential for effectively evaluating TAT radiopharmaceuticals. Given the current suboptimal supply chain of these radionuclides, animal studies must be refined to produce the most translatable TAT agents with the greatest clinical potential. Vector design is pivotal, emphasizing harmonious physical and biological characteristics among the vector, target, and radionuclide. The scarcity of alpha-emitting radionuclides remains a significant consideration. Actinium-225 and lead-212 appear as the most readily available radionuclides at this stage. Available animal models for researchers encompass xenografts, allografts, and PDX (patient-derived xenograft) models. Emerging strategies for imaging alpha-emitters are also briefly explored. Ultimately, preclinical research must address two critical aspects: (1) offering valuable insights into balancing safety and efficacy, and (2) providing guidance on the optimal dosing of the TAT agent.
Collapse
Affiliation(s)
- Janke Kleynhans
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, 3000, Belgium
| | - Thomas Ebenhan
- Department of Nuclear Medicine, University of Pretoria, and Steve Biko Academic Hospital, Pretoria, 0001, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, 0001, South Africa
| | - Frederik Cleeren
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, 3000, Belgium
| | - Mike Machaba Sathekge
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, 0001, South Africa.
- Preclinical Imaging Facility, Nuclear Medicine Research Infrastructure, Pretoria, 0001, South Africa.
| |
Collapse
|
21
|
Gao J, Li M, Yin J, Liu M, Wang H, Du J, Li J. The Different Strategies for the Radiolabeling of [ 211At]-Astatinated Radiopharmaceuticals. Pharmaceutics 2024; 16:738. [PMID: 38931860 PMCID: PMC11206656 DOI: 10.3390/pharmaceutics16060738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/10/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
Astatine-211 (211At) has emerged as a promising radionuclide for targeted alpha therapy of cancer by virtue of its favorable nuclear properties. However, the limited in vivo stability of 211At-labeled radiopharmaceuticals remains a major challenge. This review provides a comprehensive overview of the current strategies for 211At radiolabeling, including nucleophilic and electrophilic substitution reactions, as well as the recent advances in the development of novel bifunctional coupling agents and labeling approaches to enhance the stability of 211At-labeled compounds. The preclinical and clinical applications of 211At-labeled radiopharmaceuticals, including small molecules, peptides, and antibodies, are also discussed. Looking forward, the identification of new molecular targets, the optimization of 211At production and quality control methods, and the continued evaluation of 211At-labeled radiopharmaceuticals in preclinical and clinical settings will be the key to realizing the full potential of 211At-based targeted alpha therapy. With the growing interest and investment in this field, 211At-labeled radiopharmaceuticals are poised to play an increasingly important role in future cancer treatment.
Collapse
Affiliation(s)
- Jie Gao
- China Institute for Radiation Protection, National Atomic Energy Agency Nuclear Technology (Nonclinical Evaluation of Radiopharmaceuticals) Research and Development Center, CNNC Key Laboratory on Radiotoxicology and Radiopharmaceutical Preclinical Evaluation, Taiyuan 030006, China; (J.G.); (M.L.); (J.Y.); (M.L.)
- China Institute of Atomic Energy, Beijing 102413, China;
| | - Mei Li
- China Institute for Radiation Protection, National Atomic Energy Agency Nuclear Technology (Nonclinical Evaluation of Radiopharmaceuticals) Research and Development Center, CNNC Key Laboratory on Radiotoxicology and Radiopharmaceutical Preclinical Evaluation, Taiyuan 030006, China; (J.G.); (M.L.); (J.Y.); (M.L.)
| | - Jingjing Yin
- China Institute for Radiation Protection, National Atomic Energy Agency Nuclear Technology (Nonclinical Evaluation of Radiopharmaceuticals) Research and Development Center, CNNC Key Laboratory on Radiotoxicology and Radiopharmaceutical Preclinical Evaluation, Taiyuan 030006, China; (J.G.); (M.L.); (J.Y.); (M.L.)
| | - Mengya Liu
- China Institute for Radiation Protection, National Atomic Energy Agency Nuclear Technology (Nonclinical Evaluation of Radiopharmaceuticals) Research and Development Center, CNNC Key Laboratory on Radiotoxicology and Radiopharmaceutical Preclinical Evaluation, Taiyuan 030006, China; (J.G.); (M.L.); (J.Y.); (M.L.)
- China Institute of Atomic Energy, Beijing 102413, China;
| | - Hongliang Wang
- First Hospital of Shanxi Medical University, Taiyuan 030001, China;
| | - Jin Du
- China Institute of Atomic Energy, Beijing 102413, China;
- China Isotope & Radiation Corporation, Beijing 100089, China
| | - Jianguo Li
- China Institute for Radiation Protection, National Atomic Energy Agency Nuclear Technology (Nonclinical Evaluation of Radiopharmaceuticals) Research and Development Center, CNNC Key Laboratory on Radiotoxicology and Radiopharmaceutical Preclinical Evaluation, Taiyuan 030006, China; (J.G.); (M.L.); (J.Y.); (M.L.)
| |
Collapse
|
22
|
Tatsumi T, Zhao S, Kasahara A, Aoki M, Nishijima KI, Ukon N, Kodama T, Takahashi K, Sugiyama A, Washiyama K, Yamatsugu K, Kanai M. In vivo-stable bis-iminobiotin for targeted radionuclide delivery with the mutant streptavidin. Bioorg Med Chem Lett 2024; 108:129803. [PMID: 38777280 DOI: 10.1016/j.bmcl.2024.129803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/14/2024] [Accepted: 05/19/2024] [Indexed: 05/25/2024]
Abstract
Targeted delivery of radionuclides to tumors is significant in theranostics applications for precision medicine. Pre-targeting, in which a tumor-targeting vehicle and a radionuclide-loaded effector small molecule are administered separately, holds promise since it can reduce unnecessary internal radiation exposure of healthy cells and can minimize radiation decay. The success of the pre-targeting delivery requires an in vivo-stable tumor-targeting vehicle selectively binding to tumor antigens and an in vivo-stable small molecule effector selectively binding to the vehicle accumulated on the tumor. We previously reported a drug delivery system composed of a low-immunogenic streptavidin with weakened affinity to endogenous biotin and a bis-iminobiotin with high affinity to the engineered streptavidin. It was, however, unknown whether the bis-iminobiotin is stable in vivo when administered alone for the pre-targeting applications. Here we report a new in vivo-stable bis-iminobiotin derivative. The keys to success were the identification of the degradation site of the original bis-iminobiotin treated with mouse plasma and the structural modification of the degradation site. We disclosed the successful pre-targeting delivery of astatine-211 (211At), α-particle emitter, to the CEACAM5-positive tumor in xenograft mouse models.
Collapse
Affiliation(s)
- Toshifumi Tatsumi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Songji Zhao
- Advanced Clinical Research Center, Fukushima Medical University, 1 Hikariga-oka, Fukushima City, Fukushima 960-1295, Japan
| | - Akitomo Kasahara
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8675, Japan
| | - Miho Aoki
- Advanced Clinical Research Center, Fukushima Medical University, 1 Hikariga-oka, Fukushima City, Fukushima 960-1295, Japan
| | - Ken-Ichi Nishijima
- Advanced Clinical Research Center, Fukushima Medical University, 1 Hikariga-oka, Fukushima City, Fukushima 960-1295, Japan
| | - Naoyuki Ukon
- Advanced Clinical Research Center, Fukushima Medical University, 1 Hikariga-oka, Fukushima City, Fukushima 960-1295, Japan
| | - Tatsuhiko Kodama
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8904, Japan
| | - Kazuhiro Takahashi
- Advanced Clinical Research Center, Fukushima Medical University, 1 Hikariga-oka, Fukushima City, Fukushima 960-1295, Japan
| | - Akira Sugiyama
- Isotope Science Center, The University of Tokyo, 2-11-16 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan.
| | - Kohshin Washiyama
- Advanced Clinical Research Center, Fukushima Medical University, 1 Hikariga-oka, Fukushima City, Fukushima 960-1295, Japan.
| | - Kenzo Yamatsugu
- Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba 260-8675, Japan.
| | - Motomu Kanai
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan.
| |
Collapse
|
23
|
Binmujlli MA. Radiological and Molecular Analysis of Radioiodinated Anastrozole and Epirubicin as Innovative Radiopharmaceuticals Targeting Methylenetetrahydrofolate Dehydrogenase 2 in Solid Tumors. Pharmaceutics 2024; 16:616. [PMID: 38794278 PMCID: PMC11126143 DOI: 10.3390/pharmaceutics16050616] [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: 02/20/2024] [Revised: 04/13/2024] [Accepted: 04/18/2024] [Indexed: 05/26/2024] Open
Abstract
In the dynamic field of radiopharmaceuticals, innovating targeted agents for cancer diagnosis and therapy is crucial. Our study enriches this evolving landscape by evaluating the potential of radioiodinated anastrozole ([125I]anastrozole) and radioiodinated epirubicin ([125I]epirubicin) as targeting agents against MTHFD2-driven tumors. MTHFD2, which is pivotal in one-carbon metabolism, is notably upregulated in various cancers, presenting a novel target for radiopharmaceutical application. Through molecular docking and 200 ns molecular dynamics (MD) simulations, we assess the binding efficiency and stability of [125I]anastrozole and [125I]epirubicin with MTHFD2. Molecular docking illustrates that [125I]epirubicin has a superior binding free energy (∆Gbind) of -41.25 kJ/mol compared to -39.07 kJ/mol for [125I]anastrozole and -38.53 kJ/mol for the control ligand, suggesting that it has a higher affinity for MTHFD2. MD simulations reinforce this, showing stable binding, as evidenced by root mean square deviation (RMSD) values within a narrow range, underscoring the structural integrity of the enzyme-ligand complexes. The root mean square fluctuation (RMSF) analysis indicates consistent dynamic behavior of the MTHFD2 complex upon binding with [125I]anastrozole and [125I]epirubicin akin to the control. The radius of gyration (RG) measurements of 16.90 Å for MTHFD2-[125I]anastrozole and 16.84 Å for MTHFD2-[125I]epirubicin confirm minimal structural disruption upon binding. The hydrogen bond analysis reveals averages of two and three stable hydrogen bonds for [125I]anastrozole and [125I]epirubicin complexes, respectively, highlighting crucial stabilizing interactions. The MM-PBSA calculations further endorse the thermodynamic favorability of these interactions, with binding free energies of -48.49 ± 0.11 kJ/mol for [125I]anastrozole and -43.8 kJ/mol for MTHFD2-. The significant contribution of Van der Waals and electrostatic interactions to the binding affinities of [125I]anastrozole and [125I]epirubicin, respectively, underscores their potential efficacy for targeted tumor imaging and therapy. These computational findings lay the groundwork for the future experimental validation of [125I]anastrozole and [125I]epirubicin as MTHFD2 inhibitors, heralding a notable advancement in precision oncology tools. The data necessitate subsequent in vitro and in vivo assays to corroborate these results.
Collapse
Affiliation(s)
- Mazen Abdulrahman Binmujlli
- Department of Internal Medicine, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), P.O. Box 90950, Riyadh 11623, Saudi Arabia
| |
Collapse
|
24
|
Viscuse P, Devitt M, Dreicer R. Clinical Management of Advanced Prostate Cancer: Where Does Radiopharmaceutical Therapy Fit in the Treatment Algorithm? J Nucl Med 2024; 65:679-685. [PMID: 38604761 DOI: 10.2967/jnumed.123.267006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/25/2024] [Indexed: 04/13/2024] Open
Abstract
Most men with newly appreciated metastatic prostate cancer are optimally treated with a backbone consisting of androgen receptor-directed therapy with or without taxane chemotherapy. Despite improvements in disease outcomes, prostate cancer remains an extremely heterogeneous disease with variable mechanisms of therapeutic resistance. As a result, it remains a leading cause of cancer-related death in men. Radiopharmaceutical therapy has emerged as an alternative, non-androgen receptor-directed treatment modality for metastatic castration-resistant prostate cancer that impacts patient survival and represents a potentially more personalized approach. In this review, we aim to outline the current treatment landscape for metastatic prostate cancer with a focus on radiopharmaceutical therapy, specifically 177Lu-PSMA-617. In addition, we illustrate various clinical challenges with 177Lu-PSMA-617 treatment to date and explore investigative efforts to leverage radiopharmaceutical therapies as part of combination regimens or earlier in the treatment algorithm to further improve patient outcomes. Finally, we introduce ongoing studies of alternative radiopharmaceutical therapies in metastatic prostate cancer that may be incorporated into the treatment algorithm pending further study.
Collapse
Affiliation(s)
- Paul Viscuse
- Division of Hematology/Oncology, Department of Medicine, and Department of Urology, University of Virginia, Charlottesville, Virginia
| | - Michael Devitt
- Division of Hematology/Oncology, Department of Medicine, and Department of Urology, University of Virginia, Charlottesville, Virginia
| | - Robert Dreicer
- Division of Hematology/Oncology, Department of Medicine, and Department of Urology, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
25
|
Lin F, Clift R, Ehara T, Yanagida H, Horton S, Noncovich A, Guest M, Kim D, Salvador K, Richardson S, Miller T, Han G, Bhat A, Song K, Li G. Peptide Binder to Glypican-3 as a Theranostic Agent for Hepatocellular Carcinoma. J Nucl Med 2024; 65:586-592. [PMID: 38423788 DOI: 10.2967/jnumed.123.266766] [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: 10/16/2023] [Revised: 01/23/2024] [Indexed: 03/02/2024] Open
Abstract
Glypican-3 (GPC3) is a membrane-associated glycoprotein that is significantly upregulated in hepatocellular carcinomas (HCC) with minimal to no expression in normal tissues. The differential expression of GPC3 between tumor and normal tissues provides an opportunity for targeted radiopharmaceutical therapy to treat HCC, a leading cause of cancer-related deaths worldwide. Methods: DOTA-RYZ-GPC3 (RAYZ-8009) comprises a novel macrocyclic peptide binder to GPC3, a linker, and a chelator that can be complexed with different radioisotopes. The binding affinity was determined by surface plasma resonance and radioligand binding assays. Target-mediated cellular internalization was radiometrically measured at multiple time points. In vivo biodistribution, monotherapy, and combination treatments with 177Lu or 225Ac were performed on HCC xenografts. Results: RAYZ-8009 showed high binding affinity to GPC3 protein of human, mouse, canine, and cynomolgus monkey origins and no binding to other glypican family members. Potent cellular binding was confirmed in GPC3-positive HepG2 cells and was not affected by isotope switching. RAYZ-8009 achieved efficient internalization on binding to HepG2 cells. Biodistribution study of 177Lu-RAYZ-8009 showed sustained tumor uptake and fast renal clearance, with minimal or no uptake in other normal tissues. Tumor-specific uptake was also demonstrated in orthotopic HCC tumors, with no uptake in surrounding liver tissue. Therapeutically, significant and durable tumor regression and survival benefit were achieved with 177Lu- and 225Ac-labeled RAYZ-8009, as single agents and in combination with lenvatinib, in GPC3-positive HCC xenografts. Conclusion: Preclinical in vitro and in vivo data demonstrate the potential of RAYZ-8009 as a theranostic agent for the treatment of patients with GPC3-positive HCC.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Matt Guest
- RayzeBio, Inc., San Diego, California; and
| | - Daniel Kim
- RayzeBio, Inc., San Diego, California; and
| | | | | | | | | | | | | | - Gary Li
- RayzeBio, Inc., San Diego, California; and
| |
Collapse
|
26
|
Ramonaheng K, Qebetu M, Ndlovu H, Swanepoel C, Smith L, Mdanda S, Mdlophane A, Sathekge M. Activity quantification and dosimetry in radiopharmaceutical therapy with reference to 177Lutetium. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2024; 4:1355912. [PMID: 39355215 PMCID: PMC11440950 DOI: 10.3389/fnume.2024.1355912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 03/12/2024] [Indexed: 10/03/2024]
Abstract
Radiopharmaceutical therapy has been widely adopted owing primarily to the development of novel radiopharmaceuticals. To fully utilize the potential of these RPTs in the era of precision medicine, therapy must be optimized to the patient's tumor characteristics. The vastly disparate dosimetry methodologies need to be harmonized as the first step towards this. Multiple factors play a crucial role in the shift from empirical activity administration to patient-specific dosimetry-based administrations from RPT. Factors such as variable responses seen in patients with presumably similar clinical characteristics underscore the need to standardize and validate dosimetry calculations. These efforts combined with ongoing initiatives to streamline the dosimetry process facilitate the implementation of radiomolecular precision oncology. However, various challenges hinder the widespread adoption of personalized dosimetry-based activity administration, particularly when compared to the more convenient and resource-efficient approach of empiric activity administration. This review outlines the fundamental principles, procedures, and methodologies related to image activity quantification and dosimetry with a specific focus on 177Lutetium-based radiopharmaceuticals.
Collapse
Affiliation(s)
- Keamogetswe Ramonaheng
- Department of Medical Physics and Radiobiology, Nuclear Medicine Research, Infrastructure (NuMeRI) NPC, Pretoria, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
- Department of Nuclear Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Milani Qebetu
- Department of Medical Physics and Radiobiology, Nuclear Medicine Research, Infrastructure (NuMeRI) NPC, Pretoria, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
| | - Honest Ndlovu
- Department of Medical Physics and Radiobiology, Nuclear Medicine Research, Infrastructure (NuMeRI) NPC, Pretoria, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
- Department of Nuclear Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Cecile Swanepoel
- Department of Medical Physics and Radiobiology, Nuclear Medicine Research, Infrastructure (NuMeRI) NPC, Pretoria, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
| | - Liani Smith
- Department of Medical Physics and Radiobiology, Nuclear Medicine Research, Infrastructure (NuMeRI) NPC, Pretoria, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
| | - Sipho Mdanda
- Department of Medical Physics and Radiobiology, Nuclear Medicine Research, Infrastructure (NuMeRI) NPC, Pretoria, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
- Department of Nuclear Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Amanda Mdlophane
- Department of Medical Physics and Radiobiology, Nuclear Medicine Research, Infrastructure (NuMeRI) NPC, Pretoria, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
- Department of Nuclear Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Mike Sathekge
- Department of Medical Physics and Radiobiology, Nuclear Medicine Research, Infrastructure (NuMeRI) NPC, Pretoria, South Africa
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
- Department of Nuclear Medicine, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
27
|
Dyer MR, Jing Z, Duncan K, Godbe J, Shokeen M. Advancements in the development of radiopharmaceuticals for nuclear medicine applications in the treatment of bone metastases. Nucl Med Biol 2024; 130-131:108879. [PMID: 38340369 DOI: 10.1016/j.nucmedbio.2024.108879] [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: 10/20/2023] [Revised: 01/25/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024]
Abstract
Bone metastases are a painful and complex condition that overwhelmingly impacts the prognosis and quality of life of cancer patients. Over the years, nuclear medicine has made remarkable progress in the diagnosis and management of bone metastases. This review aims to provide a comprehensive overview of the recent advancements in nuclear medicine for the diagnosis and management of bone metastases. Furthermore, the review explores the role of targeted radiopharmaceuticals in nuclear medicine for bone metastases, focusing on radiolabeled molecules that are designed to selectively target biomarkers associated with bone metastases, including osteocytes, osteoblasts, and metastatic cells. The applications of radionuclide-based therapies, such as strontium-89 (Sr-89) and radium-223 (Ra-223), are also discussed. This review also highlights the potential of theranostic approaches for bone metastases, enabling personalized treatment strategies based on individual patient characteristics. Importantly, the clinical applications and outcomes of nuclear medicine in osseous metastatic disease are discussed. This includes the assessment of treatment response, predictive and prognostic value of imaging biomarkers, and the impact of nuclear medicine on patient management and outcomes. The review identifies current challenges and future perspectives on the role of nuclear medicine in treating bone metastases. It addresses limitations in imaging resolution, radiotracer availability, radiation safety, and the need for standardized protocols. The review concludes by emphasizing the need for further research and advancements in imaging technology, radiopharmaceutical development, and integration of nuclear medicine with other treatment modalities. In summary, advancements in nuclear medicine have significantly improved the diagnosis and management of osseous metastatic disease and future developements in the integration of innovative imaging modalities, targeted radiopharmaceuticals, radionuclide production, theranostic approaches, and advanced image analysis techniques hold great promise in improving patient outcomes and enhancing personalized care for individuals with bone metastases.
Collapse
Affiliation(s)
- Michael R Dyer
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Zhenghan Jing
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kathleen Duncan
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Jacqueline Godbe
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Monica Shokeen
- Edward Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA; Alvin J. Siteman Cancer Center, Washington University School of Medicine and Barnes-Jewish Hospital, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63110, USA; Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
28
|
Cao S, Kang Y, Tang H, Chen Z. Separation of lead-212 from natural thorium solution utilizing novel sulfonamide dibenzo-18-crown-6. Dalton Trans 2024; 53:3722-3730. [PMID: 38299333 DOI: 10.1039/d3dt04166b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2024]
Abstract
The extraction of lead-212 (212Pb) from radioactive thorium (Th) waste is immensely important, as it serves to mitigate environmental risks associated with radioactive waste and provides a vital source for medical isotopes. To economically extract 212Pb from thorium, we synthesized a novel extractant known as 2,13-disulfonyldiethylamine dibenzo-18-crown-6 (DSADB18C6). We assessed its performance in isolating Pb(II) by employing stable lead and optimizing the parameters of the extraction system. The results showcased an exceptional ability to extract Pb(II) efficiently. Within 10 minutes, using 20 mmol of DSADB18C6 and under conditions of 10 mg L-1 lead concentration (HNO3 = 0.5 mol L-1), the extraction efficiency reached up to 96.1%. Even after four rounds of stripping with 0.4 mol L-1 ammonium citrate, the efficiency remained at 90.2%. Moreover, the extraction stoichiometry and thermodynamics revealed that DSADB18C6 exhibited superior extraction performance compared to the commercial extractant 4',4'',(5'')-di-(tert-butyldicyclohexano)-18-crown-6 (DtBuDC18C6), in line with the density functional theory (DFT) calculation result. Furthermore, we successfully separated 212Pb from the thorium nitrate solution, maintaining radioactive equilibrium with its progeny. Gamma-spectroscopy confirmed a recovery yield of extraction exceeding 85.7%. This study presents a viable approach, underscoring the potential of DSADB18C6 as a promising extractant for the effective separation of 212Pb from radioactive thorium sources.
Collapse
Affiliation(s)
- Shiquan Cao
- School of Nuclear Science and Technology, University of Science and Technology of China, Hefei 230026, China.
| | - Yujia Kang
- School of Nuclear Science and Technology, University of Science and Technology of China, Hefei 230026, China.
| | - Huiping Tang
- School of Nuclear Science and Technology, University of Science and Technology of China, Hefei 230026, China.
| | - Zhi Chen
- School of Nuclear Science and Technology, University of Science and Technology of China, Hefei 230026, China.
| |
Collapse
|
29
|
Hooijman EL, Radchenko V, Ling SW, Konijnenberg M, Brabander T, Koolen SLW, de Blois E. Implementing Ac-225 labelled radiopharmaceuticals: practical considerations and (pre-)clinical perspectives. EJNMMI Radiopharm Chem 2024; 9:9. [PMID: 38319526 PMCID: PMC10847084 DOI: 10.1186/s41181-024-00239-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 01/25/2024] [Indexed: 02/07/2024] Open
Abstract
BACKGROUND In the past years, there has been a notable increase in interest regarding targeted alpha therapy using Ac-225, driven by the observed promising clinical anti-tumor effects. As the production and technology has advanced, the availability of Ac-225 is expected to increase in the near future, making the treatment available to patients worldwide. MAIN BODY Ac-225 can be labelled to different biological vectors, whereby the success of developing a radiopharmaceutical depends heavily on the labelling conditions, purity of the radionuclide source, chelator, and type of quenchers used to avoid radiolysis. Multiple (methodological) challenges need to be overcome when working with Ac-225; as alpha-emission detection is time consuming and highly geometry dependent, a gamma co-emission is used, but has to be in equilibrium with the mother-nuclide. Because of the high impact of alpha emitters in vivo it is highly recommended to cross-calibrate the Ac-225 measurements for used quality control (QC) techniques (radio-TLC, HPLC, HP-Ge detector, and gamma counter). More strict health physics regulations apply, as Ac-225 has a high toxicity, thereby limiting practical handling and quantities used for QC analysis. CONCLUSION This overview focuses specifically on the practical and methodological challenges when working with Ac-225 labelled radiopharmaceuticals, and underlines the required infrastructure and (detection) methods for the (pre-)clinical application.
Collapse
Affiliation(s)
- Eline L Hooijman
- Department of Radiology and Nuclear Medicine, Erasmus MC, 3015 CN, Rotterdam, The Netherlands
- Department of Hospital Pharmacy, Erasmus MC, 3015 CN, Rotterdam, The Netherlands
| | - Valery Radchenko
- Life Sciences Division, TRIUMF, Vancouver, BC, V6T 2A3, Canada
- Chemistry Department, University of British Columbia, Vancouver, BC, V6T 1Z1, Canada
| | - Sui Wai Ling
- Department of Radiology and Nuclear Medicine, Erasmus MC, 3015 CN, Rotterdam, The Netherlands
| | - Mark Konijnenberg
- Department of Radiology and Nuclear Medicine, Erasmus MC, 3015 CN, Rotterdam, The Netherlands
| | - Tessa Brabander
- Department of Radiology and Nuclear Medicine, Erasmus MC, 3015 CN, Rotterdam, The Netherlands
| | - Stijn L W Koolen
- Department of Radiology and Nuclear Medicine, Erasmus MC, 3015 CN, Rotterdam, The Netherlands
- Department of Hospital Pharmacy, Erasmus MC, 3015 CN, Rotterdam, The Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, 3015 CN, Rotterdam, The Netherlands
| | - Erik de Blois
- Department of Radiology and Nuclear Medicine, Erasmus MC, 3015 CN, Rotterdam, The Netherlands.
| |
Collapse
|
30
|
Trencsényi G, Csikos C, Képes Z. Targeted Radium Alpha Therapy in the Era of Nanomedicine: In Vivo Results. Int J Mol Sci 2024; 25:664. [PMID: 38203834 PMCID: PMC10779852 DOI: 10.3390/ijms25010664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/20/2023] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
Targeted alpha-particle therapy using radionuclides with alpha emission is a rapidly developing area in modern cancer treatment. To selectively deliver alpha-emitting isotopes to tumors, targeting vectors, including monoclonal antibodies, peptides, small molecule inhibitors, or other biomolecules, are attached to them, which ensures specific binding to tumor-related antigens and cell surface receptors. Although earlier studies have already demonstrated the anti-tumor potential of alpha-emitting radium (Ra) isotopes-Radium-223 and Radium-224 (223/224Ra)-in the treatment of skeletal metastases, their inability to complex with target-specific moieties hindered application beyond bone targeting. To exploit the therapeutic gains of Ra across a wider spectrum of cancers, nanoparticles have recently been embraced as carriers to ensure the linkage of 223/224Ra to target-affine vectors. Exemplified by prior findings, Ra was successfully bound to several nano/microparticles, including lanthanum phosphate, nanozeolites, barium sulfate, hydroxyapatite, calcium carbonate, gypsum, celestine, or liposomes. Despite the lengthened tumor retention and the related improvement in the radiotherapeutic effect of 223/224Ra coupled to nanoparticles, the in vivo assessment of the radiolabeled nanoprobes is a prerequisite prior to clinical usage. For this purpose, experimental xenotransplant models of different cancers provide a well-suited scenario. Herein, we summarize the latest achievements with 223/224Ra-doped nanoparticles and related advances in targeted alpha radiotherapy.
Collapse
Affiliation(s)
- György Trencsényi
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary; (G.T.); (C.C.)
| | - Csaba Csikos
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary; (G.T.); (C.C.)
- Gyula Petrányi Doctoral School of Clinical Immunology and Allergology, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - Zita Képes
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary; (G.T.); (C.C.)
| |
Collapse
|
31
|
Pijeira MSO, Nunes PSG, Chaviano SL, Diaz AMA, DaSilva JN, Ricci-Junior E, Alencar LMR, Chen X, Santos-Oliveira R. Medicinal (Radio) Chemistry: Building Radiopharmaceuticals for the Future. Curr Med Chem 2024; 31:5481-5534. [PMID: 37594105 DOI: 10.2174/0929867331666230818092634] [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: 03/10/2023] [Revised: 05/30/2023] [Accepted: 07/13/2023] [Indexed: 08/19/2023]
Abstract
Radiopharmaceuticals are increasingly playing a leading role in diagnosing, monitoring, and treating disease. In comparison with conventional pharmaceuticals, the development of radiopharmaceuticals does follow the principles of medicinal chemistry in the context of imaging-altered physiological processes. The design of a novel radiopharmaceutical has several steps similar to conventional drug discovery and some particularity. In the present work, we revisited the insights of medicinal chemistry in the current radiopharmaceutical development giving examples in oncology, neurology, and cardiology. In this regard, we overviewed the literature on radiopharmaceutical development to study overexpressed targets such as prostate-specific membrane antigen and fibroblast activation protein in cancer; β-amyloid plaques and tau protein in brain disorders; and angiotensin II type 1 receptor in cardiac disease. The work addresses concepts in the field of radiopharmacy with a special focus on the potential use of radiopharmaceuticals for nuclear imaging and theranostics.
Collapse
Affiliation(s)
- Martha Sahylí Ortega Pijeira
- Laboratory of Nanoradiopharmaceuticals and Synthesis of Novel Radiopharmaceuticals, Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Rio de Janeiro 21941906, Brazil
| | - Paulo Sérgio Gonçalves Nunes
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas SP13083-970, Brazil
| | - Samila Leon Chaviano
- Laboratoire de Biomatériaux pour l'Imagerie Médicale, Axe Médicine Régénératrice, Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, QC, Canada
| | - Aida M Abreu Diaz
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
- Institute de Génie Biomédical, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Jean N DaSilva
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
- Institute de Génie Biomédical, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Eduardo Ricci-Junior
- Laboratório de Desenvolvimento Galênico, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Luciana Magalhães Rebelo Alencar
- Laboratory of Biophysics and Nanosystems, Federal University of Maranhão, Av. dos Portugueses, 1966, Vila Bacanga, São Luís MA65080-805, Brazil
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore 117597, Singapore
| | - Ralph Santos-Oliveira
- Laboratory of Nanoradiopharmaceuticals and Synthesis of Novel Radiopharmaceuticals, Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Rio de Janeiro 21941906, Brazil
- Laboratory of Radiopharmacy and Nanoradiopharmaceuticals, Rio de Janeiro State University, Rio de Janeiro 23070200, Brazil
| |
Collapse
|
32
|
Echigo H, Mishiro K, Munekane M, Fuchigami T, Washiyama K, Takahashi K, Kitamura Y, Wakabayashi H, Kinuya S, Ogawa K. Development of probes for radiotheranostics with albumin binding moiety to increase the therapeutic effects of astatine-211 ( 211At). Eur J Nucl Med Mol Imaging 2024; 51:412-421. [PMID: 37819452 DOI: 10.1007/s00259-023-06457-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 09/23/2023] [Indexed: 10/13/2023]
Abstract
PURPOSE We have developed probes for multiradionuclides radiotheranostics using RGD peptide ([67Ga]Ga-DOTA-c[RGDf(4-I)K] ([67Ga]1) and Ga-DOTA-[211At]c[RGDf(4-At)K] ([211At]2)) for clinical applications. The introduction of an albumin binding moiety (ABM), such as 4-(4-iodophenyl)-butyric acid (IPBA), that has high affinity with the blood albumin and prolongs the circulation half-life can improve the pharmacokinetics of drugs. To perform more effective targeted alpha therapy (TAT), we designed and synthesized Ga-DOTA-K([211At]APBA)-c(RGDfK) ([211At]5) with 4-(4-astatophenyl)-butyric acid (APBA), which has an astato group instead of an iodo group in IPBA. We evaluated whether APBA functions as ABM and [211At]5 is effective for TAT. In addition, we prepared 67Ga-labeled RGD peptide without ABM, [67Ga]Ga-DOTA-K-c(RGDfK) ([67Ga]3), and 125I-labeled RGD peptide with ABM, Ga-DOTA-K([125I]IPBA)-c(RGDfK) ([125I]4), to compare with [211At]5. METHODS Biodistribution experiments of [67Ga]3 without ABM, [125I]4 and [211At]5 with ABM were conducted in normal mice and U-87 MG tumor-bearing mice. In addition, two doses of [211At]5 (370 or 925 kBq) were administered to U-87 MG tumor-bearing mice to confirm the therapeutic effects. RESULTS The blood retention of [125I]4 and [211At]5 was remarkably increased compared to [67Ga]3. Also, [125I]4 and [211At]5 showed similar biodistribution and significantly greater tumor accumulation and retention compared to [67Ga]3. In addition, [211At]5 inhibited tumor growth in a dose-dependent manner. CONCLUSION The functionality of APBA as ABM like IPBA, and the usefulness of [211At]5 as the radionuclide therapy agent for TAT was revealed.
Collapse
Affiliation(s)
- Hiroaki Echigo
- Graduate School of Medical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa, 920-1192, Japan
| | - Kenji Mishiro
- Institute for Frontier Science Initiative, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa, 920-1192, Japan
| | - Masayuki Munekane
- Graduate School of Medical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa, 920-1192, Japan
| | - Takeshi Fuchigami
- Graduate School of Medical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa, 920-1192, Japan
| | - Kohshin Washiyama
- Advanced Clinical Research Center, Fukushima Global Medical Science Center, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Kazuhiro Takahashi
- Advanced Clinical Research Center, Fukushima Global Medical Science Center, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Yoji Kitamura
- Research Center for Experimental Modeling of Human Disease, Kanazawa University, Takara-machi 13-1, Kanazawa, Ishikawa, 920-8640, Japan
| | - Hiroshi Wakabayashi
- Department of Nuclear Medicine, Kanazawa University Hospital, Kanazawa University, Takara-machi 13-1, Kanazawa, Ishikawa, 920-8641, Japan
| | - Seigo Kinuya
- Department of Nuclear Medicine, Kanazawa University Hospital, Kanazawa University, Takara-machi 13-1, Kanazawa, Ishikawa, 920-8641, Japan
| | - Kazuma Ogawa
- Graduate School of Medical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa, 920-1192, Japan.
- Institute for Frontier Science Initiative, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa, 920-1192, Japan.
| |
Collapse
|
33
|
Chen X, Liang R, Liu W, Ma H, Bai C, Xiong Y, Lan T, Liao J, Yang Y, Yang J, Li F, Liu N. Biocompatible conjugated polymer nanoparticles labeled with 225Ac for tumor endoradiotherapy. Bioorg Med Chem 2023; 96:117517. [PMID: 37939492 DOI: 10.1016/j.bmc.2023.117517] [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: 10/13/2023] [Revised: 11/01/2023] [Accepted: 11/03/2023] [Indexed: 11/10/2023]
Abstract
Recently, endoradiotherapy based on actinium-225 (225Ac) has attracted increasing attention, which is due to its α particles can generate maximal damage to cancer cells while minimizing unnecessary radiation effects on healthy tissues. Herein, 111In/225Ac-radiolabeled conjugated polymer nanoparticles (CPNs) coated with amphiphilic polymer DSPE-PEG-DOTA have been developed as a new injectable nano-radiopharmaceuticals for cancer endoradiotherapy under the guidance of nuclear imaging. Single photon emission computed tomography/computed tomography (SPECT/CT) using 111In-DOTA-PEG-CPNs as nano probe indicates a prolonged retention of radiolabeled nanocarriers, which was consistent with the in vivo biodistribution examined by direct radiometry analysis. Significant inhibition of tumor growth has been observed in murine 4T1 models treated with 225Ac-DOTA-PEG-CPNs when compared to mice treated with PBS or DOTA-PEG-CPNs. The 225Ac-DOTA-PEG-CPNs group experienced no single death within 24 days with the median survival considerably extended to 35 days, while all the mice treated with PBS or DOTA-PEG-CPNs died at 20 days post injection. Additionally, the histopathology studies demonstrated no obvious side effects on healthy tissues after treatment with 225Ac-DOTA-PEG-CPNs. All these results reveal that the new 225Ac-labeled DOTA-PEG-CPNs is promising as paradigm for endoradiotherapy.
Collapse
Affiliation(s)
- Xijian Chen
- Key Laboratory of Radiation Physics and Technology of the Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, PR China
| | - Ranxi Liang
- Key Laboratory of Radiation Physics and Technology of the Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, PR China
| | - Weihao Liu
- Key Laboratory of Radiation Physics and Technology of the Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, PR China
| | - Huan Ma
- Key Laboratory of Radiation Physics and Technology of the Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, PR China
| | - Chiyao Bai
- Chengdu New Radiomedicine Technology CO. LTD., Chengdu 610064, PR China
| | - Yao Xiong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, PR China
| | - Tu Lan
- Key Laboratory of Radiation Physics and Technology of the Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, PR China
| | - Jiali Liao
- Key Laboratory of Radiation Physics and Technology of the Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, PR China
| | - Yuanyou Yang
- Key Laboratory of Radiation Physics and Technology of the Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, PR China
| | - Jijun Yang
- Key Laboratory of Radiation Physics and Technology of the Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, PR China
| | - Feize Li
- Key Laboratory of Radiation Physics and Technology of the Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, PR China.
| | - Ning Liu
- Key Laboratory of Radiation Physics and Technology of the Ministry of Education, Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, PR China.
| |
Collapse
|
34
|
Jalloul W, Ghizdovat V, Stolniceanu CR, Ionescu T, Grierosu IC, Pavaleanu I, Moscalu M, Stefanescu C. Targeted Alpha Therapy: All We Need to Know about 225Ac's Physical Characteristics and Production as a Potential Theranostic Radionuclide. Pharmaceuticals (Basel) 2023; 16:1679. [PMID: 38139806 PMCID: PMC10747780 DOI: 10.3390/ph16121679] [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: 10/31/2023] [Revised: 11/24/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023] Open
Abstract
The high energy of α emitters, and the strong linear energy transfer that goes along with it, lead to very efficient cell killing through DNA damage. Moreover, the degree of oxygenation and the cell cycle state have no impact on these effects. Therefore, α radioisotopes can offer a treatment choice to individuals who are not responding to β- or gamma-radiation therapy or chemotherapy drugs. Only a few α-particle emitters are suitable for targeted alpha therapy (TAT) and clinical applications. The majority of available clinical research involves 225Ac and its daughter nuclide 213Bi. Additionally, the 225Ac disintegration cascade generates γ decays that can be used in single-photon emission computed tomography (SPECT) imaging, expanding the potential theranostic applications in nuclear medicine. Despite the growing interest in applying 225Ac, the restricted global accessibility of this radioisotope makes it difficult to conduct extensive clinical trials for many radiopharmaceutical candidates. To boost the availability of 225Ac, along with its clinical and potential theranostic applications, this review attempts to highlight the fundamental physical properties of this α-particle-emitting isotope, as well as its existing and possible production methods.
Collapse
Affiliation(s)
- Wael Jalloul
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- North East Regional Innovative Cluster for Structural and Molecular Imaging (Imago-Mol), 700115 Iasi, Romania
| | - Vlad Ghizdovat
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- North East Regional Innovative Cluster for Structural and Molecular Imaging (Imago-Mol), 700115 Iasi, Romania
| | - Cati Raluca Stolniceanu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- North East Regional Innovative Cluster for Structural and Molecular Imaging (Imago-Mol), 700115 Iasi, Romania
| | - Teodor Ionescu
- Department of Morpho-Functional Sciences (Pathophysiology), “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Irena Cristina Grierosu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Ioana Pavaleanu
- Department of Mother and Child, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Mihaela Moscalu
- Department of Preventive Medicine and Interdisciplinarity, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Cipriana Stefanescu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- North East Regional Innovative Cluster for Structural and Molecular Imaging (Imago-Mol), 700115 Iasi, Romania
| |
Collapse
|
35
|
Nelson BJB, Wilson J, Andersson JD, Wuest F. Theranostic Imaging Surrogates for Targeted Alpha Therapy: Progress in Production, Purification, and Applications. Pharmaceuticals (Basel) 2023; 16:1622. [PMID: 38004486 PMCID: PMC10674391 DOI: 10.3390/ph16111622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/08/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
This article highlights recent developments of SPECT and PET diagnostic imaging surrogates for targeted alpha particle therapy (TAT) radiopharmaceuticals. It outlines the rationale for using imaging surrogates to improve diagnostic-scan accuracy and facilitate research, and the properties an imaging-surrogate candidate should possess. It evaluates the strengths and limitations of each potential imaging surrogate. Thirteen surrogates for TAT are explored: 133La, 132La, 134Ce/134La, and 226Ac for 225Ac TAT; 203Pb for 212Pb TAT; 131Ba for 223Ra and 224Ra TAT; 123I, 124I, 131I and 209At for 211At TAT; 134Ce/134La for 227Th TAT; and 155Tb and 152Tb for 149Tb TAT.
Collapse
Affiliation(s)
- Bryce J. B. Nelson
- Department of Oncology, University of Alberta, 11560 University Ave., Edmonton, AB T6G 1Z2, Canada; (B.J.B.N.); (J.W.); (J.D.A.)
| | - John Wilson
- Department of Oncology, University of Alberta, 11560 University Ave., Edmonton, AB T6G 1Z2, Canada; (B.J.B.N.); (J.W.); (J.D.A.)
| | - Jan D. Andersson
- Department of Oncology, University of Alberta, 11560 University Ave., Edmonton, AB T6G 1Z2, Canada; (B.J.B.N.); (J.W.); (J.D.A.)
- Edmonton Radiopharmaceutical Center, Alberta Health Services, 11560 University Ave., Edmonton, AB T6G 1Z2, Canada
| | - Frank Wuest
- Department of Oncology, University of Alberta, 11560 University Ave., Edmonton, AB T6G 1Z2, Canada; (B.J.B.N.); (J.W.); (J.D.A.)
- Cancer Research Institute of Northern Alberta, University of Alberta, Edmonton, AB T6G 2E1, Canada
| |
Collapse
|
36
|
Franchi S, Asti M, Di Marco V, Tosato M. The Curies' element: state of the art and perspectives on the use of radium in nuclear medicine. EJNMMI Radiopharm Chem 2023; 8:38. [PMID: 37947909 PMCID: PMC10638329 DOI: 10.1186/s41181-023-00220-4] [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: 09/27/2023] [Accepted: 10/19/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND The alpha-emitter radium-223 (223Ra) is presently used in nuclear medicine for the palliative treatment of bone metastases from castration-resistant prostate cancer. This application arises from its advantageous decay properties and its intrinsic ability to accumulate in regions of high bone turnover when injected as a simple chloride salt. The commercial availability of [223Ra]RaCl2 as a registered drug (Xofigo®) is a further additional asset. MAIN BODY The prospect of extending the utility of 223Ra to targeted α-therapy of non-osseous cancers has garnered significant interest. Different methods, such as the use of bifunctional chelators and nanoparticles, have been explored to incorporate 223Ra in proper carriers designed to precisely target tumor sites. Nevertheless, the search for a suitable scaffold remains an ongoing challenge, impeding the diffusion of 223Ra-based radiopharmaceuticals. CONCLUSION This review offers a comprehensive overview of the current role of radium radioisotopes in nuclear medicine, with a specific focus on 223Ra. It also critically examines the endeavors conducted so far to develop constructs capable of incorporating 223Ra into cancer-targeting drugs. Particular emphasis is given to the chemical aspects aimed at providing molecular scaffolds for the bifunctional chelator approach.
Collapse
Affiliation(s)
- Sara Franchi
- Department of Chemical Sciences, University of Padova, Via Marzolo 1, 35131, Padua, Italy
| | - Mattia Asti
- Radiopharmaceutical Chemistry Section, Nuclear Medicine Unit, AUSL di Reggio Emilia: Azienda Unità Sanitaria Locale - IRCCS Tecnologie Avanzate e Modelli Assistenziali in Oncologia di Reggio Emilia, Via Amendola 2, 42122, Reggio Emilia, Italy
| | - Valerio Di Marco
- Department of Chemical Sciences, University of Padova, Via Marzolo 1, 35131, Padua, Italy
| | - Marianna Tosato
- Radiopharmaceutical Chemistry Section, Nuclear Medicine Unit, AUSL di Reggio Emilia: Azienda Unità Sanitaria Locale - IRCCS Tecnologie Avanzate e Modelli Assistenziali in Oncologia di Reggio Emilia, Via Amendola 2, 42122, Reggio Emilia, Italy.
| |
Collapse
|
37
|
McIntosh LA, Burns JD, Tereshatov EE, Muzzioli R, Hagel K, Jinadu NA, McCann LA, Picayo GA, Pisaneschi F, Piwnica-Worms D, Schultz SJ, Tabacaru GC, Abbott A, Green B, Hankins T, Hannaman A, Harvey B, Lofton K, Rider R, Sorensen M, Tabacaru A, Tobin Z, Yennello SJ. Production, isolation, and shipment of clinically relevant quantities of astatine-211: A simple and efficient approach to increasing supply. Nucl Med Biol 2023; 126-127:108387. [PMID: 37837782 DOI: 10.1016/j.nucmedbio.2023.108387] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/04/2023] [Accepted: 09/18/2023] [Indexed: 10/16/2023]
Abstract
The alpha emitter astatine-211 (211At) is a promising candidate for cancer treatment based on Targeted Alpha (α) Therapy (TAT). A small number of facilities, distributed across the United States, are capable of accelerating α-particle beams to produce 211At. However, challenges remain regarding strategic methods for shipping 211At in a form adaptable to advanced radiochemistry reactions and other uses of the radioisotope. PURPOSE Our method allows shipment of 211At in various quantities in a form convenient for further radiochemistry. PROCEDURES For this study, a 3-octanone impregnated Amberchrom CG300M resin bed in a column cartridge was used to separate 211At from the bismuth matrix on site at the production accelerator (Texas A&M) in preparation for shipping. Aliquots of 6 M HNO3 containing up to ≈2.22 GBq of 211At from the dissolved target were successfully loaded and retained on columns. Exempt packages (<370 MBq) were shipped to a destination radiochemistry facility, University of Texas MD Anderson Cancer Center, in the form of a convenient air-dried column. Type A packages have been shipped overnight to University of Alabama at Birmingham. MAIN FINDINGS Air-dried column hold times of various lengths did not inhibit simple and efficient recovery of 211At. Solution eluted from the column was sufficiently high in specific activity to successfully radiolabel a model compound, 4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)aniline (1), with 211At. The method to prepare and ship 211At described in this manuscript has also been used to ship larger quantities of 211At a greater distance to University of Alabama at Birmingham. PRINCIPAL CONCLUSIONS The successful proof of this method paves the way for the distribution of 211At from Texas A&M University to research institutions and clinical oncology centers in Texas and elsewhere. Use of this simple method at other facilities has the potential increase the overall availability of 211At for preclinical and clinical studies.
Collapse
Affiliation(s)
- Lauren A McIntosh
- Cyclotron Institute, Texas A&M University, College Station, TX 77843, USA.
| | - Jonathan D Burns
- Chemistry Department, The University of Alabama at Birmingham, Birmingham, AL 35924, USA.
| | | | - Riccardo Muzzioli
- Department of Cancer System Imaging, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Kris Hagel
- Cyclotron Institute, Texas A&M University, College Station, TX 77843, USA
| | - Noimat A Jinadu
- Chemistry Department, The University of Alabama at Birmingham, Birmingham, AL 35924, USA
| | - Laura A McCann
- Cyclotron Institute, Texas A&M University, College Station, TX 77843, USA; Chemistry Department, Texas A&M University, College Station, TX 77843, USA
| | - Gabriela A Picayo
- Cyclotron Institute, Texas A&M University, College Station, TX 77843, USA; Chemistry Department, Texas A&M University, College Station, TX 77843, USA
| | - Federica Pisaneschi
- Department of Cancer System Imaging, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Center for Translational Cancer Research, The Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases (IMM) at The University of Texas Health Science Center at Houston, USA
| | - David Piwnica-Worms
- Department of Cancer System Imaging, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Steven J Schultz
- Cyclotron Institute, Texas A&M University, College Station, TX 77843, USA; Chemistry Department, Texas A&M University, College Station, TX 77843, USA
| | - Gabriel C Tabacaru
- Cyclotron Institute, Texas A&M University, College Station, TX 77843, USA
| | - Austin Abbott
- Cyclotron Institute, Texas A&M University, College Station, TX 77843, USA; Chemistry Department, Texas A&M University, College Station, TX 77843, USA
| | - Brooklyn Green
- Cyclotron Institute, Texas A&M University, College Station, TX 77843, USA; Chemistry Department, Texas A&M University, College Station, TX 77843, USA
| | - Travis Hankins
- Cyclotron Institute, Texas A&M University, College Station, TX 77843, USA; Chemistry Department, Texas A&M University, College Station, TX 77843, USA
| | - Andrew Hannaman
- Cyclotron Institute, Texas A&M University, College Station, TX 77843, USA; Chemistry Department, Texas A&M University, College Station, TX 77843, USA
| | - Bryan Harvey
- Cyclotron Institute, Texas A&M University, College Station, TX 77843, USA; Physics Department, Texas A&M University, College Station, TX 77843, USA
| | - Kylie Lofton
- Cyclotron Institute, Texas A&M University, College Station, TX 77843, USA; Chemistry Department, Texas A&M University, College Station, TX 77843, USA
| | - Robert Rider
- Cyclotron Institute, Texas A&M University, College Station, TX 77843, USA; Chemistry Department, Texas A&M University, College Station, TX 77843, USA
| | - Maxwell Sorensen
- Cyclotron Institute, Texas A&M University, College Station, TX 77843, USA; Chemistry Department, Texas A&M University, College Station, TX 77843, USA
| | - Alexandra Tabacaru
- Cyclotron Institute, Texas A&M University, College Station, TX 77843, USA
| | - Zachary Tobin
- Cyclotron Institute, Texas A&M University, College Station, TX 77843, USA; Chemistry Department, Texas A&M University, College Station, TX 77843, USA
| | - Sherry J Yennello
- Cyclotron Institute, Texas A&M University, College Station, TX 77843, USA; Chemistry Department, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
38
|
Mourtada F, Tomiyoshi K, Sims-Mourtada J, Mukai-Sasaki Y, Yagihashi T, Namiki Y, Murai T, Yang DJ, Inoue T. Actinium-225 Targeted Agents: Where Are We Now? Brachytherapy 2023; 22:697-708. [PMID: 37690972 PMCID: PMC10840862 DOI: 10.1016/j.brachy.2023.06.228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 06/26/2023] [Indexed: 09/12/2023]
Abstract
α-particle targeted radionuclide therapy has shown promise for optimal cancer management, an exciting new era for brachytherapy. Alpha-emitting nuclides can have significant advantages over gamma- and beta-emitters due to their high linear energy transfer (LET). While their limited path length results in more specific tumor 0kill with less damage to surrounding normal tissues, their high LET can produce substantially more lethal double strand DNA breaks per radiation track than beta particles. Over the last decade, the physical and chemical attributes of Actinium-225 (225Ac) including its half-life, decay schemes, path length, and straightforward chelation ability has peaked interest for brachytherapy agent development. However, this has been met with challenges including source availability, accurate modeling for standardized dosimetry for brachytherapy treatment planning, and laboratory space allocation in the hospital setting for on-demand radiopharmaceuticals production. Current evidence suggests that a simple empirical approach based on 225Ac administered radioactivity may lead to inconsistent outcomes and toxicity. In this review article, we highlight the recent advances in 225Ac source production, dosimetry modeling, and current clinical studies.
Collapse
Affiliation(s)
- Firas Mourtada
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA.
| | - Katsumi Tomiyoshi
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura, Japan
| | | | - Yuki Mukai-Sasaki
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura, Japan; Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Takayuki Yagihashi
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura, Japan
| | - Yuta Namiki
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura, Japan
| | - Taro Murai
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura, Japan
| | - David J Yang
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura, Japan
| | - Tomio Inoue
- Advanced Medical Center, Shonan Kamakura General Hospital, Kamakura, Japan
| |
Collapse
|
39
|
Hassan M, Bokhari TH, Lodhi NA, Khosa MK, Usman M. A review of recent advancements in Actinium-225 labeled compounds and biomolecules for therapeutic purposes. Chem Biol Drug Des 2023; 102:1276-1292. [PMID: 37715360 DOI: 10.1111/cbdd.14311] [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: 03/16/2022] [Revised: 06/03/2023] [Accepted: 07/17/2023] [Indexed: 09/17/2023]
Abstract
In nuclear medicine, cancers that cannot be cured or can only be treated partially by traditional techniques like surgery or chemotherapy are killed by ionizing radiation as a form of therapeutic treatment. Actinium-225 is an alpha-emitting radionuclide that is highly encouraging as a therapeutic approach and more promising for targeted alpha therapy (TAT). Actinium-225 is the best candidate for tumor cells treatment and has physical characteristics such as high (LET) linear energy transfer (150 keV per μm), half-life (t1/2 = 9.92d), and short ranges (400-100 μm) which prevent the damage of normal healthy tissues. The introduction of various new radiopharmaceuticals and radioisotopes has significantly assisted the advancement of nuclear medicine. Ac-225 radiopharmaceuticals continuously demonstrate their potential as targeted alpha therapeutics. 225 Ac-labeled radiopharmaceuticals have confirmed their importance in medical and clinical areas by introducing [225 Ac]Ac-PSMA-617, [225 Ac]Ac-DOTATOC, [225 Ac]Ac-DOTA-substance-P, reported significantly improved response in patients with prostate cancer, neuroendocrine, and glioma, respectively. The development of these radiopharmaceuticals required a suitable buffer, incubation time, optimal pH, and reaction temperature. There is a growing need to standardize quality control (QC) testing techniques such as radiochemical purity (RCP). This review aims to summarize the development of the Ac-225 labeled compounds and biomolecules. The current state of their reported resulting clinical applications is also summarized as well.
Collapse
Affiliation(s)
- Maria Hassan
- Department of Chemistry, Government College University, Faisalabad, Pakistan
| | | | - Nadeem Ahmed Lodhi
- Isotope Production Division, Pakistan institute of Nuclear Science & Technology (PINSTECH), Islamabad, Pakistan
| | | | - Muhammad Usman
- Department of Chemistry, Government College University, Faisalabad, Pakistan
| |
Collapse
|
40
|
Sharma S, Pandey MK. Radiometals in Imaging and Therapy: Highlighting Two Decades of Research. Pharmaceuticals (Basel) 2023; 16:1460. [PMID: 37895931 PMCID: PMC10610335 DOI: 10.3390/ph16101460] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/03/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
The present article highlights the important progress made in the last two decades in the fields of molecular imaging and radionuclide therapy. Advancements in radiometal-based positron emission tomography, single photon emission computerized tomography, and radionuclide therapy are illustrated in terms of their production routes and ease of radiolabeling. Applications in clinical diagnostic and radionuclide therapy are considered, including human studies under clinical trials; their current stages of clinical translations and findings are summarized. Because the metalloid astatine is used for imaging and radionuclide therapy, it is included in this review. In regard to radionuclide therapy, both beta-minus (β-) and alpha (α)-emitting radionuclides are discussed by highlighting their production routes, targeted radiopharmaceuticals, and current clinical translation stage.
Collapse
Affiliation(s)
| | - Mukesh K. Pandey
- Division of Nuclear Medicine, Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA;
| |
Collapse
|
41
|
Jang A, Kendi AT, Johnson GB, Halfdanarson TR, Sartor O. Targeted Alpha-Particle Therapy: A Review of Current Trials. Int J Mol Sci 2023; 24:11626. [PMID: 37511386 PMCID: PMC10380274 DOI: 10.3390/ijms241411626] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/08/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Radiopharmaceuticals are rapidly developing as a field, with the successful use of targeted beta emitters in neuroendocrine tumors and prostate cancer serving as catalysts. Targeted alpha emitters are in current development for several potential oncologic indications. Herein, we review the three most prevalently studied conjugated/chelated alpha emitters (225actinium, 212lead, and 211astatine) and focus on contemporary clinical trials in an effort to more fully appreciate the breadth of the current evaluation. Phase I trials targeting multiple diseases are now underway, and at least one phase III trial (in selected neuroendocrine cancers) is currently in the initial stages of recruitment. Combination trials are now also emerging as alpha emitters are integrated with other therapies in an effort to create solutions for those with advanced cancers. Despite the promise of targeted alpha therapies, many challenges remain. These challenges include the development of reliable supply chains, the need for a better understanding of the relationships between administered dose and absorbed dose in both tissue and tumor and how that predicts outcomes, and the incomplete understanding of potential long-term deleterious effects of the alpha emitters. Progress on multiple fronts is necessary to bring the potential of targeted alpha therapies into the clinic.
Collapse
Affiliation(s)
- Albert Jang
- Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Ayse T Kendi
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Geoffrey B Johnson
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Oliver Sartor
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Urology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
42
|
Hurley K, Cao M, Huang H, Wang Y. Targeted Alpha Therapy (TAT) with Single-Domain Antibodies (Nanobodies). Cancers (Basel) 2023; 15:3493. [PMID: 37444603 DOI: 10.3390/cancers15133493] [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: 05/30/2023] [Revised: 06/23/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
The persistent threat of cancer necessitates the development of improved and more efficient therapeutic strategies that limit damage to healthy tissues. Targeted alpha therapy (TαT), a novel form of radioimmuno-therapy (RIT), utilizes a targeting vehicle, commonly antibodies, to deliver high-energy, but short-range, alpha-emitting particles specifically to cancer cells, thereby reducing toxicity to surrounding normal tissues. Although full-length antibodies are often employed as targeting vehicles for TαT, their high molecular weight and the presence of an Fc-region lead to a long blood half-life, increased bone marrow toxicity, and accumulation in other tissues such as the kidney, liver, and spleen. The discovery of single-domain antibodies (sdAbs), or nanobodies, naturally occurring in camelids and sharks, has introduced a novel antigen-specific vehicle for molecular imaging and TαT. Given that nanobodies are the smallest naturally occurring antigen-binding fragments, they exhibit shorter relative blood half-lives, enhanced tumor uptake, and equivalent or superior binding affinity and specificity. Nanobody technology could provide a viable solution for the off-target toxicity observed with full-length antibody-based TαT. Notably, the pharmacokinetic properties of nanobodies align better with the decay characteristics of many short-lived α-emitting radionuclides. This review aims to encapsulate recent advancements in the use of nanobodies as a vehicle for TαT.
Collapse
Affiliation(s)
- Kate Hurley
- Radiobiology and Health, Canadian Nuclear Laboratories, Chalk River, ON K0J 1J0, Canada
| | - Meiyun Cao
- Radiobiology and Health, Canadian Nuclear Laboratories, Chalk River, ON K0J 1J0, Canada
| | - Haiming Huang
- Research Center, Forlong Biotechnology Inc., Suzhou 215004, China
| | - Yi Wang
- Radiobiology and Health, Canadian Nuclear Laboratories, Chalk River, ON K0J 1J0, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
43
|
Zhang Y, Li F, Cui Z, Li K, Guan J, Tian L, Wang Y, Liu N, Wu W, Chai Z, Wang S. A Radioluminescent Metal-Organic Framework for Monitoring 225Ac in Vivo. J Am Chem Soc 2023. [PMID: 37366004 DOI: 10.1021/jacs.3c02325] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
225Ac is considered as one of the most promising radioisotopes for alpha-therapy because its emitted high-energy α-particles can efficiently damage tumor cells. However, it also represents a significant threat to healthy tissues owing to extremely high radiotoxicity if targeted therapy fails. This calls for a pressing requirement of monitoring the biodistribution of 225Ac in vivo during the treatment of tumors. However, the lack of imageable photons or positrons from therapeutic doses of 225Ac makes this task currently quite challenging. We report here a nanoscale luminescent europium-organic framework (EuMOF) that allows for fast, simple, and efficient labeling of 225Ac in its crystal structure with sufficient 225Ac-retention stability based on similar coordination behaviors between Ac3+ and Eu3+. After labeling, the short distance between 225Ac and Eu3+ in the structure leads to exceedingly efficient energy transduction from225Ac-emitted α-particles to surrounding Eu3+ ions, which emits red luminescence through a scintillation process and produces sufficient photons for clearcut imaging. The in vivo intensity distribution of radioluminescence signal originating from the 225Ac-labeled EuMOF is consistent with the dose of 225Ac dispersed among the various organs determined by the radioanalytical measurement ex vivo, certifying the feasibility of in vivo directly monitoring 225Ac using optical imaging for the first time. In addition, 225Ac-labeled EuMOF displays notable efficiency in treating the tumor. These results provide a general design principle for fabricating 225Ac-labeled radiopharmaceuticals with imaging photons and propose a simple way to in vivo track radionuclides with no imaging photons, including but not limited to 225Ac.
Collapse
Affiliation(s)
- Yugang Zhang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Feize Li
- Key Laboratory of Radiation Physics and Technology of the Ministry of Education Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, China
| | - Zhencun Cui
- Frontiers Science Center for Rare Isotopes, Lanzhou University, Lanzhou 730000, China
| | - Kai Li
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Jingwen Guan
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Longlong Tian
- Frontiers Science Center for Rare Isotopes, Lanzhou University, Lanzhou 730000, China
| | - Yaxing Wang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Ning Liu
- Key Laboratory of Radiation Physics and Technology of the Ministry of Education Institute of Nuclear Science and Technology, Sichuan University, Chengdu 610064, China
| | - Wangsuo Wu
- Frontiers Science Center for Rare Isotopes, Lanzhou University, Lanzhou 730000, China
| | - Zhifang Chai
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| | - Shuao Wang
- State Key Laboratory of Radiation Medicine and Protection, School for Radiological and Interdisciplinary Sciences (RAD-X) and Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, China
| |
Collapse
|
44
|
Iizuka Y, Manabe Y, Ooe K, Toyoshima A, Yin X, Haba H, Kabayama K, Fukase K. Exploring a Nuclear-Selective Radioisotope Delivery System for Efficient Targeted Alpha Therapy. Int J Mol Sci 2023; 24:ijms24119593. [PMID: 37298546 DOI: 10.3390/ijms24119593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/15/2023] [Accepted: 05/30/2023] [Indexed: 06/12/2023] Open
Abstract
Targeted alpha therapy (TAT) has garnered significant interest as an innovative cancer therapy. Owing to their high energy and short range, achieving selective α-particle accumulation in target tumor cells is crucial for obtaining high potency without adverse effects. To meet this demand, we fabricated an innovative radiolabeled antibody, specifically designed to selectively deliver 211At (α-particle emitter) to the nuclei of cancer cells. The developed 211At-labeled antibody exhibited a superior effect compared to its conventional counterparts. This study paves the way for organelle-selective drug delivery.
Collapse
Affiliation(s)
- Yuki Iizuka
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
| | - Yoshiyuki Manabe
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
- Forefront Research Center, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
- Division of Science, Institute for Radiation Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
| | - Kazuhiro Ooe
- Radioisotope Research Center, Institute for Radiation Sciences, Osaka University, 2-4 Yamadaoka, Suita 565-0871, Osaka, Japan
| | - Atsushi Toyoshima
- Division of Science, Institute for Radiation Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
| | - Xiaojie Yin
- RIKEN Nishina Center for Accelerator-Based Science, 2-1 Hirosawa, Wako 351-0198, Saitama, Japan
| | - Hiromitsu Haba
- RIKEN Nishina Center for Accelerator-Based Science, 2-1 Hirosawa, Wako 351-0198, Saitama, Japan
| | - Kazuya Kabayama
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
- Forefront Research Center, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
- Division of Science, Institute for Radiation Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
| | - Koichi Fukase
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
- Forefront Research Center, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
- Division of Science, Institute for Radiation Sciences, Osaka University, 1-1 Machikaneyama, Toyonaka 560-0043, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, 1-1 Yamadaoka, Suita 565-0871, Osaka, Japan
| |
Collapse
|
45
|
Funeh CN, Bridoux J, Ertveldt T, De Groof TWM, Chigoho DM, Asiabi P, Covens P, D'Huyvetter M, Devoogdt N. Optimizing the Safety and Efficacy of Bio-Radiopharmaceuticals for Cancer Therapy. Pharmaceutics 2023; 15:pharmaceutics15051378. [PMID: 37242621 DOI: 10.3390/pharmaceutics15051378] [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: 03/31/2023] [Revised: 04/20/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
The precise delivery of cytotoxic radiation to cancer cells through the combination of a specific targeting vector with a radionuclide for targeted radionuclide therapy (TRT) has proven valuable for cancer care. TRT is increasingly being considered a relevant treatment method in fighting micro-metastases in the case of relapsed and disseminated disease. While antibodies were the first vectors applied in TRT, increasing research data has cited antibody fragments and peptides with superior properties and thus a growing interest in application. As further studies are completed and the need for novel radiopharmaceuticals nurtures, rigorous considerations in the design, laboratory analysis, pre-clinical evaluation, and clinical translation must be considered to ensure improved safety and effectiveness. Here, we assess the status and recent development of biological-based radiopharmaceuticals, with a focus on peptides and antibody fragments. Challenges in radiopharmaceutical design range from target selection, vector design, choice of radionuclides and associated radiochemistry. Dosimetry estimation, and the assessment of mechanisms to increase tumor uptake while reducing off-target exposure are discussed.
Collapse
Affiliation(s)
- Cyprine Neba Funeh
- Laboratory for In Vivo Cellular and Molecular Imaging, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K.001, 1090 Brussels, Belgium
| | - Jessica Bridoux
- Laboratory for In Vivo Cellular and Molecular Imaging, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K.001, 1090 Brussels, Belgium
| | - Thomas Ertveldt
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Timo W M De Groof
- Laboratory for In Vivo Cellular and Molecular Imaging, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K.001, 1090 Brussels, Belgium
| | - Dora Mugoli Chigoho
- Laboratory for In Vivo Cellular and Molecular Imaging, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K.001, 1090 Brussels, Belgium
| | - Parinaz Asiabi
- Laboratory for In Vivo Cellular and Molecular Imaging, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K.001, 1090 Brussels, Belgium
| | - Peter Covens
- Laboratory for In Vivo Cellular and Molecular Imaging, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K.001, 1090 Brussels, Belgium
| | - Matthias D'Huyvetter
- Laboratory for In Vivo Cellular and Molecular Imaging, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K.001, 1090 Brussels, Belgium
| | - Nick Devoogdt
- Laboratory for In Vivo Cellular and Molecular Imaging, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K.001, 1090 Brussels, Belgium
| |
Collapse
|
46
|
Radiochemistry with {Al18F}2+: Current status and optimization perspectives for efficient radiofluorination by complexation. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2023.215028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
47
|
Monte Carlo simulation study to explore optimum conditions for Astatine-211 SPECT. Radiol Phys Technol 2023; 16:102-108. [PMID: 36719548 DOI: 10.1007/s12194-023-00702-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 01/20/2023] [Accepted: 01/21/2023] [Indexed: 02/01/2023]
Abstract
211At is a promising nuclide for targeted radioisotope therapy. Direct imaging of this nuclide is important for in vivo evaluation of its distribution. We investigated suitable conditions for single-photon emission computed tomography (SPECT) imaging of 211At and assessed their feasibility using a homemade Monte Carlo simulation code, MCEP-SPECT. Radioactivity concentrations of 5, 10, or 20 kBq/mL were distributed in six spheres in a National Electrical Manufactures Association (NEMA) body phantom with a background of 1 kBq/mL. The energy window, projection number, and acquisition time were 71-88 keV, 60, and 60 s, respectively, per projection. A medium-energy collimator and three low-energy collimators were tested. SPECT images were reconstructed using the ordered subset expectation maximization (OSEM) method with attenuation correction (Chang method) and scatter correction (triple-energy-windows method). Image quality was evaluated using the contrast-to-noise ratio (CNR) for detectability and the contrast recovery coefficient (CRC) for quantitavity. The low-energy, high-sensitivity collimator exhibited the best detectability among the four types of collimators, with a maximum CNR value of 43. In contrast, the low-energy, high-resolution collimator exhibited excellent quantitavity, with a maximum CRC value of 102%. Scatter correction improved the image quality. In particular, the CRC value almost doubled after scatter correction. The detection of spheres smaller than 20 mm in diameter was difficult. In summary, low-energy collimators were suitable for the SPECT imaging of 211At. In addition, scatter correction was extremely effective in improving the image quality. The feasibility of 211At SPECT was demonstrated for lesions larger than 20 mm.
Collapse
|
48
|
Ostuni E, Taylor MRG. Commercial and business aspects of alpha radioligand therapeutics. Front Med (Lausanne) 2023; 9:1070497. [PMID: 36816719 PMCID: PMC9932801 DOI: 10.3389/fmed.2022.1070497] [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: 10/14/2022] [Accepted: 12/29/2022] [Indexed: 02/05/2023] Open
Abstract
Radioligand therapy (RLT) is gaining traction as a safe and effective targeted approach for the treatment of many cancer types, reflected by a substantial and growing commercial market (valued at $7.78 billion in 2021, with a projected value of $13.07 billion by 2030). Beta-emitting RLTs have a long history of clinical success dating back to the approval of Zevalin and Bexxar in the early 2000s, later followed by Lutathera and Pluvicto. Alpha radioligand therapeutics (ARTs) offer the potential for even greater success. Driven by ground-breaking clinical results in early trials, improved isotope availability, and better understanding of isotope and disease characteristics, the global market for alpha emitters was estimated at $672.3 million for the year 2020, with projected growth to $5.2 billion by 2027. New company formations, promising clinical trial data, and progression for many radioligand therapy products, as well as an inflow of investor capital, are contributing to this expanding field. Future growth will be fueled by further efficacy and safety data from ART clinical trials and real-world results, but challenges remain. Radionuclide supply, manufacturing, and distribution are key obstacles for growth of the field. New models of delivery are needed, along with cross-disciplinary training of specialized practitioners, to ensure patient access and avoid challenges faced by early RLT candidates such as Zevalin and Bexxar. Understanding of the history of radiation medicine is critical to inform what may be important to the success of ART-most past projections were inaccurate and it is important to analyze the reasons for this. Practical considerations in how radiation medicine is delivered and administered are important to understand in order to inform future approaches.
Collapse
Affiliation(s)
- Emanuele Ostuni
- ARTbio Inc., Cambridge, MA, United States,*Correspondence: Emanuele Ostuni,
| | | |
Collapse
|
49
|
Bruland ØS, Larsen RH, Baum RP, Juzeniene A. Editorial: Targeted alpha particle therapy in oncology. Front Med (Lausanne) 2023; 10:1165747. [PMID: 36960341 PMCID: PMC10029265 DOI: 10.3389/fmed.2023.1165747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 03/09/2023] Open
Affiliation(s)
- Øyvind Sverre Bruland
- Department of Oncology, The Norwegian Radium Hospital, Oslo University Hospital, Oslo, Norway
- Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Richard Paul Baum
- Curanosticum Wiesbaden-Frankfurt, Center for Advanced Radiomolecular Precision Oncology, Wiesbaden, Germany
| | - 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
- *Correspondence: Asta Juzeniene
| |
Collapse
|
50
|
Nelson BJB, Wilson J, Schultz MK, Andersson JD, Wuest F. High-yield cyclotron production of 203Pb using a sealed 205Tl solid target. Nucl Med Biol 2023; 116-117:108314. [PMID: 36708660 DOI: 10.1016/j.nucmedbio.2023.108314] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/15/2022] [Accepted: 01/02/2023] [Indexed: 01/15/2023]
Abstract
INTRODUCTION 203Pb (t1/2 = 51.9 h, 279 keV (81 %)) is a diagnostic SPECT imaging radionuclide ideally suited for theranostic applications in combination with 212Pb for targeted alpha particle therapy. Our objectives were to develop a high-yield solid target 203Pb cyclotron production route using isotopically enriched 205Tl target material and the 205Tl(p,3n)203Pb reaction as an alternative to lower energy production via the 203Tl(p,n)203Pb reaction. METHODS 250 mg 205Tl metal (99.9 % isotopic enrichment) was pressed using a hardened stainless steel die. Aluminum target discs were machined with a central depression and annulus groove. The flattened 205Tl pellet was placed into the central depression of the Al disc and a circle of indium wire was laid in the machined annulus surrounding the pellet. An aluminum foil cover was then pressed onto the target disc to create an airtight bond. Targets were irradiated at 23.3 MeV for up to 516 min on a TR-24 cyclotron at currents up to 60 μA to produce 203Pb via the 205Tl(p,3n)203Pb nuclear reaction. Following a cool-down period of >12 h, the target was removed and 205Tl dissolved in 4 M HNO3. A NEPTIS Mosaic-LC synthesis unit performed automated separation using Eichrom Pb resin, and 203Pb was eluted using 8 M HCl or 1 M NH4OAc. 205Tl was diverted to a vial for recovery in an electrolytic cell. 203Pb product radionuclidic purity was assessed by HPGe gamma spectroscopy, while elemental purity was assessed by ICP-OES. Radiolabeling and stability studies were performed with PSC, TCMC, and DOTA chelators, and 203Pb incorporation was verified by radio-TLC analysis. RESULTS Cyclotron irradiations performed at 60 μA proton beam current and 23.3 MeV (205Tl incident energy) had a 203Pb saturated yield of 4658 ± 62 MBq/μA (n = 3). Automated NEPTIS separation took <4 h from the start of target dissolution to product elution, yielding >85 % decay-corrected [203Pb]PbCl2 with a radionuclidic purity of >99.9 %. Purified [203Pb]PbCl2 yields of up to 12 GBq 203Pb were attained (15.8 GBq at EOB). The [203Pb]PbCl2 and [203Pb]Pb(OAc)2 products contained no detectable radionuclidic impurities besides 201Pb (<0.1 %), and <0.4 ppm stable Pb. 205Tl metal was recovered with a 92 % batch yield. Aliquots of 100 μL [203Pb]Pb(OAc)2 were used for radiolabeling PSC-Bn-NCS, TCMC-NCS, and DOTA-NCS chelators at pH 4.5 and 22 °C for 30 min, with maximum respective molar activities of 461 ± 30 GBq/μmol, 195 ± 37 GBq/μmol, and 83 ± 12 GBq/μmol. PSC, TCMC, and DOTA chelators exhibited >99.9 % incorporation after a 120-hour incubation in human serum at 37 °C. CONCLUSIONS Nuclear medicine centers with access to higher energy cyclotrons can produce large 203Pb activities sufficient for clinical applications, with a convenient separation technique producing highly pure [203Pb]PbCl2 or [203Pb]Pb(OAc)2 for direct radiolabeling. This represents an attractive route to produce 203Pb for diagnostic SPECT imaging alongside 212Pb targeted alpha particle therapy. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE Our high-yield 203Pb production technique significantly enhances 203Pb production capabilities to meet the growing preclinical and clinical demand for 203Pb radiopharmaceuticals alongside 212Pb target alpha particle therapy.
Collapse
Affiliation(s)
- Bryce J B Nelson
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta T6G 1Z2, Canada
| | - John Wilson
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, Alberta T6G 1Z2, Canada
| | - Michael K Schultz
- Viewpoint Molecular Targeting, Inc., Coralville, IA 52241, USA; Department of Radiology, The University of Iowa, Iowa City, IA 52240, 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.
| |
Collapse
|