1
|
Zhang Y, Coghi P, Ren Z, Hosmane NS, Zhu Y. Comparison of Radionuclide Drug Conjugates With Boron Neutron Capture Therapy: An Overview of Targeted Charged Particle Radiation Therapy. Med Res Rev 2025; 45:867-886. [PMID: 39690514 DOI: 10.1002/med.22093] [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: 06/24/2024] [Revised: 09/11/2024] [Accepted: 11/25/2024] [Indexed: 12/19/2024]
Abstract
Targeted charged alpha- and beta-particle therapies are currently being used in clinical radiation treatments as newly developed methods for either killing or controlling tumor cell growth. The alpha particles can be generated either through a nuclear decay reaction or in situ by a nuclear fission reaction such as the boron neutron capture reaction. Different strategies have been employed to improve the selectivity and delivery of radiation dose to tumor cells based on the source of the clinically used alpha particles. As a result, the side effects of the treatment can be minimized. The increasing attention and research efforts on targeted alpha-particle therapy have been fueled by exciting results of both academic research and clinical trials. It is highly anticipated that alpha-particle therapy will improve the efficacy of treating malignant tumors. In this overview, we compare radionuclide drug conjugates (RDC) with boron neutron capture therapy (BNCT) to present recent developments in targeted alpha-particle therapy.
Collapse
Affiliation(s)
| | - Paolo Coghi
- Laboratory for Drug Discovery from Natural Resources & Industrialization, School of Pharmacy, Macau University of Science and Technology, Macau, China
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Zimo Ren
- Laboratory for Drug Discovery from Natural Resources & Industrialization, School of Pharmacy, Macau University of Science and Technology, Macau, China
| | - Narayan S Hosmane
- Department of Chemistry and Biochemistry, Northern Illinois University, DeKalb, Illinois, USA
| | | |
Collapse
|
2
|
Yamaguchi N, Wei JJ, Isomoto H. Clinical application of targeted α-emitter therapy in gastroenteropancreatic neuroendocrine neoplasms. J Gastroenterol 2025:10.1007/s00535-025-02241-z. [PMID: 40220045 DOI: 10.1007/s00535-025-02241-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/28/2025] [Indexed: 04/14/2025]
Abstract
Effective therapeutic strategies for advanced gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) remain challenging, including a lack of response to therapy and post-treatment relapse. The rapid development of targeted radionuclide therapy (TRT) offers promising data for patients with somatostatin receptor (SSTR)-expressing tumors. This approach exhibits more advantages than somatostatin analog (SSA) therapy, which is primarily effective for well-differentiated and slow-growing GEP-NENs. Fortunately, some clinical studies on peptide receptor radionuclide therapy (PRRT) labeled with α-emitting radionuclides for GEP-NENs patients showed effective results for those with more advanced GEP-NENs, or those with malignant metastasis. For the improvement of clinical efficacy and the decline in the incidence of treatment-related relapse, recent progress in developing novel techniques and effective disease management strategies for optimal targeting has led to the emergence of targeted alpha therapy (TAT) in GEP-NENs patients. For instance, labeled technology and combination therapy could contribute to significantly improved long-term outcomes. However, the exact dosimetry for precision oncology, the shortage of radionuclides, and the stability of disease control are still under careful consideration. More high-quality, large-scale prospective studies are essential for obtaining valuable evidence on challenging problems and for further exploration.
Collapse
Affiliation(s)
- Naoyuki Yamaguchi
- Department of Endoscopy, Nagasaki University Hospital, 1-7-1 Sakamoto, Nagasaki, Nagasaki, 852-8501, Japan
| | - Jing-Jing Wei
- Department of Endoscopy, the First Affiliated Hospital of Fujian Medical University, Cha Zhong Road No.20, Tai Jiang District, Fuzhou, 350004, Fujian, China.
- Department of Endoscopy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, Fujian, China.
- Division of Gastroenterology and Nephrology, Department of Multidisciplinary Internal Medicine, School of Medicine, Faculty of Medicine, Tottori University, Yonago, 683-8504, Japan.
| | - Hajime Isomoto
- Division of Gastroenterology and Nephrology, Department of Multidisciplinary Internal Medicine, School of Medicine, Faculty of Medicine, Tottori University, Yonago, 683-8504, Japan
| |
Collapse
|
3
|
Bogdanovic B, Hugonnet F, Montemagno C. Theranostics in Hematological Malignancies: Cutting-Edge Advances in Diagnosis and Targeted Therapy. Cancers (Basel) 2025; 17:1247. [PMID: 40227793 PMCID: PMC11987953 DOI: 10.3390/cancers17071247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 04/15/2025] Open
Abstract
Hematologic malignancies, including leukemia, lymphoma, and multiple myeloma, continue to challenge clinicians with complex treatment regimens that often involve significant side effects and limited success, especially in advanced stages. Recent advancements in nuclear medicine have introduced theranostic strategies that merge diagnostic imaging with targeted therapeutic approaches, offering the potential for more precise and personalized treatment. A key area of progress lies in the development of alpha-emitting radiopharmaceuticals, such as 225Ac, 211At, or 212Pb, which can deliver potent radiation directly to tumor cells, sparing healthy tissue and minimizing collateral damage. In parallel with these therapeutic advancements, molecular imaging using radiolabeled agents enables better disease monitoring, assessment of treatment efficacy, and personalized management of patients with hematologic malignancies. The integration of diagnostic imaging with radiotherapy allows for a more tailored approach, where treatment can be adjusted based on real-time information about tumor progression and response. This review examines the recent strides made in both the development of radiopharmaceuticals and their applications in molecular imaging, with a focus on the potential to improve precision, reduce toxicity, and optimize patient outcomes. The synergy between targeted therapy and molecular imaging represents a transformative shift in the management of hematologic malignancies. As these technologies evolve, they are poised to redefine treatment paradigms, offering new hope for patients and potentially improving survival rates with more effective and less toxic treatment options.
Collapse
Affiliation(s)
- Bojana Bogdanovic
- Laboratory of Bioclinical Radiopharmaceutics, University Grenoble Alpes, INSERM, CHU Grenoble Alpes, 38000 Grenoble, France;
| | - Florent Hugonnet
- Nuclear Medicine Department, Centre Hospitalier Princesse Grace, 98000 Monaco, Monaco;
| | - Christopher Montemagno
- Laboratory of Bioclinical Radiopharmaceutics, University Grenoble Alpes, INSERM, CHU Grenoble Alpes, 38000 Grenoble, France;
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco
| |
Collapse
|
4
|
Batool A, Kopp I, Kubeil M, Bachmann M, Andrews PC, Stephan H. Targeted bismuth-based materials for cancer. Dalton Trans 2025; 54:5614-5639. [PMID: 40040450 DOI: 10.1039/d5dt00163c] [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: 03/06/2025]
Abstract
The use of bismuth and its compounds in biomedicine has developed rapidly in recent years. Due to their unique properties, there are great opportunities for the development of new non-invasive strategies for the early diagnosis and effective treatment of cancers. This perspective highlights key fabrication methods to generate well-defined and clinically relevant bismuth materials of varying characteristics. On the one hand, this opens up a wide range of possibilities for unimodal and multimodal imaging. On the other hand, effective treatment strategies, which are increasingly based on combinatorial therapies, are given a great deal of attention. One of the biggest challenges remains the selective tumour targeting, whether active or passive. Here we present an overview on new developments of bismuth based materials moving forward from a simple enrichment at the tumour site via uptake by the mononuclear phagocytic system (MPS) to a more active tumour specific targeting via covalent modification with tumour-seeking molecules based on either small or antibody-derived molecules.
Collapse
Affiliation(s)
- Amna Batool
- School of Chemistry, Monash University, Clayton, Melbourne, VIC 3800, Australia.
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany.
| | - Ina Kopp
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany.
| | - Manja Kubeil
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany.
| | - Michael Bachmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany.
| | - Philip C Andrews
- School of Chemistry, Monash University, Clayton, Melbourne, VIC 3800, Australia.
| | - Holger Stephan
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany.
| |
Collapse
|
5
|
Rousseau C, Baumgartner P, Heymann MF, Taupin M, Geffroy M, Chatal JF, Gautier G, Allam N, Gaschet J, Eychenne R, Guérard F, Gestin JF, Varmenot N, Chérel M. Preclinical and Clinical Feasibility Studies as the First Step Before Forthcoming Intravesical Instillation of [ 211At]At-anti-CA-IX Antibody (ATO-101™) Study in Patients with Non-Muscle-Invasive Bladder Cancer Unresponsive to Standard of Care. Cancers (Basel) 2025; 17:1190. [PMID: 40227816 PMCID: PMC11987989 DOI: 10.3390/cancers17071190] [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: 02/10/2025] [Revised: 03/19/2025] [Accepted: 03/25/2025] [Indexed: 04/15/2025] Open
Abstract
INTRODUCTION Recently, alpha-emitting radionuclides like astatine-211 have offered promising results in clinical development. Non-muscle-invasive bladder cancer (NMIBC) presents a need for novel therapies. One promising approach is radioimmunotherapy targeting Carbonic Anhydrase IX (CA-IX), which is supported by preclinical and clinical evidence. The aim of our preclinical and clinical studies was to evaluate the [211At]At-anti-CA-IX antibody (ATO-101™) for future use in NMIBC patient care. METHODS The anti-CA-IX antibody, girentuximab (TLX250), was labeled with lutetium-177 and astatine-211 for in vitro studies. Affinity constant measurements of [211At]At-girentuximab in RT-112 cells were taken, and toxicity evaluations were conducted in vitro and in healthy mice. Additionally, a clinical proof-of-concept study, PERTINENCE, that used [89Zr]Zr-girentuximab for PET/CT imaging in bladder cancer patients was conducted. RESULTS The measurement of the affinity constant of [211At]At-girentuximab in RT112 cells revealed high binding affinity and significant cytotoxicity compared to [177Lu]Lu-girentuximab. Biodistribution studies in healthy mice indicated low systemic radioactivity uptake, and a bladder post-instillation examination showed no abnormalities in bladder mucosa, suggesting safety. In the PERTINENCE study, which involved patients with NMIBC tumors expressing CA-IX, [89Zr]Zr-girentuximab PET/CT showed no extravesical leakage. Wall bladder uptake spots correlated with recurrence or inflammatory reaction. A dosimetric study suggested the potential efficacy and favorable safety profile of intravesical alpha therapy with the [211At]At-anti-CA-IX antibody (ATO-101™) in NMIBC treatment. CONCLUSIONS Preclinical and clinical data demonstrate the promising therapeutic role of 211At-targeted alpha agents in NMIBC, and the [211At]At-anti-CA-IX antibody (ATO-101™) could fulfill this role. A phase I FIH clinical trial is in preparation, and results are expected within the next years.
Collapse
Affiliation(s)
- Caroline Rousseau
- Institut de Cancérologie de l’Ouest, F-44800 Saint-Herblain, France; (P.B.); (M.-F.H.); (M.T.); (M.G.); (N.A.); (J.G.); (J.-F.G.); (N.V.); (M.C.)
- Nantes Université, INSERM, CNRS, CRCI2NA, Univ Angers, F-44000 Nantes, France; (R.E.); (F.G.)
| | - Pierre Baumgartner
- Institut de Cancérologie de l’Ouest, F-44800 Saint-Herblain, France; (P.B.); (M.-F.H.); (M.T.); (M.G.); (N.A.); (J.G.); (J.-F.G.); (N.V.); (M.C.)
| | - Marie-Françoise Heymann
- Institut de Cancérologie de l’Ouest, F-44800 Saint-Herblain, France; (P.B.); (M.-F.H.); (M.T.); (M.G.); (N.A.); (J.G.); (J.-F.G.); (N.V.); (M.C.)
- Research Pathology Platform, Tumor Heterogeneity and Precision Medicine, F-44800 Saint-Herblain, France
| | - Manon Taupin
- Institut de Cancérologie de l’Ouest, F-44800 Saint-Herblain, France; (P.B.); (M.-F.H.); (M.T.); (M.G.); (N.A.); (J.G.); (J.-F.G.); (N.V.); (M.C.)
- Research Pathology Platform, F-44800 Saint-Herblain, France
| | - Maïwenn Geffroy
- Institut de Cancérologie de l’Ouest, F-44800 Saint-Herblain, France; (P.B.); (M.-F.H.); (M.T.); (M.G.); (N.A.); (J.G.); (J.-F.G.); (N.V.); (M.C.)
| | - Jean-François Chatal
- Atonco, Pôle Bio-Ouest Laennec, Rue du Moulin de la Rousselière, F-44800 Saint-Herblain, France;
| | - Gaëlle Gautier
- Chelatec, 1 Rue Aronnax, F-44817 Saint-Herblain, France;
| | - Nadia Allam
- Institut de Cancérologie de l’Ouest, F-44800 Saint-Herblain, France; (P.B.); (M.-F.H.); (M.T.); (M.G.); (N.A.); (J.G.); (J.-F.G.); (N.V.); (M.C.)
| | - Joëlle Gaschet
- Institut de Cancérologie de l’Ouest, F-44800 Saint-Herblain, France; (P.B.); (M.-F.H.); (M.T.); (M.G.); (N.A.); (J.G.); (J.-F.G.); (N.V.); (M.C.)
- Nantes Université, INSERM, CNRS, CRCI2NA, Univ Angers, F-44000 Nantes, France; (R.E.); (F.G.)
| | - Romain Eychenne
- Nantes Université, INSERM, CNRS, CRCI2NA, Univ Angers, F-44000 Nantes, France; (R.E.); (F.G.)
- Groupement d’Intérêt Public ARRONAX, 1 Rue Aronnax, F-44817 Saint-Herblain, France
| | - François Guérard
- Nantes Université, INSERM, CNRS, CRCI2NA, Univ Angers, F-44000 Nantes, France; (R.E.); (F.G.)
| | - Jean-François Gestin
- Institut de Cancérologie de l’Ouest, F-44800 Saint-Herblain, France; (P.B.); (M.-F.H.); (M.T.); (M.G.); (N.A.); (J.G.); (J.-F.G.); (N.V.); (M.C.)
- Nantes Université, INSERM, CNRS, CRCI2NA, Univ Angers, F-44000 Nantes, France; (R.E.); (F.G.)
| | - Nicolas Varmenot
- Institut de Cancérologie de l’Ouest, F-44800 Saint-Herblain, France; (P.B.); (M.-F.H.); (M.T.); (M.G.); (N.A.); (J.G.); (J.-F.G.); (N.V.); (M.C.)
- Nantes Université, INSERM, CNRS, CRCI2NA, Univ Angers, F-44000 Nantes, France; (R.E.); (F.G.)
- Groupement d’Intérêt Public ARRONAX, 1 Rue Aronnax, F-44817 Saint-Herblain, France
| | - Michel Chérel
- Institut de Cancérologie de l’Ouest, F-44800 Saint-Herblain, France; (P.B.); (M.-F.H.); (M.T.); (M.G.); (N.A.); (J.G.); (J.-F.G.); (N.V.); (M.C.)
- Nantes Université, INSERM, CNRS, CRCI2NA, Univ Angers, F-44000 Nantes, France; (R.E.); (F.G.)
| |
Collapse
|
6
|
Salas-Ramirez M, Lassmann M, Eberlein U. In silico analysis of radiation-induced double-strand breaks by internal ex vivo irradiation of lymphocytes for 45 alpha- and beta/gamma-emitting radionuclides. EJNMMI Res 2025; 15:21. [PMID: 40063302 PMCID: PMC11893945 DOI: 10.1186/s13550-025-01214-w] [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: 11/12/2024] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND The aim of this study is to evaluate the induction of DNA damage by 45 radionuclides, including those used in medical applications and others relevant to radiation protection. The research focuses on understanding the differential effects of irradiating lymphocytes with beta/gamma- and alpha-emitting radionuclides using Monte Carlo simulations. A validated Monte Carlo simulation model was used to assess radiation-induced DNA damage in lymphocytes. The model integrates GATE for macroscopic radiation transport and Geant4-DNA for microscopic simulations at the cellular level. For the study, 45 radionuclides were selected and their S-values and DNA double-strand break (DSB) induction were investigated. For beta- and gamma-emitting radionuclides, DSBs per cell per mGy were quantified, while for alpha-emitters, alpha tracks per cell per mGy, DSBs per cell per mGy, and DSBs per micrometer of alpha track were calculated. RESULT For beta/gamma emitters, the lowest number of DSBs was observed with 125I at 0.006 ± 0.003 DSBs·cell⁻¹·mGy⁻¹, while 99mTc had the highest at approximately 0.015 ± 0.005 DSBs·cell⁻¹·mGy⁻¹. The S-value for lymphocyte nuclei ranked from 0.91 ± 0.14 mGy∙h⁻¹∙MBq⁻¹ (63Ni) and 1.06 ± 0.15 mGy∙h⁻¹∙MBq⁻¹ (125I) to 61.83 ± 1.17 mGy∙h⁻¹∙MBq⁻¹ (90Sr). For alpha-emitting radionuclides, 213Bi produced 0.0677 ± 0.0005 DSB·cell⁻¹·mGy⁻¹ while 232Th yielded 0.0914 ± 0.0004 DSB·cell⁻¹·mGy⁻¹. The DSB linear density for alpha tracks ranged from 7.4 ± 0.1 DSBs/µm for 252Cf to 16.8 ± 0.1 DSBs/µm for 232Th. The S-values for lymphocyte nuclei for alpha emitters varied, from 232Th (0.29 ± 0.21 Gy∙h⁻¹∙MBq⁻¹) to 227Th having the highest at 2.22 ± 0.16 Gy∙h⁻¹∙MBq⁻¹, due to cumulative energy deposition. CONCLUSIONS Differences were observed in DNA damage induced by beta/gamma- and alpha-emitting radionuclides. High-energy beta emitters induced DSBs similarly to gamma emitters, but with greater fluctuations in low-energy beta and gamma emitters due to heterogeneous energy deposition and varying interaction probabilities at the cellular level. This study highlights that long half-life alpha-emitting radionuclides may cause more extensive DNA damage due to their higher LET. This work provides a comprehensive S-values database for future experimental studies on radiation-induced DNA damage in lymphocytes.
Collapse
Affiliation(s)
- Maikol Salas-Ramirez
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany.
| | - Michael Lassmann
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Uta Eberlein
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
7
|
Mapanao AK, Busslinger SD, Mehta A, Kegler K, Favaretto C, Grundler PV, Talip Z, Köster U, Johnston K, Schibli R, van der Meulen NP, Müller C. Preclinical investigation of [ 149Tb]Tb-DOTATATE and [ 149Tb]Tb-DOTA-LM3 for tumor-targeted alpha therapy. Eur J Nucl Med Mol Imaging 2025; 52:1383-1398. [PMID: 39743617 DOI: 10.1007/s00259-024-07035-8] [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: 09/13/2024] [Accepted: 12/15/2024] [Indexed: 01/04/2025]
Abstract
PURPOSE Terbium-149 is a short-lived α-particle emitter, potentially useful for tumor-targeted therapy. The aim of this study was to investigate terbium-149 in combination with the somatostatin receptor (SSTR) agonist DOTATATE and the SSTR antagonist DOTA-LM3. The radiopeptides were evaluated to compare their therapeutic efficacy in vitro and in vivo. METHODS Terbium-149 was produced at ISOLDE/CERN and chemically purified at the Paul Scherrer Institute. Radiolabeling of somatostatin analogues with [149Tb]TbCl3 was performed under standard labeling conditions at pH 4.5. Cell viability (MTT) and survival assays (colony forming) assays were performed after 16-18 h exposure of SSTR-positive AR42J rat pancreatic tumor cells to various activity concentrations of [149Tb]Tb-DOTATATE and [149Tb]Tb-DOTA-LM3. DNA double-strand breaks were determined using immunofluorescence imaging of γ-H2A.X and 53BP1. Therapy studies were performed with AR42J tumor-bearing mice injected with 1 × 5 MBq or 2 × 5 MBq of the respective radiopeptide. The tolerability of up to 40 MBq [149Tb]Tb-DOTATATE or 40 MBq [149Tb]Tb-DOTA-LM3 was assessed with regard to undesired effects to the bone marrow and kidneys in immunocompetent mice without tumors. RESULTS The radiolabeling of peptides was achieved at molar activities of up to 20 MBq/nmol at ≥ 98% radiochemical purity. AR42J cell viability was reduced in an activity-dependent manner, with [149Tb]Tb-DOTA-LM3 being slightly more potent than [149Tb]Tb-DOTATATE (EC50: 0.5 vs. 1.2 kBq/mL). Both radiopeptides induced a similar number of γ-H2A.X and 53BP1 foci per nuclei, which indicated DNA damage in AR42J tumor cells. Injection of tumor-bearing mice with 1 × 5 MBq radiopeptide resulted in median survival times of 16.5 days and 19 days for [149Tb]Tb-DOTATATE and [149Tb]Tb-DOTA-LM3, respectively, as compared to only 8 days for untreated control mice. Application of 2 × 5 MBq of the radiopeptides further extended the median survival times to 30 days and 29 days, respectively. The blood cell counts and values for blood plasma biomarkers of treated mice without tumors were similar to those of untreated controls. Renal accumulation of [99mTc]Tc-DMSA was similar in all mice, indicating normal kidney function. CONCLUSION 149Tb-based radiopeptides effectively reduced the viability of tumor cells in vitro as well as the tumor growth in mice without causing relevant adverse events, irrespective of whether the SSTR agonist or antagonist was employed. These data encourage further preclinical application of terbium-149 to evaluate its potential in combination with other tumor-targeting agents.
Collapse
Affiliation(s)
- Ana Katrina Mapanao
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland
| | - Sarah D Busslinger
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland
| | - Avni Mehta
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland
| | | | - Chiara Favaretto
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland
- Division of Nuclear Medicine, University Hospital Basel, Basel, 4031, Switzerland
| | - Pascal V Grundler
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland
| | - Zeynep Talip
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland
- Laboratory of Radiochemistry, PSI Center for Nuclear Engineering and Sciences, Villigen-PSI, 5232, Switzerland
| | - Ulli Köster
- Institut Laue-Langevin, Grenoble, 38042, France
| | - Karl Johnston
- Physics Department, ISOLDE/CERN, Geneva, 1211, Switzerland
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, 8093, Switzerland
| | - Nicholas P van der Meulen
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland
- Laboratory of Radiochemistry, PSI Center for Nuclear Engineering and Sciences, Villigen-PSI, 5232, Switzerland
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland.
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, 8093, Switzerland.
| |
Collapse
|
8
|
Navalkissoor S, Grossman A. Somatostatin receptor-linked α-particle therapy in neuroendocrine tumours. J Neuroendocrinol 2025; 37:e13463. [PMID: 39529416 DOI: 10.1111/jne.13463] [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: 06/19/2024] [Revised: 09/19/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
The incidence and prevalence of neuroendocrine tumours (NETs) are on the rise, but to date, only complete surgical resection is curative. Among the various therapeutic options for metastatic disease, peptide receptor radionuclide therapy (PRRT), linking a radioactive moiety to an octreotide derivative, has been shown to be highly efficacious and a well-tolerated therapy, improving progression-free survival and prolonging overall survival. Nevertheless, complete responses are rare, and the current β-particle emitters have non-optimal radiobiological properties. A new generation of α-particle-emitting radionuclides is being developed, with the advantages of very high energy and a short path length. We survey the most recent developments in this field, summarising the result of currently performed studies in this potentially ground-breaking novel form of therapy for NETs.
Collapse
Affiliation(s)
- Shaunak Navalkissoor
- Department of Nuclear Medicine, Royal Free London NHS Foundation Trust, London, UK
| | - Ashley Grossman
- NET UNIT, ENETS Centre of Excellence, Royal Free London NHS Foundation Trust, London, UK
| |
Collapse
|
9
|
Cangut B, Akinlusi R, Mohseny A, Ghesani N, Ghesani M. Evolving Paradigms in Lung Cancer: Latest Trends in Diagnosis, Management, and Radiopharmaceuticals. Semin Nucl Med 2025; 55:264-276. [PMID: 40055050 DOI: 10.1053/j.semnuclmed.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 03/17/2025]
Abstract
Lung cancer is one of the most common and deadliest forms of cancer worldwide. Over the past two decades, significant changes have occurred in the classification of lung cancer, involving multidisciplinary input and emphasizing the growing contribution of immunohistochemistry and molecular techniques to morphology in the classification scheme. This comprehensive review will cover the background and epidemiology of lung cancer as well as advancements in its staging and management, including discussions of new surgical techniques, targeted therapies, and immunotherapy. The review will detail the role of 18F-FDG-PET-CT in lung cancer, highlighting its importance in staging, treatment response assessment, and recurrence detection. While immunotherapy has transformed lung cancer management and improved patient outcomes, it presents major challenges and opportunities for optimal assessment of treatment response in lung cancer patients using 18F-FDG-PET-CT. This review will also explore future directions, including a discussion of promising new targeted diagnostic radiopharmaceuticals for PET/CT imaging. Additionally, there will be a brief discussion of evolving and exciting treatment options for lung cancer using targeted therapeutic radiopharmaceuticals. Several case-based illustrations are included to exemplify the role of 18F-FDG-PET-CT in various clinical scenarios.
Collapse
Affiliation(s)
- Busra Cangut
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Rahman Akinlusi
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ava Mohseny
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Nasrin Ghesani
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Munir Ghesani
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
10
|
Lepareur N. An overview of current phase 3 radiopharmaceutical therapy clinical trials. Front Med (Lausanne) 2025; 12:1549676. [PMID: 40041466 PMCID: PMC11876376 DOI: 10.3389/fmed.2025.1549676] [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: 12/21/2024] [Accepted: 02/04/2025] [Indexed: 03/06/2025] Open
Abstract
Over the past few years, radiopharmaceutical therapy has emerged as a groundbreaking therapeutic modality, taking advantage of the unique properties of radionuclides to deliver molecularly targeted therapy with high precision and transforming the landscape of precision oncology and personalized medicine. Its development reflects decades of advances in nuclear medicine, chemistry, and cancer biology. However, until recently, definitive clinical evidence was lacking to establish it into treatment plans, with few large randomized controlled clinical studies. The last two decades witnessed a paradigm shift, with three successful phase 3 studies which shed light on radiopharmaceutical therapy. This paper offers a brief overview of currently active phase 3 studies to highlight the dynamism and promise of this clinical domain, as well as the large variety of cancers being treated.
Collapse
Affiliation(s)
- Nicolas Lepareur
- Comprehensive Cancer Center Eugène Marquis, Rennes, France
- Univ Rennes, Inrae, Inserm, Institut NUMECAN (Nutrition, Métabolismes et Cancer) – UMR_A 1341, UMR_S 1241, Rennes, France
| |
Collapse
|
11
|
Woods JJ, Rigby A, Wacker JN, Arino T, Alvarenga Vasquez JV, Cosby A, Martin KE, Abergel RJ. Synthesis and Evaluation of a Bifunctional Chelator for Thorium-227 Targeted Radiotherapy. J Med Chem 2025; 68:1682-1692. [PMID: 39752149 DOI: 10.1021/acs.jmedchem.4c02423] [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: 01/04/2025]
Abstract
Thorium-227 (227Th) is an α-emitting radionuclide currently under investigation for targeted alpha therapy. Available chelators used for this isotope suffer from challenging multistep syntheses. Here, we present the synthesis and preclinical evaluation of a novel bifunctional chelator, p-SCN-Bn-DOTHOPO, which contains an isothiocyanate group that is suitable for conjugation to biological molecules. This bifunctional chelator was prepared with a 26% overall yield in four steps and conjugated to the human epidermal growth factor receptor 2 targeting antibody, trastuzumab. The resulting immunoconjugate was labeled with [227Th]ThIV (pH 5.5, room temperature, 60 min) with ≥95% radiochemical yield and purity. The conjugate was also labeled with zirconium-89 (89Zr), which can be used for positron emission tomography imaging. The radiometal complexes were subsequently investigated for their biological stability. The results described here provide insight into ligand design strategies and optimization of chelators for the development of the next generation of 89Zr and 227Th radiopharmaceuticals.
Collapse
Affiliation(s)
- Joshua J Woods
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Alex Rigby
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Jennifer N Wacker
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Trevor Arino
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
- Department of Nuclear Engineering, University of California Berkeley, Berkeley, California 94720, United States
| | | | - Alexia Cosby
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Kirsten E Martin
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Rebecca J Abergel
- Chemical Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
- Department of Nuclear Engineering, University of California Berkeley, Berkeley, California 94720, United States
- Department of Chemistry, University of California Berkeley, Berkeley, California 94720, United States
| |
Collapse
|
12
|
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
|
13
|
Yang M, Li J, Han Z, Luan X, Zhang X, Gao J, Qin S, Yu F. Layered Double Hydroxides for Radium-223 Targeted Alpha Therapy with Elicitation of the Immune Response. Adv Healthc Mater 2025; 14:e2403175. [PMID: 39618118 DOI: 10.1002/adhm.202403175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 10/18/2024] [Indexed: 01/29/2025]
Abstract
Targeted Alpha therapy (TAT) has promising application prospects in tumor therapy. It is very appealing to design alpha-emitting radiopharmaceuticals that can modulate the immune microenvironment to overcome the limitations of immunotherapy. Herein, Mg/Al layered double hydroxide nanomaterials (LDH) are utilized to load the alpha-emitting nuclide Radium-223 (223Ra), achieving precise delivery of 223Ra to the tumor microenvironment. Dual-modal imaging is employed to dynamically monitor the in vivo distribution of 223Ra-LDH, ensuring its prolonged retention at the tumor site. In vitro experimentsshowed that ionizing radiation from alpha-emitting nuclides effectively reduced glutathione (GSH) and produced large amounts of reactive oxygen species (ROS), which damaged mitochondria and released free calcium (Ca2+), thereby aggravating tumor cell death. Additionally, DNA double-strand breaks induced by alpha-emitting radiation triggered the STING signaling pathway, which in turn effectively induced immunogenic cell death (ICD) and promoted immune cell maturation and activation. The synergistic effect with immunotherapy triggered a powerful systemic antitumor immune response. Overall, this study develops a novel TAT therapeutic strategy with sufficient antitumor immunity.
Collapse
Affiliation(s)
- Mengdie Yang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, P. R. China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, 200072, P. R. 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 Radio-toxicology and Radiopharmaceutical Preclinical Evaluation, Taiyuan, Shanxi, 030006, P. R. China
| | - Zongtai Han
- China Institute for Radiation Protection, National Atomic Energy Agency Nuclear Technology (Nonclinical Evaluation of Radiopharmaceuticals) Research and Development Center, CNNC Key Laboratory on Radio-toxicology and Radiopharmaceutical Preclinical Evaluation, Taiyuan, Shanxi, 030006, P. R. China
| | - Xiaohui Luan
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, P. R. China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, 200072, P. R. China
| | - Xiaoyi Zhang
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, P. R. China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, 200072, P. R. China
| | - 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 Radio-toxicology and Radiopharmaceutical Preclinical Evaluation, Taiyuan, Shanxi, 030006, P. R. China
| | - Shanshan Qin
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, P. R. China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, 200072, P. R. China
| | - Fei Yu
- Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, P. R. China
- Institute of Nuclear Medicine, Tongji University School of Medicine, Shanghai, 200072, P. R. China
| |
Collapse
|
14
|
Ermolaev SV, Vasiliev AN, Skasyrskaya AK, Lapshina EV, Khaliullina DR, Libanova ON. 225Aс/ 213Bi generator for direct synthesis of 213Bi-labeled bioconjugates. Nucl Med Biol 2025; 140-141:108975. [PMID: 39603113 DOI: 10.1016/j.nucmedbio.2024.108975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 11/16/2024] [Accepted: 11/21/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND 213Bi is a short-lived radionuclide currently trialed for alpha therapy of various oncological diseases. A serious obstacle to the wide medical use is decay losses of 213Bi during a conventional synthesis of radiopharmaceuticals. In this work, we aimed to develop a two-column 225Aс/213Bi generator providing the accumulation of 213Bi separately from the parent 225Ac via continuous circular separation and decay of intermediate 221Fr. When attaining the transient equilibrium, 213Bi could be promptly extracted from the generator with an appropriate complexing agent, including chelator-protein bioconjugates. METHODS Sorption behavior of Bi(III) ions onto the cross-linked dextran gel Sephadex G-25 was studied from solutions of hydrochloric and nitric acid, and from sodium chloride, sodium acetate and DTPA solutions. A bifunctional chelating agent p-SCN-Bn-DTPA was conjugated to an antibody Nimotuzumab specific to the epidermal growth factor receptor, and the procedure of 207,213Bi-DTPA-Nimotuzumab synthesis in the dextran gel medium was developed. The parameters of 225Aс/213Bi generator system were evaluated. RESULTS The weight distribution ratios of Bi(III) adsorbed onto the Sephadex G-25 gel were obtained. Up to 85 % of 213Bi was accumulated in the second Sephadex-filled column of 225Aс/213Bi generator after four-hour circulation of 0.15 M NaCl (pH 5.5) solution. Having passed the solution of DTPA-Nimotuzumab bioconjugate through the second column, a fraction of 213Bi-DTPA-Nimotuzumab radioimmunoconjugate was produced with the radiochemical yield of 64 % ± 3 % (n = 6). High radionuclidic and radiochemical purity of product was achieved. CONCLUSIONS The circulating 225Aс/213Bi generator provides a 213Bi-labeled bioconjugate as a final product. While a conventional synthesis route including generator milking, bioconjugate labeling and size-exclusion purification takes >20 min, the duration of 213Bi-DTPA-Nimotuzumab production by the method proposed in this work is reduced to 6-8 min.
Collapse
Affiliation(s)
| | - Aleksandr N Vasiliev
- Institute for Nuclear Research of Russian Academy of Sciences, Moscow, Russia; Faculty of Materials Science, Shenzhen MSU-BIT University, Shenzhen, China.
| | - Aino K Skasyrskaya
- Institute for Nuclear Research of Russian Academy of Sciences, Moscow, Russia
| | - Elena V Lapshina
- Institute for Nuclear Research of Russian Academy of Sciences, Moscow, Russia
| | - Daria R Khaliullina
- Institute for Nuclear Research of Russian Academy of Sciences, Moscow, Russia; Department of Chemistry, Lomonosov Moscow State University, Moscow, Russia
| | - Olga N Libanova
- Institute for Nuclear Research of Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
15
|
Ding J, Qin S, Hou X, Zhang J, Yang M, Ma S, Zhu H, Feng Y, Yu F. Recent advances in emerging radiopharmaceuticals and the challenges in radiochemistry and analytical chemistry. Trends Analyt Chem 2025; 182:118053. [DOI: 10.1016/j.trac.2024.118053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
16
|
Lugat A, Chouin N, Chocteau F, Esnault M, Marionneau-Lambot S, Gouard S, Frampas É, Faivre-Chauvet A, Bourgeois M, Morgenstern A, Bruchertseifer F, Chérel M, Kraeber-Bodéré F, Ansquer C, Gaschet J. Survival impact of [ 225Ac]Ac-DOTATOC alpha-therapy in a preclinical model of pancreatic neuroendocrine tumor liver micrometastases. Eur J Nucl Med Mol Imaging 2025; 52:730-743. [PMID: 39269657 DOI: 10.1007/s00259-024-06918-0] [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: 04/23/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024]
Abstract
Although peptide radionuclide therapy (PRRT) using a somatostatin analog (SSA) radiolabeled with a beta- emitter: [177Lu]Lu-DOTATATE has shown a good clinical efficacy in neuroendocrine tumors (NETs), most of the patients only achieved tumoral stabilization and rare but severe long-term hematological toxicities have been reported. One of the promising options to improve PRRT is targeted alpha therapy. It is therefore essential to propose animal models that can mimic systemic spread disease, especially microscopic disease such as early stage of NET liver metastases to explore targeted alpha therapy. Herein, we report the evaluation of efficacy and toxicity of [225Ac]Ac-DOTATOC in an original preclinical murine model simulating the development of well-characterized liver metastases of pancreatic NETs with SSTR overexpression. METHODS A mouse model of liver metastases of pancreatic NETs was developed by intraportal injection of AR42J cells and explored using [68 Ga]Ga-DOTATOC and [18F]F-FDG PET/MRI. Biodistribution study and radiation dosimetry of [225Ac]Ac-DOTATOC were determined in subcutaneous tumor-bearing NMRI-nude mice. Efficacy and toxicity were determined by intravenous injection of increasing activities of [225Ac]Ac-DOTATOC 10 days after intraportal graft. RESULTS Liver tumors showed a high uptake of [68 Ga]Ga-DOTATOC and no uptake of [18F]F-FDG confirming the well-differentiated phenotype. All groups treated with [225Ac]Ac-DOTATOC showed a significant increase in overall survival compared with DOTATOC-treated mice, especially those treated with the highest activities: 53 days with 240 kBq (p = 0.0001), and 58 days with 2 × 120 kBq (p < 0.0001) vs 28 days with non-radiolabeled DOTATOC. On blood tests, a transient and moderate decreased in white blood cells count after treatment and no severe hepatic or renal toxicity were observed after treatment which was consistent with pathological and radiation dosimetry findings. CONCLUSION [225Ac]Ac-DOTATOC exhibit a favorable efficacy and toxicity profile in a mouse model of liver micrometastatic pancreatic NET.
Collapse
Affiliation(s)
- Alexandre Lugat
- Medical Oncology Department, Nantes University Hospital, 44000, Nantes, France
- Nuclear Medicine Department, Nantes University Hospital, 1, Place Alexis Ricordeau, 44000, Nantes, France
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 8 Quai Moncousu, BP70721, Cedex 1, 44007, Nantes, France
| | - Nicolas Chouin
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 8 Quai Moncousu, BP70721, Cedex 1, 44007, Nantes, France
| | - Florian Chocteau
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 8 Quai Moncousu, BP70721, Cedex 1, 44007, Nantes, France
| | - Mathilde Esnault
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 8 Quai Moncousu, BP70721, Cedex 1, 44007, Nantes, France
| | - Séverine Marionneau-Lambot
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 8 Quai Moncousu, BP70721, Cedex 1, 44007, Nantes, France
| | - Sébastien Gouard
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 8 Quai Moncousu, BP70721, Cedex 1, 44007, Nantes, France
| | - Éric Frampas
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 8 Quai Moncousu, BP70721, Cedex 1, 44007, Nantes, France
- Central Department of Radiology and Medical Imaging, Nantes University Hospital, 44000, Nantes, France
| | - Alain Faivre-Chauvet
- Nuclear Medicine Department, Nantes University Hospital, 1, Place Alexis Ricordeau, 44000, Nantes, France
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 8 Quai Moncousu, BP70721, Cedex 1, 44007, Nantes, France
| | - Mickaël Bourgeois
- Nuclear Medicine Department, Nantes University Hospital, 1, Place Alexis Ricordeau, 44000, Nantes, France
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 8 Quai Moncousu, BP70721, Cedex 1, 44007, Nantes, France
| | - Alfred Morgenstern
- European Commission, Joint Research Centre, Directorate for Nuclear Safety and Security, Karlsruhe, Germany
| | - Frank Bruchertseifer
- European Commission, Joint Research Centre, Directorate for Nuclear Safety and Security, Karlsruhe, Germany
| | - Michel Chérel
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 8 Quai Moncousu, BP70721, Cedex 1, 44007, Nantes, France
- Department of Nuclear Medicine, Institut de Cancérologie de L'Ouest (ICO) - Site Gauducheau, Saint-Herblain, France
| | - Françoise Kraeber-Bodéré
- Nuclear Medicine Department, Nantes University Hospital, 1, Place Alexis Ricordeau, 44000, Nantes, France
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 8 Quai Moncousu, BP70721, Cedex 1, 44007, Nantes, France
| | - Catherine Ansquer
- Nuclear Medicine Department, Nantes University Hospital, 1, Place Alexis Ricordeau, 44000, Nantes, France
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 8 Quai Moncousu, BP70721, Cedex 1, 44007, Nantes, France
| | - Joëlle Gaschet
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 8 Quai Moncousu, BP70721, Cedex 1, 44007, Nantes, France.
| |
Collapse
|
17
|
Zhu H, Heinitz S, Binnemans K, Mullens S, Cardinaels T. A Preliminary Study of Spherical Surface-Modified Carbon Materials for Potential Application in La 3+ (Ac 3+) and Bi 3+ Separation. ACS OMEGA 2024; 9:51009-51021. [PMID: 39758655 PMCID: PMC11696413 DOI: 10.1021/acsomega.4c05457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/12/2024] [Accepted: 11/27/2024] [Indexed: 01/07/2025]
Abstract
Separation of high-activity 213Bi from 225Ac for targeted alpha therapy is challenging due to the instability of existing sorbents. Surface-modified carbon materials have shown promise for use in inverse 225Ac/213Bi generators. However, previously reported materials with irregular shapes may limit their applications in column separations. In contrast, spherical particles are expected to be more suitable for column chromatography compared to irregular powders as they can ensure uniform flow patterns, lower pressure drop, and effective packing. To address this limitation, a method was developed for the synthesis of spherical carbon beads via the carbonization of cellulose beads. Subsequently, surface modification on the spherical carbon beads was performed via sulfonation or oxidation of the carbon beads. Batch sorption experiments were conducted to assess their selective sorption toward Bi3+ over La3+ (as a surrogate of Ac3+) by varying the concentrations of HNO3 and NaNO3. It was found that the selective sorption of Bi3+ onto spherical surface-modified carbon beads could be achieved by adjusting the concentrations of HNO3 and NaNO3. Furthermore, the sorption capacity of Bi3+ decreased as the concentration of HCl increased due to the formation of bichloride complexes and the H+ competition. This implies that Bi3+ can be effectively eluted from the spherical surface-modified carbon beads when using HCl as the eluate. Consequently, spherical surface-modified carbon beads show potential as alternative adsorbents for inverse 225Ac/213Bi generators.
Collapse
Affiliation(s)
- Hongshan Zhu
- Belgian
Nuclear Research Centre (SCK CEN), Institute for Nuclear Materials
Science, Boeretang 200, Mol B-2400, Belgium
- KU
Leuven, Department of Chemistry, Celestijnenlaan 200F, P.O. 2404, Leuven B-3001, Belgium
- Flemish
Institute for Technological Research (VITO NV), Sustainable Materials
Management, Boeretang 200, Mol B-2400, Belgium
| | - Stephan Heinitz
- Belgian
Nuclear Research Centre (SCK CEN), Institute for Nuclear Materials
Science, Boeretang 200, Mol B-2400, Belgium
| | - Koen Binnemans
- KU
Leuven, Department of Chemistry, Celestijnenlaan 200F, P.O. 2404, Leuven B-3001, Belgium
| | - Steven Mullens
- Flemish
Institute for Technological Research (VITO NV), Sustainable Materials
Management, Boeretang 200, Mol B-2400, Belgium
| | - Thomas Cardinaels
- Belgian
Nuclear Research Centre (SCK CEN), Institute for Nuclear Materials
Science, Boeretang 200, Mol B-2400, Belgium
- KU
Leuven, Department of Chemistry, Celestijnenlaan 200F, P.O. 2404, Leuven B-3001, Belgium
| |
Collapse
|
18
|
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
|
19
|
Alcocer-Ávila M, Levrague V, Delorme R, Testa É, Beuve M. Biophysical modeling of low-energy ion irradiations with NanOx. Med Phys 2024; 51:9358-9371. [PMID: 39287463 DOI: 10.1002/mp.17407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 07/22/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Targeted radiotherapies with low-energy ions show interesting possibilities for the selective irradiation of tumor cells, a strategy particularly appropriate for the treatment of disseminated cancer. Two promising examples are boron neutron capture therapy (BNCT) and targeted radionuclide therapy with α $\alpha$ -particle emitters (TAT). The successful clinical translation of these radiotherapies requires the implementation of accurate radiation dosimetry approaches able to take into account the impact on treatments of the biological effectiveness of ions and the heterogeneity in the therapeutic agent distribution inside the tumor cells. To this end, biophysical models can be applied to translate the interactions of radiations with matter into biological endpoints, such as cell survival. PURPOSE The NanOx model was initially developed for predicting the cell survival fractions resulting from irradiations with the high-energy ion beams encountered in hadrontherapy. We present in this work a new implementation of the model that extends its application to irradiations with low-energy ions, as the ones found in TAT and BNCT. METHODS The NanOx model was adapted to consider the energy loss of primary ions within the sensitive volume (i.e., the cell nucleus). Additional assumptions were introduced to simplify the practical implementation of the model and reduce computation time. In particular, for low-energy ions the narrow-track approximation allowed to neglect the energy deposited by secondary electrons outside the sensitive volume, increasing significantly the performance of simulations. Calculations were performed to compare the original hadrontherapy implementation of the NanOx model with the present one in terms of the inactivation cross sections of human salivary gland cells as a function of the kinetic energy of incident α $\alpha$ -particles. RESULTS The predictions of the previous and current versions of NanOx agreed for incident energies higher than 1 MeV/n. For lower energies, the new NanOx implementation predicted a decrease in the inactivation cross sections that depended on the length of the sensitive volume. CONCLUSIONS We reported in this work an extension of the NanOx biophysical model to consider irradiations with low-energy ions, such as the ones found in TAT and BNCT. The excellent agreement observed at intermediate and high energies between the original hadrontherapy implementation and the present one showed that NanOx offers a consistent, self-integrated framework for describing the biological effects induced by ion irradiations. Future work will focus on the application of the latest version of NanOx to cases closer to the clinical setting.
Collapse
Affiliation(s)
- Mario Alcocer-Ávila
- Université Claude Bernard Lyon 1, CNRS/IN2P3, IP2I Lyon, UMR 5822, Villeurbanne, France
| | - Victor Levrague
- University of Grenoble Alpes, CNRS, Grenoble INP, LPSC-IN2P3, Grenoble, France
| | - Rachel Delorme
- University of Grenoble Alpes, CNRS, Grenoble INP, LPSC-IN2P3, Grenoble, France
| | - Étienne Testa
- Université Claude Bernard Lyon 1, CNRS/IN2P3, IP2I Lyon, UMR 5822, Villeurbanne, France
| | - Michaël Beuve
- Université Claude Bernard Lyon 1, CNRS/IN2P3, IP2I Lyon, UMR 5822, Villeurbanne, France
| |
Collapse
|
20
|
Au M, Nies L, Stegemann S, Athanasakis-Kaklamanakis M, Cocolios TE, Fischer P, Giesel PF, Johnson JD, Köster U, Lange D, Mougeot M, Reilly J, Schlaich M, Schweiger C, Schweikhard L, Wienholtz F, Wojtaczka W, Düllmann CE, Rothe S. Production and purification of molecular 225Ac at CERN-ISOLDE. J Radioanal Nucl Chem 2024; 334:367-379. [PMID: 39901925 PMCID: PMC11787200 DOI: 10.1007/s10967-024-09811-0] [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: 06/21/2024] [Accepted: 09/26/2024] [Indexed: 02/05/2025]
Abstract
The radioactive nuclide 225Ac is one of the few promising candidates for cancer treatment by targeted- α -therapy, but worldwide production of 225Ac faces significant limitations. In this work, the Isotope Separation On-Line method was used to produce actinium by irradiating targets made of uranium carbide and thorium carbide with 1.4-GeV protons. Actinium fluoride molecules were formed, ionized through electron impact, then extracted and mass-separated as a beam of molecular ions. The composition of the mass-selected ion beam was verified using time-of-flight mass spectrometry, α - and γ -ray decay spectrometry. Extracted quantities of225 Ac 19 F 2 + particles per μ C of incident protons were 3.9 ( 3 ) × 10 7 from a uranium carbide target and 4.3 ( 4 ) × 10 7 for a thorium carbide target. Using a magnetic mass separator, the long-lived contamination 227 Ac is suppressed to < 5.47 × 10 - 7 (95% confidence interval) with respect to 225Ac by activity. Measured rates scale to collections of 108 kBq μ A- 1 h- 1 of directly produced225 Ac 19 F 2 + .
Collapse
Affiliation(s)
- M. Au
- CERN, Meyrin, 1211 Geneva Switzerland
- Johannes Gutenberg University Mainz, 55099 Mainz, Germany
| | - L. Nies
- CERN, Meyrin, 1211 Geneva Switzerland
- University of Greifswald, 17489 Greifswald, Germany
| | | | - M. Athanasakis-Kaklamanakis
- CERN, Meyrin, 1211 Geneva Switzerland
- KU Leuven, 3001 Leuven, Belgium
- Present Address: Centre for Cold Matter, Imperial College London, SW7 2AZ London, United Kingdom
| | - T. E. Cocolios
- CERN, Meyrin, 1211 Geneva Switzerland
- KU Leuven, 3001 Leuven, Belgium
| | - P. Fischer
- University of Greifswald, 17489 Greifswald, Germany
| | - P. F. Giesel
- University of Greifswald, 17489 Greifswald, Germany
| | | | - U. Köster
- CERN, Meyrin, 1211 Geneva Switzerland
- Institut Laue-Langevin, 38000 Grenoble, France
| | - D. Lange
- Max Planck Institute for Nuclear Physics, 69221 Heidelberg, Germany
| | - M. Mougeot
- CERN, Meyrin, 1211 Geneva Switzerland
- Present Address: Univeristy of Jyväskylä, 40014 Jyväskylä, Finland
| | - J. Reilly
- University of Manchester, Manchester, M13 9PL UK
| | - M. Schlaich
- Technical University of Darmstadt, 64289 Darmstadt, Germany
| | - Ch. Schweiger
- CERN, Meyrin, 1211 Geneva Switzerland
- Max Planck Institute for Nuclear Physics, 69221 Heidelberg, Germany
| | | | - F. Wienholtz
- Technical University of Darmstadt, 64289 Darmstadt, Germany
| | | | - Ch. E. Düllmann
- Johannes Gutenberg University Mainz, 55099 Mainz, Germany
- GSI Helmholtzzentrum für Schwerionenforschung, 64291 Darmstadt, Germany
- Helmholtz Institute Mainz, 55099 Mainz, Germany
| | - S. Rothe
- CERN, Meyrin, 1211 Geneva Switzerland
| |
Collapse
|
21
|
Zubenko AD, Pashanova AV, Mosaleva SP, Chernikova EY, Karnoukhova VA, Fedyanin IV, Egorova BV, Shchukina AA, Fedorov YV, Fedorova OA. Double-Armed 18- and 21-Membered Macrocycles as Potential Chelators for Lead and Bismuth Radiopharmaceuticals. Inorg Chem 2024; 63:21652-21669. [PMID: 39475211 DOI: 10.1021/acs.inorgchem.4c03116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2024]
Abstract
With increasing clinical applications and interest in targeted alpha therapy, there is growing interest in developing alternative chelating agents for [212Pb]Pb2+ and [212/213Bi]Bi3+ that exhibit rapid radiolabeling kinetics and kinetic inertness. Herein we report the synthesis and detailed investigation of diacetate and dipicolinate 18- and 21-membered macrocyclic chelators BADA-18, BADA-21, BADPA-18, and BADPA-21 for the complexation of Pb2+ and Bi3+ ions with potential use in the preparation of radiopharmaceuticals. The formation of mononuclear complexes was established by using ESI-mass spectrometry, and their stability constants were determined by potentiometric titration. A thorough study of the structure of the metal complexes was carried out by using X-ray diffraction and NMR spectroscopy. It was shown how the stability of the complex is influenced by an increase in the size of the macrocycle, the replacement of acetate arms with picolinate ones, the rigidity of the ligand, as well as the type of conformation (syn- or anti-) of the metal complex. The new ligands were radiolabeled with [210Pb]Pb2+ and [207Bi]Bi3+, and the in vitro stability of the resulting complexes in a competitive environment of serum and biologically significant metal ions was assessed. Rapid complex formation in 1-2 min at room temperature, as well as the high kinetic inertness of the complexes Pb(BADPA-18) and Bi(BADPA-18) in biological media, demonstrate its potential for use in targeted radionuclide therapy.
Collapse
Affiliation(s)
- Anastasia D Zubenko
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 st. Vavilova, 28, GSP-1, Moscow 119991, Russian Federation
| | - Anna V Pashanova
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 st. Vavilova, 28, GSP-1, Moscow 119991, Russian Federation
| | - Sofia P Mosaleva
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 st. Vavilova, 28, GSP-1, Moscow 119991, Russian Federation
- D. I. Mendeleev University of Chemistry and Technology of Russia, 125047 Miusskaya sqr., 9, Moscow 119991, Russian Federation
| | - Ekaterina Y Chernikova
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 st. Vavilova, 28, GSP-1, Moscow 119991, Russian Federation
| | - Valentina A Karnoukhova
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 st. Vavilova, 28, GSP-1, Moscow 119991, Russian Federation
| | - Ivan V Fedyanin
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 st. Vavilova, 28, GSP-1, Moscow 119991, Russian Federation
| | - Bayirta V Egorova
- Lomonosov Moscow State University, 119991 Leninskie Gory, 1/3, Moscow 119991, Russian Federation
| | - Anna A Shchukina
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 st. Vavilova, 28, GSP-1, Moscow 119991, Russian Federation
| | - Yury V Fedorov
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 st. Vavilova, 28, GSP-1, Moscow 119991, Russian Federation
| | - Olga A Fedorova
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 st. Vavilova, 28, GSP-1, Moscow 119991, Russian Federation
- D. I. Mendeleev University of Chemistry and Technology of Russia, 125047 Miusskaya sqr., 9, Moscow 119991, Russian Federation
| |
Collapse
|
22
|
Le Saux L, Haddad F, Gestin JF, Eychenne R, Guérard F. Sydnone-based prosthetic groups for radioiodination. Bioorg Med Chem 2024; 113:117904. [PMID: 39265508 DOI: 10.1016/j.bmc.2024.117904] [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: 06/25/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 09/14/2024]
Abstract
The potential of Strained-Promoted Sydnone-Alkyne Cycloaddition (SPSAC) for radioiodination was evaluated with model cyclooctyne-conjugated peptides. Starting with a series of sydnones with varying N3 and C4 substitution, a preliminary kinetic study with non-radioactive iodinated compounds highlighted the superiority of an arylsydnone substituted by a chlorine atom in C4 position. Interestingly, reaction rate up to 11 times higher than using an azide was achieved with the best system. Access to 125I-labelled sydnones was granted with high efficiency from arylboronic acid precursors by copper catalyzed nucleophilic substitution. Application of SPSAC on the model peptide in radiotracer conditions showed the same trend than in non-radioactive kinetic study and complete reactions could be achieved within less than an hour for the best systems. These results are favorable for use in the production of radiopharmaceuticals with heavy halogens and increase the diversity of available bioorthogonal reaction for nuclear imaging and therapy.
Collapse
Affiliation(s)
- Ludovic Le Saux
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA Nantes, France; Groupement d'Intérêt Public ARRONAX, 1 rue Aronnax, F-44817 Saint-Herblain, France
| | - Ferid Haddad
- Groupement d'Intérêt Public ARRONAX, 1 rue Aronnax, F-44817 Saint-Herblain, France; Laboratoire Subatech, IN2P3-CNRS, IMT Atlantique, Nantes Université, 4 rue Alfred Kastler, F-44307 Nantes, France
| | | | - Romain Eychenne
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA Nantes, France; Groupement d'Intérêt Public ARRONAX, 1 rue Aronnax, F-44817 Saint-Herblain, France.
| | - François Guérard
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA Nantes, France.
| |
Collapse
|
23
|
Abe K, Watabe T, Kaneda-Nakashima K, Shirakami Y, Kadonaga Y, Naka S, Ooe K, Toyoshima A, Giesel F, Usui T, Masunaga N, Mishima C, Tsukabe M, Yoshinami T, Sota Y, Miyake T, Tanei T, Shimoda M, Shimazu K. Evaluation of Targeted Alpha Therapy Using [ 211At]FAPI1 in Triple-Negative Breast Cancer Xenograft Models. Int J Mol Sci 2024; 25:11567. [PMID: 39519118 PMCID: PMC11547022 DOI: 10.3390/ijms252111567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/14/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Triple-negative breast cancer (TNBC) presents limited therapeutic options and is associated with poor prognosis. Early detection and the development of novel therapeutic agents are therefore imperative. Fibroblast activation protein (FAP) is a membrane protein expressed on cancer-associated fibroblasts (CAFs) that plays an essential role in TNBC proliferation, migration, and invasion. Consequently, it is hypothesized that the Astatine (211At)-labeled FAP inhibitor (FAPI) selectively exerts anti-tumor effects through alpha-particle emission. In this study, we aimed to assess its theranostic capabilities by integrating [18F]FAPI-74 PET imaging with targeted alpha therapy using [211At]FAPI1 in TNBC models. Mice xenografts were established by transplanting MDA-MB-231 and HT1080 cells (control). As a parallel diagnostic method, [18F]FAPI-74 was administered for PET imaging to validate FAP expression. A single dose of [211At]FAPI1 (1.04 ± 0.10 MBq) was administered to evaluate the therapeutic efficacy. [18F]FAPI-74 exhibited high accumulation in MDA-MB-231 xenografts, and FAP expression was pathologically confirmed via immunostaining. The group that received [211At]FAPI1 (n = 11) demonstrated a significantly enhanced anti-tumor effect compared with the control group (n = 7) (p = 0.002). In conclusion, [18F]FAPI-74 PET imaging was successfully used to diagnose FAP expression, and as [211At]FAPI1 showed promising therapeutic efficacy in TNBC models, it is expected to be a viable therapeutic option.
Collapse
Affiliation(s)
- Kaori Abe
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (K.A.)
| | - Tadashi Watabe
- Department of Radiology, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan
- Institute for Radiation Sciences, Osaka University, Suita 565-0871, Japan
| | - Kazuko Kaneda-Nakashima
- Institute for Radiation Sciences, Osaka University, Suita 565-0871, Japan
- Core for Medicine and Science Collaborative Research and Education, Forefront Research Center, Graduate School of Medicine, Osaka University, Suita 560-0043, Japan
| | | | - Yuichiro Kadonaga
- Institute for Radiation Sciences, Osaka University, Suita 565-0871, Japan
| | - Sadahiro Naka
- Department of Pharmacy, Osaka University Hospital, Suita 565-0871, Japan
| | - Kazuhiro Ooe
- Institute for Radiation Sciences, Osaka University, Suita 565-0871, Japan
| | - Atsushi Toyoshima
- Institute for Radiation Sciences, Osaka University, Suita 565-0871, Japan
| | - Frederik Giesel
- Institute for Radiation Sciences, Osaka University, Suita 565-0871, Japan
- Department of Nuclear Medicine, University Hospital Duesseldorf, Medical Faculty, Heinrich-Heine-University, 40225 Duesseldorf, Germany
| | - Takeshi Usui
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (K.A.)
| | - Nanae Masunaga
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (K.A.)
| | - Chieko Mishima
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (K.A.)
| | - Masami Tsukabe
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (K.A.)
| | - Tetsuhiro Yoshinami
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (K.A.)
| | - Yoshiaki Sota
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (K.A.)
| | - Tomohiro Miyake
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (K.A.)
| | - Tomonori Tanei
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (K.A.)
| | - Masafumi Shimoda
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (K.A.)
| | - Kenzo Shimazu
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (K.A.)
| |
Collapse
|
24
|
Vanermen M, Ligeour M, Oliveira MC, Gestin JF, Elvas F, Navarro L, Guérard F. Astatine-211 radiolabelling chemistry: from basics to advanced biological applications. EJNMMI Radiopharm Chem 2024; 9:69. [PMID: 39365487 PMCID: PMC11452365 DOI: 10.1186/s41181-024-00298-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 09/17/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND 211At-radiopharmaceuticals are currently the subject of growing studies for targeted alpha therapy of cancers, which leads to the widening of the scope of the targeting vectors, from small molecules to peptides and proteins. This has prompted, during the past decade, to a renewed interest in developing novel 211At-labelling approaches and novel prosthetic groups to address the diverse scenarios and to reach improved efficiency and robustness of procedures as well as an appropriate in vivo stability of the label. MAIN BODY Translated from the well-known (radio)iodine chemistry, the long preferred electrophilic astatodemetallation using trialkylaryltin precursors is now complemented by new approaches using electrophilic or nucleophilic At. Alternatives to the astatoaryl moiety have been proposed to improve labelling stability, and the range of prosthetic groups available to label proteins has expanded. CONCLUSION In this report, we cover the evolution of radiolabelling chemistry, from the initial strategies developed in the late 1970's to the most recent findings.
Collapse
Affiliation(s)
- Maarten Vanermen
- Molecular Imaging and Radiology (MIRA), University of Antwerp, Wilrijk, Belgium
| | - Mathilde Ligeour
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, Nantes, France
| | - Maria-Cristina Oliveira
- Departamento de Engenharia e Ciências Nucleares and Centro de Ciências e Tecnologias Nucleares, IST, Universidade de Lisboa, Estrada Nacional 10 (km 139,7), 2695-066, Bobadela LRS, Portugal
| | | | - Filipe Elvas
- Molecular Imaging and Radiology (MIRA), University of Antwerp, Wilrijk, Belgium
| | | | - François Guérard
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, Nantes, France.
| |
Collapse
|
25
|
Alkhani L, Luce JP, Mínguez Gabiña P, Roeske JC. Calculation of alpha particle single-event spectra using a neural network. Front Oncol 2024; 14:1394671. [PMID: 39416463 PMCID: PMC11480074 DOI: 10.3389/fonc.2024.1394671] [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: 03/01/2024] [Accepted: 08/30/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction A neural network was trained to accurately predict the entire single-event specific energy spectra for use in alpha-particle microdosimetry calculations. Methods The network consisted of 4 inputs and 21 outputs and was trained on data calculated using Monte Carlo simulation where input parameters originated both from previously published data as well as randomly generated parameters that fell within a target range. The 4 inputs consisted of the source-target configuration (consisting of both cells in suspension and in tissue-like geometries), alpha particle energy (3.97-8.78 MeV), nuclei radius (2-10 μm), and cell radius (2.5-20 μm). The 21 output values consisted of the maximum specific energy (zmax), and 20 values of the single-event spectra, which were expressed as fractional values of zmax. The neural network consisted of two hidden layers with 10 and 26 nodes, respectively, with the loss function characterized as the mean square error (MSE) between the actual and predicted values for zmax and the spectral outputs. Results For the final network, the root mean square error (RMSE) values of zmax for training, validation and testing were 1.57 x10-2, 1.51 x 10-2 and 1.35 x 10-2, respectively. Similarly, the RMSE values of the spectral outputs were 0.201, 0.175 and 0.199, respectively. The correlation coefficient, R2, was > 0.98 between actual and predicted values from the neural network. Discussion In summary, the network was able to accurately reproduce alpha-particle single-event spectra for a wide range of source-target geometries.
Collapse
Affiliation(s)
- Layth Alkhani
- Department of Bioengineering, Stanford University, Stanford, CA, United States
| | - Jason P. Luce
- Department of Radiation Oncology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
| | - Pablo Mínguez Gabiña
- Department of Medical Physics and Radiation Protection, Gurutzeta/Cruces University Hospital, Biocruces Health Research Institute, Barakaldo, Spain
| | - John C. Roeske
- Department of Radiation Oncology, Stritch School of Medicine, Loyola University Chicago, Maywood, IL, United States
| |
Collapse
|
26
|
Shah A, Dabhade A, Bharadia H, Parekh PS, Yadav MR, Chorawala MR. Navigating the landscape of theranostics in nuclear medicine: current practice and future prospects. Z NATURFORSCH C 2024; 79:235-266. [PMID: 38807355 DOI: 10.1515/znc-2024-0043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 05/10/2024] [Indexed: 05/30/2024]
Abstract
Theranostics refers to the combination of diagnostic biomarkers with therapeutic agents that share a specific target expressed by diseased cells and tissues. Nuclear medicine is an exciting component explored for its applicability in theranostic concepts in clinical and research investigations. Nuclear theranostics is based on the employment of radioactive compounds delivering ionizing radiation to diagnose and manage certain diseases employing binding with specifically expressed targets. In the realm of personalized medicine, nuclear theranostics stands as a beacon of potential, potentially revolutionizing disease management. Studies exploring the theranostic profile of radioactive compounds have been presented in this review along with a detailed explanation of radioactive compounds and their theranostic applicability in several diseases. It furnishes insights into their applicability across diverse diseases, elucidating the intricate interplay between these compounds and disease pathologies. Light is shed on the important milestones of nuclear theranostics beginning with radioiodine therapy in thyroid carcinomas, MIBG labelled with iodine in neuroblastoma, and several others. Our perspectives have been put forth regarding the most important theranostic agents along with emerging trends and prospects.
Collapse
Affiliation(s)
- Aayushi Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Akshada Dabhade
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Hetvi Bharadia
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Priyajeet S Parekh
- AV Pharma LLC, 1545 University Blvd N Ste A, Jacksonville, FL, 32211, USA
| | - Mayur R Yadav
- Department of Pharmacy Practice and Administration, Western University of Health Science, 309 E Second St, Pomona, CA, 91766, USA
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| |
Collapse
|
27
|
Aneheim E, Bäck T, Jensen H, Palm S, Lindegren S. A proposed production method for astatinated (At-211) Trastuzumab for use in a Phase I clinical trial. PLoS One 2024; 19:e0307543. [PMID: 39316568 PMCID: PMC11421776 DOI: 10.1371/journal.pone.0307543] [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: 10/16/2023] [Accepted: 07/08/2024] [Indexed: 09/26/2024] Open
Abstract
Astatine-211 is a nuclide with a short half-life of 7.2 h, that show promise for targeted alpha therapy of disseminated cancer. Despite nuclide production being straight-forward using a medium energy cyclotron and an uncomplicated target, not many cyclotrons are currently producing the nuclide. In this work we propose a stream-lined method to produce astatine labelled antibodies that enable production of clinical doses at other sites, remote from the nuclide producing cyclotron. Preconjugating the antibody prior to labelling, quick and efficient astatine recovery from the irradiated target in combination with optimized nuclide production logistics and an efficient synthesis for labelling are all key components to produce a clinical amount, > 300 MBq, of astatinated Trastuzumab.
Collapse
Affiliation(s)
- Emma Aneheim
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Oncology, Region Västra Götaland, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Tom Bäck
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Holger Jensen
- Department of Clinical Physiology and Nuclear Medicine, Cyclotron and Radiochemistry unit, Copenhagen University Hospital, Copenhagen, Denmark
| | - Stig Palm
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Sture Lindegren
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
28
|
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
|
29
|
Ramdhani D, Watabe H, Hardianto A, Janitra RS. Complexation of 3p- C-NETA with radiometal ions: A density functional theory study for targeted radioimmunotherapy. Heliyon 2024; 10:e34875. [PMID: 39144950 PMCID: PMC11320446 DOI: 10.1016/j.heliyon.2024.e34875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 07/13/2024] [Accepted: 07/17/2024] [Indexed: 08/16/2024] Open
Abstract
Bifunctional chelators (BFCs) are vital in the design of effective radiopharmaceuticals, as they are able to bind to both a radiometal ion and a targeting vector. The 3p-C-NETA or 4-[2-(bis-carboxy-methylamino)-5-(4-nitrophenyl)-entyl])-7-carboxymethyl-[1,4,7]tri-azonan-1-yl acetic acid is a novel and promising BFC, developed for diagnostic and therapeutic purposes. The binding affinity between the BFC and radiometal ion significantly impacts their effectiveness. Predicting the equilibrium constants for the formation of 1:1 radiometals/chelator complexes (log K1 values) is crucial for designing BFCs with improved affinity and selectivity for radiometals. The purpose of this study is to evaluate the complexation of Ga3+, Tb3+, Bi3+, and Ac3+ radiometal ions with 3p-C-NETA using density functional theory (B3LYP and M06-HF functional) and 6-311G(d)/SDD basis sets, where the 1,4,7,10-tetrazacyclodecane-1,4,7,10-tetracetic acid (DOTA) was employed as a benchmark. Formation of the [Ac3+(3p-C-NETA)(H2O)]- complexes is predicted to be markedly less stable compared to the other complexes, exhibiting the lowest chemical hardness and the highest chemical softness. Additionally, the chelation stability of the complexes is mainly determined by ligand-ion and ion-water interactions, which depend on the atomic charge and atomic radius of the metal ion.
Collapse
Affiliation(s)
- Danni Ramdhani
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
- Division of Radiation Protection and Safety Control, Cyclotron and Radioisotope Center (CYRIC), Tohoku University, Sendai, Japan
| | - Hiroshi Watabe
- Division of Radiation Protection and Safety Control, Cyclotron and Radioisotope Center (CYRIC), Tohoku University, Sendai, Japan
| | - Ari Hardianto
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Sumedang, Indonesia
| | - Regaputra S. Janitra
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Sumedang, Indonesia
| |
Collapse
|
30
|
Brühlmann SA, Walther M, Blei MK, Mamat C, Kopka K, Freudenberg R, Kreller M. Scalability study on [ 133La]LaCl 3 production with a focus on potential clinical applications. EJNMMI Radiopharm Chem 2024; 9:60. [PMID: 39147960 PMCID: PMC11327230 DOI: 10.1186/s41181-024-00292-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/05/2024] [Indexed: 08/17/2024] Open
Abstract
BACKGROUND In recent years, targeted alpha therapy has gained importance in the clinics, and in particular, the alpha-emitter 225Ac plays a fundamental role in this clinical development. Nevertheless, depending on the chelating system no real diagnostic alternative has been established which shares similar chemical properties with this alpha-emitting radionuclide. In fact, the race to launch a diagnostic radionuclide to form a matched pair with 225Ac is still open, and 133La features attractive radiation properties to claim this place. However, in order to enable its translation into clinical use, upscaling of the production of this PET radionuclide is needed. RESULTS A study on optimal irradiation parameters, separation conditions and an exhaustive product characterization was carried out. In this framework, a proton irradiation of 2 h, 60 µA and 18.7 MeV produced 133La activities of up to 10.7 GBq at end of bombardment. In addition, the performance of four different chromatographic resins were tested and two optimized purification methods presented, taking approximately 20 min with a 133La recovery efficiencies of over 98%, decay corrected. High radionuclide purity and apparent molar activity was proved, of over 99.5% and 120 GBq/µmol, respectively, at end of purification. Furthermore, quantitative complexation of PSMA-617 and mcp-M-PSMA were obtained with molar activities up to 80 GBq/µmol. In addition, both 133La-radioconjugates offered high stability in serum, of over (98.5 ± 0.3)% and (99.20 ± 0.08)%, respectively, for up to 24 h. A first dosimetry estimation was also performed and it was calculated that an 133La application for imaging with between 350 and 750 MBq would only have an effective dose of 2.1-4.4 mSv, which is comparable to that of 18F and 68Ga based radiopharmaceuticals. CONCLUSIONS In this article we present an overarching study on 133La production, from the radiation parameters optimization to a clinical dose estimation. Lanthanum-133 activities in the GBq range could be produced, formulated as [133La]LaCl3 with high quality regarding radiolabeling and radionuclide purity. We believe that increasing the 133La availability will further promote the development of radiopharmaceuticals based on macropa or other chelators suitable for 225Ac.
Collapse
Affiliation(s)
- Santiago Andrés Brühlmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, 01062, Dresden, Germany
| | - Martin Walther
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany.
| | - Magdalena Kerstin Blei
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, 01062, Dresden, Germany
| | - Constantin Mamat
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, 01062, Dresden, Germany
| | - Klaus Kopka
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, 01062, Dresden, Germany
- National Center for Tumor Diseases (NCT) Dresden, University Hospital Carl Gustav Carus, Fetscherstraße 74, 01307, Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Robert Freudenberg
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Fetscherstraße 74, 01307, Dresden, Germany
| | - Martin Kreller
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
| |
Collapse
|
31
|
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
|
32
|
Hartmann S, Taubner K, Vogt T, Meisenberg O, Schkade UK, Steyer C, Meckel M, Kesenheimer C. Actinium-225 as an example for monitoring of internal exposure of occupational intakes of radionuclides in face of new nuclear-medical applications for short-lived alpha emitting particles. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2024; 63:385-394. [PMID: 39031187 PMCID: PMC11341604 DOI: 10.1007/s00411-024-01081-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 07/02/2024] [Indexed: 07/22/2024]
Abstract
Monitoring of internal exposure to short-lived alpha-emitting radionuclides such as actinium-225 (225Ac), which are becoming increasingly important in nuclear medicine, plays an important role in the radiation protection of occupationally exposed persons. After having tested gamma spectrometry, liquid scintillation counting and alpha spectrometry for monitoring of internal exposure, the focus of the present study was on solid phase extraction of 225Ac from urine in combination with alpha spectrometry. The development of the method was based on recent findings from the literature on this topic. The method was used in a pilot phase to monitor internal exposure of four workers who were directly or indirectly involved in the manufacture and/or use of 225Ac. The monitoring protocol allowed a relatively short 24-hour urine sample analysis with excellent recovery of the internal standard, but it did not allow for a detection limit of less than 1 mBq nor a sufficient yield of 225Ac. Based on these results it is concluded that an in vitro excretion analysis alone is not appropriate for monitoring internal exposure to 225Ac. Instead, different radiation monitoring techniques have to be combined to ensure the radiation protection of employees.
Collapse
Affiliation(s)
- Sven Hartmann
- Federal Office for Radiation Protection, Medical and Occupational Radiation Protection, Incorporation Monitoring MB 5, Berlin, Germany.
| | - Kerstin Taubner
- Federal Office for Radiation Protection, Medical and Occupational Radiation Protection, Incorporation Monitoring MB 5, Berlin, Germany
| | - Tobias Vogt
- Federal Office for Radiation Protection, Medical and Occupational Radiation Protection, Incorporation Monitoring MB 5, Berlin, Germany
| | - Oliver Meisenberg
- Federal Office for Radiation Protection, Medical and Occupational Radiation Protection, Incorporation Monitoring MB 5, Berlin, Germany
| | - Uwe-Karsten Schkade
- Federal Office for Radiation Protection, Environmental Radioactivity, Dosimetry and Spectrometry UR 5, Berlin, Germany
| | | | | | | |
Collapse
|
33
|
Meisenberg O. Reduction of detection limits in monitoring of internal exposures by a combined evaluation of emissions and spectra. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2024; 63:371-383. [PMID: 38980362 PMCID: PMC11341640 DOI: 10.1007/s00411-024-01079-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 06/21/2024] [Indexed: 07/10/2024]
Abstract
Routine monitoring of internal exposures requires the detection of effective doses of at most 1 mSv per calendar year. For some radionuclides, this requirement cannot be satisfied by a conventional evaluation of the spectra that are gained in alpha or gamma spectrometry. However, since several measurements are conducted per calendar year on a regular basis, a combined evaluation of measurements, i.e. the evaluation of sum spectra, is possible. Additionally, radionuclides that feature several emissions of alpha or gamma radiation allow a combined evaluation of their emissions. Both methods can lead to significantly smaller detection limits as compared to a separate evaluation of spectra in many cases. However, the variation of parameters that influence the evaluation such as the measurement efficiency, abundance and chemical yield requires specific calculations and treatments of the spectra as well as a manipulation of the channel contents: In a combination of emissions, energy regions are summed and evaluated with a combined efficiency that is weighted by the abundances. In a combination of spectra, the channel contents must be scaled by the ratio of the calibration factors before the summation of the spectra. In the routine monitoring of short-lived radionuclides that feature a variety of emissions such as 225Ac, these combinations are particularly effective in reducing the detectable annual effective dose. For alpha spectrometry of 225Ac, both methods applied together can lead to a detectable effective dose of about 1 mSv per year as compared to a dose of about 90 mSv with a conventional separate evaluation.
Collapse
Affiliation(s)
- Oliver Meisenberg
- Federal Office for Radiation Protection (BfS), Medical and Occupational Radiation Protection, 85764, Oberschleißheim, Germany.
| |
Collapse
|
34
|
Koniar H, Wharton L, Ingham A, Rodríguez-Rodríguez C, Kunz P, Radchenko V, Yang H, Rahmim A, Uribe C, Schaffer P. In vivoquantitative SPECT imaging of actinium-226: feasibility and proof-of-concept. Phys Med Biol 2024; 69:155003. [PMID: 38925140 DOI: 10.1088/1361-6560/ad5c37] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 06/26/2024] [Indexed: 06/28/2024]
Abstract
Objective.225Ac radiopharmaceuticals have tremendous potential for targeted alpha therapy, however,225Ac (t1/2= 9.9 d) lacks direct gamma emissions forin vivoimaging.226Ac (t1/2= 29.4 h) is a promising element-equivalent matched diagnostic radionuclide for preclinical evaluation of225Ac radiopharmaceuticals.226Ac has two gamma emissions (158 keV and 230 keV) suitable for SPECT imaging. This work is the first feasibility study forin vivoquantitative226Ac SPECT imaging and validation of activity estimation.Approach.226Ac was produced at TRIUMF (Vancouver, Canada) with its Isotope Separator and Accelerator (ISAC) facility. [226Ac]Ac3+was radiolabelled with the bioconjugate crown-TATE developed for therapeutic targeting of neuroendocrine tumours. Mice with AR42J tumour xenografts were injected with either 2 MBq of [226Ac]Ac-crown-TATE or 4 MBq of free [226Ac]Ac3+activity and were scanned at 1, 2.5, 5, and 24 h post injection in a preclinical microSPECT/CT. Quantitative SPECT images were reconstructed from the 158 keV and 230 keV photopeaks with attenuation, background, and scatter corrections. Image-based226Ac activity measurements were assessed from volumes of interest within tumours and organs of interest. Imaging data was compared withex vivobiodistribution measured via gamma counter.Main results. We present, to the best of our knowledge, the first everin vivoquantitative SPECT images of226Ac activity distributions. Time-activity curves derived from SPECT images quantify thein vivobiodistribution of [226Ac]Ac-crown-TATE and free [226Ac]Ac3+activity. Image-based activity measurements in the tumours and organs of interest corresponded well withex vivobiodistribution measurements.Significance. Here in, we established the feasibility ofin vivo226Ac quantitative SPECT imaging for accurate measurement of actinium biodistribution in a preclinical model. This imaging method could facilitate more efficient development of novel actinium labelled compounds by providing accurate quantitativein vivopharmacokinetic information essential for estimating toxicities, dosimetry, and therapeutic potency.
Collapse
Affiliation(s)
- Helena Koniar
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
- Department of Physics and Astronomy, University of British Columbia, 6224 Agricultural Road, Vancouver, BC V6T 1Z1, Canada
| | - Luke Wharton
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
| | - Aidan Ingham
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
| | - Cristina Rodríguez-Rodríguez
- Department of Physics and Astronomy, University of British Columbia, 6224 Agricultural Road, Vancouver, BC V6T 1Z1, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC V6T 1Z3, Canada
| | - Peter Kunz
- Accelerator Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
| | - Valery Radchenko
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1Z1, Canada
| | - Hua Yang
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
| | - Arman Rahmim
- Department of Physics and Astronomy, University of British Columbia, 6224 Agricultural Road, Vancouver, BC V6T 1Z1, Canada
- Department of Radiology, University of British Columbia, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
- Department of Integrative Oncology, BC Cancer Research Centre, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada
| | - Carlos Uribe
- Department of Radiology, University of British Columbia, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
- Department of Integrative Oncology, BC Cancer Research Centre, 675 W 10th Ave, Vancouver, BC, V5Z 1L3, Canada
- Department of Functional Imaging, BC Cancer, 600 West 10th Avenue, Vancouver, BC, V5Z 4E6, Canada
| | - Paul Schaffer
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
- Department of Chemistry, Simon Fraser University, 8888 University Drive, Burnaby, BC V5A 1S6, Canada
- Department of Radiology, University of British Columbia, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
| |
Collapse
|
35
|
Goswami M, Toro-González M, Moon J, Davern S. Precision Atomistic Structures of Actinium-/Radium-/Barium-Doped Lanthanide Nanoconstructs for Radiotherapeutic Applications. ACS NANO 2024; 18:16577-16588. [PMID: 38885179 PMCID: PMC11223473 DOI: 10.1021/acsnano.3c13213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 04/29/2024] [Accepted: 05/07/2024] [Indexed: 06/20/2024]
Abstract
Lanthanide vanadate (LnVO4) nanoconstructs have generated considerable interest in radiotherapeutic applications as a medium for nanoscale-targeted drug delivery. For cancer treatment, LnVO4 nanoconstructs have shown promise in encapsulating and retaining radionuclides that emit alpha-particles. In this work, we examined the structure formation of LnVO4 nanoconstructs doped with actinium (Ac) and radium (Ra), both experimentally and using large-scale atomistic molecular dynamics simulations. LnVO4 nanoconstructs were synthesized via a precipitation method in aqueous media. The reaction conditions and elemental compositions were varied to control the structure, fluorescence properties, and size distribution of the LnVO4 nanoconstructs. LnVO4 nanoconstructs were characterized by X-ray diffraction, Raman spectroscopy, and fluorescence spectroscopy. Molecular dynamics simulations were performed to obtain a fundamental understanding of the structure-property relationship between radionuclides and LnVO4 nanoconstructs at the atomic length scale. Molecular dynamics simulations with well-established force field (FF) parameters show that Ra atoms tend to distribute across the nanoconstructs' surface in a broader coordination shell, while the Ac atoms are arranged inside a smaller coordination shell within the nanocluster. The Ba atoms prefer to self-assemble around the surface. These theoretical/simulation predictions of the atomistic structures and an understanding of the relationship between radionuclides and LnVO4 nanoconstructs at the atomic scale are important because they provide design principles for the future development of nanoconstructs for targeted radionuclide delivery.
Collapse
Affiliation(s)
- Monojoy Goswami
- Chemical
Sciences Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee 37831, United States
| | - Miguel Toro-González
- Radioisotope
Science & Technology Division, Oak Ridge
National Laboratory, Oak Ridge, Tennessee 37831, United States
| | - Jisue Moon
- Radioisotope
Science & Technology Division, Oak Ridge
National Laboratory, Oak Ridge, Tennessee 37831, United States
| | - Sandra Davern
- Radioisotope
Science & Technology Division, Oak Ridge
National Laboratory, Oak Ridge, Tennessee 37831, United States
| |
Collapse
|
36
|
Roncali L, Marionneau-Lambot S, Roy C, Eychenne R, Gouard S, Avril S, Chouin N, Riou J, Allard M, Rousseau A, Guérard F, Hindré F, Chérel M, Garcion E. Brain intratumoural astatine-211 radiotherapy targeting syndecan-1 leads to durable glioblastoma remission and immune memory in female mice. EBioMedicine 2024; 105:105202. [PMID: 38905749 PMCID: PMC11246004 DOI: 10.1016/j.ebiom.2024.105202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/31/2024] [Accepted: 05/31/2024] [Indexed: 06/23/2024] Open
Abstract
BACKGROUND Glioblastoma (GB), the most aggressive brain cancer, remains a critical clinical challenge due to its resistance to conventional treatments. Here, we introduce a locoregional targeted-α-therapy (TAT) with the rat monoclonal antibody 9E7.4 targeting murine syndecan-1 (SDC1) coupled to the α-emitter radionuclide astatine-211 (211At-9E7.4). METHODS We orthotopically transplanted 50,000 GL261 cells of murine GB into the right striatum of syngeneic female C57BL/6JRj mice using stereotaxis. After MRI validation of tumour presence at day 11, TAT was injected at the same coordinates. Biodistribution, efficacy, toxicity, local and systemic responses were assessed following application of this protocol. The 9E7.4 monoclonal antibody was labelled with iodine-125 (125I) for biodistribution and with astatine-211 (211At) for the other experiments. FINDINGS The 211At-9E7.4 TAT demonstrated robust efficacy in reducing orthotopic tumours and achieved improved survival rates in the C57BL/6JRj model, reaching up to 70% with a minimal activity of 100 kBq. Targeting SDC1 ensured the cerebral retention of 211At over an optimal time window, enabling low-activity administration with a minimal toxicity profile. Moreover, TAT substantially reduced the occurrence of secondary tumours and provided resistance to new tumour development after contralateral rechallenge, mediated through the activation of central and effector memory T cells. INTERPRETATION The locoregional 211At-9E7.4 TAT stands as one of the most efficient TAT across all preclinical GB models. This study validates SDC1 as a pertinent therapeutic target for GB and underscores 211At-9E7.4 TAT as a promising advancement to improve the treatment and quality of life for patients with GB. FUNDING This work was funded by the French National Agency for Research (ANR) "France 2030 Investment Plan" Labex Iron [ANR-11-LABX-18-01], The SIRIC ILIAD [INCa-DGOS-INSERM-18011], the French program "Infrastructure d'Avenir en Biologie-Santé" (France Life Imaging) [ANR-11-INBS-0006], the PIA3 of the ANR, integrated to the "France 2030 Investment Plan" [ANR-21-RHUS-0012], and support from Inviscan SAS (Strasbourg, France). It was also related to: the ANR under the frame of EuroNanoMed III (project GLIOSILK) [ANR-19-ENM3-0003-01]; the "Région Pays-de-la-Loire" under the frame of the Target'In project; the "Ligue Nationale contre le Cancer" and the "Comité Départemental de Maine-et-Loire de la Ligue contre le Cancer" (CD49) under the frame of the FusTarG project and the "Tumour targeting, imaging and radio-therapies network" of the "Cancéropôle Grand-Ouest" (France). This work was also funded by the Institut National de la Santé et de la Recherche Médicale (INSERM), the University of Nantes, and the University of Angers.
Collapse
Affiliation(s)
- Loris Roncali
- Université d'Angers, INSERM, CNRS, CRCI(2)NA, F-49000, Angers, France; Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France
| | - Séverine Marionneau-Lambot
- Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France; CHU Nantes, Nantes Université, Service de médecine nucléaire, F-44000, Nantes, France; CIMA (Centre d'Imagerie Multimodale Appliquée), Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France
| | - Charlotte Roy
- Université d'Angers, INSERM, CNRS, CRCI(2)NA, F-49000, Angers, France; PRIMEX (Plateforme de Radiobiologie et d'Imageries Expérimentales), Université d'Angers, SFR 4208, F-49000, Angers, France
| | - Romain Eychenne
- Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France; GIP ARRONAX, F-44160, Saint-Herblain, France
| | - Sébastien Gouard
- Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France; CIMA (Centre d'Imagerie Multimodale Appliquée), Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France
| | - Sylvie Avril
- Université d'Angers, INSERM, CNRS, CRCI(2)NA, F-49000, Angers, France
| | - Nicolas Chouin
- Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France; ONIRIS, F-44000, Nantes, France
| | - Jérémie Riou
- CHU Angers, Université d'Angers, F-49000, Angers, France
| | - Mathilde Allard
- Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France
| | - Audrey Rousseau
- Université d'Angers, INSERM, CNRS, CRCI(2)NA, F-49000, Angers, France; CHU Angers, Université d'Angers, F-49000, Angers, France
| | - François Guérard
- Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France
| | - François Hindré
- Université d'Angers, INSERM, CNRS, CRCI(2)NA, F-49000, Angers, France; PRIMEX (Plateforme de Radiobiologie et d'Imageries Expérimentales), Université d'Angers, SFR 4208, F-49000, Angers, France
| | - Michel Chérel
- Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France; CIMA (Centre d'Imagerie Multimodale Appliquée), Nantes Université, INSERM, CNRS, CRCI(2)NA, F-44000, Nantes, France; Institut de Cancérologie de l'Ouest, Service de médecine nucléaire, F-44160, Saint-Herblain, France.
| | - Emmanuel Garcion
- Université d'Angers, INSERM, CNRS, CRCI(2)NA, F-49000, Angers, France; PRIMEX (Plateforme de Radiobiologie et d'Imageries Expérimentales), Université d'Angers, SFR 4208, F-49000, Angers, France; PACEM (Plateforme d'Analyse Cellulaire et Moléculaire), Université d'Angers, SFR 4208, F-49000, Angers, France.
| |
Collapse
|
37
|
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
|
38
|
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
|
39
|
Timperanza C, Jensen H, Hansson E, Bäck T, Lindegren S, Aneheim E. In vitro and in vivo evaluation of a tetrazine-conjugated poly-L-lysine effector molecule labeled with astatine-211. EJNMMI Radiopharm Chem 2024; 9:43. [PMID: 38775973 PMCID: PMC11111624 DOI: 10.1186/s41181-024-00273-z] [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: 01/25/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND A significant challenge in cancer therapy lies in eradicating hidden disseminated tumor cells. Within Nuclear Medicine, Targeted Alpha Therapy is a promising approach for cancer treatment tackling disseminated cancer. As tumor size decreases, alpha-particles gain prominence due to their high Linear Energy Transfer (LET) and short path length. Among alpha-particle emitters, 211At stands out with its 7.2 hour half-life and 100% alpha emission decay. However, optimizing the pharmacokinetics of radiopharmaceuticals with short lived radionuclides such as 211At is pivotal, and in this regard, pretargeting is a valuable tool. This method involves priming the tumor with a modified monoclonal antibody capable of binding both the tumor antigen and the radiolabeled carrier, termed the "effector molecule. This smaller, faster-clearing molecule improves efficacy. Utilizing the Diels Alder click reaction between Tetrazine (Tz) and Trans-cyclooctene (TCO), the Tz-substituted effector molecule combines seamlessly with the TCO-modified antibody. This study aims to evaluate the in vivo biodistribution of two Poly-L-Lysine-based effector molecule sizes (10 and 21 kDa), labelled with 211At, and the in vitro binding of the most favorable polymer size, in order to optimize the pretargeted radioimmunotherapy with 211At. RESULTS In vivo results favor the smaller polymer's biodistribution pattern over the larger one, which accumulates in organs like the liver and spleen. This is especially evident when comparing the biodistribution of the smaller polymer to a directly labelled monoclonal antibody. The smaller variant also shows rapid and efficient binding to SKOV-3 cells preloaded with TCO-modified Trastuzumab in vitro, emphasizing its potential. Both polymer sizes showed equal or better in vivo stability of the astatine-carbon bond compared to a monoclonal antibody labelled with the same prosthetic group. CONCLUSIONS Overall, the small Poly-L-Lysine-based effector molecule (10 kDa) holds the most promise for future research, exhibiting significantly lower uptake in the kidneys and spleen compared to the larger effector (21 kDa) while maintaining an in vivo stability of the astatine-carbon bond comparable to or better than intact antibodies. A proof of concept in vitro cell study demonstrates rapid reaction between the small astatinated effector and a TCO-labelled antibody, indicating the potential of this novel Poly-L-Lysine-based pretargeting system for further investigation in an in vivo tumor model.
Collapse
Affiliation(s)
- Chiara Timperanza
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 413 45, Sweden.
| | - Holger Jensen
- Department of Clinical Physiology and Nuclear Medicine, Cyclotron and Radiochemistry unit, Rigshospitalet, Blegdamsvej 9, Copenhagen, 2100, Denmark
| | - Ellinor Hansson
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 413 45, Sweden
- Atley Solutions AB, Gothenburg, 413 27, Sweden
| | - Tom Bäck
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 413 45, Sweden
| | - Sture Lindegren
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 413 45, Sweden
| | - Emma Aneheim
- Department of Medical Radiation Sciences, Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 413 45, Sweden
- Department of Oncology, Sahlgrenska University Hospital, Region Västra Götaland, Gothenburg, 413 45, Sweden
| |
Collapse
|
40
|
Sallam M, Nguyen NT, Sainsbury F, Kimizuka N, Muyldermans S, Benešová-Schäfer M. PSMA-targeted radiotheranostics in modern nuclear medicine: then, now, and what of the future? Theranostics 2024; 14:3043-3079. [PMID: 38855174 PMCID: PMC11155394 DOI: 10.7150/thno.92612] [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: 11/26/2023] [Accepted: 04/04/2024] [Indexed: 06/11/2024] Open
Abstract
In 1853, the perception of prostate cancer (PCa) as a rare ailment prevailed, was described by the eminent Londoner surgeon John Adams. Rapidly forward to 2018, the landscape dramatically altered. Currently, men face a one-in-nine lifetime risk of PCa, accentuated by improved diagnostic methods and an ageing population. With more than three million men in the United States alone grappling with this disease, the overall risk of succumbing to stands at one in 39. The intricate clinical and biological diversity of PCa poses serious challenges in terms of imaging, ongoing monitoring, and disease management. In the field of theranostics, diagnostic and therapeutic approaches that harmoniously merge targeted imaging with treatments are integrated. A pivotal player in this arena is radiotheranostics, employing radionuclides for both imaging and therapy, with prostate-specific membrane antigen (PSMA) at the forefront. Clinical milestones have been reached, including FDA- and/or EMA-approved PSMA-targeted radiodiagnostic agents, such as [18F]DCFPyL (PYLARIFY®, Lantheus Holdings), [18F]rhPSMA-7.3 (POSLUMA®, Blue Earth Diagnostics) and [68Ga]Ga-PSMA-11 (Locametz®, Novartis/ ILLUCCIX®, Telix Pharmaceuticals), as well as PSMA-targeted radiotherapeutic agents, such as [177Lu]Lu-PSMA-617 (Pluvicto®, Novartis). Concurrently, ligand-drug and immune therapies designed to target PSMA are being advanced through rigorous preclinical research and clinical trials. This review delves into the annals of PSMA-targeted radiotheranostics, exploring its historical evolution as a signature molecule in PCa management. We scrutinise its clinical ramifications, acknowledge its limitations, and peer into the avenues that need further exploration. In the crucible of scientific inquiry, we aim to illuminate the path toward a future where the enigma of PCa is deciphered and where its menace is met with precise and effective countermeasures. In the following sections, we discuss the intriguing terrain of PCa radiotheranostics through the lens of PSMA, with the fervent hope of advancing our understanding and enhancing clinical practice.
Collapse
Affiliation(s)
- Mohamed Sallam
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
- School of Environment and Science (ESC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
- Griffith Institute for Drug Discovery (GRIDD), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
| | - Nam-Trung Nguyen
- Queensland Micro- and Nanotechnology Centre (QMNC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
| | - Frank Sainsbury
- School of Environment and Science (ESC), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
- Griffith Institute for Drug Discovery (GRIDD), Griffith University, Nathan Campus, Nathan, QLD 4111, Australia
| | - Nobuo Kimizuka
- Department of Applied Chemistry, Graduate School of Engineering, Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka 819-0395, Japan
- Center for Molecular Systems (CMS), Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka 819-0395, Japan
- Research Center for Negative Emissions Technologies (K-NETs), Kyushu University, 744 Moto-oka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Serge Muyldermans
- Laboratory of Cellular and Molecular Immunology (CMIM), Vrije Universiteit Brussel, 1050 Brussels, Belgium
| | - Martina Benešová-Schäfer
- Research Group Molecular Biology of Systemic Radiotherapy, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| |
Collapse
|
41
|
Suzuki H, Kannaka K, Uehara T. Approaches to Reducing Normal Tissue Radiation from Radiolabeled Antibodies. Pharmaceuticals (Basel) 2024; 17:508. [PMID: 38675468 PMCID: PMC11053530 DOI: 10.3390/ph17040508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/02/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Radiolabeled antibodies are powerful tools for both imaging and therapy in the field of nuclear medicine. Radiolabeling methods that do not release radionuclides from parent antibodies are essential for radiolabeling antibodies, and practical radiolabeling protocols that provide high in vivo stability have been established for many radionuclides, with a few exceptions. However, several limitations remain, including undesirable side effects on the biodistribution profiles of antibodies. This review summarizes the numerous efforts made to tackle this problem and the recent advances, mainly in preclinical studies. These include pretargeting approaches, engineered antibody fragments and constructs, the secondary injection of clearing agents, and the insertion of metabolizable linkages. Finally, we discuss the potential of these approaches and their prospects for further clinical application.
Collapse
Affiliation(s)
- Hiroyuki Suzuki
- Laboratory of Molecular Imaging and Radiotherapy, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan; (K.K.); (T.U.)
| | | | | |
Collapse
|
42
|
Rybczyńska M, Sikorski A. Structural insight and in silico prediction of the pharmacokinetic parameters and toxicity of alkaline earth metal compounds strontium and barium with the non-steroidal anti-inflammatory drug nimesulide. Dalton Trans 2024; 53:6501-6506. [PMID: 38511607 DOI: 10.1039/d4dt00446a] [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: 03/22/2024]
Abstract
In the crystals of alkaline earth metal compounds strontium and barium with the non-steroidal anti-inflammatory drug nimesulide, the strontium cation is nine-coordinated with a distorted tricapped trigonal prismatic geometry TCTPR-9, whereas the ten-coordinated barium ion exhibits a distorted tetracapped trigonal prismatic geometry TCTPR-10.
Collapse
Affiliation(s)
| | - Artur Sikorski
- Faculty of Chemistry, University of Gdansk, W. Stwosza 63, 80-308 Gdansk, Poland.
| |
Collapse
|
43
|
Sathekge M, Morgenstern A. Advances in targeted alpha therapy of cancer. Eur J Nucl Med Mol Imaging 2024; 51:1205-1206. [PMID: 38376807 DOI: 10.1007/s00259-024-06658-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2024]
Affiliation(s)
- Mike Sathekge
- Steve Biko Academic Hospital, Nuclear Medicine Research Infrastructure (NuMeRI), University of Pretoria, Pretoria, South Africa.
| | | |
Collapse
|
44
|
Song Y, Zou J, Castellanos EA, Matsuura N, Ronald JA, Shuhendler A, Weber WA, Gilad AA, Müller C, Witney TH, Chen X. Theranostics - a sure cure for cancer after 100 years? Theranostics 2024; 14:2464-2488. [PMID: 38646648 PMCID: PMC11024861 DOI: 10.7150/thno.96675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/23/2024] Open
Abstract
Cancer has remained a formidable challenge in medicine and has claimed an enormous number of lives worldwide. Theranostics, combining diagnostic methods with personalized therapeutic approaches, shows huge potential to advance the battle against cancer. This review aims to provide an overview of theranostics in oncology: exploring its history, current advances, challenges, and prospects. We present the fundamental evolution of theranostics from radiotherapeutics, cellular therapeutics, and nanotherapeutics, showcasing critical milestones in the last decade. From the early concept of targeted drug delivery to the emergence of personalized medicine, theranostics has benefited from advances in imaging technologies, molecular biology, and nanomedicine. Furthermore, we emphasize pertinent illustrations showcasing that revolutionary strategies in cancer management enhance diagnostic accuracy and provide targeted therapies customized for individual patients, thereby facilitating the implementation of personalized medicine. Finally, we describe future perspectives on current challenges, emerging topics, and advances in the field.
Collapse
Affiliation(s)
- Yangmeihui Song
- Department of Nuclear Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, 81675, Germany
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 43000, China
| | - Jianhua Zou
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| | | | - Naomi Matsuura
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON, Canada
- Department of Materials Science & Engineering, University of Toronto, Toronto, ON, Canada
- Department of Medical Imaging, University of Toronto, Toronto, ON, Canada
| | - John A. Ronald
- Imaging Laboratories, Department of Medical Biophysics, Robarts Research Institute, University of Western Ontario, London, ON, Canada
- Lawson Health Research Institute, London, ON, Canada
| | - Adam Shuhendler
- University of Ottawa Heart Institute, Ottawa, ON, Canada
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Wolfgang A Weber
- Department of Nuclear Medicine, Klinikum Rechts der Isar, Technical University of Munich, Munich, 81675, Germany
| | - Assaf A. Gilad
- Department of Chemical Engineering and Materials Sciences, Michigan State University, East Lansing, MI, USA
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, USA
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences, Paul Scherrer Institute, 5232 Villigen-PSI, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zurich, Switzerland
| | - Timothy H. Witney
- School of Biomedical Engineering & Imaging Sciences, King's College London, London, UK
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
- Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore
| |
Collapse
|
45
|
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
|
46
|
Sathekge MM, Lawal IO, Bal C, Bruchertseifer F, Ballal S, Cardaci G, Davis C, Eiber M, Hekimsoy T, Knoesen O, Kratochwil C, Lenzo NP, Mahapane J, Maserumule LC, Mdlophane AH, Mokoala KMG, Ndlovu H, Pant V, Rathke H, Reed J, Sen IB, Singh A, Sood A, Tauber R, Thakral P, Yadav MP, Morgenstern A. Actinium-225-PSMA radioligand therapy of metastatic castration-resistant prostate cancer (WARMTH Act): a multicentre, retrospective study. Lancet Oncol 2024; 25:175-183. [PMID: 38218192 DOI: 10.1016/s1470-2045(23)00638-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/06/2023] [Accepted: 12/06/2023] [Indexed: 01/15/2024]
Abstract
BACKGROUND Actinium-225 (225Ac) prostate-specific membrane antigen (PSMA) radioligand therapy (RLT) is a novel therapy for metastatic castration-resistant prostate cancer (mCRPC). We aimed to report the safety and antitumour activity of 225Ac-PSMA RLT of mCRPC in a large cohort of patients treated at multiple centres across the world. METHODS This retrospective study included patients treated at seven centres in Australia, India, Germany, and South Africa. We pooled data of consecutive patients of any age and Eastern Cooperative Oncology Group performance status with histopathologically confirmed adenocarcinoma of the prostate who were treated with one or more cycles of 8 MBq 225Ac-PSMA RLT administered intravenously for mCRPC. Previous lines of mCRPC treatment included taxane-based chemotherapy, androgen-receptor-axis inhibitors, lutetium-177 (177Lu) PSMA RLT, and radium-223 dichloride. The primary outcomes were overall survival and progression-free survival. FINDINGS Between Jan 1, 2016, and May 31, 2023, 488 men with mCRPC received 1174 cycles of 225Ac-PSMA RLT (median two cycles, IQR 2-4). The mean age of the patients was 68·1 years (SD 8·8), and the median baseline prostate-specific antigen was 169·5 ng/mL (IQR 34·6-519·8). Previous lines of treatment were docetaxel in 324 (66%) patients, cabazitaxel in 103 (21%) patients, abiraterone in 191 (39%) patients, enzalutamide in 188 (39%) patients, 177Lu-PSMA RLT in 154 (32%) patients, and radium-223 dichloride in 18 (4%) patients. The median follow-up duration was 9·0 months (IQR 5·0-17·5). The median overall survival was 15·5 months (95% CI 13·4-18·3) and median progression-free survival was 7·9 months (6·8-8·9). In 347 (71%) of 488 patients, information regarding treatment-induced xerostomia was available, and 236 (68%) of the 347 patients reported xerostomia after the first cycle of 225Ac-PSMA RLT. All patients who received more than seven cycles of 225Ac-PSMA RLT reported xerostomia. Grade 3 or higher anaemia occurred in 64 (13%) of 488 patients, leukopenia in 19 (4%), thrombocytopenia in 32 (7%), and renal toxicity in 22 (5%). No serious adverse events or treatment-related deaths were recorded. INTERPRETATION 225Ac-PSMA RLT shows a substantial antitumour effect in mCRPC and represents a viable therapy option in patients treated with previous lines of approved agents. Xerostomia is a common side-effect. Severe bone marrow and renal toxicity are less common adverse events. FUNDING None.
Collapse
Affiliation(s)
- Mike M Sathekge
- Department of Nuclear Medicine, University of Pretoria, Pretoria, South Africa; Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa; Nuclear Medicine Research Infrastructure, Pretoria, South Africa.
| | - Ismaheel O Lawal
- Department of Nuclear Medicine, University of Pretoria, Pretoria, South Africa; Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, USA
| | - Chandrasekhar Bal
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | | | - Sajana Ballal
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | | | - Cindy Davis
- Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa
| | - Mathias Eiber
- Department of Nuclear Medicine, Klinikum Rechts Der Isar, Technical University of Munich and Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Türkay Hekimsoy
- Department of Nuclear Medicine, Klinikum Rechts Der Isar, Technical University of Munich and Bavarian Cancer Research Center (BZKF), Munich, Germany
| | - Otto Knoesen
- Nuclear Technology Products (NTP), Division of the South African Nuclear Energy Cooperation (NECSA), Pelindaba, South Africa
| | - Clemens Kratochwil
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Johncy Mahapane
- Department of Radiography, University of Pretoria, Pretoria, South Africa
| | - Letjie C Maserumule
- Department of Nuclear Medicine, University of Pretoria, Pretoria, South Africa; Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa; Nuclear Medicine Research Infrastructure, Pretoria, South Africa
| | - Amanda H Mdlophane
- Department of Nuclear Medicine, University of Pretoria, Pretoria, South Africa; Nuclear Medicine Research Infrastructure, Pretoria, South Africa
| | - Kgomotso M G Mokoala
- Department of Nuclear Medicine, University of Pretoria, Pretoria, South Africa; Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa; Nuclear Medicine Research Infrastructure, Pretoria, South Africa
| | - Honest Ndlovu
- Department of Nuclear Medicine, University of Pretoria, Pretoria, South Africa; Department of Nuclear Medicine, Steve Biko Academic Hospital, Pretoria, South Africa; Nuclear Medicine Research Infrastructure, Pretoria, South Africa
| | - Vineet Pant
- Royal Liverpool University Hospital, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Hendrik Rathke
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany; Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Janet Reed
- Department of Nuclear Medicine, University of Pretoria, Pretoria, South Africa
| | - Ishita B Sen
- Department of Nuclear Medicine, Fortis Memorial Research Institute, Gurugram, India
| | | | - Ashwani Sood
- Department of Nuclear Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Robert Tauber
- Department of Urology, School of Medicine, and Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Parul Thakral
- Department of Nuclear Medicine, Fortis Memorial Research Institute, Gurugram, India
| | - Madhav Prasad Yadav
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Alfred Morgenstern
- Department of Nuclear Medicine, University of Pretoria, Pretoria, South Africa; European Commission, Joint Research Centre, Karlsruhe, Germany
| |
Collapse
|
47
|
White FD, Thiele NA, Simms ME, Cary SK. Structure and bonding of a radium coordination compound in the solid state. Nat Chem 2024; 16:168-172. [PMID: 37945833 DOI: 10.1038/s41557-023-01366-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 10/11/2023] [Indexed: 11/12/2023]
Abstract
The structure and bonding of radium (Ra) is poorly understood because of challenges arising from its scarcity and radioactivity. Here we report the synthesis of a molecular Ra2+ complex using 226Ra and the organic ligand dibenzo-30-crown-10, and its characterization in the solid state by single-crystal X-ray diffraction. The crystal structure of the Ra2+ complex shows an 11-coordinate arrangement comprising the 10 donor O atoms of dibenzo-30-crown-10 and that of a bound water molecule. Under identical crystallization conditions, barium (Ba2+) yielded a 10-coordinate 'Pac-Man'-shaped structure lacking water. Furthermore, the bond distance between the Ra centre and the O atom of the coordinated water is substantially longer than would be predicted from the ionic radius of Ra2+ and by analogy with Ba2+, supporting greater water lability in Ra2+ complexes than in their Ba2+ counterparts. Barium often serves as a non-radioactive surrogate for radium, but our findings show that Ra2+ chemistry cannot always be predicted using Ba2+.
Collapse
Affiliation(s)
- Frankie D White
- Radioisotope Science and Technology Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA.
| | - Nikki A Thiele
- Chemical Sciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA.
| | - Megan E Simms
- Chemical Sciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Samantha K Cary
- Radioisotope Science and Technology Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| |
Collapse
|
48
|
Harriswangler C, Lucio-Martínez F, Rodríguez-Rodríguez A, Esteban-Gómez D, Platas-Iglesias C. Unravelling the 6sp ← 6s absorption spectra of Bi(III) complexes. Dalton Trans 2024; 53:2275-2285. [PMID: 38197124 DOI: 10.1039/d3dt03744d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
We report a spectroscopic and computational study that investigates the absorption spectra of Bi(III) complexes, which often show an absorption band in the UV region (∼270-350 nm) due to 6sp ← 6s transitions. We investigated the spectra of three simple complexes, [BiCl5]2-, [BiCl6]3- and [Bi(DMSO)8]3+, which show absorption maxima at 334, 326 and 279 nm due to 3P1 ← 1S0 transitions. Theoretical calculations based on quasi-degenerate N-electron valence perturbation theory to second order (QD-NEVPT2) provide an accurate description of the absorption spectra when employing CAS(2,9) wave functions. We next investigated the absorption spectra of the [Bi(NOTA)] complex (H3NOTA = 1,4,7-triazacyclononane-1,4,7-triacetic acid), which forms ternary complexes [Bi(NOTA)X]- (X = Cl, Br or I) in the presence of excess halide in aqueous solutions. Halide binding has an important impact on the position of the 3P1 ← 1S0 transition, which shifts progressively to longer wavelengths from 282 nm ([Bi(NOTA)]) to 298 nm (X = Cl), 305 nm (X = Br) and 325 nm (X = I). Subsequent QD-NEVPT2 calculations indicate that this effect is related to the progressive stabilization of the spin-orbit free states associated with the 6s16p1 configuration on increasing the covalent character of the metal-ligand(s) bonds, rather than with significant differences in spin-orbit coupling (SOC). These studies provide valuable insight into the coordination chemistry of Bi(III), an ion with increasing interest in targeted alpha therapy due to the possible application of bismuth isotopes bismuth-212 (212Bi, t1/2 = 60.6 min) and bismuth-213 (213Bi, t1/2 = 45.6 min).
Collapse
Affiliation(s)
- Charlene Harriswangler
- Universidade da Coruña, Centro de Interdisciplinar de Química e Bioloxía (CICA) and Departamento de Química, Facultade de Ciencias, 15071, A Coruña, Galicia, Spain.
| | - Fátima Lucio-Martínez
- Universidade da Coruña, Centro de Interdisciplinar de Química e Bioloxía (CICA) and Departamento de Química, Facultade de Ciencias, 15071, A Coruña, Galicia, Spain.
| | - Aurora Rodríguez-Rodríguez
- Universidade da Coruña, Centro de Interdisciplinar de Química e Bioloxía (CICA) and Departamento de Química, Facultade de Ciencias, 15071, A Coruña, Galicia, Spain.
| | - David Esteban-Gómez
- Universidade da Coruña, Centro de Interdisciplinar de Química e Bioloxía (CICA) and Departamento de Química, Facultade de Ciencias, 15071, A Coruña, Galicia, Spain.
| | - Carlos Platas-Iglesias
- Universidade da Coruña, Centro de Interdisciplinar de Química e Bioloxía (CICA) and Departamento de Química, Facultade de Ciencias, 15071, A Coruña, Galicia, Spain.
| |
Collapse
|
49
|
Yssartier T, Liu L, Pardoue S, Le Questel JY, Guérard F, Montavon G, Galland N. In vivo stability of 211At-radiopharmaceuticals: on the impact of halogen bond formation. RSC Med Chem 2024; 15:223-233. [PMID: 38283213 PMCID: PMC10809332 DOI: 10.1039/d3md00579h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 11/22/2023] [Indexed: 01/30/2024] Open
Abstract
211At, when coupled to a targeting agent, is one of the most promising radionuclides for therapeutic applications. The main labelling approach consists in the formation of astatoaryl compounds, which often show a lack of in vivo stability. The hypothesis that halogen bond (XB) interactions with protein functional groups initiate a deastatination mechanism is investigated through radiochemical experiments and DFT modelling. Several descriptors agree on the known mechanism of iodoaryl substrates dehalogenation by iodothyronine deiodinases, supporting the higher in vivo dehalogenation of N-succinimidyl 3-[211At]astatobenzoate (SAB) conjugates in comparison with their iodinated counterparts. The guanidinium group in 3-[211At]astato-4-guanidinomethylbenzoate (SAGMB) prevents the formation of At-mediated XBs with the selenocysteine active site in iodothyronine deiodinases. The initial step of At-aryl bond dissociation is inhibited, elucidating the better in vivo stability of SAGMB conjugates compared with those of SAB. The impact of astatine's ability to form XB interactions on radiopharmaceutical degradation may not be limited to the case of aryl radiolabeling.
Collapse
Affiliation(s)
- Thibault Yssartier
- CNRS, CEISAM UMR 6230, Nantes Université F-44000 Nantes France
- CNRS, SUBATECH UMR 6457, IMT Atlantique F-44307 Nantes France
| | - Lu Liu
- CNRS, IPHC UMR 7178, Université de Strasbourg F-67037 Strasbourg France
| | - Sylvain Pardoue
- CNRS, SUBATECH UMR 6457, IMT Atlantique F-44307 Nantes France
| | | | - François Guérard
- Inserm UMR 1307, CNRS UMR 6075, CRCI2NA, Nantes Université, Université d'Angers F-44000 Nantes France
| | - Gilles Montavon
- CNRS, SUBATECH UMR 6457, IMT Atlantique F-44307 Nantes France
| | - Nicolas Galland
- CNRS, CEISAM UMR 6230, Nantes Université F-44000 Nantes France
| |
Collapse
|
50
|
Miederer M, Benešová-Schäfer M, Mamat C, Kästner D, Pretze M, Michler E, Brogsitter C, Kotzerke J, Kopka K, Scheinberg DA, McDevitt MR. Alpha-Emitting Radionuclides: Current Status and Future Perspectives. Pharmaceuticals (Basel) 2024; 17:76. [PMID: 38256909 PMCID: PMC10821197 DOI: 10.3390/ph17010076] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 12/27/2023] [Accepted: 12/28/2023] [Indexed: 01/24/2024] Open
Abstract
The use of radionuclides for targeted endoradiotherapy is a rapidly growing field in oncology. In particular, the focus on the biological effects of different radiation qualities is an important factor in understanding and implementing new therapies. Together with the combined approach of imaging and therapy, therapeutic nuclear medicine has recently made great progress. A particular area of research is the use of alpha-emitting radionuclides, which have unique physical properties associated with outstanding advantages, e.g., for single tumor cell targeting. Here, recent results and open questions regarding the production of alpha-emitting isotopes as well as their chemical combination with carrier molecules and clinical experience from compassionate use reports and clinical trials are discussed.
Collapse
Affiliation(s)
- Matthias Miederer
- Department of Translational Imaging in Oncology, National Center for Tumor Diseases (NCT/UCC), 01307 Dresden, Germany
- Medizinische Fakultät and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany
| | - Martina Benešová-Schäfer
- Research Group Molecular Biology of Systemic Radiotherapy, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany;
| | - Constantin Mamat
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstr, 400, 01328 Dresden, Germany
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, 01062 Dresden, Germany
| | - David Kästner
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany; (D.K.); (C.B.)
| | - Marc Pretze
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany; (D.K.); (C.B.)
| | - Enrico Michler
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany; (D.K.); (C.B.)
| | - Claudia Brogsitter
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany; (D.K.); (C.B.)
| | - Jörg Kotzerke
- Medizinische Fakultät and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, 01307 Dresden, Germany; (D.K.); (C.B.)
| | - Klaus Kopka
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstr, 400, 01328 Dresden, Germany
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, 01062 Dresden, Germany
- National Center for Tumor Diseases (NCT) Dresden, University Hospital Carl Gustav Carus, Fetscherstraße 74, 01307 Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, Fetscherstraße 74, 01307 Dresden, Germany
| | - David A. Scheinberg
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY 10065, USA;
| | - Michael R. McDevitt
- Molecular Imaging and Therapy Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Department of Radiology, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|