1
|
Sancho L, Roteta A, Torres I, de Arcocha M, Ramos R, Domínguez ML, Rosales JJ, Prieto E, Quincoces G. State of the art and future perspectives of new radionuclides in Nuclear Medicine. Part II. Rev Esp Med Nucl Imagen Mol 2025; 44:500128. [PMID: 40147757 DOI: 10.1016/j.remnie.2025.500128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/29/2025]
Abstract
The state of the art and future perspectives of new radionuclides in Nuclear Medicine continue to evolve, driven by the development of isotopes with innovative applications in theragnostics. In this second part of the continuing education series, the clinical and therapeutic applications of terbium, actinium, and bismuth are analyzed in depth. The use of the four terbium isotopes (terbium-149, terbium-152, terbium-155, and terbium-161) is described, offering a versatile system for both diagnosis and treatment due to their chemical similarity to lutetium-177, along with the challenges related to their production and availability. Additionally, actinium-225, a powerful alpha-emitting radionuclide, is reviewed for its growing role in Targeted Alpha Therapy (TAT), particularly in prostate cancer and neuroendocrine tumors. Finally, bismuth-213, derived from actinium-225, is analyzed for its short half-life, making it a viable option for localized and selective therapies. Despite technical and production challenges, these radionuclides are driving the evolution of precision medicine, expanding therapeutic and diagnostic possibilities in Nuclear Medicine.
Collapse
Affiliation(s)
- L Sancho
- Servicio de Medicina Nuclear, Clínica Universidad de Navarra, Madrid, Spain
| | - A Roteta
- Servicio de Medicina Nuclear, Hospital Universitario de Donostia, San Sebastián, Spain
| | - I Torres
- Servicio de Medicina Nuclear, Hospital La Fe, Valencia, Spain
| | - M de Arcocha
- Unidad de Radiofarmacia, Servicio de Medicina Nuclear, Hospital Universitario Marqués de Valdecilla, Santander, Spain; Grupo de Imagen Molecular IDIVAL, Santander, Spain
| | - R Ramos
- Unidad de Radiofarmacia, Servicio de Medicina Nuclear, Clínica Universidad de Navarra, Madrid, Spain
| | - M L Domínguez
- Servicio de Medicina Nuclear, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - J J Rosales
- Servicio de Medicina Nuclear, Clínica Universidad de Navarra, Pamplona, Spain
| | - E Prieto
- Servicio de Radiofísica y Protección Radiológica, Clínica Universidad de Navarra, Pamplona, Spain.
| | - G Quincoces
- Unidad de Radiofarmacia, Servicio de Medicina Nuclear, Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
2
|
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
|
3
|
Grundmane A, Ramogida CF, Radchenko V. Novel dibutyl ether resin for the purification of the Auger emitter antimony-119 from tin targets. Nucl Med Biol 2025; 146-147:109022. [PMID: 40279962 DOI: 10.1016/j.nucmedbio.2025.109022] [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: 04/13/2025] [Accepted: 04/16/2025] [Indexed: 04/29/2025]
Abstract
Antimony-119 (119Sb, t1/2 = 38.19 h) is an Auger electron emitting radionuclide of interest for radiopharmaceutical therapy (RPT). The potential of 119Sb has only been explored theoretically, due to the absence of a suitable bifunctional chelator that enables the attachment of the radionuclide onto a radiopharmaceutical. Meanwhile, potential chelators are difficult to evaluate given that the production and radiochemical purification of 119Sb has not been optimized for radiopharmaceutical applications. 119Sb can be produced from proton bombardment of tin-119 (119Sn) targets on medical cyclotrons, subsequently the nanograms of radioantimony must be efficiently separated from the macroscopic (> mg) amount of target material while being recovered in a matrix suitable for chelation. To this end, a solid phase extraction (SPE) method employing a novel dibutyl ether (DBE) resin was developed to separate radioantimony from tin targets. The DBE resin was synthesized and characterized using thermogravimetric analysis, total organic carbon, and 1H nuclear magnetic resonance spectroscopy. The DBE resin exhibited excellent capacity (>8 mg Sb per gram) and integrity. Distribution coefficients (KD) for Sb(V) (KD up to 4600) and Sn(IV) (KD <0.3) showed good separation (Separation factor of >15,000) of both elements at high concentrations of HCl. Finally, dynamic separations with the DBE resin were capable of recovering up to 79 ± 2 % of radioantimony (1xxSb(III)) in 2 mL of 0.5 M sodium thioglycolate solution when separating nanogram quantities of 1xxSb from tens of milligrams of stable tin. Quantitative recovery of Sn was also achieved in just 1.5 mL of concentrated hydrochloric acid, indicating the potential for target recycling of enriched 119Sn required for pre-clinical evaluation of 119Sb.
Collapse
Affiliation(s)
- Aivija Grundmane
- Simon Fraser University, Department of Chemistry, 8888 University Dr W, Burnaby, BC V5A 1S6, Canada; TRIUMF, Life Sciences Division, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada.
| | - Caterina F Ramogida
- Simon Fraser University, Department of Chemistry, 8888 University Dr W, Burnaby, BC V5A 1S6, Canada; TRIUMF, Life Sciences Division, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada.
| | - Valery Radchenko
- TRIUMF, Life Sciences Division, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada; University of British Columbia, Department of Chemistry, Vancouver, BC V6T 1Z4, Canada.
| |
Collapse
|
4
|
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
|
5
|
Plichta KA, Buatti JM. The Emerging Potential of Lead-212 Theranostics. Hematol Oncol Clin North Am 2025; 39:221-236. [PMID: 39827042 DOI: 10.1016/j.hoc.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The field of theranostics uses radiopharmaceuticals to diagnose and treat disease, allowing for a personalized approach to treatment. Most theranostic therapies involve the use of beta-emitting radiopharmaceuticals. Because of their higher energies and decreased range, the use of alpha-emitting radiopharmaceuticals offers potential advantages over beta-emitting radiopharmaceuticals, including the potential for improved cell kill and decreased toxicity to normal tissues. This article focuses on the potential use of lead-212 as a theranostic treatment agent.
Collapse
Affiliation(s)
- Kristin A Plichta
- Department of Radiation Oncology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - John M Buatti
- Department of Radiation Oncology, University of Iowa Carver College of Medicine, Iowa City, IA, USA.
| |
Collapse
|
6
|
Roncali L, Hindré F, Samarut E, Lacoeuille F, Rousseau A, Lemée JM, Garcion E, Chérel M. Current landscape and future directions of targeted-alpha-therapy for glioblastoma treatment. Theranostics 2025; 15:4861-4889. [PMID: 40303349 PMCID: PMC12036880 DOI: 10.7150/thno.106081] [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: 10/30/2024] [Accepted: 03/02/2025] [Indexed: 05/02/2025] Open
Abstract
Glioblastoma (GB) is the most aggressive malignancy of the central nervous system. Despite two decades of intensive research since the establishment of the standard of care, emerging strategies have yet to produce consistent satisfactory outcomes. Because of its specific localisation and intricate characteristics, GB is a uniquely regulated solid tumour with a strong resistance to therapy. Advances in targeted radionuclide therapy (TRT), particularly with the introduction of a-emitting radionuclides, have unveiled potential avenues for the management of GB. Recent preclinical and clinical studies underscored promising advancements for targeted-α-therapy (TAT), but these therapeutic approaches exhibit a vast design heterogeneity, encompassing diverse radionuclides, vectors, target molecules, and administration modalities. This review seeks to critically assess the therapeutic landscape of GB through the perspective of TAT. Here, the focus is made on the advancements and limitations of in vivo explorations, pilot studies, and clinical trials, to determine the best directions for future investigations. In doing so, we hope to identify existing challenges and draw insights that might pave the way towards a more effective therapeutic approach.
Collapse
Affiliation(s)
- Loris Roncali
- Centre for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela; E-15782 Santiago de Compostela, Spain
- University of Angers, INSERM, CNRS, CRCI 2 NA; F-49000 Angers, France
- Nantes University, INSERM, CNRS, CRCI 2 NA; F-44000 Nantes, France
| | - François Hindré
- University of Angers, INSERM, CNRS, CRCI 2 NA; F-49000 Angers, France
- PRIMEX (Experimental Imagery and Radiobiology Platform), University of Angers, SFR 4208; F-49000 Angers, France
| | - Edouard Samarut
- Nantes University, INSERM, CNRS, CRCI 2 NA; F-44000 Nantes, France
- Department of Neurosurgery & Neurotraumatology, University Hospital of Nantes; F-44093 Nantes, France
| | - Franck Lacoeuille
- University of Angers, INSERM, CNRS, CRCI 2 NA; F-49000 Angers, France
- Department of Nuclear Medicine, University Hospital of Angers; F-49000 Angers, France
| | - Audrey Rousseau
- University of Angers, INSERM, CNRS, CRCI 2 NA; F-49000 Angers, France
- Department of Pathology, University Hospital of Angers; F-49000 Angers, France
| | - Jean-Michel Lemée
- University of Angers, INSERM, CNRS, CRCI 2 NA; F-49000 Angers, France
- Department of Neurosurgery, University Hospital of Angers; F-49000 Angers, France
| | - Emmanuel Garcion
- University of Angers, INSERM, CNRS, CRCI 2 NA; F-49000 Angers, France
- PACEM (Platform of Cellular and Molecular Analysis), University of Angers, SFR 4208; F-49000 Angers, France
| | - Michel Chérel
- Nantes University, INSERM, CNRS, CRCI 2 NA; F-44000 Nantes, France
- Institut de Cancérologie de l'Ouest, Department of Nuclear Medicine; F-44160 Saint-Herblain, France
| |
Collapse
|
7
|
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
|
8
|
Hooijman EL, de Jong JR, Ntihabose CM, Bruchertseifer F, Morgenstern A, Seimbille Y, Brabander T, Koolen SLW, de Blois E. Ac-225 radiochemistry through the lens of [ 225Ac]Ac-DOTA-TATE. EJNMMI Radiopharm Chem 2025; 10:9. [PMID: 39976810 PMCID: PMC11842643 DOI: 10.1186/s41181-025-00332-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 02/05/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND Targeted alpha therapy with Ac-225 showed to be effective in treating metastatic cancers. However, the complex decay chain requires optimized radiolabeling and quality control. This study aims to determine critical parameters and establish optimal labeling and accurate measuring techniques for radiochemical yield and purity with DOTA-TATE as a model molecule. Ac-225 sources were analyzed for metals (ΣFe, Zn, Cu) and quantified by UPLC. Optimization of radiolabeling kinetics for clinical conditions was performed in regards to temperature (20-90 °C), heating time (5-60 min), pH (2.5-10, with/without excess of metal ions), buffers, quenchers, volume (0.1-10 mL) and molar activity (90-540 kBq/nmol). The quality control was investigated using radio-TLC/HPLC by changing gradient to evaluate peak separation, radiolysed peptide and impurity separation. RESULTS Metal ingrowth was observed in Ac-225 stocks (n = 3), (time of arrival: 17.9, 36.8 and 101.4 nmol per 10 MBq). Optimal radiochemical yields were achieved with > 80 °C (20 min) at pH 8.5 (15 mM TRIS) up to 270 kBq. Labeling at a high pH showed a higher RCY, even in presence of an excess of metals. High stability (RCP > 90%) was achieved after addition of quenchers (cysteine, methionine, ascorbate, histidine, or gentisic acid (35 mM)) up to 24 h. For optimal determination of the radiochemical purity (indirect HPLC) fifty fractions are required. CONCLUSION The quality of Ac-225 labeled DOTA-radiopharmaceuticals is highly dependent on the pH and stabilization (buffer/quencher). Within this research it is demonstrated that optimized quality control methods and accurate measurement of the radiolabeling kinetics are crucial to ensure safe implementation for patient treatment.
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
| | - Jan R de Jong
- Department of Radiology and Nuclear Medicine, Erasmus MC, 3015 CN, Rotterdam, The Netherlands
| | - Carolline M Ntihabose
- Department of Radiology and Nuclear Medicine, Erasmus MC, 3015 CN, Rotterdam, The Netherlands
- Department of Hospital Pharmacy, Erasmus MC, 3015 CN, Rotterdam, The Netherlands
| | | | | | - Yann Seimbille
- Department of Radiology and Nuclear Medicine, Erasmus MC, 3015 CN, Rotterdam, The Netherlands
- Division of Life Sciences, TRIUMF, Vancouver, BC, V6T 2A3, Canada
| | - 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
|
9
|
Jin ZH, Degardin M, Furukawa T, Uehara T, Tsuji AB, Suzuki H, Wakizaka H, Sugyo A, Aung W, Suzuki H, Nagatsu K, Zhang MR, Dumy P, Boturyn D, Higashi T. Evaluation of the Gly-Phe-Lys Linker to Reduce the Renal Radioactivity of a [ 64Cu]Cu-Labeled Multimeric cRGD Peptide. ACS OMEGA 2025; 10:4102-4120. [PMID: 39926504 PMCID: PMC11799997 DOI: 10.1021/acsomega.4c10621] [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: 11/22/2024] [Revised: 01/02/2025] [Accepted: 01/08/2025] [Indexed: 02/11/2025]
Abstract
Radiometal-labeled peptide-based radiopharmaceuticals (RLPB-radiopharmaceuticals) are promising for cancer imaging and targeted radiotherapy; however, their effectiveness is often compromised by the high retention of nonspecific radioactivity in the kidneys due to renal excretion pathways. Current strategies to address this issue have limitations, highlighting the need for innovative approaches to improve targeting specificity and therapeutic efficacy. We aimed to evaluate the applicability of the Gly-Phe-Lys (GFK) tripeptide, a renal brush border (RBB) enzyme-cleavable linkage, to reduce renal radioactivity in RLPB-radiopharmaceuticals using the integrin-targeting radiopeptide [64Cu]Cu-cyclam-RAFT-c(-RGDfK-)4 ([64Cu]Cu-cyclam-RaftRGD). We designed and synthesized the model compound [64Cu]Cu-cyclam-GFK(benzoyl [Bz]), its predictive metabolites, and GFK-incorporated [64Cu]Cu-cyclam-RaftRGD derivatives [64Cu]Cu-cyclam-GFK-RaftRGD and [64Cu]Cu-cyclam-GFK(beta-alanine [βA])3-RaftRGD. In vitro studies showed that dual radiometabolites, namely, [64Cu]Cu-cyclam-G and [64Cu]Cu-cyclam-GF, were simultaneously released from [64Cu]Cu-cyclam-GFK(Bz) by different RBB enzymes, whereas both RaftRGD derivatives released only [64Cu]Cu-cyclam-GF. When injected into mice, [64Cu]Cu-cyclam-GFK(Bz) and the two RaftRGD derivatives led to the urinary excretion of [64Cu]Cu-cyclam-G and [64Cu]Cu-cyclam-GF, respectively. PET imaging and biodistribution studies showed the increased rates of reduction in renal radioactivity levels for the two RaftRGD derivatives compared to the parental [64Cu]Cu-cyclam-RaftRGD (e.g., PET: 1 to 24 h postinjection, 73.0 ± 2.3 and 75.6 ± 1.8 vs 43.0 ± 4.5%, p < 0.0001; biodistribution: 3 to 24 h, 61.1 and 74.4 vs 22.8%). Taken together, these results indicate that the designed renal cleavage occurred in vivo. We also noted the steric interference of the RaftRGD moiety on enzyme access, the spacer effect of the trimeric βA sequence (reduced steric hindrance), and the altered radiopharmacokinetics (e.g., initially increased renal accumulation) of the RaftRGD compounds upon linker incorporation. These findings provide important insights into the chemical design of RLPB-radiopharmaceuticals with reduced renal retention based on the RBB strategy.
Collapse
Affiliation(s)
- Zhao-Hui Jin
- Department
of Molecular Imaging and Theranostics, Institute for Quantum Medical
Science, National Institutes for Quantum
Science and Technology (QST), Chiba 263-8555, Japan
| | - Mélissa Degardin
- Département
de Chimie Moléculaire, CNRS, Université
Grenoble Alpes, cedex 9, Grenoble 38058, France
| | - Takako Furukawa
- Department
of Molecular Imaging and Theranostics, Institute for Quantum Medical
Science, National Institutes for Quantum
Science and Technology (QST), Chiba 263-8555, Japan
- Department
of Integrated Health Sciences, Graduate School of Medicine, Nagoya University, Nagoya 461-8673, Japan
| | - Tomoya Uehara
- Laboratory
of Molecular Imaging and Radiotherapy, Graduate School of Pharmaceutical
Sciences, Chiba University, Chiba 260-8675, Japan
| | - Atsushi B. Tsuji
- Department
of Molecular Imaging and Theranostics, Institute for Quantum Medical
Science, National Institutes for Quantum
Science and Technology (QST), Chiba 263-8555, Japan
| | - Hiroyuki Suzuki
- Laboratory
of Molecular Imaging and Radiotherapy, Graduate School of Pharmaceutical
Sciences, Chiba University, Chiba 260-8675, Japan
| | - Hidekatsu Wakizaka
- Department
of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical
Science, National Institutes for Quantum
Science and Technology (QST), Chiba 263-8555, Japan
| | - Aya Sugyo
- Department
of Molecular Imaging and Theranostics, Institute for Quantum Medical
Science, National Institutes for Quantum
Science and Technology (QST), Chiba 263-8555, Japan
| | - Winn Aung
- Department
of Molecular Imaging and Theranostics, Institute for Quantum Medical
Science, National Institutes for Quantum
Science and Technology (QST), Chiba 263-8555, Japan
| | - Hisashi Suzuki
- Department
of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical
Science, National Institutes for Quantum
Science and Technology (QST), Chiba 263-8555, Japan
| | - Kotaro Nagatsu
- Department
of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical
Science, National Institutes for Quantum
Science and Technology (QST), Chiba 263-8555, Japan
| | - Ming-Rong Zhang
- Department
of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical
Science, National Institutes for Quantum
Science and Technology (QST), Chiba 263-8555, Japan
| | - Pascal Dumy
- Institut
des Biomolécules Max Mousseron, École Nationale Supérieure
de Chimie de Montpellier, Université
de Montpellier, cedex 5, Montpellier 34296, France
| | - Didier Boturyn
- Département
de Chimie Moléculaire, CNRS, Université
Grenoble Alpes, cedex 9, Grenoble 38058, France
| | - Tatsuya Higashi
- Department
of Molecular Imaging and Theranostics, Institute for Quantum Medical
Science, National Institutes for Quantum
Science and Technology (QST), Chiba 263-8555, Japan
| |
Collapse
|
10
|
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]
|
11
|
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
|
12
|
Pretze M, Michler E, Kästner D, Kunkel F, Sagastume EA, Schultz MK, Kotzerke J. Lead-it-EAZY! GMP-compliant production of [ 212Pb]Pb-PSC-PEG 2-TOC. EJNMMI Radiopharm Chem 2024; 9:81. [PMID: 39604560 PMCID: PMC11602913 DOI: 10.1186/s41181-024-00305-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Recently, radiotheranostics comprising the true matched radionuclide pair 203/212Pb could serve as real dosimetric planning utility using 203Pb-radiolabelled pharmaceuticals before therapy with 212Pb-radiolabelled counterparts. 212Pb might act as the missing radionuclide therapy between standard β- therapies (e.g. with 177Lu or 90Y), in some cases leading to β- resistance and highly cytotoxic α therapies (e.g. with 225Ac) leading in some cases to renal insufficiency or even renal failure, due to the daughter nuclide 213Bi, which accumulates in > 90% within the kidneys during 225Ac therapy. 212Pb converts to 212Bi by β--decay and the following pathway of decay bears in sum only one α decay, which certainly happens within the targeted tumour tissue. Following daughter nuclides (e.g. 208Tl), which could distribute in organs at risk have only β- or γ decay, which is not as cytotoxic as α decay. RESULTS By ingenious customization of the standard cassettes of the ML EAZY it was possible to adapt the manual radiosynthesis of [212Pb]Pb-PSC-PEG2-TOC ([212Pb]Pb-VMT-α-NET) to a GMP-compliant synthesis module. The whole process of production, namely conditioning of C18 cartridge for purification, elution of the 224Ra/212Pb-generator, radiolabelling, C18 purification and sterile filtration performed automatically within one hour to access [212Pb]Pb-VMT-α-NET for patient application. [212Pb]Pb-VMT-α-NET was radiolabelled with high radiochemical purity > 95% and high radiochemical yield > 95% with molar activity ~ 15.8 MBq/nmol. CONCLUSIONS The Lead-it-EAZY process performed stable and robust over ten radiosyntheses and yielded sterile [212Pb]Pb-VMT-α-NET in high purity for patient application. By changing the precursor this process could easily be adapted to other 212Pb-radiopharmaceuticals.
Collapse
Affiliation(s)
- Marc Pretze
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstr. 74, 01307, Dresden, Germany.
| | - Enrico Michler
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - David Kästner
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| | - Falk Kunkel
- Eckert & Ziegler Eurotope, 13125, Berlin, Germany
| | | | - Michael K Schultz
- Perspective Therapeutics, Coralville, IA, 52241, USA
- Department of Radiology, The University of Iowa, Iowa City, IA, 52240, USA
- University of Iowa, Iowa City, IA, 52241, USA
- Department of Chemistry, The University of Iowa, Iowa City, IA, 52242, USA
| | - Jörg Kotzerke
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstr. 74, 01307, Dresden, Germany
| |
Collapse
|
13
|
Taïeb D, Deandreis D, Hicks RJ. Reimagining Biologically Adapted Somatostatin Receptor-Targeted Radionuclide Therapy: Perspectives Based on Personal Experience and Observations on Recent Trials. J Nucl Med 2024; 65:1685-1688. [PMID: 39299784 DOI: 10.2967/jnumed.124.268136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 08/02/2024] [Indexed: 09/22/2024] Open
Affiliation(s)
- David Taïeb
- Department of Nuclear Medicine, La Timone University Hospital, CERIMED, Aix-Marseille University, Marseille, France;
- ERL INSERM U1326 "RNAnoTher", CNRS, Aix-Marseille University, Marseille, France
| | | | - Rodney J Hicks
- Department of Medicine, St Vincent's Hospital, University of Melbourne, Melbourne, Victoria, Australia; and
- Department of Medicine, Central Clinical School, Alfred Hospital, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
14
|
Winter RC, Amghar M, Wacker AS, Bakos G, Taş H, Roscher M, Kelly JM, Benešová-Schäfer M. Future Treatment Strategies for Cancer Patients Combining Targeted Alpha Therapy with Pillars of Cancer Treatment: External Beam Radiation Therapy, Checkpoint Inhibition Immunotherapy, Cytostatic Chemotherapy, and Brachytherapy. Pharmaceuticals (Basel) 2024; 17:1031. [PMID: 39204136 PMCID: PMC11359268 DOI: 10.3390/ph17081031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 09/03/2024] Open
Abstract
Cancer is one of the most complex and challenging human diseases, with rising incidences and cancer-related deaths despite improved diagnosis and personalized treatment options. Targeted alpha therapy (TαT) offers an exciting strategy emerging for cancer treatment which has proven effective even in patients with advanced metastatic disease that has become resistant to other treatments. Yet, in many cases, more sophisticated strategies are needed to stall disease progression and overcome resistance to TαT. The combination of two or more therapies which have historically been used as stand-alone treatments is an approach that has been pursued in recent years. This review aims to provide an overview on TαT and the four main pillars of therapeutic strategies in cancer management, namely external beam radiation therapy (EBRT), immunotherapy with checkpoint inhibitors (ICI), cytostatic chemotherapy (CCT), and brachytherapy (BT), and to discuss their potential use in combination with TαT. A brief description of each therapy is followed by a review of known biological aspects and state-of-the-art treatment practices. The emphasis, however, is given to the motivation for combination with TαT as well as the pre-clinical and clinical studies conducted to date.
Collapse
Affiliation(s)
- Ruth Christine Winter
- Research Group Molecular Biology of Systemic Radiotherapy/Translational Radiotheranostics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; (R.C.W.); (M.A.); (G.B.); (H.T.)
| | - Mariam Amghar
- Research Group Molecular Biology of Systemic Radiotherapy/Translational Radiotheranostics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; (R.C.W.); (M.A.); (G.B.); (H.T.)
| | - Anja S. Wacker
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; (A.S.W.); (J.M.K.)
| | - Gábor Bakos
- Research Group Molecular Biology of Systemic Radiotherapy/Translational Radiotheranostics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; (R.C.W.); (M.A.); (G.B.); (H.T.)
| | - Harun Taş
- Research Group Molecular Biology of Systemic Radiotherapy/Translational Radiotheranostics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; (R.C.W.); (M.A.); (G.B.); (H.T.)
| | - Mareike Roscher
- Service Unit for Radiopharmaceuticals and Preclinical Studies, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany;
| | - James M. Kelly
- Department of Radiology, Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, 413 East 69th Street, New York, NY 10021, USA; (A.S.W.); (J.M.K.)
| | - Martina Benešová-Schäfer
- Research Group Molecular Biology of Systemic Radiotherapy/Translational Radiotheranostics, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; (R.C.W.); (M.A.); (G.B.); (H.T.)
| |
Collapse
|
15
|
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
|
16
|
Rathke H, Winter E, Bruchertseifer F, Röhrich M, Giesel FL, Haberkorn U, Morgenstern A, Kratochwil C. Deescalated 225Ac-PSMA-617 Versus 177Lu/ 225Ac-PSMA-617 Cocktail Therapy: A Single-Center Retrospective Analysis of 233 Patients. J Nucl Med 2024; 65:1057-1063. [PMID: 38844358 DOI: 10.2967/jnumed.123.267206] [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: 12/17/2023] [Accepted: 04/25/2024] [Indexed: 07/03/2024] Open
Abstract
The aim of this work is to evaluate our clinical real-world data obtained with 225Ac-PSMA-617 (AcPSMA), which were acquired under compassionate care regulations in patients with advanced-stage prostate cancer. The objective parameters that could be derived from this evaluation are compared with previous literature about AcPSMA and 177Lu-PSMA-617 (LuPSMA). Methods: The medical files of all patients who had received AcPSMA on an individual patient basis at the Heidelberg University Hospital since January 2014 were analyzed retrospectively. Previously published patients were excluded. The remaining patients were tailored into 2 subgroups with different treatment strategies: group 1 received AcPSMA as a deescalated monotherapy, and group 2 received LuPSMA plus AcPSMA as a cocktail regimen. Baseline characteristics, serum prostate-specific antigen (PSA) response, and overall survival were compared with the most appropriate historical controls. Results: Of 287 patients treated, 54 were excluded because of previous publication and 233 were evaluated, 104 of whom received AcPSMA monotherapy (median, 6 MBq). In this group, 55 patients (53%) presented with a best PSA response of at least 50%. The other 129 patients received a cocktail therapy of AcPSMA (median, 4 MBq) plus LuPSMA (4 GBq). In this group, a best PSA response of at least 50% was observed in 74 patients (57%). The median overall survival in the monogroup was 9 mo and in the cocktail group was 15 mo. If adjusted for prognostic baseline characteristics, the efficacy of both regimens was not significantly different. Conclusion: Deescalated treatment activities of AcPSMA or AcPSMA and LuPSMA cocktail regimens present better tolerability with regard to xerostomia than previous regimens of at least 100 kBq/kg while retaining high antitumor activity in poor-prognosis prostate cancer patients.
Collapse
Affiliation(s)
- Hendrik Rathke
- Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany;
- Department of Nuclear Medicine, Bern University Hospital, Bern, Switzerland
| | - Erik Winter
- Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Manuel Röhrich
- Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany
- Department of Nuclear Medicine, Mainz University Hospital, Mainz, Germany; and
| | - Frederik Lars Giesel
- Department of Nuclear Medicine, University Hospital Duesseldorf, Duesseldorf, Germany
| | - Uwe Haberkorn
- Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Clemens Kratochwil
- Department of Nuclear Medicine, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
17
|
Song H, Sgouros G. Alpha and Beta Radiation for Theragnostics. PET Clin 2024; 19:307-323. [PMID: 38688775 DOI: 10.1016/j.cpet.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Targeted radionuclide therapy (TRT) has significantly evolved from its beginnings with iodine-131 to employing carrier molecules with beta emitting isotopes like lutetium-177. With the success of Lu-177-DOTATATE for neuroendocrine tumors and Lu-177-PSMA-617 for prostate cancer, several other beta emitting radioisotopes, such as Cu-67 and Tb-161, are being explored for TRT. The field has also expanded into targeted alpha therapy (TAT) with agents like radium-223 for bone metastases in prostate cancer, and several other alpha emitter radioisotopes with carrier molecules, such as Ac-225, and Pb-212 under clinical trials. Despite these advancements, the scope of TRT in treating diverse solid tumors and integration with other therapies like immunotherapy remains under investigation. The success of antibody-drug conjugates further complements treatments with TRT, though challenges in treatment optimization continue.
Collapse
Affiliation(s)
- Hong Song
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Stanford University, Stanford, CA 94305, USA.
| | - George Sgouros
- Division of Radiological Physics, Department of Radiology and Radiological Sciences, The Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
18
|
Yang H, Zhang Y, Li H, Zhang Y, Feng Y, Yang X, Chen Y. Efficacy and Safety of 225 Ac-DOTATATE in the Treatment of Neuroendocrine Neoplasms With High SSTR Expression. Clin Nucl Med 2024; 49:505-512. [PMID: 38498615 DOI: 10.1097/rlu.0000000000005149] [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: 03/20/2024]
Abstract
PURPOSE We aimed to evaluate the efficacy and safety of 225 Ac-DOTATATE targeted α therapy (TAT) in various neuroendocrine neoplasms (NENs) with high somatostatin receptor (SSTR) expression. PATIENTS AND METHODS This single-center prospective study included 10 patients with histologically diagnosed NENs that exhibited increased SSTR expression on 68 Ga-DOTATATE PET/CT imaging. All patients received 225 Ac-DOTATATE TAT. The primary end points were molecular imaging-based response and disease control rate (DCR), measured using the slightly modified Positron Emission Tomography Response Criteria in Solid Tumors 1.0. The secondary end points were adverse event profiles and clinical responses. The adverse event profile was determined according to the Common Terminology Criteria for Adverse Events version 5.0. Clinical response was assessed using the EORTC QLQ-C30 v3.0 (European Organization for Research and Treatment of Cancer Core Quality of Life questionnaire version 3.0). RESULTS A molecular imaging-based partial response was observed in 40% of all patients, SD in 40%, PD in 20%, and DCR in 80%. The DCR was 83.3% (5/6) in patients who were previously treated with 177 Lu-DOTATATE. According to the EORTC QLQ-C30 v3.0 score, most symptoms improved after 225 Ac-DOTATATE treatment, with only diarrhea showing no improvement. Grade III/IV hematological, kidney, and liver toxicities were not observed. The median follow-up time was 14 months (7-22 months), and no deaths were reported. CONCLUSIONS This initial study suggests that 225 Ac-DOTATATE is a potentially promising option for treating NENs with elevated SSTR expression, with an acceptable toxicity profile and well-tolerated adverse effects.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiqun Yang
- Department of Dermatology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | | |
Collapse
|
19
|
Lawal IO, Abubakar SO, Ndlovu H, Mokoala KMG, More SS, Sathekge MM. Advances in Radioligand Theranostics in Oncology. Mol Diagn Ther 2024; 28:265-289. [PMID: 38555542 DOI: 10.1007/s40291-024-00702-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2024] [Indexed: 04/02/2024]
Abstract
Theranostics with radioligands (radiotheranostics) has played a pivotal role in oncology. Radiotheranostics explores the molecular targets expressed on tumor cells to target them for imaging and therapy. In this way, radiotheranostics entails non-invasive demonstration of the in vivo expression of a molecular target of interest through imaging followed by the administration of therapeutic radioligand targeting the tumor-expressed molecular target. Therefore, radiotheranostics ensures that only patients with a high likelihood of response are treated with a particular radiotheranostic agent, ensuring the delivery of personalized care to cancer patients. Within the last decades, a couple of radiotheranostics agents, including Lutetium-177 DOTATATE (177Lu-DOTATATE) and Lutetium-177 prostate-specific membrane antigen (177Lu-PSMA), were shown to prolong the survival of cancer patients compared to the current standard of care leading to the regulatory approval of these agents for routine use in oncology care. This recent string of successful approvals has broadened the interest in the development of different radiotheranostic agents and their investigation for clinical translation. In this work, we present an updated appraisal of the literature, reviewing the recent advances in the use of established radiotheranostic agents such as radioiodine for differentiated thyroid carcinoma and Iodine-131-labeled meta-iodobenzylguanidine therapy of tumors of the sympathoadrenal axis as well as the recently approved 177Lu-DOTATATE and 177Lu-PSMA for differentiated neuroendocrine tumors and advanced prostate cancer, respectively. We also discuss the radiotheranostic agents that have been comprehensively characterized in preclinical studies and have shown some clinical evidence supporting their safety and efficacy, especially those targeting fibroblast activation protein (FAP) and chemokine receptor 4 (CXCR4) and those still being investigated in preclinical studies such as those targeting poly (ADP-ribose) polymerase (PARP) and epidermal growth factor receptor 2.
Collapse
Affiliation(s)
- Ismaheel O Lawal
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Road, NE, Atlanta, GA, 30322, USA.
- Department of Nuclear Medicine, University of Pretoria, Pretoria, 0001, South Africa.
| | - Sofiullah O Abubakar
- Department of Radiology and Nuclear Medicine, Sultan Qaboos Comprehensive Cancer Care and Research Center, Muscat, Oman
| | - Honest Ndlovu
- Department of Nuclear Medicine, University of Pretoria, Pretoria, 0001, South Africa
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria, 0001, South Africa
| | - Kgomotso M G Mokoala
- Department of Nuclear Medicine, University of Pretoria, Pretoria, 0001, South Africa
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria, 0001, South Africa
| | - Stuart S More
- Department of Nuclear Medicine, University of Pretoria, Pretoria, 0001, South Africa
- Division of Nuclear Medicine, Department of Radiation Medicine, University of Cape Town, Cape Town, 7700, South Africa
| | - Mike M Sathekge
- Department of Nuclear Medicine, University of Pretoria, Pretoria, 0001, South Africa
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria, 0001, South Africa
| |
Collapse
|
20
|
Munekane M, Fuchigami T, Ogawa K. Recent advances in the development of 225Ac- and 211At-labeled radioligands for radiotheranostics. ANAL SCI 2024; 40:803-826. [PMID: 38564087 PMCID: PMC11035452 DOI: 10.1007/s44211-024-00514-w] [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: 11/28/2023] [Accepted: 01/16/2024] [Indexed: 04/04/2024]
Abstract
Radiotheranostics utilizes a set of radioligands incorporating diagnostic or therapeutic radionuclides to achieve both diagnosis and therapy. Imaging probes using diagnostic radionuclides have been used for systemic cancer imaging. Integration of therapeutic radionuclides into the imaging probes serves as potent agents for radionuclide therapy. Among them, targeted alpha therapy (TAT) is a promising next-generation cancer therapy. The α-particles emitted by the radioligands used in TAT result in a high linear energy transfer over a short range, inducing substantial damage to nearby cells surrounding the binding site. Therefore, the key to successful cancer treatment with minimal side effects by TAT depends on the selective delivery of radioligands to their targets. Recently, TAT agents targeting biomolecules highly expressed in various cancer cells, such as sodium/iodide symporter, norepinephrine transporter, somatostatin receptor, αvβ3 integrin, prostate-specific membrane antigen, fibroblast-activation protein, and human epidermal growth factor receptor 2 have been developed and have made remarkable progress toward clinical application. In this review, we focus on two radionuclides, 225Ac and 211At, which are expected to have a wide range of applications in TAT. We also introduce recent fundamental and clinical studies of radiopharmaceuticals labeled with these radionuclides.
Collapse
Affiliation(s)
- Masayuki Munekane
- Graduate School of Medical Sciences, Kanazawa University, Kakuma-Machi, Kanazawa, Ishikawa, 920-1192, Japan
| | - Takeshi Fuchigami
- Graduate School of Medical Sciences, Kanazawa University, Kakuma-Machi, Kanazawa, Ishikawa, 920-1192, Japan.
| | - Kazuma Ogawa
- Graduate School of Medical Sciences, Kanazawa University, Kakuma-Machi, Kanazawa, Ishikawa, 920-1192, Japan.
- Institute for Frontier Science Initiative, Kanazawa University, Kakuma-Machi, Kanazawa, Ishikawa, 920-1192, Japan.
| |
Collapse
|
21
|
Li M, Robles-Planells C, Liu D, Graves SA, Vasquez-Martinez G, Mayoral-Andrade G, Lee D, Rastogi P, Marks BM, Sagastume EA, Weiss RM, Linn-Peirano SC, Johnson FL, Schultz MK, Zepeda-Orozco D. Pre-clinical evaluation of biomarkers for the early detection of nephrotoxicity following alpha-particle radioligand therapy. Eur J Nucl Med Mol Imaging 2024; 51:1395-1408. [PMID: 38095674 PMCID: PMC10957612 DOI: 10.1007/s00259-023-06559-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 12/01/2023] [Indexed: 12/26/2023]
Abstract
PURPOSE Cancer treatment with alpha-emitter-based radioligand therapies (α-RLTs) demonstrates promising tumor responses. Radiolabeled peptides are filtered through glomeruli, followed by potential reabsorption of a fraction by proximal tubules, which may cause acute kidney injury (AKI) and chronic kidney disease (CKD). Because tubular cells are considered the primary site of radiopeptides' renal reabsorption and potential injury, the current use of kidney biomarkers of glomerular functional loss limits the evaluation of possible nephrotoxicity and its early detection. This study aimed to investigate whether urinary secretion of tubular injury biomarkers could be used as an additional non-invasive sensitive diagnostic tool to identify unrecognizable tubular damage and risk of long-term α-RLT nephrotoxicity. METHODS A bifunctional cyclic peptide, melanocortin 1 ligand (MC1L), labeled with [203Pb]Pb-MC1L, was used for [212Pb]Pb-MC1L biodistribution and absorbed dose measurements in CD-1 Elite mice. Mice were treated with [212Pb]Pb-MC1L in a dose-escalation study up to levels of radioactivity intended to induce kidney injury. The approach enabled prospective kidney functional and injury biomarker evaluation and late kidney histological analysis to validate these biomarkers. RESULTS Biodistribution analysis identified [212Pb]Pb-MC1L reabsorption in kidneys with a dose deposition of 2.8, 8.9, and 20 Gy for 0.9, 3.0, and 6.7 MBq injected [212Pb]Pb-MC1L doses, respectively. As expected, mice receiving 6.7 MBq had significant weight loss and CKD evidence based on serum creatinine, cystatin C, and kidney histological alterations 28 weeks after treatment. A dose-dependent urinary neutrophil gelatinase-associated lipocalin (NGAL, tubular injury biomarker) urinary excretion the day after [212Pb]Pb-MC1L treatment highly correlated with the severity of late tubulointerstitial injury and histological findings. CONCLUSION Urine NGAL secretion could be a potential early diagnostic tool to identify unrecognized tubular damage and predict long-term α-RLT-related nephrotoxicity.
Collapse
Affiliation(s)
- Mengshi Li
- Viewpoint Molecular Targeting, Inc. Dba Perspective Therapeutics, Coralville, IA, USA
| | - Claudia Robles-Planells
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH, USA
| | - Dijie Liu
- Viewpoint Molecular Targeting, Inc. Dba Perspective Therapeutics, Coralville, IA, USA
| | - Stephen A Graves
- Department of Radiology, The University of Iowa, Iowa City, IA, USA
| | - Gabriela Vasquez-Martinez
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH, USA
| | - Gabriel Mayoral-Andrade
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH, USA
| | - Dongyoul Lee
- Department of Physics and Chemistry, Korea Military Academy, Seoul, Republic of Korea
| | - Prerna Rastogi
- Department of Pathology, The University of Iowa, Iowa City, IA, USA
| | - Brenna M Marks
- Viewpoint Molecular Targeting, Inc. Dba Perspective Therapeutics, Coralville, IA, USA
| | - Edwin A Sagastume
- Viewpoint Molecular Targeting, Inc. Dba Perspective Therapeutics, Coralville, IA, USA
| | - Robert M Weiss
- Department of Internal Medicine, The University of Iowa, Iowa City, IA, USA
| | - Sarah C Linn-Peirano
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH, USA
- Department of Veterinary Biosciences, The Ohio State University College of Veterinary Medicine Columbus, Columbus, OH, USA
| | - Frances L Johnson
- Viewpoint Molecular Targeting, Inc. Dba Perspective Therapeutics, Coralville, IA, USA
| | - Michael K Schultz
- Viewpoint Molecular Targeting, Inc. Dba Perspective Therapeutics, Coralville, IA, USA.
- Department of Radiology, The University of Iowa, Iowa City, IA, USA.
- Department of Radiation Oncology, Free Radical, and Radiation Biology Program, The University of Iowa, Iowa City, IA, USA.
| | - Diana Zepeda-Orozco
- Kidney and Urinary Tract Center, Abigail Wexner Research Institute at Nationwide Children's, Columbus, OH, USA.
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA.
- Division of Nephrology and Hypertension, Nationwide Children's Hospital, Columbus, OH, USA.
| |
Collapse
|
22
|
Gape PMD, Schultz MK, Stasiuk GJ, Terry SYA. Towards Effective Targeted Alpha Therapy for Neuroendocrine Tumours: A Review. Pharmaceuticals (Basel) 2024; 17:334. [PMID: 38543120 PMCID: PMC10974115 DOI: 10.3390/ph17030334] [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: 02/08/2024] [Revised: 02/22/2024] [Accepted: 02/26/2024] [Indexed: 04/01/2024] Open
Abstract
This review article explores the evolving landscape of Molecular Radiotherapy (MRT), emphasizing Peptide Receptor Radionuclide Therapy (PRRT) for neuroendocrine tumours (NETs). The primary focus is on the transition from β-emitting radiopharmaceuticals to α-emitting agents in PRRT, offering a critical analysis of the radiobiological basis, clinical applications, and ongoing developments in Targeted Alpha Therapy (TAT). Through an extensive literature review, the article delves into the mechanisms and effectiveness of PRRT in targeting somatostatin subtype 2 receptors, highlighting both its successes and limitations. The discussion extends to the emerging paradigm of TAT, underlining its higher potency and specificity with α-particle emissions, which promise enhanced therapeutic efficacy and reduced toxicity. The review critically evaluates preclinical and clinical data, emphasizing the need for standardised dosimetry and a deeper understanding of the dose-response relationship in TAT. The review concludes by underscoring the significant potential of TAT in treating SSTR2-overexpressing cancers, especially in patients refractory to β-PRRT, while also acknowledging the current challenges and the necessity for further research to optimize treatment protocols.
Collapse
Affiliation(s)
- Paul M. D. Gape
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London SE1 7EP, UK; (G.J.S.); (S.Y.A.T.)
| | - Michael K. Schultz
- Departments of Radiology, Radiation Oncology, Free Radical and Radiation Biology Program, University of Iowa, Iowa City, IA 52242, USA;
- Perspective Therapeutics, Coralville, IA 52241, USA
| | - Graeme J. Stasiuk
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London SE1 7EP, UK; (G.J.S.); (S.Y.A.T.)
| | - Samantha Y. A. Terry
- School of Biomedical Engineering & Imaging Sciences, King’s College London, London SE1 7EP, UK; (G.J.S.); (S.Y.A.T.)
| |
Collapse
|
23
|
Lee D, Li M, Liu D, Baumhover NJ, Sagastume EA, Marks BM, Rastogi P, Pigge FC, Menda Y, Johnson FL, Schultz MK. Structural modifications toward improved lead-203/lead-212 peptide-based image-guided alpha-particle radiopharmaceutical therapies for neuroendocrine tumors. Eur J Nucl Med Mol Imaging 2024; 51:1147-1162. [PMID: 37955792 PMCID: PMC10881741 DOI: 10.1007/s00259-023-06494-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/26/2023] [Indexed: 11/14/2023]
Abstract
PURPOSE The lead-203 (203Pb)/lead-212 (212Pb) elementally identical radionuclide pair has gained significant interest in the field of image-guided targeted alpha-particle therapy for cancer. Emerging evidence suggests that 212Pb-labeled peptide-based radiopharmaceuticals targeting somatostatin receptor subtype 2 (SSTR2) may provide improved effectiveness compared to beta-particle-based therapies for neuroendocrine tumors (NETs). This study aims to improve the performance of SSTR2-targeted radionuclide imaging and therapy through structural modifications to Tyr3-octreotide (TOC)-based radiopharmaceuticals. METHODS New SSTR2-targeted peptides were designed and synthesized with the goal of optimizing the incorporation of Pb isotopes through the use of a modified cyclization technique; the introduction of a Pb-specific chelator (PSC); and the insertion of polyethylene glycol (PEG) linkers. The binding affinity of the peptides and the cellular uptake of 203Pb-labeled peptides were evaluated using pancreatic AR42J (SSTR2+) tumor cells and the biodistribution and imaging of the 203Pb-labeled peptides were assessed in an AR42J tumor xenograft mouse model. A lead peptide was identified (i.e., PSC-PEG2-TOC), which was then further evaluated for efficacy in 212Pb therapy studies. RESULTS The lead radiopeptide drug conjugate (RPDC) - [203Pb]Pb-PSC-PEG2-TOC - significantly improved the tumor-targeting properties, including receptor binding and tumor accumulation and retention as compared to [203Pb]Pb-DOTA0-Tyr3-octreotide (DOTATOC). Additionally, the modified RPDC exhibited faster renal clearance than the DOTATOC counterpart. These advantageous characteristics of [212Pb]Pb-PSC-PEG2-TOC resulted in a dose-dependent therapeutic effect with minimal signs of toxicity in the AR42J xenograft model. Fractionated administrations of 3.7 MBq [212Pb]Pb-PSC-PEG2-TOC over three doses further improved anti-tumor effectiveness, resulting in 80% survival (70% complete response) over 120 days in the mouse model. CONCLUSION Structural modifications to chelator and linker compositions improved tumor targeting and pharmacokinetics (PK) of 203/212Pb peptide-based radiopharmaceuticals for NET theranostics. These findings suggest that PSC-PEG2-TOC is a promising candidate for Pb-based targeted radionuclide therapy for NETs and other types of cancers that express SSTR2.
Collapse
Affiliation(s)
- Dongyoul Lee
- Department of Physics and Chemistry, Korea Military Academy, Seoul, Republic of Korea
| | - Mengshi Li
- Perspective Therapeutics, Inc., Coralville, IA, USA
| | - Dijie Liu
- Perspective Therapeutics, Inc., Coralville, IA, USA
| | | | | | | | - Prerna Rastogi
- Department of Pathology, The University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - F Christopher Pigge
- Department of Chemistry, The University of Iowa, ML B180 FRRBP, 500 Newton Road, Iowa City, IA, 52240, USA
| | - Yusuf Menda
- Department of Radiology, The University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | | | - Michael K Schultz
- Perspective Therapeutics, Inc., Coralville, IA, USA.
- Department of Chemistry, The University of Iowa, ML B180 FRRBP, 500 Newton Road, Iowa City, IA, 52240, USA.
- Department of Radiology, The University of Iowa Hospitals and Clinics, Iowa City, IA, USA.
- Department of Radiation Oncology, The University of Iowa Hospitals and Clinics, Iowa City, IA, USA.
| |
Collapse
|
24
|
Van Laere C, Koole M, Deroose CM, de Voorde MV, Baete K, Cocolios TE, Duchemin C, Ooms M, Cleeren F. Terbium radionuclides for theranostic applications in nuclear medicine: from atom to bedside. Theranostics 2024; 14:1720-1743. [PMID: 38389843 PMCID: PMC10879862 DOI: 10.7150/thno.92775] [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: 11/30/2023] [Accepted: 02/07/2024] [Indexed: 02/24/2024] Open
Abstract
Terbium features four clinically interesting radionuclides for application in nuclear medicine: terbium-149, terbium-152, terbium-155, and terbium-161. Their identical chemical properties enable the synthesis of radiopharmaceuticals with the same pharmacokinetic character, while their distinctive decay characteristics make them valuable for both imaging and therapeutic applications. In particular, terbium-152 and terbium-155 are useful candidates for positron emission tomography (PET) and single photon emission computed tomography (SPECT) imaging, respectively; whereas terbium-149 and terbium-161 find application in α- and β--/Auger electron therapy, respectively. This unique characteristic makes the terbium family ideal for the "matched-pair" principle of theranostics. In this review, the advantages and challenges of terbium-based radiopharmaceuticals are discussed, covering the entire chain from radionuclide production to bedside administration. It elaborates on the fundamental properties of terbium, the production routes of the four interesting radionuclides and gives an overview of the available bifunctional chelators. Finally, we discuss the preclinical and clinical studies as well as the prospects of this promising development in nuclear medicine.
Collapse
Affiliation(s)
- Camille Van Laere
- Belgian Nuclear Research Centre (SCK CEN), Institute for Nuclear Medical Applications, Mol, Belgium
- Radiopharmaceutical Research, Department of Pharmacy and Pharmacology, KU Leuven, Leuven, Belgium
| | - Michel Koole
- Nuclear Medicine, University Hospitals Leuven, Belgium
- Nuclear Medicine & Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Christophe M. Deroose
- Nuclear Medicine, University Hospitals Leuven, Belgium
- Nuclear Medicine & Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Michiel Van de Voorde
- Belgian Nuclear Research Centre (SCK CEN), Institute for Nuclear Medical Applications, Mol, Belgium
| | - Kristof Baete
- Nuclear Medicine, University Hospitals Leuven, Belgium
- Nuclear Medicine & Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Thomas E. Cocolios
- KU Leuven, Institute for Nuclear and Radiation Physics, Department of Physics and Astronomy, Leuven, Belgium
| | | | - Maarten Ooms
- Belgian Nuclear Research Centre (SCK CEN), Institute for Nuclear Medical Applications, Mol, Belgium
| | - Frederik Cleeren
- Radiopharmaceutical Research, Department of Pharmacy and Pharmacology, KU Leuven, Leuven, Belgium
| |
Collapse
|
25
|
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
|
26
|
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
|
27
|
Jalloul W, Ghizdovat V, Stolniceanu CR, Ionescu T, Grierosu IC, Pavaleanu I, Moscalu M, Stefanescu C. Targeted Alpha Therapy: All We Need to Know about 225Ac's Physical Characteristics and Production as a Potential Theranostic Radionuclide. Pharmaceuticals (Basel) 2023; 16:1679. [PMID: 38139806 PMCID: PMC10747780 DOI: 10.3390/ph16121679] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/24/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023] Open
Abstract
The high energy of α emitters, and the strong linear energy transfer that goes along with it, lead to very efficient cell killing through DNA damage. Moreover, the degree of oxygenation and the cell cycle state have no impact on these effects. Therefore, α radioisotopes can offer a treatment choice to individuals who are not responding to β- or gamma-radiation therapy or chemotherapy drugs. Only a few α-particle emitters are suitable for targeted alpha therapy (TAT) and clinical applications. The majority of available clinical research involves 225Ac and its daughter nuclide 213Bi. Additionally, the 225Ac disintegration cascade generates γ decays that can be used in single-photon emission computed tomography (SPECT) imaging, expanding the potential theranostic applications in nuclear medicine. Despite the growing interest in applying 225Ac, the restricted global accessibility of this radioisotope makes it difficult to conduct extensive clinical trials for many radiopharmaceutical candidates. To boost the availability of 225Ac, along with its clinical and potential theranostic applications, this review attempts to highlight the fundamental physical properties of this α-particle-emitting isotope, as well as its existing and possible production methods.
Collapse
Affiliation(s)
- Wael Jalloul
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- North East Regional Innovative Cluster for Structural and Molecular Imaging (Imago-Mol), 700115 Iasi, Romania
| | - Vlad Ghizdovat
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- North East Regional Innovative Cluster for Structural and Molecular Imaging (Imago-Mol), 700115 Iasi, Romania
| | - Cati Raluca Stolniceanu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- North East Regional Innovative Cluster for Structural and Molecular Imaging (Imago-Mol), 700115 Iasi, Romania
| | - Teodor Ionescu
- Department of Morpho-Functional Sciences (Pathophysiology), “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Irena Cristina Grierosu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Ioana Pavaleanu
- Department of Mother and Child, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Mihaela Moscalu
- Department of Preventive Medicine and Interdisciplinarity, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
| | - Cipriana Stefanescu
- Department of Biophysics and Medical Physics-Nuclear Medicine, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania
- North East Regional Innovative Cluster for Structural and Molecular Imaging (Imago-Mol), 700115 Iasi, Romania
| |
Collapse
|
28
|
Han G, Hwang E, Lin F, Clift R, Kim D, Guest M, Bischoff E, Moran S, Li G. RYZ101 (Ac-225 DOTATATE) Opportunity beyond Gastroenteropancreatic Neuroendocrine Tumors: Preclinical Efficacy in Small-Cell Lung Cancer. Mol Cancer Ther 2023; 22:1434-1443. [PMID: 37616528 DOI: 10.1158/1535-7163.mct-23-0029] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 04/28/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023]
Abstract
Overexpression of somatostatin receptors (SSTR), particularly SSTR2, is found in gastroenteropancreatic neuroendocrine tumors (GEP-NET), and subsets of other solid tumors such as small-cell lung cancer (SCLC). SCLC accounts for approximately 13% to 15% of lung cancer and lacks effective therapeutic options. IHC analysis indicates that up to 50% of SCLC tumors are SSTR2-positive, with a substantial subset showing high and homogenous expression. Peptide receptor radionuclide therapy with radiolabeled somatostatin analogue, Lu-177 DOTATATE, has been approved for GEP-NETs. Different strategies aimed at improving outcomes, such as the use of alpha-emitting radioisotopes, are currently being investigated. RYZ101 (Ac-225 DOTATATE) is comprised of the alpha-emitting radioisotope actinium-225, chemical chelator DOTA, and octreotate (TATE), a somatostatin analogue. In the cell-based competitive radioligand binding assay, RAYZ-10001-La (lanthanum surrogate for RYZ101) showed high binding affinity (Ki = 0.057 nmol/L) to human SSTR2 and >600-fold selectivity against other SSTR subtypes. RAYZ-10001-La exhibited efficient internalization to SSTR2-positive cells. In multiple SSTR2-expressing SCLC xenograft models, single-dose intravenous RYZ101 3 μCi (0.111 MBq) or 4 μCi (0.148 MBq) significantly inhibited tumor growth, with deeper responses, including sustained regression, observed in the models with higher SSTR2 levels. The antitumor effect was further enhanced when RYZ101 was combined with carboplatin and etoposide at clinically relevant doses. In summary, RYZ101 is a highly potent, alpha-emitting radiopharmaceutical agent, and preclinical data demonstrate the potential of RYZ101 for the treatment of patients with SSTR-positive cancers.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Gary Li
- RayzeBio, Inc., San Diego, California
| |
Collapse
|
29
|
Pretze M, Michler E, Runge R, Wetzig K, Tietze K, Brandt F, Schultz MK, Kotzerke J. Influence of the Molar Activity of 203/212Pb-PSC-PEG 2-TOC on Somatostatin Receptor Type 2-Binding and Cell Uptake. Pharmaceuticals (Basel) 2023; 16:1605. [PMID: 38004470 PMCID: PMC10675797 DOI: 10.3390/ph16111605] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/06/2023] [Accepted: 11/10/2023] [Indexed: 11/26/2023] Open
Abstract
(1) Background: In neuroendocrine tumors (NETs), somatostatin receptor subtype 2 is highly expressed, which can be targeted by a radioactive ligand such as [177Lu]Lu-1,4,7,10-tetraazacyclododecane-N,N',N″,N‴,-tetraacetic acid-[Tyr3,Thr8]-octreotide (177Lu-DOTA-TOC) and, more recently, by a lead specific chelator (PSC) containing 203/212Pb-PSC-PEG2-TOC (PSC-TOC). The molar activity (AM) can play a crucial role in tumor uptake, especially in receptor-mediated uptake, such as in NETs. Therefore, an investigation of the influence of different molar activities of 203/212Pb-PSC-TOC on cell uptake was investigated. (2) Methods: Optimized radiolabeling of 203/212Pb-PSC-TOC was performed with 50 µg of precursor in a NaAc/AcOH buffer at pH 5.3-5.5 within 15-45 min at 95° C. Cell uptake was studied in AR42 J, HEK293 sst2, and ZR75-1 cells. (3) Results: 203/212Pb-PSC-TOC was radiolabeled with high radiochemical purity >95% and high radiochemical yield >95%, with AM ranging from 0.2 to 61.6 MBq/nmol. The cell uptake of 203Pb-PSC-TOC (AM = 38 MBq/nmol) was highest in AR42 J (17.9%), moderate in HEK293 sstr (9.1%) and lowest in ZR75-1 (0.6%). Cell uptake increased with the level of AM. (4) Conclusions: A moderate AM of 15-40 MBq/nmol showed the highest cell uptake. No uptake limitation was found in the first 24-48 h. Further escalation experiments with even higher AM should be performed in the future. It was shown that AM plays an important role because of its direct dependence on the cellular uptake levels, possibly due to less receptor saturation with non-radioactive ligands at higher AM.
Collapse
Affiliation(s)
- Marc Pretze
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstr. 74, 01307 Dresden, Germany; (E.M.); (R.R.); (K.W.); (K.T.); (F.B.); (J.K.)
| | - Enrico Michler
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstr. 74, 01307 Dresden, Germany; (E.M.); (R.R.); (K.W.); (K.T.); (F.B.); (J.K.)
| | - Roswitha Runge
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstr. 74, 01307 Dresden, Germany; (E.M.); (R.R.); (K.W.); (K.T.); (F.B.); (J.K.)
| | - Kerstin Wetzig
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstr. 74, 01307 Dresden, Germany; (E.M.); (R.R.); (K.W.); (K.T.); (F.B.); (J.K.)
| | - Katja Tietze
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstr. 74, 01307 Dresden, Germany; (E.M.); (R.R.); (K.W.); (K.T.); (F.B.); (J.K.)
| | - Florian Brandt
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstr. 74, 01307 Dresden, Germany; (E.M.); (R.R.); (K.W.); (K.T.); (F.B.); (J.K.)
| | - Michael K. Schultz
- Department of Radiology, University of Iowa, Iowa City, IA 52240, USA;
- Viewpoint Molecular Targeting, Inc. (DBA Perspective Therapeutics), Coralville, IA 52241, USA
- Department of Chemistry, University of Iowa, Iowa City, IA 52241, USA
- Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA
| | - Jörg Kotzerke
- Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Technical University Dresden, Fetscherstr. 74, 01307 Dresden, Germany; (E.M.); (R.R.); (K.W.); (K.T.); (F.B.); (J.K.)
| |
Collapse
|
30
|
Morgan KA, Rudd SE, Noor A, Donnelly PS. Theranostic Nuclear Medicine with Gallium-68, Lutetium-177, Copper-64/67, Actinium-225, and Lead-212/203 Radionuclides. Chem Rev 2023; 123:12004-12035. [PMID: 37796539 DOI: 10.1021/acs.chemrev.3c00456] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/06/2023]
Abstract
Molecular changes in malignant tissue can lead to an increase in the expression levels of various proteins or receptors that can be used to target the disease. In oncology, diagnostic imaging and radiotherapy of tumors is possible by attaching an appropriate radionuclide to molecules that selectively bind to these target proteins. The term "theranostics" describes the use of a diagnostic tool to predict the efficacy of a therapeutic option. Molecules radiolabeled with γ-emitting or β+-emitting radionuclides can be used for diagnostic imaging using single photon emission computed tomography or positron emission tomography. Radionuclide therapy of disease sites is possible with either α-, β-, or Auger-emitting radionuclides that induce irreversible damage to DNA. This Focus Review centers on the chemistry of theranostic approaches using metal radionuclides for imaging and therapy. The use of tracers that contain β+-emitting gallium-68 and β-emitting lutetium-177 will be discussed in the context of agents in clinical use for the diagnostic imaging and therapy of neuroendocrine tumors and prostate cancer. A particular emphasis is then placed on the chemistry involved in the development of theranostic approaches that use copper-64 for imaging and copper-67 for therapy with functionalized sarcophagine cage amine ligands. Targeted therapy with radionuclides that emit α particles has potential to be of particular use in late-stage disease where there are limited options, and the role of actinium-225 and lead-212 in this area is also discussed. Finally, we highlight the challenges that impede further adoption of radiotheranostic concepts while highlighting exciting opportunities and prospects.
Collapse
Affiliation(s)
- Katherine A Morgan
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Melbourne 3010, Australia
| | - Stacey E Rudd
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Melbourne 3010, Australia
| | - Asif Noor
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Melbourne 3010, Australia
| | - Paul S Donnelly
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Melbourne 3010, Australia
| |
Collapse
|
31
|
Li M, Robles-Planells C, Liu D, Graves SA, Vasquez-Martinez G, Mayoral-Andrade G, Lee D, Rastogi P, Marks BM, Sagastume EA, Weiss RM, Linn-Peirano SC, Johnson FL, Schultz MK, Zepeda-Orozco D. Pre-clinical Evaluation of Biomarkers for Early Detection of Nephrotoxicity Following Alpha-particle Radioligand Therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.27.559789. [PMID: 37808634 PMCID: PMC10557737 DOI: 10.1101/2023.09.27.559789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Purpose Cancer treatment with alpha-emitter-based radioligand therapies (α-RLTs) demonstrates promising tumor responses. Radiolabeled peptides are filtered through glomeruli, followed by potential reabsorption of a fraction by proximal tubules, which may cause acute kidney injury (AKI) and chronic kidney disease (CKD). Because tubular cells are considered the primary site of radiopeptides' renal reabsorption and potential injury, the current use of kidney biomarkers of glomerular functional loss limits the evaluation of possible nephrotoxicity and its early detection. This study aimed to investigate whether urinary secretion of tubular injury biomarkers could be used as additional non-invasive sensitive diagnostic tool to identify unrecognizable tubular damage and risk of long-term α-RLTs nephrotoxicity. Methods A bifunctional cyclic peptide, melanocortin ligand-1(MC1L), labeled with [ 203 Pb]Pb-MC1L, was used for [ 212 Pb]Pb-MC1L biodistribution and absorbed dose measurements in CD-1 Elite mice. Mice were treated with [ 212 Pb]Pb-MC1L in a dose escalation study up to levels of radioactivity intended to induce kidney injury. The approach enabled prospective kidney functional and injury biomarker evaluation and late kidney histological analysis to validate these biomarkers. Results Biodistribution analysis identified [ 212 Pb]Pb-MC1L reabsorption in kidneys with a dose deposition of 2.8, 8.9, and 20 Gy for 0.9, 3.0, and 6.7 MBq injected [ 212 Pb]Pb-MC1L doses, respectively. As expected, mice receiving 6.7 MBq had significant weight loss and CKD evidence based on serum creatinine, cystatin C, and kidney histological alterations 28 weeks after treatment. A dose-dependent urinary Neutrophil gelatinase-associated lipocalin (NGAL, tubular injury biomarker) urinary excretion the day after [ 212 Pb]Pb-MC1L treatment highly correlated with the severity of late tubulointerstitial injury and histological findings. Conclusion urine NGAL secretion could be a potential early diagnostic tool to identify unrecognized tubular damage and predict long-term α-RLT-related nephrotoxicity.
Collapse
|
32
|
Beydağı G, Alan Selçuk N, Kabasakal L. Alpha Peptide Receptor Radionuclide Therapy in Neuroendocrine Tumors. NUCLEAR MEDICINE SEMINARS 2023; 9:109-115. [DOI: 10.4274/nts.galenos.2023.0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
33
|
Koniar H, Miller C, Rahmim A, Schaffer P, Uribe C. A GATE simulation study for dosimetry in cancer cell and micrometastasis from the 225Ac decay chain. EJNMMI Phys 2023; 10:46. [PMID: 37525027 PMCID: PMC10390455 DOI: 10.1186/s40658-023-00564-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 07/24/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND Radiopharmaceutical therapy (RPT) with alpha-emitting radionuclides has shown great promise in treating metastatic cancers. The successive emission of four alpha particles in the 225Ac decay chain leads to highly targeted and effective cancer cell death. Quantifying cellular dosimetry for 225Ac RPT is essential for predicting cell survival and therapeutic success. However, the leading assumption that all 225Ac progeny remain localized at their target sites likely overestimates the absorbed dose to cancer cells. To address limitations in existing semi-analytic approaches, this work evaluates S-values for 225Ac's progeny radionuclides with GATE Monte Carlo simulations. METHODS The cellular geometries considered were an individual cell (10 µm diameter with a nucleus of 8 µm diameter) and a cluster of cells (micrometastasis) with radionuclides localized in four subcellular regions: cell membrane, cytoplasm, nucleus, or whole cell. The absorbed dose to the cell nucleus was scored, and self- and cross-dose S-values were derived. We also evaluated the total absorbed dose with various degrees of radiopharmaceutical internalization and retention of the progeny radionuclides 221Fr (t1/2 = 4.80 m) and 213Bi (t1/2 = 45.6 m). RESULTS For the cumulative 225Ac decay chain, our self- and cross-dose nuclear S-values were both in good agreement with S-values published by MIRDcell, with per cent differences ranging from - 2.7 to - 8.7% for the various radionuclide source locations. Source location had greater effects on self-dose S-values than the intercellular cross-dose S-values. Cumulative 225Ac decay chain self-dose S-values increased from 0.167 to 0.364 GyBq-1 s-1 with radionuclide internalization from the cell surface into the cell. When progeny migration from the target site was modelled, the cumulative self-dose S-values to the cell nucleus decreased by up to 71% and 21% for 221Fr and 213Bi retention, respectively. CONCLUSIONS Our GATE Monte Carlo simulations resulted in cellular S-values in agreement with existing MIRD S-values for the alpha-emitting radionuclides in the 225Ac decay chain. To obtain accurate absorbed dose estimates in 225Ac studies, accurate understanding of daughter migration is critical for optimized injected activities. Future work will investigate other novel preclinical alpha-emitting radionuclides to evaluate therapeutic potency and explore realistic cellular geometries corresponding to targeted cancer cell lines.
Collapse
Affiliation(s)
- Helena Koniar
- Life Sciences Division, TRIUMF, Vancouver, BC, Canada.
- Department of Physics and Astronomy, University of British Columbia, Vancouver, BC, Canada.
| | - Cassandra Miller
- Department of Physics and Astronomy, University of British Columbia, Vancouver, BC, Canada
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Arman Rahmim
- Department of Physics and Astronomy, University of British Columbia, Vancouver, BC, Canada
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC, Canada
| | - Paul Schaffer
- Life Sciences Division, TRIUMF, Vancouver, BC, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC, Canada
- Department of Chemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Carlos Uribe
- Department of Radiology, University of British Columbia, Vancouver, BC, Canada
- Functional Imaging, BC Cancer, Vancouver, BC, Canada
| |
Collapse
|
34
|
Nock BA, Kanellopoulos P, Joosten L, Mansi R, Maina T. Peptide Radioligands in Cancer Theranostics: Agonists and Antagonists. Pharmaceuticals (Basel) 2023; 16:ph16050674. [PMID: 37242457 DOI: 10.3390/ph16050674] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/25/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
The clinical success of radiolabeled somatostatin analogs in the diagnosis and therapy-"theranostics"-of tumors expressing the somatostatin subtype 2 receptor (SST2R) has paved the way for the development of a broader panel of peptide radioligands targeting different human tumors. This approach relies on the overexpression of other receptor-targets in different cancer types. In recent years, a shift in paradigm from internalizing agonists to antagonists has occurred. Thus, SST2R-antagonist radioligands were first shown to accumulate more efficiently in tumor lesions and clear faster from the background in animal models and patients. The switch to receptor antagonists was soon adopted in the field of radiolabeled bombesin (BBN). Unlike the stable cyclic octapeptides used in the case of somatostatin, BBN-like peptides are linear, fast to biodegradable and elicit adverse effects in the body. Thus, the advent of BBN-like antagonists provided an elegant way to obtain effective and safe radiotheranostics. Likewise, the pursuit of gastrin and exendin antagonist-based radioligands is advancing with exciting new outcomes on the horizon. In the present review, we discuss these developments with a focus on clinical results, commenting on challenges and opportunities for personalized treatment of cancer patients by means of state-of-the-art antagonist-based radiopharmaceuticals.
Collapse
Affiliation(s)
- Berthold A Nock
- Molecular Radiopharmacy, INRaSTES, NCSR "Demokritos", 15310 Athens, Greece
| | | | - Lieke Joosten
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Rosalba Mansi
- Division of Radiopharmaceutical Chemistry, Clinic of Radiology and Nuclear Medicine, University Hospital Basel, 4031 Basel, Switzerland
| | - Theodosia Maina
- Molecular Radiopharmacy, INRaSTES, NCSR "Demokritos", 15310 Athens, Greece
| |
Collapse
|
35
|
Feuerecker B, Kratochwil C, Ahmadzadehfar H, Morgenstern A, Eiber M, Herrmann K, Pomykala KL. Clinical Translation of Targeted α-Therapy: An Evolution or a Revolution? J Nucl Med 2023; 64:685-692. [PMID: 37055224 DOI: 10.2967/jnumed.122.265353] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/10/2023] [Indexed: 04/15/2023] Open
Abstract
The field of radioligand therapy has advanced greatly in recent years, driven largely by β-emitting therapies targeting somatostatin receptor-expressing tumors and the prostate-specific membrane antigen. Now, more clinical trials are under way to evaluate α-emitting targeted therapies as potential next-generation theranostics with even higher efficacy due to their high linear energy and short range in human tissues. In this review, we summarize the important studies ranging from the first Food and Drug Administration-approved α-therapy, 223Ra-dichloride, for treatment of bone metastases in castration-resistant prostate cancer, including concepts in clinical translation such as targeted α-peptide receptor radiotherapy and 225Ac-PSMA-617 for treatment of prostate cancer, innovative therapeutic models evaluating new targets, and combination therapies. Targeted α-therapy is one of the most promising fields in novel targeted cancer therapy, with several early- and late-stage clinical trials for neuroendocrine tumors and metastatic prostate cancer already in progress, along with significant interest and investment in additional early-phase studies. Together, these studies will help us understand the short- and long-term toxicity of targeted α-therapy and potentially identify suitable therapeutic combination partners.
Collapse
Affiliation(s)
- Benedikt Feuerecker
- Department of Nuclear Medicine, Technische Universität München, München, Germany
- Department of Radiology, Technische Universität München, München, Germany
- German Cancer Consortium, partner sites München, Heidelberg, and Essen, Germany
- Department of Radiology, University Hospital, LMU München, München, Germany
| | - Clemens Kratochwil
- Department of Nuclear Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Hojjat Ahmadzadehfar
- Department of Nuclear Medicine, Klinikum Westfalen-Knappschaftskrankenhaus, Dortmund, Germany
| | | | - Matthias Eiber
- Department of Nuclear Medicine, Technische Universität München, München, Germany
| | - Ken Herrmann
- German Cancer Consortium, partner sites München, Heidelberg, and Essen, Germany
- Department of Nuclear Medicine, University Hospital Essen, Essen, Germany; and
| | - Kelsey L Pomykala
- Institute for Artificial Intelligence in Medicine, University Hospital Essen, Essen, Germany
| |
Collapse
|
36
|
Satapathy S, Chandekar KR, Bal C. Gastro-Enteric-Pancreatic Neuroendocrine Tumor Treatment: Actinium-225-DOTATATE and Combined Therapies. PET Clin 2023; 18:215-221. [PMID: 36858746 DOI: 10.1016/j.cpet.2022.11.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
The role of lutetium-177-DOTATATE in advanced well-differentiated gastro-entero-pancreatic neuroendocrine tumors is well established. However, there is a scope for improving treatment outcomes. Actinium-225-DOTATATE is a form of targeted alpha therapy (TAT) that results in more efficient tumor cell killing owing to the substantially higher linear energy transfer of alpha particles. Systemic TAT is also safe given that the shorter path length of the alpha particles spares the surrounding healthy tissue and results in relatively fewer adverse events. Combination therapies with radiosensitizing and other chemotherapeutic agents have also gained popularity, especially in the setting of higher grade and fluorodeoxyglucose-avid tumors.
Collapse
Affiliation(s)
- Swayamjeet Satapathy
- Department of Nuclear Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| | - Kunal Ramesh Chandekar
- Department of Nuclear Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| | - Chandrasekhar Bal
- Department of Nuclear Medicine, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India.
| |
Collapse
|
37
|
Neuroendocrine Tumor Therapy Response Assessment. PET Clin 2023; 18:267-286. [PMID: 36858748 DOI: 10.1016/j.cpet.2022.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Peptide receptor radionuclide therapy has become an integral part of management of neuroendocrine neoplasms. Gallium-68- and lutetium-177-labeled somatostatin receptor analogues have replaced yttrium-90- and 111-indium-based tracers. Several newer targeted therapies are also being used in clinical and research settings. It is imperative to accurately evaluate the response to these agents. The characteristics of NENs and the response patterns of the targeted therapies make response assessment in this group challenging. This article provides an overview of the strengths and weaknesses of the various biomarkers available for response assessment.
Collapse
|
38
|
Rubira L, Deshayes E, Santoro L, Kotzki PO, Fersing C. 225Ac-Labeled Somatostatin Analogs in the Management of Neuroendocrine Tumors: From Radiochemistry to Clinic. Pharmaceutics 2023; 15:1051. [PMID: 37111537 PMCID: PMC10146019 DOI: 10.3390/pharmaceutics15041051] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/18/2023] [Accepted: 03/22/2023] [Indexed: 04/29/2023] Open
Abstract
The widespread use of peptide receptor radionuclide therapy (PRRT) represents a major therapeutic breakthrough in nuclear medicine, particularly since the introduction of 177Lu-radiolabeled somatostatin analogs. These radiopharmaceuticals have especially improved progression-free survival and quality of life in patients with inoperable metastatic gastroenteropancreatic neuroendocrine tumors expressing somatostatin receptors. In the case of aggressive or resistant disease, the use of somatostatin derivatives radiolabeled with an alpha-emitter could provide a promising alternative. Among the currently available alpha-emitting radioelements, actinium-225 has emerged as the most suitable candidate, especially regarding its physical and radiochemical properties. Nevertheless, preclinical and clinical studies on these radiopharmaceuticals are still few and heterogeneous, despite the growing momentum for their future use on a larger scale. In this context, this report provides a comprehensive and extensive overview of the development of 225Ac-labeled somatostatin analogs; particular emphasis is placed on the challenges associated with the production of 225Ac, its physical and radiochemical properties, as well as the place of 225Ac-DOTATOC and 225Ac-DOTATATE in the management of patients with advanced metastatic neuroendocrine tumors.
Collapse
Affiliation(s)
- Léa Rubira
- Nuclear Medicine Department, Institut Régional du Cancer de Montpellier (ICM), University Montpellier, 34090 Montpellier, France
| | - Emmanuel Deshayes
- Nuclear Medicine Department, Institut Régional du Cancer de Montpellier (ICM), University Montpellier, 34090 Montpellier, France
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, University Montpellier, Institut Régional du Cancer de Montpellier (ICM), 34298 Montpellier, France
| | - Lore Santoro
- Nuclear Medicine Department, Institut Régional du Cancer de Montpellier (ICM), University Montpellier, 34090 Montpellier, France
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, University Montpellier, Institut Régional du Cancer de Montpellier (ICM), 34298 Montpellier, France
| | - Pierre Olivier Kotzki
- Nuclear Medicine Department, Institut Régional du Cancer de Montpellier (ICM), University Montpellier, 34090 Montpellier, France
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, University Montpellier, Institut Régional du Cancer de Montpellier (ICM), 34298 Montpellier, France
| | - Cyril Fersing
- Nuclear Medicine Department, Institut Régional du Cancer de Montpellier (ICM), University Montpellier, 34090 Montpellier, France
- IBMM, University Montpellier, CNRS, ENSCM, 34293 Montpellier, France
| |
Collapse
|
39
|
Watabe T, Kaneda-Nakashima K, Shirakami Y, Kadonaga Y, Ooe K, Wang Y, Haba H, Toyoshima A, Cardinale J, Giesel FL, Tomiyama N, Fukase K. Targeted α-therapy using astatine ( 211At)-labeled PSMA1, 5, and 6: a preclinical evaluation as a novel compound. Eur J Nucl Med Mol Imaging 2023; 50:849-858. [PMID: 36344651 PMCID: PMC9852121 DOI: 10.1007/s00259-022-06016-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 10/18/2022] [Indexed: 11/10/2022]
Abstract
PURPOSE Targeted α-therapy (TAT) for prostate-specific membrane antigen (PSMA) is a promising treatment for metastatic castration-resistant prostate cancer (CRPC). Astatine is an α-emitter (half-life=7.2 h) that can be produced by a 30-MeV cyclotron. This study evaluated the treatment effect of 211At-labeled PSMA compounds in mouse xenograft models. METHODS Tumor xenograft models were established by subcutaneous transplantation of human prostate cancer cells (LNCaP) in NOD/SCID mouse. [211At]PSMA1, [211At]PSMA5, or [211At]PSMA6 was administered to LNCaP xenograft mice to evaluate biodistribution at 3 and 24 h. The treatment effect was evaluated by administering [211At]PSMA1 (0.40 ± 0.07 MBq), [211At]PSMA5 (0.39 ± 0.03 MBq), or saline. Histopathological evaluation was performed for the at-risk organs at 3 and 6 weeks after administration. RESULTS [211At]PSMA5 resulted in higher tumor retention compared to [211At]PSMA1 and [211At]PSMA6 (30.6 ± 17.8, 12.4 ± 4.8, and 19.1 ± 4.5 %ID/g at 3 h versus 40.7 ± 2.6, 8.7 ± 3.5, and 18.1 ± 2.2%ID/g at 24 h, respectively), whereas kidney excretion was superior in [211At]PSMA1 compared to [211At]PSMA5 and [211At]PSMA6. An excellent treatment effect on tumor growth was observed after [211At]PSMA5 administration. [211At]PSMA1 also showed a substantial treatment effect; however, the tumor size was relatively larger compared to that with [211At]PSMA5. In the histopathological evaluation, regenerated tubules were detected in the kidneys at 3 and 6 weeks after the administration of [211At]PSMA5. CONCLUSION TAT using [211At]PSMA5 resulted in excellent tumor growth suppression with minimal side effects in the normal organs. [211At]PSMA5 should be considered a new possible TAT for metastatic CRPC, and translational prospective trials are warranted.
Collapse
Affiliation(s)
- Tadashi Watabe
- Department of Nuclear Medicine and Tracer Kinetics, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Institute for Radiation Sciences, Osaka University, Osaka, Japan.
| | - Kazuko Kaneda-Nakashima
- Institute for Radiation Sciences, Osaka University, Osaka, Japan
- Core for Medicine and Science Collaborative Research and Education, Project Research Center for Fundamental Sciences, Graduate School of Science, Osaka University, Osaka, Japan
| | | | - Yuichiro Kadonaga
- Department of Nuclear Medicine and Tracer Kinetics, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Institute for Radiation Sciences, Osaka University, Osaka, Japan
| | - Kazuhiro Ooe
- Department of Nuclear Medicine and Tracer Kinetics, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
- Institute for Radiation Sciences, Osaka University, Osaka, Japan
| | - Yang Wang
- Nishina Center for Accelerator-Based Science, RIKEN, Tokyo, Japan
| | - Hiromitsu Haba
- Nishina Center for Accelerator-Based Science, RIKEN, Tokyo, Japan
| | | | - Jens Cardinale
- Department of Nuclear Medicine, Dusseldorf University, Düsseldorf, Germany
| | - Frederik L Giesel
- Institute for Radiation Sciences, Osaka University, Osaka, Japan
- Department of Nuclear Medicine, Dusseldorf University, Düsseldorf, Germany
| | - Noriyuki Tomiyama
- Institute for Radiation Sciences, Osaka University, Osaka, Japan
- Department of Radiology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Koichi Fukase
- Institute for Radiation Sciences, Osaka University, Osaka, Japan
- Department of Chemistry, Graduate School of Science, Osaka University, Osaka, Japan
| |
Collapse
|
40
|
Sartor O, Baghian A. Prostate specific membrane antigen binding radiopharmaceuticals: Current data and new concepts. Front Med (Lausanne) 2022; 9:1060922. [PMID: 36561718 PMCID: PMC9763319 DOI: 10.3389/fmed.2022.1060922] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 11/17/2022] [Indexed: 12/12/2022] Open
Abstract
Prostate specific membrane antigen (PSMA) represents a validated target for prostate cancer therapeutics. The phase III VISION study with 177lutetium (177Lu)-PSMA-617 represented a pivotal step forward and the FDA has now approved this agent in advanced metastatic castrate-resistant prostate cancer (mCRPC). A number of other PSMA targeted radiopharmaceuticals are now under development. Some of these agents are targeted to PSMA via monoclonal antibodies such as J591 and TLX591. Others are targeted to PSMA via small molecules such as PSMA-617, PSMA I&T, MIP-1095, etc. In addition to the use of various ligands, multiple isotopes are now in clinical trials. Beta emitters in development include 177Lu, 131iodide (131I), and 67copper (67Cu). Targeted alpha emitters potentially include 225actinium (225Ac), 227thorium (227Th), and 212lead (212Pb). Phase III trials are underway with both 177Lu-PSMA-617 and 177Lu-PSMA I&T in mCRPC. Single dose phase I trials are complete with 225Ac-J591 but additional data are need to launch a phase III. Data are promising with 225Ac-PSMA-617 but concerns remain over salivary and renal toxicity. Tandem therapies are also considered combining both beta and alpha-targeted therapy. Taken together the field of PSMA targeted radiopharmaceuticals is rapidly developing. The targeted alpha therapies are particularly promising and several developmental paths forward are being considered in the near future.
Collapse
Affiliation(s)
- Oliver Sartor
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States,Department of Urology, Tulane University School of Medicine, New Orleans, LA, United States,*Correspondence: Oliver Sartor,
| | - Ali Baghian
- Section of Hematology and Medical Oncology, Deming Department of Medicine, Tulane University School of Medicine, New Orleans, LA, United States,Department of Population and Public Health Sciences, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
41
|
Rodak M, Dekempeneer Y, Wojewódzka M, Caveliers V, Covens P, Miller BW, Sevenois MB, Bruchertseifer F, Morgenstern A, Lahoutte T, D'Huyvetter M, Pruszyński M. Preclinical Evaluation of 225Ac-Labeled Single-Domain Antibody for the Treatment of HER2pos Cancer. Mol Cancer Ther 2022; 21:1835-1845. [PMID: 36129807 PMCID: PMC9716241 DOI: 10.1158/1535-7163.mct-21-1021] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 05/25/2022] [Accepted: 09/16/2022] [Indexed: 01/12/2023]
Abstract
Human epidermal growth factor receptor type 2 (HER2) is overexpressed in various cancers; thus, HER2-targeting single-domain antibodies (sdAb) could offer a useful platform for radioimmunotherapy. In this study, we optimized the labeling of an anti-HER2-sdAb with the α-particle-emitter 225Ac through a DOTA-derivative. The formed radioconjugate was tested for binding affinity, specificity and internalization properties, whereas cytotoxicity was evaluated by clonogenic and DNA double-strand-breaks assays. Biodistribution studies were performed in mice bearing subcutaneous HER2pos tumors to estimate absorbed doses delivered to organs and tissues. Therapeutic efficacy and potential toxicity were assessed in HER2pos intraperitoneal ovarian cancer model and in healthy C57Bl/6 mice. [225Ac]Ac-DOTA-2Rs15d exhibited specific cell uptake and cell-killing capacity in HER2pos cells (EC50 = 3.9 ± 1.1 kBq/mL). Uptake in HER2pos lesions peaked at 3 hours (9.64 ± 1.69% IA/g), with very low accumulation in other organs (<1% IA/g) except for kidneys (11.69 ± 1.10% IA/g). α-camera imaging presented homogeneous uptake of radioactivity in tumors, although heterogeneous in kidneys, with a higher signal density in cortex versus medulla. In mice with HER2pos disseminated tumors, repeated administration of [225Ac]Ac-DOTA-2Rs15d significantly prolonged survival (143 days) compared to control groups (56 and 61 days) and to the group treated with HER2-targeting mAb trastuzumab (100 days). Histopathologic evaluation revealed signs of kidney toxicity after repeated administration of [225Ac]Ac-DOTA-2Rs15d. [225Ac]Ac-DOTA-2Rs15d efficiently targeted HER2pos cells and was effective in treatment of intraperitoneal disseminated tumors, both alone and as an add-on combination with trastuzumab, albeit with substantial signs of inflammation in kidneys. This study warrants further development of [225Ac]Ac-DOTA-2Rs15d.
Collapse
Affiliation(s)
- Magdalena Rodak
- Institute of Nuclear Chemistry and Technology, Warsaw, Poland
| | - Yana Dekempeneer
- Department of Medical Imaging, In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Vicky Caveliers
- Department of Medical Imaging, In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
- Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Peter Covens
- Department of Medical Imaging, In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - Brian W. Miller
- Department of Medical Imaging, University of Arizona, Tucson, Arizona
| | - Matthijs B. Sevenois
- Department of Medical Imaging, In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | | | | | - Tony Lahoutte
- Department of Medical Imaging, In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
- Nuclear Medicine Department, UZ Brussel, Brussels, Belgium
| | - Matthias D'Huyvetter
- Department of Medical Imaging, In Vivo Cellular and Molecular Imaging Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - Marek Pruszyński
- Institute of Nuclear Chemistry and Technology, Warsaw, Poland
- NOMATEN Centre of Excellence, National Centre for Nuclear Research, Otwock, Poland
| |
Collapse
|
42
|
Multiplexed Imaging Reveals the Spatial Relationship of the Extracellular Acidity-Targeting pHLIP with Necrosis, Hypoxia, and the Integrin-Targeting cRGD Peptide. Cells 2022; 11:cells11213499. [DOI: 10.3390/cells11213499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 10/24/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
pH (low) insertion peptides (pHLIPs) have been developed for cancer imaging and therapy targeting the acidic extracellular microenvironment. However, the characteristics of intratumoral distribution (ITD) of pHLIPs are not yet fully understood. This study aimed to reveal the details of the ITD of pHLIPs and their spatial relationship with other tumor features of concern. The fluorescent dye-labeled pHLIPs were intravenously administered to subcutaneous xenograft mouse models of U87MG and IGR-OV1 expressing αVβ3 integrins (using large necrotic tumors). The αVβ3 integrin-targeting Cy5.5-RAFT-c(-RGDfK-)4 was used as a reference. In vivo and ex vivo fluorescence imaging, whole-tumor section imaging, fluorescence microscopy, and multiplexed fluorescence colocalization analysis were performed. The ITD of fluorescent dye-labeled pHLIPs was heterogeneous, having a high degree of colocalization with necrosis. A direct one-to-one comparison of highly magnified images revealed the cellular localization of pHLIP in pyknotic, karyorrhexis, and karyolytic necrotic cells. pHLIP and hypoxia were spatially contiguous but not overlapping cellularly. The hypoxic region was found between the ITDs of pHLIP and the cRGD peptide and the Ki-67 proliferative activity remained detectable in the pHLIP-accumulated regions. The results provide a better understanding of the characteristics of ITD of pHLIPs, leading to new insights into the theranostic applications of pHLIPs.
Collapse
|
43
|
Delpassand ES, Tworowska I, Esfandiari R, Torgue J, Hurt J, Shafie A, Núñez R. Targeted α-Emitter Therapy with 212Pb-DOTAMTATE for the Treatment of Metastatic SSTR-Expressing Neuroendocrine Tumors: First-in-Humans Dose-Escalation Clinical Trial. J Nucl Med 2022; 63:1326-1333. [PMID: 34992153 PMCID: PMC9454455 DOI: 10.2967/jnumed.121.263230] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 01/04/2022] [Indexed: 01/26/2023] Open
Abstract
Peptide receptor radiotherapy with somatostatin analogs has been successfully used for years as a treatment for somatostatin-overexpressing tumors. Treatment of neuroendocrine tumors (NETs) with the β-particle emitter 177Lu-DOTATATE is currently considered the standard of care for subjects with gastroenteropancreatic NETs. Despite the success of 177Lu-DOTATATE, there remains significant room for improvement in terms of both safety and efficacy. Targeted α-emitter therapy with isotopes such as 212Pb has the potential to improve both. Here, we present the preliminary results of the phase 1 first-in-humans dose-escalation trial evaluating 212Pb-DOTAMTATE (a bifunctional metal chelator [DOTAM] and the SSTR-targeting peptide [TATE]) in patients with somatostatin receptor-positive NETs. Methods: Twenty subjects with histologically confirmed NETs, prior positive somatostatin analog scans, and no prior history of 177Lu/90Y/111In peptide receptor radiotherapy, with different primary sites of the disease, were enrolled. Treatment began with single ascending doses of 212Pb-DOTAMTATE, with subsequent cohorts receiving an incremental 30% dose increase, which was continued until a tumor response or a dose-limiting toxicity was observed. This was followed by a multiple ascending dose regimen. The recommended phase 2 dose regimen consisted of 4 cycles of 2.50 MBq/kg (67.6 μCi/kg) of 212Pb-DOTAMTATE administered at 8-wk intervals, intravenously. Results: Ten subjects received the highest dose, 2.50 MBq/kg/cycle (67.6 μCi/kg/cycle). Treatment was well tolerated, with the most common treatment-emergent adverse events being nausea, fatigue, and alopecia. No serious treatment-emergent adverse events were related to the study drug, and no subjects required treatment delay or a dose reduction. An objective radiologic response of 80% was observed for the first 10 subjects treated at the recommended phase 2 dose. Conclusion: Targeted α-therapy with 212Pb-DOTAMTATE has been shown to be well tolerated. Preliminary efficacy results are highly promising. If these results are confirmed in a larger, multicenter clinical trial, 212Pb-DOTAMTATE would provide a substantial benefit over currently Food and Drug Administration-approved therapies for patients with metastatic or inoperable SSTR-expressing NETs regardless of the grade and location of the primary tumor.
Collapse
Affiliation(s)
- Ebrahim S. Delpassand
- Excel Diagnostics and Nuclear Oncology Center, Houston, Texas;,RadioMedix, Inc., Houston, Texas; and
| | | | | | | | | | - Afshin Shafie
- Excel Diagnostics and Nuclear Oncology Center, Houston, Texas
| | - Rodolfo Núñez
- Excel Diagnostics and Nuclear Oncology Center, Houston, Texas;
| |
Collapse
|
44
|
Hope TA, Pavel M, Bergsland EK. Neuroendocrine Tumors and Peptide Receptor Radionuclide Therapy: When Is the Right Time? J Clin Oncol 2022; 40:2818-2829. [PMID: 35649195 PMCID: PMC9390818 DOI: 10.1200/jco.22.00176] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/14/2022] [Accepted: 04/18/2022] [Indexed: 12/29/2022] Open
Abstract
Since its approval in 2018 by the US Food and Drug Administration, peptide receptor radionuclide therapy (PRRT) has become a mainstay in the treatment of neuroendocrine tumors. Lutetium-177-DOTATATE, the only approved agent, is indicated for the treatment of gastroenteropancreatic-neuroendocrine tumors. Although patient selection appears straightforward with somatostatin receptor-positron emission tomography, there is considerable complexity when deciding which patients to treat and when to start PRRT. Herein, we review the many factors that affect patient selection, focusing on the optimal patients to treat. Although significant effort has been expended to determine which patients benefit the most from PRRT, a validated predictive biomarker remains elusive. Although PRRT has been used for more than 2 decades in Europe and standards of care exist for safe treatment, there remain numerous questions regarding when PRRT should be used relative to other treatments. It is important to remember that multidisciplinary discussions are essential. Currently, there are a number of ongoing studies looking to assess the efficacy of PRRT compared with other treatment options and to optimize treatment through combination therapy, different dosing strategies, or use of different radionuclides and radioligands.
Collapse
Affiliation(s)
- Thomas A. Hope
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA
- Helen Diller Family Comprehensive Cancer Centre, University of California, San Francisco, San Francisco, CA
- Department of Radiology, San Francisco VA Medical Center, San Francisco, CA
| | - Marianne Pavel
- Department of Medicine 1, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Emily K. Bergsland
- Helen Diller Family Comprehensive Cancer Centre, University of California, San Francisco, San Francisco, CA
- Division of Hematology/Oncology, Department of Medicine, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
45
|
Ballal S, Yadav MP, Tripathi M, Sahoo RK, Bal C. Survival Outcomes in Metastatic Gastroenteropancreatic Neuroendocrine Tumor Patients receiving Concomitant 225Ac-DOTATATE Targeted Alpha Therapy and Capecitabine: A Real-world Scenario Management Based Long-term Outcome Study. J Nucl Med 2022; 64:jnumed.122.264043. [PMID: 35863893 DOI: 10.2967/jnumed.122.264043] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 07/15/2022] [Indexed: 11/16/2022] Open
Abstract
Rationale: Although the short-term results of targeted alpha therapy (TAT) with 225Ac-DOTATATE in gastroenteropancreatic neuroendocrine tumors (GEP-NETs) have proven effective, none have assessed the long-term outcome results. In this study, we aimed to evaluate the long-term outcome of 225Ac-DOTATATE targeted alpha therapy (TAT) in patients with somatostatin receptor (SSTR)-expressing advanced-stage metastatic gastroenteropancreatic neuroendocrine tumors (GEP-NETs). Methods: Patients with 68Ga-DOTANOC PET/CT scans showing moderate-to-high SSTR expression were recruited. Systemic TAT was performed in 91 adults with GEP-NET [54 males, and 37 females] mean age 54 years (y) (range: 25-75y)] using 225Ac-DOTATATE (100-120 kBq/kg body weight). All patients were given capecitabine therapy as a radiosensitizer (dose 2 g/day) from day 0 to 14 of every 225Ac-DOTATATE treatment cycle. Patients were categorized into three groups based on the status of prior 177Lu-PRRT: prior 177Lu-PRRT-refractory-group; prior 177Lu-PRRT-disease-control group; and 177Lu-PRRT naïve group. Primary endpoints were overall survival (OS), and secondary endpoints included progression-free survival (PFS), objective tumour response, clinical response, and the assessment of treatment-related toxicities. Results: Among the 91 patients, 57 underwent prior 177Lu-DOTATATE therapy [24 disease controlled (PR/SD), 33 progressive diseases (PD)]. A total of 453 225Ac-DOTATATE TAT cycles were administered [median four cycles per patient; range 1-10] in a median follow-up duration of 24 months (range 5-41mo). Median OS was not attained with a 24-month overall survival probability of 70.8%. In multivariate analysis, prognostic factors associated with a poor OS included, the presence bone metastases [HR: 2.501; 95% CI: 1.826 - 5.791; P<0.032], and 225Ac-DOTATATE therapy refractory disease [HR: 8.781; 95% CI: 3.843 - 20.062; P<0.0001]. Median PFS was also not reached with a 24-month progression-free survival probability of 67.5%. The multivariate analysis revealed only 177Lu-PRRT refractory disease significantly associated with a reduced PFS. [HR: 14.338; 95% CI: 1.853 - 97.698; P = 0.011]. Two of 79 patients (2.5%) with assessable disease experienced complete response; 38 (48%) had a partial response, 23 (29%) had SD, and 16 (20.2%) had PD. PD was observed in more patients from the prior 177Lu-PRRT-refractory group (11/33; 34%) as compared to 177Lu-PRRT-naïve patients (4/24; 11%), P-0.056. Patients from the prior 177Lu-PRRT-refractory group had the highest risk of poor PFS [HR:13.91; 95% CI: 4.45 - 42.271; P = 0.0009]. A significant clinical benefit was achieved post 225Ac-DOTATATE therapy with minimal treatment-related toxicities. Conclusion: The long-term results reveal 225Ac-DOTATATE TAT has shown promising results and improves overall survival, even in patients refractory to prior 177Lu-DOTATATE treatment, with transient and acceptable adverse effects.
Collapse
|
46
|
Repair of α-particle-induced DNA damage in peripheral blood mononuclear cells after internal ex vivo irradiation with 223Ra. Eur J Nucl Med Mol Imaging 2022; 49:3981-3988. [PMID: 35759008 PMCID: PMC9525426 DOI: 10.1007/s00259-022-05860-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/30/2022] [Indexed: 11/10/2022]
Abstract
Purpose As α-emitters for radiopharmaceutical therapies are administered systemically by intravenous injection, blood will be irradiated by α-particles that induce clustered DNA double-strand breaks (DSBs). Here, we investigated the induction and repair of DSB damage in peripheral blood mononuclear cells (PBMCs) as a function of the absorbed dose to the blood following internal ex vivo irradiation with [223Ra]RaCl2. Methods Blood samples of ten volunteers were irradiated by adding [223Ra]RaCl2 solution with different activity concentrations resulting in absorbed doses to the blood of 3 mGy, 25 mGy, 50 mGy and 100 mGy. PBMCs were isolated, divided in three parts and either fixed directly (d-samples) or after 4 h or 24 h culture. After immunostaining, the induced γ-H2AX α-tracks were counted. The time-dependent decrease in α-track frequency was described with a model assuming a repair rate R and a fraction of non-repairable damage Q. Results For 25 mGy, 50 mGy and 100 mGy, the numbers of α-tracks were significantly increased compared to baseline at all time points. Compared to the corresponding d-samples, the α-track frequency decreased significantly after 4 h and after 24 h. The repair rates R were (0.24 ± 0.05) h−1 for 25 mGy, (0.16 ± 0.04) h−1 for 50 mGy and (0.13 ± 0.02) h−1 for 100 mGy, suggesting faster repair at lower absorbed doses, while Q-values were similar. Conclusion The results obtained suggest that induction and repair of the DSB damage depend on the absorbed dose to the blood. Repair rates were similar to what has been observed for irradiation with low linear energy transfer.
Collapse
|
47
|
Abstract
Theranostic applications with radio-isotopes currently are rapidly progressing and expand nuclear medicine application in clinical routine. Alpha emitting isotopes, in particular, have long been hypothesized to achieve relevant advances for the treatment of malignancies. Here, an overview of their properties and the knowledge of radiobiology is reviewed in view of clinical translation. Clinical evidence of radiopharmaceuticals based on alpha emitters is summarized with a focus on recent developments for treatment of metastasized castration resistant prostate cancer.
Collapse
Affiliation(s)
- Matthias Miederer
- Department of Nuclear Medicine, University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
48
|
Grey N, Silosky M, Lieu CH, Chin BB. Current status and future of targeted peptide receptor radionuclide positron emission tomography imaging and therapy of gastroenteropancreatic-neuroendocrine tumors. World J Gastroenterol 2022; 28:1768-1780. [PMID: 35633909 PMCID: PMC9099199 DOI: 10.3748/wjg.v28.i17.1768] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/07/2022] [Accepted: 03/27/2022] [Indexed: 02/06/2023] Open
Abstract
Theranostics is the highly targeted molecular imaging and therapy of tumors. Targeted peptide receptor radionuclide therapy has taken the lead in demonstrating the safety and effectiveness of this molecular approach to treating cancers. Metastatic, well-differentiated gastroenteropancreatic neuroendocrine tumors may be most effectively imaged and treated with DOTATATE ligands. We review the current practice, safety, advantages, and limitations of DOTATATE based theranostics. Finally, we briefly describe the exciting new areas of development and future directions of gastroenteropancreatic neuroendocrine tumor theranostics.
Collapse
Affiliation(s)
- Neil Grey
- Radiology-Nuclear Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Michael Silosky
- Department of Radiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Christopher H Lieu
- Medical Oncology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Bennett B Chin
- Department of Radiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO 80045, United States
| |
Collapse
|
49
|
Yadav MP, Ballal S, Sahoo RK, Bal C. Efficacy and safety of 225Ac-DOTATATE targeted alpha therapy in metastatic paragangliomas: a pilot study. Eur J Nucl Med Mol Imaging 2022; 49:1595-1606. [PMID: 34837103 PMCID: PMC8626283 DOI: 10.1007/s00259-021-05632-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Accepted: 11/20/2021] [Indexed: 12/16/2022]
Abstract
PURPOSE In this study, we aim to evaluate the efficacy and safety of 225AC-DOTATATE targeted alpha therapy in advanced-stage paragangliomas (PGLs). METHODS Nine (6 males and 3 females) consecutive patients with histologically proven PGLs were treated with 225Ac-DOTATATE targeted alpha therapy (TAT) and concomitant radiosensitizer, capecitabine, at 8-weekly intervals up to a cumulative activity of ~ 74 MBq. The primary endpoint included evaluating therapy response and disease control rate (DCR) using the RECIST 1.1 criteria. Additional secondary endpoints comprised clinical response assessment using EORTC QLQ-H&N35 questionnaire, Karnofsky Performance Scale (KPS), Eastern Cooperative Oncology Group performance status (ECOG), analgesic score (AS), dose alterations of anti-hypertensive drugs (anti-HTN), and the safety and side-effect profile evaluation as per CTCAE criteria version 5.0. RESULTS Following 225Ac-DOTATATE treatment, morphological response revealed partial response in 50%, stable disease in 37.5%, and disease progression in 12.5%, with a DCR of 87.5%. Similarly, the symptomatic response was remarkable, and anti-HTN drugs were stopped in 25% and reduced in 37.5%. Another significant finding in our study revealed a morphologic DCR of 66.6% (2/3) in patients who failed previous lutetium-177 peptide receptor radionuclide therapy (177Lu-PRRT). Regarding the KPS, ECOG, and AS performance scores, a notable improvement was observed post-225Ac-DOTATATE treatment. The QLQ-H&N35 symptom scores evaluated in seven H&N PGL patients showed significant improvement in all aspects. No improvement in sexual function was noted (P = 0.3559). Despite the significant reduction in the analgesic score post-treatment (P = 0.0031), the QLQ-H&N35 revealed only marginal significance concerning the intake of pain killers (P = 0.1723). No grade III/IV hematological, renal, and hepatological toxicities were noted. CONCLUSION The evidence from this study suggests 225Ac-DOTATATE therapy is effective and safe in the treatment of advanced-stage PGLs and also reports a clear benefit even in patient's refractory to the previous 177Lu-PRRT.
Collapse
Affiliation(s)
- Madhav Prasad Yadav
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Sanjana Ballal
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Ranjit Kumar Sahoo
- Department of Medical Oncology, BR Ambedkar Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Chandrasekhar Bal
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
50
|
Park EA, Graves SA, Menda Y. The Impact of Radiopharmaceutical Therapy on Renal Function. Semin Nucl Med 2022; 52:467-474. [DOI: 10.1053/j.semnuclmed.2022.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/13/2022] [Accepted: 02/20/2022] [Indexed: 11/11/2022]
|