1
|
Santos JF, Laere CV, Silva CD, Cassells I, Fernandes C, Raposinho P, Belchior A, Pinto CIG, Mendes F, Cawthorne C, Ooms M, Voorde MVD, Cleeren F, Paulo A. Mitochondria-tropic radioconjugates to enhance the therapeutic potential of terbium-161. EJNMMI Radiopharm Chem 2025; 10:18. [PMID: 40214871 PMCID: PMC11992321 DOI: 10.1186/s41181-025-00339-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Strategies that focus on delivering Auger electron emitters to highly radiosensitive intracellular targets-such as the nucleus, cell membrane, or mitochondria-are gaining attention. Targeting these organelles could enhance therapeutic efficacy while minimizing off-target toxicity by allowing lower administered doses. In this context, this study explores the therapeutic potential of 161Tb-labeled radiocomplexes that integrate the mitochondria-targeting triphenylphosphonium (TPP) moiety with a prostate-specific membrane antigen (PSMA) targeting vector. The goal is to assess these dual-targeted radiocomplexes for their ability to deliver conversion electrons (CE) and Auger electrons (AEs) to prostate cancer (PCa) cells, specifically targeting the mitochondria to enhance therapeutic efficacy. RESULTS Two novel radiocomplexes, [161Tb]Tb-TPP-PSMA and [161Tb]Tb-TPP-G3-PSMA, were synthesized with high radiochemical yield and purity. The proposed structures were validated using HPLC and ESI-MS analysis, with their natTb counterparts serving as reference compounds. In vitro experiments included cellular uptake, internalization, mitochondrial uptake, and DNA damage assays in PSMA-positive PCa cell lines. Clonogenic assays were performed to evaluate cell survival post-treatment. In vivo studies were conducted using SCID/Beige mice bearing PCa xenografts and involved µSPECT/CT imaging and radiometabolite analysis to evaluate biodistribution, pharmacokinetics, tumor uptake and in vivo stability of the radiocomplexes. Both [161Tb]Tb-TPP-PSMA and [161Tb]Tb-TPP-G3-PSMA showed high radiochemical stability and were efficiently internalized by PSMA-positive cells, while showing minimal uptake in PSMA-negative cells. These dual-targeted radiocomplexes demonstrated significantly higher mitochondrial uptake compared to the non-TPP-containing [161Tb]Tb-PSMA-617, leading to increased DNA damage and enhanced radiocytotoxicity. In vivo, the dual-targeted complexes demonstrated PSMA-specific tumor uptake and pharmacokinetics comparable to [161Tb]Tb-PSMA-617, with effective clearance from non-target tissues. CONCLUSIONS The TPP-modified 161Tb-radiocomplexes effectively targeted the mitochondria of PSMA-positive PCa cells, leading to increased DNA damage and reduced cell viability compared to single-targeted radiocomplexes. These findings suggest that dual-targeting strategies, which combine PSMA and mitochondrial targeting, can enhance the therapeutic potential of radiopharmaceuticals for prostate cancer treatment.
Collapse
Affiliation(s)
- Joana F Santos
- C2TN - Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Bobadela LRS, 2695-066, Portugal
| | - Camille Van Laere
- Nuclear Medical Applications, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, B-3000, Belgium
| | - Catarina D Silva
- C2TN - Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Bobadela LRS, 2695-066, Portugal
| | - Irwin Cassells
- Nuclear Medical Applications, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, B-3000, Belgium
| | - Célia Fernandes
- C2TN - Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Bobadela LRS, 2695-066, Portugal
- DECN - Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Bobadela LRS, 2695-066, Portugal
| | - Paula Raposinho
- C2TN - Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Bobadela LRS, 2695-066, Portugal
- DECN - Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Bobadela LRS, 2695-066, Portugal
| | - Ana Belchior
- C2TN - Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Bobadela LRS, 2695-066, Portugal
- DECN - Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Bobadela LRS, 2695-066, Portugal
| | - Catarina I G Pinto
- C2TN - Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Bobadela LRS, 2695-066, Portugal
| | - Filipa Mendes
- C2TN - Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Bobadela LRS, 2695-066, Portugal
- DECN - Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Bobadela LRS, 2695-066, Portugal
| | - Christopher Cawthorne
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, University of Leuven, Leuven, Belgium
| | - Maarten Ooms
- Nuclear Medical Applications, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Michiel Van de Voorde
- Nuclear Medical Applications, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Frederik Cleeren
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, B-3000, Belgium.
| | - António Paulo
- C2TN - Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Bobadela LRS, 2695-066, Portugal.
- DECN - Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Bobadela LRS, 2695-066, Portugal.
| |
Collapse
|
2
|
Nosrati Shanjani S, Łyczko M, Walczak R, Koźmiński P, Majka E, Narbutt J, Wojdowska W, Majkowska-Pilip A, Bilewicz A. Cytotoxicity Comparison of 99mTc-Labeled Peptide Antagonist and Agonist Targeting the SSTR2 Receptor in AR42J Cells. Molecules 2025; 30:1715. [PMID: 40333672 PMCID: PMC12029662 DOI: 10.3390/molecules30081715] [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: 02/23/2025] [Revised: 04/03/2025] [Accepted: 04/09/2025] [Indexed: 05/09/2025] Open
Abstract
Auger electrons are low-energy, high-linear-energy-transfer particles that deposit their energy over nanometers distances. Their biological impact depends heavily on where the radionuclide is localized within the cell. To verify the hypothesis that the cell membrane may be a better molecular target than the cytoplasm in Auger electron therapy, we investigated whether the radiotoxicity of 99mTc varied depending on its location in the cell. The behavior of peptide radiopharmaceuticals 99mTc-TECANT-1 targeted the cell membrane was compared with 99mTc-TEKTROTYD directed to the cytoplasm. Our findings confirmed that 99mTc-TECANT-1 displayed greater binding to AR-42-J cells than 99mTc-TEKTROTYD. Additionally, it was demonstrated that the receptor agonist 99mTc-TEKTROTYD is localized in more than 90% of the cytoplasm, while 99mTc-TECANT-1 is found in 60-80% of the cell membrane. When evaluating cell survival using the MTS assay, we observed that toxicity was significantly higher when 99mTc was targeted to the membrane compared to the cytoplasm. This indicates that, for 99mTc, as with 161Tb, the membrane is a more sensitive target for Auger electrons than the cytoplasm. Our results also suggest that receptor antagonists labelled with therapeutic doses of 99mTc may be effective in treating certain cancers. However, further detailed studies, particularly dosimetric investigations, are necessary to validate these findings.
Collapse
Affiliation(s)
- Sahar Nosrati Shanjani
- Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland; (S.N.S.); (M.Ł.); (R.W.); (P.K.); (E.M.); (J.N.)
| | - Monika Łyczko
- Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland; (S.N.S.); (M.Ł.); (R.W.); (P.K.); (E.M.); (J.N.)
| | - Rafał Walczak
- Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland; (S.N.S.); (M.Ł.); (R.W.); (P.K.); (E.M.); (J.N.)
| | - Przemysław Koźmiński
- Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland; (S.N.S.); (M.Ł.); (R.W.); (P.K.); (E.M.); (J.N.)
| | - Emilia Majka
- Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland; (S.N.S.); (M.Ł.); (R.W.); (P.K.); (E.M.); (J.N.)
| | - Jerzy Narbutt
- Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland; (S.N.S.); (M.Ł.); (R.W.); (P.K.); (E.M.); (J.N.)
| | - Wioletta Wojdowska
- National Centre for Nuclear Research, Sołtana 7/3, 05-400 Otwock, Poland;
| | - Agnieszka Majkowska-Pilip
- Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland; (S.N.S.); (M.Ł.); (R.W.); (P.K.); (E.M.); (J.N.)
| | - Aleksander Bilewicz
- Institute of Nuclear Chemistry and Technology, Dorodna 16, 03-195 Warsaw, Poland; (S.N.S.); (M.Ł.); (R.W.); (P.K.); (E.M.); (J.N.)
| |
Collapse
|
3
|
Spreckelmeyer S, Dasilva J, Decristoforo C, Mach RH, Passchier J, Carlucci G, Qhatani MA, Duatti A, Cornelissen BT, Engle J, Denkova A, Hendrikx JJMA, Seimbille Y, Yang X, Jia H, Zhang MR, Yang M, Perk L, Caravan P, Laverman P, Cheng Z, Hoehr C, Sakr T, Zeevaart JR. Highlight selection of radiochemistry and radiopharmacy developments by editorial board. EJNMMI Radiopharm Chem 2025; 10:13. [PMID: 40131608 PMCID: PMC11936851 DOI: 10.1186/s41181-025-00335-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND The Editorial Board of EJNMMI Radiopharmacy and Chemistry releases a biannual highlight commentary to update the readership on trends in the field of radiopharmaceutical development and application of radiopharmaceuticals. MAIN BODY This selection of highlights provides commentary on 24 different topics selected by each co-authoring Editorial Board member addressing a variety of aspects ranging from novel radiochemistry to first-in-human application of novel radiopharmaceuticals. CONCLUSION Trends in radiochemistry and radiopharmacy are highlighted. Hot topics cover the entire scope of EJNMMI Radiopharmacy and Chemistry, demonstrating the progress in the research field in many aspects.
Collapse
Affiliation(s)
- S Spreckelmeyer
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt- Universität zu Berlin, Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany.
| | - J Dasilva
- University of Montreal, Montreal, Canada
| | | | - R H Mach
- University of Pennsylvania, Philadelphia, USA
| | | | - G Carlucci
- University of California, Los Angeles, CA, USA
| | - M Al Qhatani
- Cyclotron and Radiopharmaceuticals Department, Research & Innovation, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - A Duatti
- University of Ferrara, Ferrara, Italy
| | - B T Cornelissen
- Oxford University, Oxford, UK
- University Medical Center Groningen, Groningen, Netherlands
| | - J Engle
- University of Wisconsin, Madison, WI, USA
| | - A Denkova
- Delft University of Technology, Delft, Netherlands
| | | | | | - X Yang
- Peking University First Hospital, Beijing, China
| | - H Jia
- Beijing Normal University, Beijing, China
| | - M-R Zhang
- National Institutes for Quantum Science and Technology, Chiba, Japan
| | - M Yang
- Jiangsu Institute of Nuclear Medicine, Jiangsu, China
| | - L Perk
- Radboud University Medical Center Nijmegen, Nijmegen, Netherlands
| | - P Caravan
- Massuchusetts General Hospital, Harvard University, Boston, USA
| | - P Laverman
- Radboud Medical Center, Nijmegen, Netherlands
| | - Z Cheng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - C Hoehr
- TRIUMF, Vancouver, BC, Canada
| | - T Sakr
- Egyptian Atomic Energy Authority, Cairo, Egypt
| | | |
Collapse
|
4
|
Saidi A, Stallons TA, Wong AG, Schatzmann AT, Soysal U, Torgue JJ. Side-by-Side Comparison of the In Vivo Performance of [ 212Pb]Pb-DOTAMTATE and Other SSTR2-Targeting Compounds. J Nucl Med 2025; 66:391-397. [PMID: 39884771 PMCID: PMC11876737 DOI: 10.2967/jnumed.124.268345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 12/16/2024] [Indexed: 02/01/2025] Open
Abstract
There are numerous versions of octreotide and octreotate, including DOTAMTATE, DOTATATE, JR11, and lead-specific chelator (PSC)-PEG2-TOC. These peptides, which can be either analogs or antagonists, are used in nuclear medicine for diagnostic imaging or targeted radionuclide therapy of neuroendocrine tumors that are positive for somatostatin receptors (SSTRs). Despite their structural and targeting similarities, they have distinct properties and clinical uses. We aimed to perform an extensive preclinical comparison of all these somatostatin analogs with 212Pb, directly studying their pharmacokinetic properties in tumors overexpressing SSTR2. Methods: All SSTR2 analogs were manufactured with the DOTAM, PSC, or DOTA chelators for appropriate comparison after radiolabeling with 212Pb. Chelation, quantification, and pharmacokinetics were compared side by side in AR42J-tumor-bearing animals. Results: These findings highlight the superior chelation efficiency and faster kinetics of DOTAM and then DOTA compared with the PSC. We also discovered a superior tumor-to-kidney area under the curve ratio for [212Pb]Pb-DOTAMTATE over other SSTR2-targeting peptides when radiolabeled with 212Pb. Conclusion: Taken together, the results indicates that [212Pb]Pb-DOTAMTATE has favorable tumor retention and a more favorable dosimetry profile, which is crucial for targeted α-therapy in treating SSTR2-positive neuroendocrine tumors.
Collapse
|
5
|
Mapanao AK, Busslinger SD, Mehta A, Kegler K, Favaretto C, Grundler PV, Talip Z, Köster U, Johnston K, Schibli R, van der Meulen NP, Müller C. Preclinical investigation of [ 149Tb]Tb-DOTATATE and [ 149Tb]Tb-DOTA-LM3 for tumor-targeted alpha therapy. Eur J Nucl Med Mol Imaging 2025; 52:1383-1398. [PMID: 39743617 DOI: 10.1007/s00259-024-07035-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 12/15/2024] [Indexed: 01/04/2025]
Abstract
PURPOSE Terbium-149 is a short-lived α-particle emitter, potentially useful for tumor-targeted therapy. The aim of this study was to investigate terbium-149 in combination with the somatostatin receptor (SSTR) agonist DOTATATE and the SSTR antagonist DOTA-LM3. The radiopeptides were evaluated to compare their therapeutic efficacy in vitro and in vivo. METHODS Terbium-149 was produced at ISOLDE/CERN and chemically purified at the Paul Scherrer Institute. Radiolabeling of somatostatin analogues with [149Tb]TbCl3 was performed under standard labeling conditions at pH 4.5. Cell viability (MTT) and survival assays (colony forming) assays were performed after 16-18 h exposure of SSTR-positive AR42J rat pancreatic tumor cells to various activity concentrations of [149Tb]Tb-DOTATATE and [149Tb]Tb-DOTA-LM3. DNA double-strand breaks were determined using immunofluorescence imaging of γ-H2A.X and 53BP1. Therapy studies were performed with AR42J tumor-bearing mice injected with 1 × 5 MBq or 2 × 5 MBq of the respective radiopeptide. The tolerability of up to 40 MBq [149Tb]Tb-DOTATATE or 40 MBq [149Tb]Tb-DOTA-LM3 was assessed with regard to undesired effects to the bone marrow and kidneys in immunocompetent mice without tumors. RESULTS The radiolabeling of peptides was achieved at molar activities of up to 20 MBq/nmol at ≥ 98% radiochemical purity. AR42J cell viability was reduced in an activity-dependent manner, with [149Tb]Tb-DOTA-LM3 being slightly more potent than [149Tb]Tb-DOTATATE (EC50: 0.5 vs. 1.2 kBq/mL). Both radiopeptides induced a similar number of γ-H2A.X and 53BP1 foci per nuclei, which indicated DNA damage in AR42J tumor cells. Injection of tumor-bearing mice with 1 × 5 MBq radiopeptide resulted in median survival times of 16.5 days and 19 days for [149Tb]Tb-DOTATATE and [149Tb]Tb-DOTA-LM3, respectively, as compared to only 8 days for untreated control mice. Application of 2 × 5 MBq of the radiopeptides further extended the median survival times to 30 days and 29 days, respectively. The blood cell counts and values for blood plasma biomarkers of treated mice without tumors were similar to those of untreated controls. Renal accumulation of [99mTc]Tc-DMSA was similar in all mice, indicating normal kidney function. CONCLUSION 149Tb-based radiopeptides effectively reduced the viability of tumor cells in vitro as well as the tumor growth in mice without causing relevant adverse events, irrespective of whether the SSTR agonist or antagonist was employed. These data encourage further preclinical application of terbium-149 to evaluate its potential in combination with other tumor-targeting agents.
Collapse
Affiliation(s)
- Ana Katrina Mapanao
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland
| | - Sarah D Busslinger
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland
| | - Avni Mehta
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland
| | | | - Chiara Favaretto
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland
- Division of Nuclear Medicine, University Hospital Basel, Basel, 4031, Switzerland
| | - Pascal V Grundler
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland
| | - Zeynep Talip
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland
- Laboratory of Radiochemistry, PSI Center for Nuclear Engineering and Sciences, Villigen-PSI, 5232, Switzerland
| | - Ulli Köster
- Institut Laue-Langevin, Grenoble, 38042, France
| | - Karl Johnston
- Physics Department, ISOLDE/CERN, Geneva, 1211, Switzerland
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, 8093, Switzerland
| | - Nicholas P van der Meulen
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland
- Laboratory of Radiochemistry, PSI Center for Nuclear Engineering and Sciences, Villigen-PSI, 5232, Switzerland
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland.
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, 8093, Switzerland.
| |
Collapse
|
6
|
Wallimann RH, Mehta A, Mapanao AK, Köster U, Kneuer R, Schindler P, van der Meulen NP, Schibli R, Müller C. Preclinical comparison of (radio)lanthanides using mass spectrometry and nuclear imaging techniques: biodistribution of lanthanide-based tumor-targeting agents and lanthanides in ionic form. Eur J Nucl Med Mol Imaging 2025; 52:1370-1382. [PMID: 39680064 PMCID: PMC11839852 DOI: 10.1007/s00259-024-07018-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 12/02/2024] [Indexed: 12/17/2024]
Abstract
PURPOSE With the growing interest in exploring radiolanthanides for nuclear medicine applications, the question arises as to whether they are generally interchangeable without affecting a biomolecule's pharmacokinetic properties. The goal of this study was to investigate similarities and differences of four (radio)lanthanides simultaneously applied as complexes of biomolecules or in ionic form. METHODS Inductively coupled plasma mass spectrometry (ICP-MS) was employed for the simultaneous detection of four lanthanides (Ln = lutetium, terbium, gadolinium and europium) in biological samples. In vitro tumor cell uptake and in vivo biodistribution studies were performed with Ln-DOTATATE, Ln-DOTA-LM3, Ln-PSMA-617 and Ln-OxFol-1. AR42J cells, PC-3 PIP cells and KB cells expressing the somatostatin receptor, the prostate-specific membrane antigen and the folate receptor, respectively, were used in vitro as well as to obtain the respective tumor mouse models for in vivo studies. The distribution of lanthanides in ionic form was investigated in immunocompetent mice. Dual-isotope SPECT/CT imaging studies were performed with mice administered with the radiolabeled biomolecules or chloride salts of lutetium-177 and terbium-161. RESULTS Similar in vitro cell uptake was observed for all four lanthanide complexes of each biomolecule into the respective tumor cell lines. AR42J tumor uptake of Ln-DOTATATE and Ln-DOTA-LM3 in mice showed similar values for all lanthanide complexes (3.8‒5.1% ID/g and 4.5‒5.0% ID/g; 1 h p.i., respectively). Accumulation of Ln-PSMA-617 in PC-3 PIP tumors (24-25% ID/g; 1 h p.i.) and of Ln-OxFol-1 in KB tumors (28-31% ID/g; 24 h p.i.) were also equal for the four lanthanide complexes of each biomolecule. After injection of lanthanide chloride salts (LnCl3; Ln = natLu, natTb, natGd, natEu), the liver uptake was different for each metal (~ 12% ID/g, ~ 22% ID/g, ~ 31% ID/g and ~ 37% ID/g; 24 h p.i., respectively) which could be ascribed to the radii of the respective lanthanide ions. In the bones, accumulation was considerably higher for lutetium than for other lanthanides (25 ± 5% ID/g vs. 14‒15% ID/g; 24 h p.i.). These data were confirmed visually by 177Lu/161Tb-based dual-isotope SPECT/CT images. CONCLUSIONS The presented study confirmed similar properties of Ln-complexes, suggesting that lutetium-177 can be replaced by other radiolanthanides, most probably without affecting the tissue distribution profile of the resultant radiopharmaceuticals. On the other hand, the different radii of the lanthanide ions affected their uptake and resorption mechanisms in liver and bones when injected in uncomplexed form.
Collapse
Affiliation(s)
- Rahel H Wallimann
- Biomedical Research, Novartis, Basel, 4056, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, 8093, Switzerland
| | - Avni Mehta
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland
| | - Ana Katrina Mapanao
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland
| | - Ulli Köster
- Institut Laue-Langevin, Grenoble, 38042, France
| | - Rainer Kneuer
- Biomedical Research, Novartis, Basel, 4056, Switzerland
| | | | - Nicholas P van der Meulen
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland
- Laboratory of Radiochemistry, PSI Center for Nuclear Engineering and Sciences, Villigen-PSI, 5232, Switzerland
| | - Roger Schibli
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, 8093, Switzerland
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland
| | - Cristina Müller
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, 8093, Switzerland.
- Center for Radiopharmaceutical Sciences, PSI Center for Life Sciences, Villigen-PSI, 5232, Switzerland.
| |
Collapse
|
7
|
Trejtnar F, Bárta P, Kozempel J, Vlk M, Ďurinová A, Kuchařová M, Pávek P. Terbium-161 in nuclear medicine: Preclinical and clinical progress in comparison with lutetium-177. Nucl Med Biol 2025; 144-145:108998. [PMID: 39978166 DOI: 10.1016/j.nucmedbio.2025.108998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/05/2025] [Accepted: 02/11/2025] [Indexed: 02/22/2025]
Abstract
The use of new radiopharmaceuticals labeled with lutetium-177 represents a successful translation of experimental results into clinical practice. Recent experimental data suggests that terbium-161 might well follow the example of lutetium-177 regarding applicability in nuclear medicine. Similarly to lutetium-177, the terbium-161 emits beta particles and gamma-radiation, although terbium-161 emits short-ranged conversion and Auger electrons, creating an effect that may eliminate smaller tumor metastases more effectively than lutetium-177. Terbium-161 may exert a higher radiobiological effect in the target tissues in comparison with lutetium-177, a difference which makes possible a reduction in the doses of radioactivity administered. Further, due to the similar chemical properties of lutetium-177 and terbium-161, similar radiolabeling techniques can be used. The differences found in preclinical experiments on radiotoxicity of the counterparts seem to be minor. Despite intensive progress, the number of preclinical studies on 161Tb-labeled agents is still not comparable to studies on lutetium-177. Clinical trials with 161Tb-labeled radiopharmaceuticals focused on the treatment of prostate cancer and selected neuroendocrine tumors have already begun, although none of them have been completed yet.
Collapse
Affiliation(s)
- František Trejtnar
- Charles University, Faculty of Pharmacy in Hradec Králové, Department of Pharmacology and Toxicology, Hradec Králové, Czech Republic.
| | - Pavel Bárta
- Charles University, Faculty of Pharmacy in Hradec Králové, Department of Biophysics and Physical Chemistry, Hradec Králové, Czech Republic.
| | - Ján Kozempel
- Czech Technical University, Faculty of Nuclear Sciences and Physical Engineering, Department of Nuclear Chemistry, Prague, Czech Republic.
| | - Martin Vlk
- Czech Technical University, Faculty of Nuclear Sciences and Physical Engineering, Department of Nuclear Chemistry, Prague, Czech Republic.
| | - Anna Ďurinová
- Charles University, Faculty of Pharmacy in Hradec Králové, Department of Pharmacology and Toxicology, Hradec Králové, Czech Republic.
| | - Monika Kuchařová
- Charles University, Faculty of Pharmacy in Hradec Králové, Department of Biophysics and Physical Chemistry, Hradec Králové, Czech Republic.
| | - Petr Pávek
- Charles University, Faculty of Pharmacy in Hradec Králové, Department of Pharmacology and Toxicology, Hradec Králové, Czech Republic.
| |
Collapse
|
8
|
Abdlkadir AS, Rosar F, Jalilian A, Moghrabi S, Al-Balooshi B, Rabei O, Kairemo K, Al-Ibraheem A. Harnessing Terbium Radioisotopes for Clinical Advancements: A Systematic Review. Nucl Med Mol Imaging 2025; 59:50-61. [PMID: 39881968 PMCID: PMC11772629 DOI: 10.1007/s13139-024-00891-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/16/2024] [Accepted: 10/24/2024] [Indexed: 01/23/2025] Open
Abstract
Background this systematic review was conducted to assess the practical application of terbium radioisotopes, utilizing systematic search methodologies to identify relevant studies. Methods the databases of PubMed, Web of Science, and Scopus were systematically scoured, targeting the research on four terbium isotopes: 149 Tb, 152 Tb, 155 Tb, and 161 Tb. Various combinations of keywords related to terbium and its four radioisotopes were used in the search process. The search encompassed studies conducted up to July 27, 2024. Results following the removal of 335 duplicate research articles, a cohort of 429 papers was curated for potential inclusion in the study. Out of 429 articles reviewed, a mere nine addressed the potential uses of 161 Tb and 152 Tb. Notably, 155 Tb and 149 Tb have yet to be examined in human subjects. Conclusions the research trajectory is now veering towards clinical studies that provide in-human data, with the goal of advancing radiotheranostics and nuclear oncology. The preliminary outcomes are stimulating and have led to the initiation of several clinical trials. The success of these trials and the establishment of production facilities will be critical for the clinical adoption of these agents.
Collapse
Affiliation(s)
- Ahmed Saad Abdlkadir
- Department of Nuclear Medicine, King Hussein Cancer Center (KHCC), Queen Rania Street, Al Jubeiha, Amman, 11942 Jordan
| | - Florian Rosar
- Department of Nuclear Medicine, Saarland University – Medical Center, 66123 Homburg, Germany
| | - Amireza Jalilian
- Division of Physical and Chemical Sciences, International Atomic Energy Agency, 1220 Vienna, Austria
| | - Serin Moghrabi
- Department of Nuclear Medicine, King Hussein Cancer Center (KHCC), Queen Rania Street, Al Jubeiha, Amman, 11942 Jordan
| | - Batool Al-Balooshi
- Dubai Nuclear Medicine and Molecular Imaging Center, Dubai Health Authority, 1853 Dubai, UAE
| | - Obayda Rabei
- Department of Nuclear Medicine, King Hussein Cancer Center (KHCC), Queen Rania Street, Al Jubeiha, Amman, 11942 Jordan
| | - Kalevi Kairemo
- Department of Molecular Radiotherapy and Nuclear Medicine, International Comprehensive Cancer Center Docrates, Saukonpaadenranta 2, 00180 Helsinki, Finland
- Department of Nuclear Medicine, University of Texas, MD Anderson Cancer Center, Holcombe Blvd, Houston, TX 77030 USA
| | - Akram Al-Ibraheem
- Department of Nuclear Medicine, King Hussein Cancer Center (KHCC), Queen Rania Street, Al Jubeiha, Amman, 11942 Jordan
- School of Medicine, The University of Jordan, Amman, 11942 Jordan
| |
Collapse
|
9
|
Salvanou EA, Apostolopoulou A, Xanthopoulos S, Koelewijn S, van Overeem P, Laurent G, Bazzi R, Denat F, Roux S, Bouziotis P. 161Terbium-Labeled Gold Nanoparticles as Nanoscale Brachytherapy Agents Against Breast Cancer. MATERIALS (BASEL, SWITZERLAND) 2025; 18:248. [PMID: 39859720 PMCID: PMC11766487 DOI: 10.3390/ma18020248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 01/27/2025]
Abstract
Due to their intriguing emission profile, Terbium-161 (161Tb) radiopharmaceuticals seem to bring significant advancement in theranostic applications to cancer treatment. The combination of 161Tb with nanoscale brachytherapy as an approach for cancer treatment is particularly advantageous and promising. Herein, we propose the application of a hybrid nanosystem comprising gold decorated (Au@TADOTAGA) iron oxide nanoflowers as a form of injectable nanobrachytherapy for the local treatment of breast cancer. More specifically, Au@TADOTAGA and NFAu@TADOTAGA NPs were efficiently radiolabeled with 161Tb, and their in vitro stability was assessed up to 21 d post-radiolabeling. Furthermore, their cytotoxic profile against 4T1 breast cancer cells was evaluated, and their ex vivo biodistribution characteristics were revealed after intratumoral injection in the same animal model. The enhanced retention at the tumor site urged us to evaluate the therapeutic effect of the [161Tb]Tb-NFAu@TADOTAGA nanosystem after intratumoral administration to 4T1-tumor-bearing mice, over a period of 24 days. Three different therapeutic protocols were performed in order to identify which therapeutic approach would offer the optimum results and identify the proposed nanosystem as a promising nanoscale brachytherapy agent.
Collapse
Affiliation(s)
- Evangelia-Alexandra Salvanou
- Radiochemical Studies Laboratory, Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Center for Scientific Research “Demokritos”, Patriarchou Grigoriou and 27 Neapoleos Street, 15341 Athens, Greece; (A.A.); (S.X.); (P.B.)
| | - Adamantia Apostolopoulou
- Radiochemical Studies Laboratory, Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Center for Scientific Research “Demokritos”, Patriarchou Grigoriou and 27 Neapoleos Street, 15341 Athens, Greece; (A.A.); (S.X.); (P.B.)
| | - Stavros Xanthopoulos
- Radiochemical Studies Laboratory, Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Center for Scientific Research “Demokritos”, Patriarchou Grigoriou and 27 Neapoleos Street, 15341 Athens, Greece; (A.A.); (S.X.); (P.B.)
| | - Stuart Koelewijn
- Terthera b.v., Minervum 7070, 4817 ZK Breda, The Netherlands; (S.K.); (P.v.O.)
| | | | - Gautier Laurent
- Laboratoire Chrono-Environnement, Université de Franche-Comté, CNRS, F-25000 Besançon, France; (G.L.); (R.B.); (S.R.)
| | - Rana Bazzi
- Laboratoire Chrono-Environnement, Université de Franche-Comté, CNRS, F-25000 Besançon, France; (G.L.); (R.B.); (S.R.)
| | - Franck Denat
- Institut de Chimie Moléculaire de l’Université de Bourgogne, UMR 6302, CNRS, Université de Bourgogne, F-21078 Dijon, France;
| | - Stéphane Roux
- Laboratoire Chrono-Environnement, Université de Franche-Comté, CNRS, F-25000 Besançon, France; (G.L.); (R.B.); (S.R.)
| | - Penelope Bouziotis
- Radiochemical Studies Laboratory, Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Center for Scientific Research “Demokritos”, Patriarchou Grigoriou and 27 Neapoleos Street, 15341 Athens, Greece; (A.A.); (S.X.); (P.B.)
| |
Collapse
|
10
|
Miller C, Klyuzhin I, Chaussé G, Brosch-Lenz J, Koniar H, Shi K, Rahmim A, Uribe C. Impact of cell geometry, cellular uptake region, and tumour morphology on 225Ac and 177Lu dose distributions in prostate cancer. EJNMMI Phys 2024; 11:97. [PMID: 39570450 PMCID: PMC11582247 DOI: 10.1186/s40658-024-00700-9] [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: 06/03/2024] [Accepted: 11/06/2024] [Indexed: 11/22/2024] Open
Abstract
BACKGROUND Radiopharmaceutical therapy with 225Ac- and 177Lu-PSMA has shown promising results for the treatment of prostate cancer. However, the distinct physical properties of alpha and beta radiation elicit varying cellular responses, which could be influenced by factors such as tumour morphology. In this study, we use simulations to examine how cell geometry, region of pharmaceutical uptake within the cell to model different internalization fractions, and the presence of tumour hypoxia and necrosis impact nucleus absorbed doses and dose heterogeneity with 225Ac and 177Lu. We also develop nucleus absorbed dose kernels for application to autoradiography images. METHODS We used the GATE Monte Carlo software to simulate three geometries of LNCaP prostate cancer cells (spherical, cubic, and ovoid) with activity of 225Ac or 177Lu internalized in the cytoplasm or bound to the extracellular membrane. Nucleus S-values were calculated for each geometry, source region, and isotope. The cell models were used to create nucleus absorbed dose kernels for each source region describing the dose to each nucleus in a cell layer, which were applied to simulated tumours composed of normoxic, hypoxic, or necrotic cancer cells to obtain dose rate maps. Absorbed doses within the tumours and dose heterogeneity were analyzed for each tumour morphology and isotope. Cell geometry made a minimal impact on S-values to the nucleus, however internalization resulted in higher nucleus doses. Applying the kernels to the simulated tumour maps showed that doses to each cell type varied between 225Ac and 177Lu depending on tumour morphology. Dose heterogeneity within tumours was slightly higher with 225Ac, however the tumour morphology made a larger impact on dose heterogeneity compared to the choice of isotope, with hypoxic and necrotic tumours having very heterogeneous dose distributions. CONCLUSIONS Cell geometry simplifications may still allow robust results in simulation studies. Furthermore, the morphology of the tumour itself may make a larger impact on treatment response compared to other variables such as ratio of internalization. Finally, nucleus absorbed dose kernels were created that could enable microdosimetric studies with autoradiography.
Collapse
Affiliation(s)
- Cassandra Miller
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Physics, University of British Columbia, Vancouver, BC, Canada
| | - Ivan Klyuzhin
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | | | - Julia Brosch-Lenz
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
| | - Helena Koniar
- Department of Physics, University of British Columbia, Vancouver, BC, Canada
| | - Kuangyu Shi
- Department of Nuclear Medicine, Bern University Hospital, The University of Bern, Bern, Switzerland
| | - Arman Rahmim
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada
- Department of Physics, University of British Columbia, Vancouver, BC, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC, Canada
| | - Carlos Uribe
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC, Canada.
- Department of Radiology, University of British Columbia, Vancouver, BC, Canada.
- Molecular Imaging and Therapy, BC Cancer Research Institute, Vancouver, BC, Canada.
| |
Collapse
|
11
|
Petitpoisson L, Mahamoud A, Mazan V, Sy M, Jeannin O, Tóth E, Charbonnière LJ, Elhabiri M, Nonat AM. Octadentate Bispidine Chelators for Tb(III) Complexation: Pyridine Carboxylate versus Pyridine Phosphonate Donors. Inorg Chem 2024. [PMID: 39558777 DOI: 10.1021/acs.inorgchem.4c03691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
With their rigid and preorganized skeleton, bispidine (3,7-diazabicyclo[3.3.1]nonane) chelators are very appealing for the preparation of metal complexes with high kinetic inertness. With the aim to develop new Tb(III)-based medical imaging probes, this study describes the synthesis and physicochemical properties of two novel terbium(III) complexes with octadentate bispidine-based ligands substituted with either pyridine-phosphonate (H6L1) or picolinate (H4L2) subunits. Thermodynamic stability constants of the corresponding Tb(III) complexes have been determined by potentiometric, UV-visible absorption spectrophotometric and spectrofluorimetric methods. Despite their apparent similarity, these two octadentate ligands differ in their most stable conformation: chair-chair conformation for H4L2 and boat-chair conformation for H6L1, as confirmed by 1H NMR studies and suggested by physicochemical investigations. This conformational change induces different protonation schemes for the two ligands. The kinetic inertness of the Tb complexes has been studied in various media and assessed by transmetalation and transchelation experiments. In particular, Tb(L2) displayed a remarkable kinetic inertness with no measurable dissociation over two months in mouse serum at 10-5 M concentration. The complex was also very inert in the presence of a 50-fold excess of Zn(II) in H2O at pH = 7.4 (7% of dissociation over two months). The complexes with ligand L1 are significantly less inert, emphasizing the influence of the ligand conformation on the kinetic inertness of the Ln(III) complexes. Finally, the luminescence properties of the isolated complexes have also been investigated. A bright green luminescence was observed, especially for Tb(L2), which displays a high quantum yield value of 50% in H2O (60% in D2O; λexc = 263 nm). In addition, luminescence lifetimes of 1.9(2) and 1.7(2) ms have been measured for Tb(L1) and Tb(L2), respectively, hence confirming the formation of nona-coordinated complexes with one inner-sphere water molecule. These data on a bispidine scaffold pave the way for developing bright, inert luminescent probes for bioimaging and for radiolabeling applications with Tb(III) radioisotopes.
Collapse
Affiliation(s)
- Lucas Petitpoisson
- Equipe de Synthèse pour l'Analyse, Université de Strasbourg, CNRS, IPHC UMR 7178, Strasbourg F-67 087, France
| | - Anli Mahamoud
- Equipe de Synthèse pour l'Analyse, Université de Strasbourg, CNRS, IPHC UMR 7178, Strasbourg F-67 087, France
| | - Valérie Mazan
- Laboratoire d'Innovation Moléculaire et Applications (LIMA), Team Bio(IN)organic and Medicinal Chemistry, European School of Chemistry, Polymers and Materials (ECPM) Université de Strasbourg/CNRS/UHA, UMR7042, 25 Rue Becquerel, Strasbourg F-67087, France
| | - Maryame Sy
- Equipe de Synthèse pour l'Analyse, Université de Strasbourg, CNRS, IPHC UMR 7178, Strasbourg F-67 087, France
| | - Olivier Jeannin
- Institut des Sciences Chimiques de Rennes, UMR-CNRS 6226, 263 Avenue du Général Leclerc, CS 74205, Rennes Cedex F-35042, France
| | - Eva Tóth
- Centre de Biophysique Moléculaire, CNRS UPR 4301, Université d'Orléans, Rue Charles Sadron, Orleans F-45071, United States
| | - Loïc J Charbonnière
- Equipe de Synthèse pour l'Analyse, Université de Strasbourg, CNRS, IPHC UMR 7178, Strasbourg F-67 087, France
| | - Mourad Elhabiri
- Laboratoire d'Innovation Moléculaire et Applications (LIMA), Team Bio(IN)organic and Medicinal Chemistry, European School of Chemistry, Polymers and Materials (ECPM) Université de Strasbourg/CNRS/UHA, UMR7042, 25 Rue Becquerel, Strasbourg F-67087, France
| | - Aline M Nonat
- Equipe de Synthèse pour l'Analyse, Université de Strasbourg, CNRS, IPHC UMR 7178, Strasbourg F-67 087, France
| |
Collapse
|
12
|
De Nardo L, Santi S, Dalla Pietà A, Ferro-Flores G, Azorín-Vega E, Nascimbene E, Barbieri V, Zorz A, Rosato A, Meléndez-Alafort L. Comparison of the dosimetry and cell survival effect of 177Lu and 161Tb somatostatin analog radiopharmaceuticals in cancer cell clusters and micrometastases. EJNMMI Phys 2024; 11:94. [PMID: 39535653 PMCID: PMC11561253 DOI: 10.1186/s40658-024-00696-2] [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: 07/10/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND 177Lu-based radiopharmaceuticals (RPs) are the most used for targeted radionuclide therapy (TRT) due to their good response rates. However, the worldwide availability of 177Lu is limited. 161Tb represents a potential alternative for TRT, as it emits photons for SPECT imaging, β--particles for therapy, and also releases a significant yield of internal conversion (IE) and Auger electrons (AE). This research aimed to evaluate cell dosimetry with the MIRDcell code considering a realistic localization of three 161Tb- and 177Lu-somatostatin (SST) analogs in different subcellular regions as reported in the literature, various cell cluster sizes (25-1000 µm of radius) and percentage of labeled cells. Experimental values of the α- and β-survival coefficients determined by external beam photon irradiation were used to estimate the survival fraction (SF) of AR42J pancreatic cell clusters and micrometastases. RESULTS The different localization of RPs labeled with the same radionuclide within the cells, resulted in only slight variations in the dose absorbed by the nuclei (ADN) of the labeled cells with no differences observed in either the unlabeled cells or the SF. ADN of labeled cells (MDLC) produced by 161Tb-RPs were from 2.8-3.7 times higher than those delivered by 177Lu-RPs in cell clusters with a radius lower than 0.1 mm and 10% of labeled cells, due to the higher amount of energy emitted by 161Tb-disintegration in form of IE and AE. However, the 161Tb-RPs/177Lu-RPs MDLC ratio decreased below 1.6 in larger cell clusters (0.5-1 mm) with > 40% labeled cells, due to the significantly higher 177Lu-RPs cross-irradiation contribution. Using a fixed number of disintegrations, SFs of 161Tb-RPs in clusters with > 40% labeled cells were lower than those of 177Lu-RPs, but when the same amount of emitted energy was used no significant differences in SF were observed between 177Lu- and 161Tb-RPs, except for the smallest cluster sizes. CONCLUSIONS Despite the emissions of IE and AE from 161Tb-RPs, their localization within different subcellular regions exerted a negligible influence on the ADN. The same cell damage produced by 177Lu-RPs could be achieved using smaller quantities of 161Tb-RPs, thus making 161Tb a suitable alternative for TRT.
Collapse
Affiliation(s)
- Laura De Nardo
- Department of Physics and Astronomy, University of Padua, Via Marzolo 8, 35131, Padua, Italy
- INFN-Padua, National Institute of Nuclear Physics, Via Marzolo 8, 35131, Padua, Italy
| | - Sara Santi
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Via Gattamelata 64, 35128, Padua, Italy
| | - Anna Dalla Pietà
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Via Gattamelata 64, 35128, Padua, Italy
| | - Guillermina Ferro-Flores
- Department of Radioactive Materials, National Institute of Nuclear Research (ININ), Carretera México-Toluca S/N, La Marquesa, 52750, Ocoyoacac, Mexico
| | - Erika Azorín-Vega
- Department of Radioactive Materials, National Institute of Nuclear Research (ININ), Carretera México-Toluca S/N, La Marquesa, 52750, Ocoyoacac, Mexico
| | - Emma Nascimbene
- Immunology and Molecular Oncology Diagnostics Unit, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35128, Padua, Italy
| | - Vito Barbieri
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Via Gattamelata 64, 35128, Padua, Italy
| | - Alessandra Zorz
- Medical Physics Department, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35128, Padua, Italy
| | - Antonio Rosato
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Via Gattamelata 64, 35128, Padua, Italy
- Immunology and Molecular Oncology Diagnostics Unit, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35128, Padua, Italy
| | - Laura Meléndez-Alafort
- Immunology and Molecular Oncology Diagnostics Unit, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata 64, 35128, Padua, Italy.
| |
Collapse
|
13
|
Busslinger SD, Mapanao AK, Kegler K, Bernhardt P, Flühmann F, Fricke J, Zeevaart JR, Köster U, van der Meulen NP, Schibli R, Müller C. Comparison of the tolerability of 161Tb- and 177Lu-labeled somatostatin analogues in the preclinical setting. Eur J Nucl Med Mol Imaging 2024; 51:4049-4061. [PMID: 39046521 PMCID: PMC11527941 DOI: 10.1007/s00259-024-06827-2] [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: 04/29/2024] [Accepted: 06/30/2024] [Indexed: 07/25/2024]
Abstract
PURPOSE [177Lu]Lu-DOTATATE is an established somatostatin receptor (SSTR) agonist for the treatment of metastasized neuroendocrine neoplasms, while the SSTR antagonist [177Lu]Lu-DOTA-LM3 has only scarcely been employed in clinics. Impressive preclinical data obtained with [161Tb]Tb-DOTA-LM3 in tumor-bearing mice indicated the potential of terbium-161 as an alternative to lutetium-177. The aim of the present study was to compare the tolerability of 161Tb- and 177Lu-based DOTA-LM3 and DOTATATE in immunocompetent mice. METHODS Dosimetry calculations were performed based on biodistribution data of the radiopeptides in immunocompetent mice. Treatment-related effects on blood cell counts were assessed on Days 10, 28 and 56 after application of [161Tb]Tb-DOTA-LM3 or [161Tb]Tb-DOTATATE at 20 MBq per mouse. These radiopeptides were also applied at 100 MBq per mouse and the effects compared to those observed after application of the 177Lu-labeled counterparts. Bone marrow smears, blood plasma parameters and organ histology were assessed at the end of the study. RESULTS The absorbed organ dose was commonly higher for the SSTR antagonist than for the SSTR agonist and for terbium-161 over lutetium-177. Application of a therapeutic activity level of 20 MBq [161Tb]Tb-DOTA-LM3 or [161Tb]Tb-DOTATATE was well tolerated without major hematological changes. The injection of 100 MBq of the 161Tb- and 177Lu-based somatostatin analogues affected the blood cell counts, however. The lymphocytes were 40-50% lower in treated mice compared to the untreated controls on Day 10 irrespective of the radionuclide employed. At the same timepoint, thrombocyte and erythrocyte counts were 30-50% and 6-12% lower, respectively, after administration of the SSTR antagonist (p < 0.05) while changes were less pronounced in mice injected with the SSTR agonist. All blood cell counts were in the normal range on Day 56. Histological analyses revealed minimal abnormalities in the kidneys, liver and spleen of treated mice. No correlation was observed between the organ dose and frequency of the occurrence of abnormalities. CONCLUSION Hematologic changes were more pronounced in mice treated with the SSTR antagonist than in those treated with the SSTR agonist. Despite the increased absorbed dose delivered by terbium-161 over lutetium-177, [161Tb]Tb-DOTA-LM3 and [161Tb]Tb-DOTATATE should be safe at activity levels that are recommended for their respective 177Lu-based analogues.
Collapse
Affiliation(s)
- Sarah D Busslinger
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, 5232, Switzerland
| | - Ana Katrina Mapanao
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, 5232, Switzerland
| | | | - Peter Bernhardt
- Department of Medical Radiation Sciences, Institution of Clinical Science, Sahlgrenska Academy, University of Gothenburg, Gothenburg, 41345, Sweden
- Department of Medical Physics and Biomedical Engineering (MFT), Sahlgrenska University Hospital, Gothenburg, 41345, Sweden
| | - Fabienne Flühmann
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, 5232, Switzerland
| | - Julia Fricke
- Division of Nuclear Medicine, University Hospital Basel, Basel, 4031, Switzerland
| | - Jan Rijn Zeevaart
- Radiochemistry, South African Nuclear Energy Corporation (Necsa), Brits, 0240, South Africa
| | - Ulli Köster
- Institut Laue-Langevin, Grenoble, 38042, France
| | - Nicholas P van der Meulen
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, 5232, Switzerland
- Laboratory of Radiochemistry, Paul Scherrer Institute, Villigen-PSI, 5232, Switzerland
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, 5232, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, 8093, Switzerland
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, 5232, Switzerland.
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, 8093, Switzerland.
| |
Collapse
|
14
|
Hierlmeier I, Marino N, Schreck MV, Schneider L, Maus S, Barrett K, Kretowicz M, Engle JW, Pierri G, Ezziddin S, Bartholomä MD. Radiochemistry and Complex Formation of the Cyclen-Derived Chelator DOTI-Me with Mn 2+, Cu 2+, Zn 2+, Ga 3+, In 3+, Tb 3+, and Lu 3. Inorg Chem 2024; 63:19468-19481. [PMID: 39352711 DOI: 10.1021/acs.inorgchem.4c03571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
In this work, we describe the complex formation and radiochemistry of the cyclen-based chelator DOTI-Me bearing four methylimidazole arms. Radiolabeling properties were evaluated for 52gMn, 64Cu, 68Ga, 111In, 161Tb, and 177Lu, and DOTI-Me showed distinct differences to the structurally related H4DOTA. While radiochemical conversions (RCCs) for 52gMn and 111In were comparable to those of H4DOTA, DOTI-Me was not suited for 68Ga. Conversely, quantitative RCCs were achieved for 64Cu at ambient temperature, while elevated temperatures were required for complexation with H4DOTA. For 161Tb and 177Lu, good but not quantitative RCCs were obtained with DOTI-Me. With the exemption of 68Ga3+, radiolabeled complexes showed high stability in ligand challenge experiments and in human serum. X-ray analysis of the nonradioactive complexes revealed the formation of 8-coordinate Mn2+ and In3+ DOTI-Me complexes. Cu2+ adopted a unique distorted square-pyramidal 2 + 3 with the neutral DOTI-Me ligand and a Jahn-Teller distorted 4 + 2 coordination geometry for the diprotonated H2DOTI-Me2+ cation, respectively. For Zn2+, the complex with HDOTI-Me+ showed a distorted 4 + 3 pentagonal bipyramidal geometry. Summarizing, the ligand DOTI-Me may be an interesting alternative to H4DOTA for 52gMn, 64Cu, 111In, 161Tb, and 177Lu, covering diagnostic as well as therapeutic radionuclides. Further studies of targeted radiopharmaceuticals based on the DOTI-Me scaffold in combination with the set of radiometals presented herein are thus warranted.
Collapse
Affiliation(s)
- Ina Hierlmeier
- Department of Nuclear Medicine, Saarland University-Medical Center, Kirrbergerstrasse, D-66421 Homburg, Germany
| | - Nadia Marino
- Department of Chemistry and Chemical Technologies, University of Calabria, 87036 Arcavacata di Rende, CS, Italy
| | - Moritz-Valentin Schreck
- Department of Nuclear Medicine, Saarland University-Medical Center, Kirrbergerstrasse, D-66421 Homburg, Germany
| | - Lukas Schneider
- Department of Nuclear Medicine, Saarland University-Medical Center, Kirrbergerstrasse, D-66421 Homburg, Germany
| | - Stephan Maus
- Department of Nuclear Medicine, Saarland University-Medical Center, Kirrbergerstrasse, D-66421 Homburg, Germany
| | - Kendall Barrett
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Margarita Kretowicz
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Jonathan W Engle
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Giovanni Pierri
- Department of Chemistry and Biology "A. Zambelli", University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, SA, Italy
| | - Samer Ezziddin
- Department of Nuclear Medicine, Saarland University-Medical Center, Kirrbergerstrasse, D-66421 Homburg, Germany
| | - Mark D Bartholomä
- Department of Nuclear Medicine, Saarland University-Medical Center, Kirrbergerstrasse, D-66421 Homburg, Germany
| |
Collapse
|
15
|
Spoormans K, Struelens L, Vermeulen K, De Saint-Hubert M, Koole M, Crabbé M. The Emission of Internal Conversion Electrons Rather Than Auger Electrons Increased the Nucleus-Absorbed Dose for 161Tb Compared with 177Lu with a Higher Dose Response for [ 161Tb]Tb-DOTA-LM3 Than for [ 161Tb]Tb-DOTATATE. J Nucl Med 2024; 65:1619-1625. [PMID: 39209546 DOI: 10.2967/jnumed.124.267873] [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: 04/08/2024] [Accepted: 07/17/2024] [Indexed: 09/04/2024] Open
Abstract
Preclinical data have shown that 161Tb-labeled peptides targeting the somatostatin receptor are therapeutically more effective for peptide receptor radionuclide therapy than are their 177Lu-labeled counterparts. To further substantiate this enhanced therapeutic effect, we performed cellular dosimetry to quantify the absorbed dose to the cell nucleus and compared dose-response curves to evaluate differences in relative biological effectiveness in vitro. Methods: CA20948 cell survival was assessed after treatment with [161Tb]Tb- and [177Lu]Lu-DOTATATE (agonist) and with [161Tb]Tb- and [177Lu]Lu-DOTA-LM3 (antagonist) via a clonogenic assay. Cell binding, internalization, and dissociation assays were performed up to 7 d to acquire time-integrated activity coefficients. Separate S values for each type of particle emission (Auger/internal conversion [IC] electrons and β- particles) were computed via Monte Carlo simulations, while considering spheric cells. Once the absorbed dose to the cell nucleus was calculated, survival curves were fitted to the appropriate linear or linear-quadratic model and corresponding relative biological effectiveness was evaluated. Results: Although the radiopeptide uptake was independent of the radionuclide, [161Tb]Tb-DOTATATE and [161Tb]Tb-DOTA-LM3 delivered a 3.6 and 3.8 times higher dose to the nucleus, respectively, than their 177Lu-labeled counterparts on saturated receptor binding. This increased nucleus-absorbed dose was mainly due to the additional emission of IC and not Auger electrons by 161Tb. When activity concentrations were considered, both [161Tb]Tb-DOTATATE and [161Tb]Tb-DOTA-LM3 showed a lower survival fraction than did labeling with 177Lu. When the absorbed dose to the nucleus was considered, no significant difference could be observed between the dose-response curves for [161Tb]Tb- and [177Lu]Lu-DOTATATE. [161Tb]Tb-DOTA-LM3 showed a linear-quadratic dose response, whereas [161Tb]Tb-DOTATATE showed only a linear dose response within the observed dose range, suggesting additional cell membrane damage by Auger electrons. Conclusion: The IC, rather than Auger, electrons emitted by 161Tb resulted in a higher absorbed dose to the cell nucleus and lower clonogenic survival for [161Tb]Tb-DOTATATE and [161Tb]Tb-DOTA-LM3 than for the 177Lu-labeled analogs. In contrast, [161Tb]Tb-DOTATATE showed no higher dose response than [177Lu]Lu-DOTATATE, whereas for [161Tb]Tb-DOTA-LM3 an additional quadratic response was observed. Because of this quadratic response, potentially caused by cell membrane damage, [161Tb]Tb-DOTA-LM3 is a more effective radiopeptide than [161Tb]Tb-DOTATATE for labeling with 161Tb.
Collapse
Affiliation(s)
- Kaat Spoormans
- Nuclear Medical Applications, Belgian Nuclear Research Center, Mol, Belgium; and
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Lara Struelens
- Nuclear Medical Applications, Belgian Nuclear Research Center, Mol, Belgium; and
| | - Koen Vermeulen
- Nuclear Medical Applications, Belgian Nuclear Research Center, Mol, Belgium; and
| | | | - Michel Koole
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Melissa Crabbé
- Nuclear Medical Applications, Belgian Nuclear Research Center, Mol, Belgium; and
| |
Collapse
|
16
|
Hindié E, Larouze A, Alcocer-Ávila M, Morgat C, Champion C. Palladium-103 ( 103Pd/ 103mRh), a promising Auger-electron emitter for targeted radionuclide therapy of disseminated tumor cells - absorbed doses in single cells and clusters, with comparison to 177Lu and 161Tb. Theranostics 2024; 14:4318-4330. [PMID: 39113794 PMCID: PMC11303077 DOI: 10.7150/thno.95436] [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: 02/18/2024] [Accepted: 07/02/2024] [Indexed: 08/10/2024] Open
Abstract
Early use of targeted radionuclide therapy (TRT) to eradicate disseminated tumor cells (DTCs) might offer cure. Selection of appropriate radionuclides is required. This work highlights the potential of 103Pd (T1/2 = 16.991 d) which decays to 103mRh (T1/2 = 56.12 min) then to stable 103Rh with emission of Auger and conversion electrons. Methods: The Monte Carlo track structure code CELLDOSE was used to assess absorbed doses in single cells (14-μm diameter; 10-μm nucleus) and clusters of 19 cells. The radionuclide was distributed on the cell surface, within the cytoplasm, or in the nucleus. Absorbed doses from 103Pd, 177Lu and 161Tb were compared after energy normalization. The impact of non-uniform cell targeting, and the potential benefit from dual-targeting was investigated. Additional results related to 103mRh, if used directly, are provided. Results: In the single cell, and depending on radionuclide distribution, 103Pd delivered 7- to 10-fold higher nuclear absorbed dose and 9- to 25-fold higher membrane dose than 177Lu. In the 19-cell clusters, 103Pd absorbed doses also largely exceeded 177Lu. In both situations, 161Tb stood in-between 103Pd and 177Lu. Non-uniform targeting, considering four unlabeled cells within the cluster, resulted in moderate-to-severe dose heterogeneity. For example, with intranuclear 103Pd, unlabeled cells received only 14% of the expected nuclear dose. Targeting with two 103Pd-labeled radiopharmaceuticals minimized dose heterogeneity. Conclusion: 103Pd, a next-generation Auger emitter, can deliver substantially higher absorbed doses than 177Lu to single tumor cells and cell clusters. This may open new horizons for the use of TRT in adjuvant or neoadjuvant settings, or for targeting minimal residual disease.
Collapse
Affiliation(s)
- Elif Hindié
- Service de Médecine Nucléaire, CHU de Bordeaux, Université de Bordeaux, UMR CNRS 5287, INCIA, F-33400, Talence, France
- Institut Universitaire de France, 1 rue Descartes, 75231 Paris cedex 05, France
| | - Alexandre Larouze
- Université de Bordeaux-CNRS-CEA, Centre Lasers Intenses et Applications, UMR 5107, 33405 Talence, France
| | - Mario Alcocer-Ávila
- Université de Bordeaux-CNRS-CEA, Centre Lasers Intenses et Applications, UMR 5107, 33405 Talence, France
| | - Clément Morgat
- Service de Médecine Nucléaire, CHU de Bordeaux, Université de Bordeaux, UMR CNRS 5287, INCIA, F-33400, Talence, France
| | - Christophe Champion
- Université de Bordeaux-CNRS-CEA, Centre Lasers Intenses et Applications, UMR 5107, 33405 Talence, France
| |
Collapse
|
17
|
Lapi SE, Scott PJH, Scott AM, Windhorst AD, Zeglis BM, Abdel-Wahab M, Baum RP, Buatti JM, Giammarile F, Kiess AP, Jalilian A, Knoll P, Korde A, Kunikowska J, Lee ST, Paez D, Urbain JL, Zhang J, Lewis JS. Recent advances and impending challenges for the radiopharmaceutical sciences in oncology. Lancet Oncol 2024; 25:e236-e249. [PMID: 38821098 PMCID: PMC11340123 DOI: 10.1016/s1470-2045(24)00030-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 06/02/2024]
Abstract
This paper is the first of a Series on theranostics that summarises the current landscape of the radiopharmaceutical sciences as they pertain to oncology. In this Series paper, we describe exciting developments in radiochemistry and the production of radionuclides, the development and translation of theranostics, and the application of artificial intelligence to our field. These developments are catalysing growth in the use of radiopharmaceuticals to the benefit of patients worldwide. We also highlight some of the key issues to be addressed in the coming years to realise the full potential of radiopharmaceuticals to treat cancer.
Collapse
Affiliation(s)
- Suzanne E Lapi
- Departments of Radiology and Chemistry, O'Neal Comprehensive Cancer Center, University of Alabama, Birmingham, AL, USA
| | - Peter J H Scott
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Andrew M Scott
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, VIC, Australia; Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia; School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia; Department of Surgery, Faculty of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - Albert D Windhorst
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, Amsterdam, Netherlands; Cancer Center Amsterdam, Vrije Universiteit, Amsterdam, Netherlands
| | - Brian M Zeglis
- Department of Chemistry, Hunter College, City University of New York, New York City, NY, USA; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA; Department of Radiology, Weill Cornell Medical College, New York City, NY, USA
| | - May Abdel-Wahab
- Division of Human Health, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| | - Richard P Baum
- Deutsche Klinik für Diagnostik (DKD Helios Klinik) Wiesbaden, Curanosticum MVZ Wiesbaden-Frankfurt, Center for Advanced Radiomolecular Precision Oncology, Germany
| | - John M Buatti
- Department of Radiation Oncology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Francesco Giammarile
- Nuclear Medicine and Diagnostic Imaging Section, Division of Human Health, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria; Centre Leon Bérard, Lyon, France
| | - Ana P Kiess
- Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Amirreza Jalilian
- Radiochemistry and Radiotechnology Section, Division of Physical and Chemical Sciences, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| | - Peter Knoll
- Dosimetry and Medical Radiation Physics Section, Division of Human Health, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| | - Aruna Korde
- Radiochemistry and Radiotechnology Section, Division of Physical and Chemical Sciences, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| | - Jolanta Kunikowska
- Nuclear Medicine Department, Medical University of Warsaw, Warsaw, Poland
| | - Sze Ting Lee
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, VIC, Australia; Olivia Newton-John Cancer Research Institute, Melbourne, VIC, Australia; School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia; Department of Surgery, Faculty of Medicine, University of Melbourne, Melbourne, VIC, Australia
| | - Diana Paez
- Nuclear Medicine and Diagnostic Imaging Section, Division of Human Health, Department of Nuclear Sciences and Applications, International Atomic Energy Agency, Vienna, Austria
| | - Jean-Luc Urbain
- Department of Radiology-Nuclear Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Jingjing Zhang
- Department of Diagnostic Radiology, National University of Singapore, Singapore; Clinical Imaging Research Centre, Nanomedicine Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York City, NY, USA; Department of Radiology, Weill Cornell Medical College, New York City, NY, USA; Department of Pharmacology, Weill Cornell Medical College, New York City, NY, USA.
| |
Collapse
|
18
|
Mohanto S, Biswas A, Gholap AD, Wahab S, Bhunia A, Nag S, Ahmed MG. Potential Biomedical Applications of Terbium-Based Nanoparticles (TbNPs): A Review on Recent Advancement. ACS Biomater Sci Eng 2024; 10:2703-2724. [PMID: 38644798 DOI: 10.1021/acsbiomaterials.3c01969] [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: 04/23/2024]
Abstract
The scientific world is increasingly focusing on rare earth metal oxide nanomaterials due to their consequential biological prospects, navigated by breakthroughs in biomedical applications. Terbium belongs to rare earth elements (lanthanide series) and possesses remarkably strong luminescence at lower energy emission and signal transduction properties, ushering in wide applications for diagnostic measurements (i.e., bioimaging, biosensors, fluorescence imaging, etc.) in the biomedical sectors. In addition, the theranostic applications of terbium-based nanoparticles further permit the targeted delivery of drugs to the specific site of the disease. Furthermore, the antimicrobial properties of terbium nanoparticles induced via reactive oxygen species (ROS) cause oxidative damage to the cell membrane and nuclei of living organisms, ion release, and surface charge interaction, thus further creating or exhibiting excellent antioxidant characteristics. Moreover, the recent applications of terbium nanoparticles in tissue engineering, wound healing, anticancer activity, etc., due to angiogenesis, cell proliferation, promotion of growth factors, biocompatibility, cytotoxicity mitigation, and anti-inflammatory potentials, make this nanoparticle anticipate a future epoch of nanomaterials. Terbium nanoparticles stand as a game changer in the realm of biomedical research, proffering a wide array of possibilities, from revolutionary imaging techniques to advanced drug delivery systems. Their unique properties, including luminescence, magnetic characteristics, and biocompatibility, have redefined the boundaries of what can be achieved in biomedicine. This review primarily delves into various mechanisms involved in biomedical applications via terbium-based nanoparticles due to their physicochemical characteristics. This review article further explains the potential biomedical applications of terbium nanoparticles with in-depth significant mechanisms from the individual literature. This review additionally stands as the first instance to furnish a "single-platted" comprehensive acquaintance of terbium nanoparticles in shaping the future of healthcare as well as potential limitations and overcoming strategies that require exploration before being trialed in clinical settings.
Collapse
Affiliation(s)
- Sourav Mohanto
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Aritra Biswas
- Department of Microbiology, Ramakrishna Mission Vivekananda Centenary College, P.O. Rahara, Kolkata, West Bengal 700118, India
| | - Amol Dilip Gholap
- Department of Pharmaceutics, St. John Institute of Pharmacy and Research, Palghar, Maharashtra 401404, India
| | - Shadma Wahab
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Adrija Bhunia
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| | - Sagnik Nag
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Selangor , Malaysia
| | - Mohammed Gulzar Ahmed
- Department of Pharmaceutics, Yenepoya Pharmacy College & Research Centre, Yenepoya (Deemed to be University), Mangalore, Karnataka 575018, India
| |
Collapse
|
19
|
Timmers HJLM, Taïeb D, Pacak K, Lenders JWM. Imaging of Pheochromocytomas and Paragangliomas. Endocr Rev 2024; 45:414-434. [PMID: 38206185 PMCID: PMC11074798 DOI: 10.1210/endrev/bnae001] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 12/11/2023] [Accepted: 01/10/2024] [Indexed: 01/12/2024]
Abstract
Pheochromocytomas/paragangliomas are unique in their highly variable molecular landscape driven by genetic alterations, either germline or somatic. These mutations translate into different clusters with distinct tumor locations, biochemical/metabolomic features, tumor cell characteristics (eg, receptors, transporters), and disease course. Such tumor heterogeneity calls for different imaging strategies in order to provide proper diagnosis and follow-up. This also warrants selection of the most appropriate and locally available imaging modalities tailored to an individual patient based on consideration of many relevant factors including age, (anticipated) tumor location(s), size, and multifocality, underlying genotype, biochemical phenotype, chance of metastases, as well as the patient's personal preference and treatment goals. Anatomical imaging using computed tomography and magnetic resonance imaging and functional imaging using positron emission tomography and single photon emission computed tomography are currently a cornerstone in the evaluation of patients with pheochromocytomas/paragangliomas. In modern nuclear medicine practice, a multitude of radionuclides with relevance to diagnostic work-up and treatment planning (theranostics) is available, including radiolabeled metaiodobenzylguanidine, fluorodeoxyglucose, fluorodihydroxyphenylalanine, and somatostatin analogues. This review amalgamates up-to-date imaging guidelines, expert opinions, and recent discoveries. Based on the rich toolbox for anatomical and functional imaging that is currently available, we aim to define a customized approach in patients with (suspected) pheochromocytomas/paragangliomas from a practical clinical perspective. We provide imaging algorithms for different starting points for initial diagnostic work-up and course of the disease, including adrenal incidentaloma, established biochemical diagnosis, postsurgical follow-up, tumor screening in pathogenic variant carriers, staging and restaging of metastatic disease, theranostics, and response monitoring.
Collapse
Affiliation(s)
- Henri J L M Timmers
- Department of Internal Medicine, Radboud University Medical Centre, 6525 GA Nijmegen, The Netherlands
| | - David Taïeb
- Department of Nuclear Medicine, La Timone University Hospital, Aix-Marseille University, Marseille, France and European Center for Research in Medical Imaging, Aix-Marseille University, 13005 Marseille, France
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892-1583, USA
| | - Jacques W M Lenders
- Department of Internal Medicine, Radboud University Medical Centre, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
20
|
Kong G, Buteau JP, Hofman MS. Is 161Tb Really Happening? J Nucl Med 2024; 65:686-687. [PMID: 38604760 DOI: 10.2967/jnumed.124.267611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 03/15/2024] [Indexed: 04/13/2024] Open
Affiliation(s)
- Grace Kong
- Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Centre Melbourne, and Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - James P Buteau
- Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Centre Melbourne, and Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Michael S Hofman
- Molecular Imaging and Therapeutic Nuclear Medicine, Cancer Imaging, Peter MacCallum Centre Melbourne, and Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
21
|
Holzleitner N, Cwojdzinski T, Beck R, Urtz-Urban N, Hillhouse CC, Grundler PV, van der Meulen NP, Talip Z, Ramaekers S, Van de Voorde M, Ponsard B, Casini A, Günther T. Preclinical Evaluation of Gastrin-Releasing Peptide Receptor Antagonists Labeled with 161Tb and 177Lu: A Comparative Study. J Nucl Med 2024; 65:481-484. [PMID: 38124121 PMCID: PMC10924159 DOI: 10.2967/jnumed.123.266233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 11/09/2023] [Accepted: 11/09/2023] [Indexed: 12/23/2023] Open
Abstract
To elucidate potential benefits of the Auger-electron-emitting radionuclide 161Tb, we compared the preclinical performance of the gastrin-releasing peptide receptor antagonists RM2 (DOTA-Pip5-d-Phe6-Gln7-Trp8-Ala9-Val10-Gly11-His12-Sta13-Leu14-NH2) and AMTG (α-Me-Trp8-RM2), each labeled with both 177Lu and 161Tb. Methods: 161Tb/177Lu labeling (90°C, 5 min) and cell-based experiments (PC-3 cells) were performed. In vivo stability (30 min after injection) and biodistribution studies (1-72 h after injection) were performed on PC-3 tumor-bearing CB17-SCID mice. Results: Gastrin-releasing peptide receptor affinity was high for all compounds (half-maximal inhibitory concentration [nM]: [161Tb]Tb-RM2, 2.46 ± 0.16; [161Tb]Tb-AMTG, 2.16 ± 0.09; [177Lu]Lu-RM2, 3.45 ± 0.18; [177Lu]Lu-AMTG, 3.04 ± 0.08), and 75%-84% of cell-associated activity was receptor-bound. In vivo, both AMTG analogs displayed distinctly higher stability (30 min after injection) and noticeably higher tumor retention than their RM2 counterparts. Conclusion: On the basis of preclinical results, [161Tb]Tb-/[177Lu]Lu-AMTG might reveal a higher therapeutic efficacy than [161Tb]Tb-/[177Lu]Lu-RM2, particularly [161Tb]Tb-AMTG because of additional Auger-electron emissions at the cell membrane level.
Collapse
Affiliation(s)
- Nadine Holzleitner
- Department of Chemistry, School of Natural Sciences, Technical University of Munich, Garching, Germany
| | - Tatjana Cwojdzinski
- Department of Chemistry, School of Natural Sciences, Technical University of Munich, Garching, Germany
| | - Roswitha Beck
- Department of Chemistry, School of Natural Sciences, Technical University of Munich, Garching, Germany
| | - Nicole Urtz-Urban
- Department of Chemistry, School of Natural Sciences, Technical University of Munich, Garching, Germany
| | - Colin C Hillhouse
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen, Switzerland; and
| | - Pascal V Grundler
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen, Switzerland; and
| | - Nicholas P van der Meulen
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen, Switzerland; and
| | - Zeynep Talip
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen, Switzerland; and
| | - Stijn Ramaekers
- Nuclear Medical Applications, Belgian Nuclear Research Centre, Mol, Belgium
| | | | - Bernard Ponsard
- Nuclear Medical Applications, Belgian Nuclear Research Centre, Mol, Belgium
| | - Angela Casini
- Department of Chemistry, School of Natural Sciences, Technical University of Munich, Garching, Germany;
| | - Thomas Günther
- Department of Chemistry, School of Natural Sciences, Technical University of Munich, Garching, Germany;
| |
Collapse
|
22
|
Palma E, Santos JF, Fernandes C, Paulo A. DNA-Targeted Complexes of Tc and Re for Biomedical Applications. Chemistry 2024; 30:e202303591. [PMID: 38038361 DOI: 10.1002/chem.202303591] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 11/30/2023] [Accepted: 12/01/2023] [Indexed: 12/02/2023]
Abstract
Due to their favorable chemical features, Re and Tc complexes have been widely used for the development of new therapeutic agents and imaging probes to solve problems of biomedical relevance. This review provides an update of the most relevant research efforts towards the development of novel cancer theranostic agents using Re and Tc-based compounds interacting with specific DNA structures. This includes a variety of homometallic complexes, namely those containing M(CO)3 (M=Re, Tc) moieties, that exhibit different modes of interaction with DNA, such as covalent binding, intercalation, groove binding or G-quadruplex DNA binding. Additionally, heterometallic complexes, designed to potentiate synergistic effects of different metal centers to improve DNA-targeting, cytotoxicity and fluorescence properties, are also reviewed. Particular attention is also given to 99m Tc- and 188 Re-labeled oligonucleotides that have been widely explored to develop imaging and therapeutic radiopharmaceuticals through the in vivo hybridization with a specific complementary DNA or RNA target sequence to provide useful molecular tools in precision medicine for cancer diagnosis and treatment. Finally, the need for further improvement of DNA-targeted Re and Tc-based compounds as potential therapeutic and diagnostic agents is highlighted, and future directions are discussed.
Collapse
Affiliation(s)
- Elisa Palma
- C2TN - Centro de Ciências e Tecnologias, Nucleares Instituto Superior Técnico, Universidade de Lisboa, Portugal
| | - Joana F Santos
- C2TN - Centro de Ciências e Tecnologias, Nucleares Instituto Superior Técnico, Universidade de Lisboa, Portugal
| | - Célia Fernandes
- C2TN - Centro de Ciências e Tecnologias, Nucleares Instituto Superior Técnico, Universidade de Lisboa, Portugal
- DECN - Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Portugal
| | - António Paulo
- C2TN - Centro de Ciências e Tecnologias, Nucleares Instituto Superior Técnico, Universidade de Lisboa, Portugal
- DECN - Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Portugal
| |
Collapse
|
23
|
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
|
24
|
Santos JF, Braz MT, Raposinho P, Cleeren F, Cassells I, Leekens S, Cawthorne C, Mendes F, Fernandes C, Paulo A. Synthesis and Preclinical Evaluation of PSMA-Targeted 111In-Radioconjugates Containing a Mitochondria-Tropic Triphenylphosphonium Carrier. Mol Pharm 2024; 21:216-233. [PMID: 37992229 DOI: 10.1021/acs.molpharmaceut.3c00787] [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: 11/24/2023]
Abstract
Nuclear DNA is the canonical target for biological damage induced by Auger electrons (AE) in the context of targeted radionuclide therapy (TRT) of cancer, but other subcellular components might also be relevant for this purpose, such as the energized mitochondria of tumor cells. Having this in mind, we have synthesized novel DOTA-based chelators carrying a prostate-specific membrane antigen (PSMA) inhibitor and a triphenyl phosphonium (TPP) group that were used to obtain dual-targeted 111In-radioconjugates ([111In]In-TPP-DOTAGA-PSMA and [111In]In-TPP-DOTAGA-G3-PSMA), aiming to promote a selective uptake of an AE-emitter radiometal (111In) by PSMA+ prostate cancer (PCa) cells and an enhanced accumulation in the mitochondria. These dual-targeted 111In-radiocomplexes are highly stable under physiological conditions and in cell culture media. The complexes showed relatively similar binding affinities toward the PSMA compared to the reference tracer [111In]In-PSMA-617, in line with their high cellular uptake and internalization in PSMA+ PCa cells. The complexes compromised cell survival in a dose-dependent manner and in the case of [111In]In-TPP-DOTAGA-G3-PSMA to a higher extent than observed for the single-targeted congener [111In]In-PSMA-617. μSPECT imaging studies in PSMA+ PCa xenografts showed that the TPP pharmacophore did not interfere with the excellent in vivo tumor uptake of the "golden standard" [111In]In-PSMA-617, although it led to a higher kidney retention. Such kidney retention does not necessarily compromise their usefulness as radiotherapeutics due to the short tissue range of the Auger/conversion electrons emitted by 111In. Overall, our results provide valuable insights into the potential use of mitochondrial targeting by PSMA-based radiocomplexes for efficient use of AE-emitting radionuclides in TRT, giving impetus to extend the studies to other AE-emitting trivalent radiometals (e.g., 161Tb or 165Er) and to further optimize the designed dual-targeting constructs.
Collapse
Affiliation(s)
- Joana F Santos
- C2TN - Centro de Ciências e Tecnologias Nucleares Instituto Superior Técnico, Universidade de Lisboa, 2695-066 Bobadela LRS, Portugal
| | - Maria T Braz
- C2TN - Centro de Ciências e Tecnologias Nucleares Instituto Superior Técnico, Universidade de Lisboa, 2695-066 Bobadela LRS, Portugal
| | - Paula Raposinho
- C2TN - Centro de Ciências e Tecnologias Nucleares Instituto Superior Técnico, Universidade de Lisboa, 2695-066 Bobadela LRS, Portugal
- DECN - Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, 2695-066 Bobadela LRS, Portugal
| | - Frederik Cleeren
- Laboratory for Radiopharmaceutical Research, Department of Pharmacy and Pharmacology, University of Leuven, B-3000 Leuven, Belgium
| | - Irwin Cassells
- Laboratory for Radiopharmaceutical Research, Department of Pharmacy and Pharmacology, University of Leuven, B-3000 Leuven, Belgium
- Nuclear Medical Applications, Belgian Nuclear Research Centre (SCK CEN), 2400 Mol, Belgium
| | - Simon Leekens
- Laboratory for Radiopharmaceutical Research, Department of Pharmacy and Pharmacology, University of Leuven, B-3000 Leuven, Belgium
| | - Christopher Cawthorne
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, University of Leuven, 3000 Leuven, Belgium
| | - Filipa Mendes
- C2TN - Centro de Ciências e Tecnologias Nucleares Instituto Superior Técnico, Universidade de Lisboa, 2695-066 Bobadela LRS, Portugal
- DECN - Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, 2695-066 Bobadela LRS, Portugal
| | - Célia Fernandes
- C2TN - Centro de Ciências e Tecnologias Nucleares Instituto Superior Técnico, Universidade de Lisboa, 2695-066 Bobadela LRS, Portugal
- DECN - Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, 2695-066 Bobadela LRS, Portugal
| | - António Paulo
- C2TN - Centro de Ciências e Tecnologias Nucleares Instituto Superior Técnico, Universidade de Lisboa, 2695-066 Bobadela LRS, Portugal
- DECN - Departamento de Engenharia e Ciências Nucleares, Instituto Superior Técnico, Universidade de Lisboa, 2695-066 Bobadela LRS, Portugal
| |
Collapse
|
25
|
Uygur E, Sezgin C, Parlak Y, Karatay KB, Arikbasi B, Avcibasi U, Toklu T, Barutca S, Harmansah C, Sozen TS, Maus S, Scher H, Aras O, Gumuser FG, Muftuler FZB. The Radiolabeling of [161Tb]-PSMA-617 by a Novel Radiolabeling Method and Preclinical Evaluation by In Vitro/In Vivo Methods. RESEARCH SQUARE 2023:rs.3.rs-3415703. [PMID: 37961521 PMCID: PMC10635383 DOI: 10.21203/rs.3.rs-3415703/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Background Prostate cancer (PC) is the most common type of cancer in elderly men, with a positive correlation with age. As resistance to treatment has developed, particularly in the progressive stage of the disease and in the presence of microfocal multiple bone metastases, new generation radionuclide therapies have emerged. Recently, [161Tb], a radiolanthanide introduced for treating micrometastatic foci, has shown great promise for treating prostate cancer. Results In this study, Terbium-161 [161Tb]Tb was radiolabeled with prostate-specific membrane antigen (PSMA)-617 ([161Tb]-PSMA-617) and the therapeutic efficacy of the radiolabeled compound investigated in vitro and in vivo. [161Tb]-PSMA-617 was found to have a radiochemical yield of 97.99 ± 2.01% and was hydrophilic. [161Tb]-PSMA-617 was also shown to have good stability, with a radiochemical yield of over 95% up to 72 hours. In vitro, [161Tb]-PSMA-617 showed a cytotoxic effect on LNCaP cells but not on PC-3 cells. In vivo, scintigraphy imaging visualized the accumulation of [161Tb]-PSMA-617 in the prostate, kidneys, and bladder. Conclusions The results suggest that [161Tb]-PSMA-617 can be an effective radiolabeled agent for the treatment of PSMA positive foci in prostate cancer.
Collapse
Affiliation(s)
- Emre Uygur
- Manisa Celal Bayar University: Manisa Celal Bayar Universitesi
| | | | - Yasemin Parlak
- Manisa Celal Bayar University: Manisa Celal Bayar Universitesi
| | - Kadriye Busra Karatay
- Ege University Institute of Nuclear Sciences: Ege Universitesi Nukleer Bilimler Enstitusu
| | | | - Ugur Avcibasi
- Manisa Celal Bayar Üniversitesi: Manisa Celal Bayar Universitesi
| | | | - Sabri Barutca
- Adnan Menderes Üniversitesi Tıp Fakültesi: Adnan Menderes Universitesi Tip Fakultesi
| | | | | | - Stephan Maus
- Saarland University Hospital and Saarland University Faculty of Medicine: Universitatsklinikum des Saarlandes und Medizinische Fakultat der Universitat des Saarlandes
| | - Howard Scher
- Memorial Sloan-Kettering Cancer Center Inpatient Hospital and Main Campus: Memorial Sloan Kettering Cancer Center
| | - Omer Aras
- Memorial Sloan-Kettering Cancer Center Inpatient Hospital and Main Campus: Memorial Sloan Kettering Cancer Center
| | | | | |
Collapse
|
26
|
DaSilva J, Decristoforo C, Mach RH, Bormans G, Carlucci G, Al-Qahtani M, Duatti A, Gee AD, Szymanski W, Rubow S, Hendrikx J, Yang X, Jia H, Zhang J, Caravan P, Yang H, Zeevaart JR, Rodriquez MA, Oliveira RS, Zubillaga M, Sakr T, Spreckelmeyer S. Highlight selection of radiochemistry and radiopharmacy developments by editorial board. EJNMMI Radiopharm Chem 2023; 8:35. [PMID: 37889361 PMCID: PMC10611660 DOI: 10.1186/s41181-023-00218-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 10/13/2023] [Indexed: 10/28/2023] Open
Abstract
BACKGROUND The Editorial Board of EJNMMI Radiopharmacy and Chemistry releases a biannual highlight commentary to update the readership on trends in the field of radiopharmaceutical development. MAIN BODY This selection of highlights provides commentary on 21 different topics selected by each coauthoring Editorial Board member addressing a variety of aspects ranging from novel radiochemistry to first-in-human application of novel radiopharmaceuticals. CONCLUSION Trends in radiochemistry and radiopharmacy are highlighted. Hot topics cover the entire scope of EJNMMI Radiopharmacy and Chemistry, demonstrating the progress in the research field in many aspects.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Xing Yang
- Peking University First Hospital, Beijing, China
| | | | | | - Peter Caravan
- Massuchusetts General Hospital, Harvard University, Boston, USA
| | | | | | | | - Ralph Santos Oliveira
- Brazilian Association of Radiopharmacy Brazil, Brazilian Nuclear Energy Commission - Nuclear Engineering Institute, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Tamer Sakr
- Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Sarah Spreckelmeyer
- Department of Nuclear Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität Zu Berlin, and Berlin Institute of Health, Augustenburger Platz 1, 13353, Berlin, Germany.
| |
Collapse
|
27
|
Renaldin E, Dellepiane G, Braccini S, Sommerhalder A, Zhang H, van der Meulen NP, Eichler R, Talip Z. Study of thulium-167 cyclotron production: a potential medically-relevant radionuclide. Front Chem 2023; 11:1288588. [PMID: 37927558 PMCID: PMC10620610 DOI: 10.3389/fchem.2023.1288588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 10/04/2023] [Indexed: 11/07/2023] Open
Abstract
Introduction: Targeted Radionuclide Therapy is used for the treatment of tumors in nuclear medicine, while sparing healthy tissues. Its application to cancer treatment is expanding. In particular, Auger-electron emitters potentially exhibit high efficacy in treating either small metastases or single tumor cells due to their short range in tissue. The aim of this paper is to study the feasibility of a large-scale production of thulium-167, an Auger-electron emitter radionuclide, in view of eventual systematic preclinical studies. Methods: Proton-irradiated enriched erbium-167 and erbium-168 oxides were used to measure the production cross sections of thulium-165, thulium-166, thulium-167, and thulium-168 utilizing an 18-MeV medical cyclotron equipped with a Beam Transport Line (BTL) at the Bern medical cyclotron laboratory. The comparison between the experimental and the TENDL 2021 theoretical cross-section results were in good agreement. Additional experiments were performed to assess the production yields of thulium radioisotopes in the BTL. Thulium-167 production yield was also measured irradiating five different target materials (167 Er 2 O 3, 168 Er 2 O 3, nat Tm 2 O 3, nat Yb 2 O 3, 171 Yb 2 O 3) with proton beams up to 63 MeV at the Injector II cyclotron of Paul Scherrer Institute. Results and Discussion: Our experiments showed that an 8-h irradiation of enriched ytterbium-171 oxide produced about 420 MBq of thulium-167 with a radionuclidic purity of 99.95% after 5 days of cooling time with a proton beam of about 53 MeV. Larger activities of thulium-167 can be achieved using enriched erbium-168 oxide with a 23-MeV proton beam, obtaining about 1 GBq after 8-h irradiation with a radionuclidic purity of < 99.5% 5 days post end of bombardment.
Collapse
Affiliation(s)
- Edoardo Renaldin
- Center for Radiopharmaceutical Sciences (CRS), Paul Scherrer Institute, Villigen-PSI, Switzerland
- Department of Chemistry, Biochemistry and Pharmaceutical sciences (DCBP), University of Bern, Bern, Switzerland
| | - Gaia Dellepiane
- Albert Einstein Center for Fundamental Physics (AEC), Laboratory of High Energy Physics (LHEP), University of Bern, Bern, Switzerland
| | - Saverio Braccini
- Albert Einstein Center for Fundamental Physics (AEC), Laboratory of High Energy Physics (LHEP), University of Bern, Bern, Switzerland
| | - Alexander Sommerhalder
- Center for Radiopharmaceutical Sciences (CRS), Paul Scherrer Institute, Villigen-PSI, Switzerland
| | - Hui Zhang
- Beam Physics, Proton Facilities, Accelerator Operation and Development, Large Research Facilities, Paul Scherrer Institute, Villigen-PSI, Switzerland
| | - Nicholas P. van der Meulen
- Center for Radiopharmaceutical Sciences (CRS), Paul Scherrer Institute, Villigen-PSI, Switzerland
- Laboratory of Radiochemistry (LRC), Paul Scherrer Institute, Villigen-PSI, Switzerland
| | - Robert Eichler
- Department of Chemistry, Biochemistry and Pharmaceutical sciences (DCBP), University of Bern, Bern, Switzerland
- Laboratory of Radiochemistry (LRC), Paul Scherrer Institute, Villigen-PSI, Switzerland
| | - Zeynep Talip
- Center for Radiopharmaceutical Sciences (CRS), Paul Scherrer Institute, Villigen-PSI, Switzerland
| |
Collapse
|
28
|
Echavidre W, Fagret D, Faraggi M, Picco V, Montemagno C. Recent Pre-Clinical Advancements in Nuclear Medicine: Pioneering the Path to a Limitless Future. Cancers (Basel) 2023; 15:4839. [PMID: 37835533 PMCID: PMC10572076 DOI: 10.3390/cancers15194839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 10/15/2023] Open
Abstract
The theranostic approach in oncology holds significant importance in personalized medicine and stands as an exciting field of molecular medicine. Significant achievements have been made in this field in recent decades, particularly in treating neuroendocrine tumors using 177-Lu-radiolabeled somatostatin analogs and, more recently, in addressing prostate cancer through prostate-specific-membrane-antigen targeted radionuclide therapy. The promising clinical results obtained in these indications paved the way for the further development of this approach. With the continuous discovery of new molecular players in tumorigenesis, the development of novel radiopharmaceuticals, and the potential combination of theranostics agents with immunotherapy, nuclear medicine is poised for significant advancements. The strategy of theranostics in oncology can be categorized into (1) repurposing nuclear medicine agents for other indications, (2) improving existing radiopharmaceuticals, and (3) developing new theranostics agents for tumor-specific antigens. In this review, we provide an overview of theranostic development and shed light on its potential integration into combined treatment strategies.
Collapse
Affiliation(s)
- William Echavidre
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (W.E.); (V.P.)
| | - Daniel Fagret
- Laboratory of Bioclinical Radiopharmaceutics, Universite Grenoble Alpes, CHU Grenoble Alpes, Inserm, 38000 Grenoble, France;
| | - Marc Faraggi
- Nuclear Medicine Department, Centre Hospitalier Princesse Grace, 98000 Monaco, Monaco;
| | - Vincent Picco
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (W.E.); (V.P.)
| | - Christopher Montemagno
- Biomedical Department, Centre Scientifique de Monaco, 98000 Monaco, Monaco; (W.E.); (V.P.)
| |
Collapse
|
29
|
Koller L, Joksch M, Schwarzenböck S, Kurth J, Heuschkel M, Holzleitner N, Beck R, von Amsberg G, Wester HJ, Krause BJ, Günther T. Preclinical Comparison of the 64Cu- and 68Ga-Labeled GRPR-Targeted Compounds RM2 and AMTG, as Well as First-in-Humans [ 68Ga]Ga-AMTG PET/CT. J Nucl Med 2023; 64:1654-1659. [PMID: 37934025 PMCID: PMC10586479 DOI: 10.2967/jnumed.123.265771] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 06/12/2023] [Indexed: 07/22/2023] Open
Abstract
Despite the recent success of prostate-specific membrane antigen (PSMA)-targeted compounds for theranostic use in prostate cancer (PCa), alternative options for the detection and treatment of PSMA-negative lesions are needed. We have recently developed a novel gastrin-releasing peptide receptor (GRPR) ligand with improved metabolic stability, which might improve diagnostic and therapeutic efficacy and could be valuable for PSMA-negative PCa patients. Our aim was to examine its suitability for theranostic use. We performed a comparative preclinical study on [64Cu]Cu-/[68Ga]Ga-AMTG ([64Cu]Cu-/[68Ga]Ga-α-Me-l-Trp8-RM2) using [64Cu]Cu-/[68Ga]Ga-RM2 ([64Cu]Cu-/[68Ga]Ga-DOTA-Pip5-Phe6-Gln7-Trp8-Ala9-Val10-Gly11-His12-Sta13-Leu14-NH2) as a reference compound and investigated [68Ga]Ga-AMTG in a proof-of-concept study in a PCa patient. Methods: Peptides were labeled with 64Cu (80 °C, 1.0 M NaOAc, pH 5.50) and 68Ga (90 °C, 0.25 M NaOAc, pH 4.50). GRPR affinity (half-maximal inhibitory concentration, room temperature, 2 h) and GRPR-mediated internalization (37 °C, 60 min) were examined on PC-3 cells. Biodistribution studies were performed at 1 h after injection in PC-3 tumor-bearing mice. For a first-in-humans application, 173 MBq of [68Ga]Ga-AMTG were administered intravenously and whole-body PET/CT scans were acquired at 75 min after injection. Results: 64Cu- and 68Ga-labeling proceeded almost quantitatively (>98%). All compounds revealed similarly high GRPR affinity (half-maximal inhibitory concentration, 1.5-4.0 nM) and high receptor-bound fractions (79%-84% of cell-associated activity). In vivo, high activity levels (percentage injected dose per gram) were found in the PC-3 tumor (14.1-15.1 %ID/g) and the pancreas (12.6-30.7 %ID/g), whereas further off-target accumulation was low at 1 h after injection, except for elevated liver uptake observed for both 64Cu-labeled compounds. Overall biodistribution profiles and tumor-to-background ratios were comparable but slightly enhanced for the 68Ga-labeled analogs in most organs. [68Ga]Ga-AMTG confirmed the favorable pharmacokinetics-as evident from preclinical studies-in a patient with metastasized castration-resistant PCa showing intense uptake in several lesions. Conclusion: AMTG is eligible for theranostic use, as labeling with 64Cu and 68Ga, as well as 177Lu (known from previous study), does not have a negative influence on its favorable biodistribution pattern. For this reason, further clinical evaluation is warranted.
Collapse
Affiliation(s)
- Lena Koller
- Pharmaceutical Radiochemistry, Technical University of Munich, Garching, Germany
| | - Markus Joksch
- Department of Nuclear Medicine, Rostock University Medical Center, Rostock, Germany; and
| | - Sarah Schwarzenböck
- Department of Nuclear Medicine, Rostock University Medical Center, Rostock, Germany; and
| | - Jens Kurth
- Department of Nuclear Medicine, Rostock University Medical Center, Rostock, Germany; and
| | - Martin Heuschkel
- Department of Nuclear Medicine, Rostock University Medical Center, Rostock, Germany; and
| | - Nadine Holzleitner
- Pharmaceutical Radiochemistry, Technical University of Munich, Garching, Germany
| | - Roswitha Beck
- Pharmaceutical Radiochemistry, Technical University of Munich, Garching, Germany
| | - Gunhild von Amsberg
- Department of Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans-Jürgen Wester
- Pharmaceutical Radiochemistry, Technical University of Munich, Garching, Germany
| | - Bernd Joachim Krause
- Department of Nuclear Medicine, Rostock University Medical Center, Rostock, Germany; and
| | - Thomas Günther
- Pharmaceutical Radiochemistry, Technical University of Munich, Garching, Germany;
| |
Collapse
|
30
|
Tschan VJ, Busslinger SD, Bernhardt P, Grundler PV, Zeevaart JR, Köster U, van der Meulen NP, Schibli R, Müller C. Albumin-Binding and Conventional PSMA Ligands in Combination with 161Tb: Biodistribution, Dosimetry, and Preclinical Therapy. J Nucl Med 2023; 64:1625-1631. [PMID: 37442604 DOI: 10.2967/jnumed.123.265524] [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] [Received: 01/28/2023] [Revised: 05/31/2023] [Indexed: 07/15/2023] Open
Abstract
The favorable decay characteristics of 161Tb attracted the interest of clinicians in using this novel radionuclide for radioligand therapy (RLT). 161Tb decays with a similar half-life to 177Lu, but beyond the emission of β--particles and γ-rays, 161Tb also emits conversion and Auger electrons, which may be particularly effective to eliminate micrometastases. The aim of this study was to compare the dosimetry and therapeutic efficacy of 161Tb and 177Lu in tumor-bearing mice using SibuDAB and PSMA-I&T, which differ in their blood residence time and tumor uptake. Methods: [161Tb]Tb-SibuDAB and [161Tb]Tb-PSMA-I&T were evaluated in vitro and investigated in biodistribution, imaging, and therapy studies using PC-3 PIP tumor-bearing mice. The 177Lu-labeled counterparts served for dose calculations and comparison of therapeutic efficacy. The tolerability of RLT in mice was monitored on the basis of body mass, blood plasma parameters, blood cell counts, and the histology of relevant organs and tissues. Results: The prostate-specific membrane antigen (PSMA)-targeting radioligands, irrespective of whether labeled with 161Tb or 177Lu, showed similar in vitro data and comparable tissue distribution profiles. As a result of the albumin-binding properties, [161Tb]Tb/[177Lu]Lu-SibuDAB had an enhanced blood residence time and higher tumor uptake (62%-69% injected activity per gram at 24 h after injection) than [161Tb]Tb/[177Lu]Lu-PSMA-I&T (30%-35% injected activity per gram at 24 h after injection). [161Tb]Tb-SibuDAB inhibited tumor growth more effectively than [161Tb]Tb-PSMA-I&T, as can be ascribed to its 4-fold increased absorbed tumor dose. At any of the applied activities, the 161Tb-based radioligands were therapeutically more effective than their 177Lu-labeled counterparts, as agreed with the approximately 40% increased tumor dose of 161Tb compared with that of 177Lu. Under the given experimental conditions, no obvious adverse events were observed. Conclusion: The data of this study indicate the promising potential of 161Tb in combination with SibuDAB for RLT of prostate cancer. Future clinical studies using 161Tb-based RLT will shed light on a potential clinical benefit of 161Tb over 177Lu.
Collapse
Affiliation(s)
- Viviane J Tschan
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, Switzerland
| | - Sarah D Busslinger
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, Switzerland
| | - Peter Bernhardt
- Department of Radiation Physics, Institution of Clinical Science, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Pascal V Grundler
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, Switzerland
| | - Jan Rijn Zeevaart
- Radiochemistry, South African Nuclear Energy Corporation (Necsa), Brits, South Africa
| | | | - Nicholas P van der Meulen
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, Switzerland
- Laboratory of Radiochemistry, Paul Scherrer Institute, Villigen-PSI, Switzerland; and
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Villigen-PSI, Switzerland;
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
31
|
Larouze A, Alcocer-Ávila M, Morgat C, Champion C, Hindié E. Membrane and Nuclear Absorbed Doses from 177Lu and 161Tb in Tumor Clusters: Effect of Cellular Heterogeneity and Potential Benefit of Dual Targeting-A Monte Carlo Study. J Nucl Med 2023; 64:1619-1624. [PMID: 37321819 DOI: 10.2967/jnumed.123.265509] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 05/11/2023] [Indexed: 06/17/2023] Open
Abstract
Early use of targeted radionuclide therapy to eradicate tumor cell clusters and micrometastases might offer cure. However, there is a need to select appropriate radionuclides and assess the potential impact of heterogeneous targeting. Methods: The Monte Carlo code CELLDOSE was used to assess membrane and nuclear absorbed doses from 177Lu and 161Tb (β--emitter with additional conversion and Auger electrons) in a cluster of 19 cells (14-μm diameter, 10-μm nucleus). The radionuclide distributions considered were cell surface, intracytoplasmic, or intranuclear, with 1,436 MeV released per labeled cell. To model heterogeneous targeting, 4 of the 19 cells were unlabeled, their position being stochastically determined. We simulated situations of single targeting, as well as dual targeting, with the 2 radiopharmaceuticals aiming at different targets. Results: 161Tb delivered 2- to 6-fold higher absorbed doses to cell membranes and 2- to 3-fold higher nuclear doses than 177Lu. When all 19 cells were targeted, membrane and nuclear absorbed doses were dependent mainly on radionuclide location. With cell surface location, membrane absorbed doses were substantially higher than nuclear absorbed doses, both with 177Lu (38-41 vs. 4.7-7.2 Gy) and with 161Tb (237-244 vs. 9.8-15.1 Gy). However, when 4 cells were not targeted by the cell surface radiopharmaceutical, the membranes of these cells received on average only 9.6% of the 177Lu absorbed dose and 2.9% of the 161Tb dose, compared with a cluster with uniform cell targeting, whereas the impact on nuclear absorbed doses was moderate. With an intranuclear radionuclide location, the nuclei of unlabeled cells received only 17% of the 177Lu absorbed dose and 10.8% of the 161Tb dose, compared with situations with uniform targeting. With an intracytoplasmic location, nuclear and membrane absorbed doses to unlabeled cells were one half to one quarter those obtained with uniform targeting, both for 177Lu and for 161Tb. Dual targeting was beneficial in minimizing absorbed dose heterogeneities. Conclusion: To eradicate tumor cell clusters, 161Tb may be a better candidate than 177Lu. Heterogeneous cell targeting can lead to substantial heterogeneities in absorbed doses. Dual targeting was helpful in reducing dose heterogeneity and should be explored in preclinical and clinical studies.
Collapse
Affiliation(s)
- Alexandre Larouze
- Université de Bordeaux-CNRS-CEA, Centre Lasers Intenses et Applications, UMR 5107, Talence, France
| | - Mario Alcocer-Ávila
- Université de Bordeaux-CNRS-CEA, Centre Lasers Intenses et Applications, UMR 5107, Talence, France
| | - Clément Morgat
- Service de Médecine Nucléaire, CHU de Bordeaux, Université de Bordeaux, UMR CNRS 5287, INCIA, Talence, France; and
| | - Christophe Champion
- Université de Bordeaux-CNRS-CEA, Centre Lasers Intenses et Applications, UMR 5107, Talence, France;
| | - Elif Hindié
- Service de Médecine Nucléaire, CHU de Bordeaux, Université de Bordeaux, UMR CNRS 5287, INCIA, Talence, France; and
- Institut Universitaire de France, Paris, France
| |
Collapse
|
32
|
Kersting D, Sandach P, Sraieb M, Wiesweg M, Metzenmacher M, Darwiche K, Oezkan F, Bölükbas S, Stuschke M, Umutlu L, Nader M, Hamacher R, Fendler WP, Wienker J, Eberhardt WEE, Schuler M, Herrmann K, Hautzel H. 68Ga-SSO-120 PET for Initial Staging of Small Cell Lung Cancer Patients: A Single-Center Retrospective Study. J Nucl Med 2023; 64:1540-1549. [PMID: 37474272 DOI: 10.2967/jnumed.123.265664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/24/2023] [Indexed: 07/22/2023] Open
Abstract
PET imaging using the somatostatin receptor 2 (SSTR2) antagonist satoreotide trizoxetan (SSO-120, previously OPS-202) could offer accurate tumor detection and screening for SSTR2-antagonist radionuclide therapy in patients with SSTR2-expressing small cell lung cancer (SCLC). The aim of this single-center study was to investigate tumor uptake and detection rates of 68Ga-SSO-120 in comparison to 18F-FDG PET in the initial staging of SCLC patients. Methods: Patients with newly diagnosed SCLC who underwent additional whole-body 68Ga-SSO-120 PET/CT during the initial diagnostic workup were retrospectively included. The mean administered activity was 139 MBq, and the mean uptake time was 60 min. Gold-standard staging 18F-FDG PET/CT was evaluated if available within 2 wk before or after 68Ga-SSO-120 PET if morphologic differences in CT images were absent. 68Ga-SSO-120- or 18F-FDG-positive lesions were reported in 7 anatomic regions (primary tumor, thoracic lymph node metastases, and distant metastases including pleural, contralateral pulmonary, liver, bone, and other) according to the TNM classification for lung cancer (eighth edition). Consensus TNM staging (derived from CT, endobronchial ultrasound-guided transbronchial needle aspiration, PET, and brain MRI) by a clinical tumor board served as the reference standard. Results: Thirty-one patients were included, 12 with limited and 19 with extensive disease according to the Veterans Administration Lung Study Group classification. 68Ga-SSO-120-positive tumor was detected in all patients (100%) and in 90 of the 217 evaluated regions (41.5%). Thirteen patients (42.0%) had intense average 68Ga-SSO-120 uptake (region-based mean SUVmax ≥ 10); 28 patients (90.3%) had average 68Ga-SSO-120 uptake greater than liver uptake (region-based mean peak tumor-to-liver ratio > 1). In 25 patients with evaluable 18F-FDG PET, primary tumor, thoracic lymph node metastases, and distant metastases were detected in 100%, 92%, and 64%, respectively, of all investigated patients by 68Ga-SSO-120 and in 100%, 92%, and 56%, respectively, by 18F-FDG PET. 68Ga-SSO-120 PET detected additional contralateral lymph node, liver, and brain metastases in 1, 1, and 2 patients, respectively (no histopathology available), and 18F-FDG PET detected additional contralateral lymph node metastases in 3 patients (1 confirmed, 1 systematic endobronchial ultrasound-guided transbronchial needle aspiration-negative, and 1 without available histopathology). None of these differences altered Veterans Administration Lung Study Group staging. The region-based monotonic correlation between 68Ga-SSO-120 and 18F-FDG uptake was low (Spearman ρ = 0.26-0.33). Conclusion: 68Ga-SSO-120 PET offers high diagnostic precision with comparable detection rates and additional complementary information to the gold standard, 18F-FDG PET. Consistent uptake in most patients warrants exploration of SSTR2-directed radionuclide therapy.
Collapse
Affiliation(s)
- David Kersting
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany;
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - Patrick Sandach
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - Miriam Sraieb
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - Marcel Wiesweg
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Martin Metzenmacher
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Kaid Darwiche
- Department of Pulmonary Medicine, Section of Interventional Pulmonology, West German Cancer Center, University Medicine Essen-Ruhrlandklinik, University of Duisburg-Essen, Essen, Germany
| | - Filiz Oezkan
- Department of Pulmonary Medicine, Section of Interventional Pulmonology, West German Cancer Center, University Medicine Essen-Ruhrlandklinik, University of Duisburg-Essen, Essen, Germany
| | - Servet Bölükbas
- Department of Thoracic Surgery and Thoracic Endoscopy, West German Cancer Center, University Medicine Essen-Ruhrlandklinik, University of Duisburg-Essen, Essen, Germany
| | - Martin Stuschke
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
- Department of Radiotherapy, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Lale Umutlu
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
- Institute of Diagnostic and Interventional Radiology and Neuroradiology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany; and
| | - Michael Nader
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - Rainer Hamacher
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Wolfgang P Fendler
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - Johannes Wienker
- Department of Pulmonary Medicine, Section of Interventional Pulmonology, West German Cancer Center, University Medicine Essen-Ruhrlandklinik, University of Duisburg-Essen, Essen, Germany
- Division of Thoracic Oncology, West German Lung Center, University Medicine Essen-Ruhrlandklinik, University of Duisburg-Essen, Essen, Germany
| | - Wilfried E E Eberhardt
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Division of Thoracic Oncology, West German Lung Center, University Medicine Essen-Ruhrlandklinik, University of Duisburg-Essen, Essen, Germany
| | - Martin Schuler
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
- Department of Medical Oncology, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Division of Thoracic Oncology, West German Lung Center, University Medicine Essen-Ruhrlandklinik, University of Duisburg-Essen, Essen, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - Hubertus Hautzel
- Department of Nuclear Medicine, West German Cancer Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| |
Collapse
|
33
|
Müller C, van der Meulen NP, Schibli R. Opportunities and potential challenges of using terbium-161 for targeted radionuclide therapy in clinics. Eur J Nucl Med Mol Imaging 2023; 50:3181-3184. [PMID: 37436459 DOI: 10.1007/s00259-023-06316-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2023]
Affiliation(s)
- Cristina Müller
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Forschungsstrasse 111, Villigen-PSI, Switzerland.
- Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, Zurich, Switzerland.
| | - Nicholas P van der Meulen
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Forschungsstrasse 111, Villigen-PSI, Switzerland
- Laboratory of Radiochemistry, Paul Scherrer Institute, Forschungsstrasse 111, Villigen-PSI, Switzerland
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences ETH-PSI, Paul Scherrer Institute, Forschungsstrasse 111, Villigen-PSI, Switzerland
- Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, Zurich, Switzerland
| |
Collapse
|
34
|
Bolcaen J, Gizawy MA, Terry SYA, Paulo A, Cornelissen B, Korde A, Engle J, Radchenko V, Howell RW. Marshalling the Potential of Auger Electron Radiopharmaceutical Therapy. J Nucl Med 2023; 64:1344-1351. [PMID: 37591544 PMCID: PMC10478825 DOI: 10.2967/jnumed.122.265039] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/05/2023] [Indexed: 08/19/2023] Open
Abstract
Auger electron (AE) radiopharmaceutical therapy (RPT) may have the same therapeutic efficacy as α-particles for oncologic small disease, with lower risks of normal-tissue toxicity. The seeds of using AE emitters for RPT were planted several decades ago. Much knowledge has been gathered about the potency of the biologic effects caused by the intense shower of these low-energy AEs. Given their short range, AEs deposit much of their energy in the immediate vicinity of their site of decay. However, the promise of AE RPT has not yet been realized, with few agents evaluated in clinical trials and none becoming part of routine treatment so far. Instigated by the 2022 "Technical Meeting on Auger Electron Emitters for Radiopharmaceutical Developments" at the International Atomic Energy Agency, this review presents the current status of AE RPT based on the discussions by experts in the field. A scoring system was applied to illustrate hurdles in the development of AE RPT, and we present a selected list of well-studied and emerging AE-emitting radionuclides. Based on the number of AEs and other emissions, physical half-life, radionuclide production, radiochemical approaches, dosimetry, and vector availability, recommendations are put forward to enhance and impact future efforts in AE RPT research.
Collapse
Affiliation(s)
- Julie Bolcaen
- SSC Laboratory, Radiation Biophysics, NRF iThemba LABS, Cape Town, South Africa
| | - Mohamed A Gizawy
- Egyptian Second Research Reactor Complex, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Samantha Y A Terry
- School of Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | - António Paulo
- Centro de Ciências e Tecnologias Nucleares, Instituto Superior Técnico, Universidade de Lisboa, Campus Tecnológico e Nuclear, Bobadela, Portugal
| | - Bart Cornelissen
- Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Aruna Korde
- Division of Physical and Chemical Sciences, Department of Nuclear Sciences and Application, International Atomic Energy Agency, Vienna, Austria
| | - Jonathan Engle
- University of Wisconsin Cyclotron Research Group, Departments of Medical Physics and Radiology, Madison, Wisconsin
| | - Valery Radchenko
- TRIUMF, Life Sciences Division, Vancouver, British Columbia, Canada;
- University of British Columbia, Chemistry Department, Vancouver, British Columbia, Canada; and
| | - Roger W Howell
- Division of Radiation Research, Department of Radiology and Center for Cell Signaling, New Jersey Medical School, Rutgers University, Newark, New Jersey
| |
Collapse
|
35
|
Favaretto C, Grundler PV, Talip Z, Landolt S, Sepini L, Köster U, Müller C, Schibli R, Geistlich S, van der Meulen NP. 161Tb-DOTATOC Production Using a Fully Automated Disposable Cassette System: A First Step Toward the Introduction of 161Tb into the Clinic. J Nucl Med 2023:jnumed.122.265268. [PMID: 37201956 DOI: 10.2967/jnumed.122.265268] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/01/2023] [Indexed: 05/20/2023] Open
Abstract
161Tb is an interesting radionuclide for application in the treatment of neuroendocrine neoplasms' small metastases and single cancer cells because of its conversion and Auger-electron emission. Tb has coordination chemistry similar to that of Lu; therefore, like 177Lu, it can stably radiolabel DOTATOC, one of the leading peptides used for the treatment of neuroendocrine neoplasms. However, 161Tb is a recently developed radionuclide that has not yet been specified for clinical use. Therefore, the aim of the current work was to characterize and specify 161Tb and to develop a protocol for the synthesis and quality control of 161Tb-DOTATOC with a fully automated process conforming to good-manufacturing-practice guidelines, in view of its clinical use. Methods: 161Tb, produced by neutron irradiation of 160Gd in high-flux reactors followed by radiochemical separation from its target material, was characterized regarding its radionuclidic purity, chemical purity, endotoxin level, and radiochemical purity (RCP) in analogy to what is described in the European Pharmacopoeia for no-carrier-added 177Lu. In addition, 161Tb was introduced into a fully automated cassette-module synthesis to produce 161Tb-DOTATOC, as used for 177Lu-DOTATOC. The quality and stability of the produced radiopharmaceutical in terms of identity, RCP, and ethanol and endotoxin content were assessed by means of high-performance liquid chromatography, gas chromatography, and an endotoxin test, respectively. Results: 161Tb produced under the described conditions showed, as the no-carrier-added 177Lu, a pH of 1-2, radionuclidic purity and RCP of more than 99.9%, and an endotoxin level below the permitted range (175 IU/mL), indicating its appropriate quality for clinical use. In addition, an efficient and robust procedure for the automated production and quality control of 161Tb-DOTATOC with clinically applicable specifications and activity levels, that is, 1.0-7.4 GBq in 20 mL, was developed. The radiopharmaceutical's quality control was also developed using chromatographic methods, which confirmed the product's stability (RCP ≥ 95%) over 24 h. Conclusion: The current study demonstrated that 161Tb has appropriate features for clinical use. The developed synthesis protocol guarantees high yields and safe preparation of injectable 161Tb-DOTATOC. The investigated approach could be translated to other DOTA-derivatized peptides; thus, 161Tb could be successfully applied in clinical practice for radionuclide therapy.
Collapse
Affiliation(s)
- Chiara Favaretto
- Center for Radiopharmaceutical Sciences, ETH-Paul Scherrer Institute, Villigen-PSI, Switzerland
- Department of Chemistry and Applied Biosciences, ETH, Zurich, Switzerland
| | - Pascal V Grundler
- Center for Radiopharmaceutical Sciences, ETH-Paul Scherrer Institute, Villigen-PSI, Switzerland
| | - Zeynep Talip
- Center for Radiopharmaceutical Sciences, ETH-Paul Scherrer Institute, Villigen-PSI, Switzerland
| | - Stefan Landolt
- Center for Radiopharmaceutical Sciences, ETH-Paul Scherrer Institute, Villigen-PSI, Switzerland
| | - Lebogang Sepini
- Radiochemistry, South African Nuclear Energy Corp., Brits, South Africa
| | - Ulli Köster
- Institut Laue-Langevin, Grenoble, France; and
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences, ETH-Paul Scherrer Institute, Villigen-PSI, Switzerland
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences, ETH-Paul Scherrer Institute, Villigen-PSI, Switzerland
- Department of Chemistry and Applied Biosciences, ETH, Zurich, Switzerland
| | - Susanne Geistlich
- Center for Radiopharmaceutical Sciences, ETH-Paul Scherrer Institute, Villigen-PSI, Switzerland
| | - Nicholas P van der Meulen
- Center for Radiopharmaceutical Sciences, ETH-Paul Scherrer Institute, Villigen-PSI, Switzerland;
- Laboratory of Radiochemistry, Paul Scherrer Institute, Villigen-PSI, Switzerland
| |
Collapse
|
36
|
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
|
37
|
Wharton L, McNeil SW, Merkens H, Yuan Z, Van de Voorde M, Engudar G, Ingham A, Koniar H, Rodríguez-Rodríguez C, Radchenko V, Ooms M, Kunz P, Bénard F, Schaffer P, Yang H. Preclinical Evaluation of [155/161Tb]Tb-Crown-TATE—A Novel SPECT Imaging Theranostic Agent Targeting Neuroendocrine Tumours. Molecules 2023; 28:molecules28073155. [PMID: 37049918 PMCID: PMC10095901 DOI: 10.3390/molecules28073155] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 04/05/2023] Open
Abstract
Terbium radioisotopes (149Tb, 152Tb, 155Tb, 161Tb) offer a unique class of radionuclides which encompass all four medicinally relevant nuclear decay modalities (α, β+, γ, β−/e−), and show high potential for the development of element-matched theranostic radiopharmaceuticals. The goal of this study was to design, synthesise, and evaluate the suitability of crown-TATE as a new peptide-conjugate for radiolabelling of [155Tb]Tb3+ and [161Tb]Tb3+, and to assess the imaging and pharmacokinetic properties of each radiotracer in tumour-bearing mice. [155Tb]Tb-crown-TATE and [161Tb]Tb-crown-TATE were prepared efficiently under mild conditions, and exhibited excellent stability in human serum (>99.5% RCP over 7 days). Longitudinal SPECT/CT images were acquired for 155Tb- and 161Tb- labelled crown-TATE in male NRG mice bearing AR42J tumours. The radiotracers, [155Tb]Tb-crown-TATE and [161Tb]Tb-crown-TATE, showed high tumour targeting (32.6 and 30.0 %ID/g, respectively) and minimal retention in non-target organs at 2.5 h post-administration. Biodistribution studies confirmed the SPECT/CT results, showing high tumour uptake (38.7 ± 8.0 %ID/g and 38.5 ± 3.5 %ID/g, respectively) and favourable tumour-to-background ratios. Blocking studies further confirmed SSTR2-specific tumour accumulation. Overall, these findings suggest that crown-TATE has great potential for element-matched molecular imaging and radionuclide therapy using 155Tb and 161Tb.
Collapse
Affiliation(s)
- Luke Wharton
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
| | - Scott W. McNeil
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
| | - Helen Merkens
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
| | - Zheliang Yuan
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
| | - Michiel Van de Voorde
- Nuclear Medicine Applications, Belgium Nuclear Research Center (SCK CEN), Boeretang, 200, 2400 Mol, Belgium
| | - Gokce Engudar
- 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
| | - Helena Koniar
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
- Department of Physics and Astronomy, University of British Columbia, 6224 Agronomy Road, Vancouver, BC V6T 1Z1, Canada
| | - Cristina Rodríguez-Rodríguez
- Department of Physics and Astronomy, University of British Columbia, 6224 Agronomy Road, Vancouver, BC V6T 1Z1, Canada
- Faculty of Pharmaceutical Sciences, University of British Columbia, 2405 Wesbrook Mall, Vancouver, BC V6T 1Z3, 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
| | - Maarten Ooms
- Nuclear Medicine Applications, Belgium Nuclear Research Center (SCK CEN), Boeretang, 200, 2400 Mol, Belgium
| | - Peter Kunz
- Accelerator Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
- Department of Chemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - François Bénard
- Department of Molecular Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 1L3, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Paul Schaffer
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
- Department of Chemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
- Department of Radiology, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Hua Yang
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3, Canada
- Department of Chemistry, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| |
Collapse
|
38
|
Kiss OC, Scott PJH, Behe M, Penuelas I, Passchier J, Rey A, Patt M, Aime S, Jalilian A, Laverman P, Cheng Z, Chauvet AF, Engle J, Cleeren F, Zhu H, Vercouillie J, van Dam M, Zhang MR, Perk L, Guillet B, Alves F. Highlight selection of radiochemistry and radiopharmacy developments by editorial board. EJNMMI Radiopharm Chem 2023; 8:6. [PMID: 36952073 PMCID: PMC10036721 DOI: 10.1186/s41181-023-00192-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 03/15/2023] [Indexed: 03/24/2023] Open
Abstract
BACKGROUND The Editorial Board of EJNMMI Radiopharmacy and Chemistry releases a biannual highlight commentary to update the readership on trends in the field of radiopharmaceutical development. MAIN BODY This selection of highlights provides commentary on 21 different topics selected by each coauthoring Editorial Board member addressing a variety of aspects ranging from novel radiochemistry to first-in-human application of novel radiopharmaceuticals. CONCLUSION Trends in radiochemistry and radiopharmacy are highlighted. Hot topics cover the entire scope of EJNMMI Radiopharmacy and Chemistry, demonstrating the progress in the research field, and include new PET-labelling methods for 11C and 18F, the importance of choosing the proper chelator for a given radioactive metal ion, implications of total body PET on use of radiopharmaceuticals, legislation issues and radionuclide therapy including the emerging role of 161Tb.
Collapse
Affiliation(s)
- Oliver C Kiss
- Helmholtz Zentrum Dresden Rossendorf, Dresden, Germany.
| | | | - Martin Behe
- Paul Scherrer Institute, Villigen, Switzerland
| | | | | | - Ana Rey
- Universidad de la Rebublica, Montevideo, Uruguay
| | | | | | | | - Peter Laverman
- Radboud University Medical Center, Nijmegen, The Netherlands
| | - Zhen Cheng
- Shanghai Institute of Materia Medica, Shanghai, China
| | | | | | | | - Hua Zhu
- Peking University Cancer Hospital, Beijing, China
| | | | | | | | - Lars Perk
- Radboud University Medical Center, Nijmegen, The Netherlands
| | | | | |
Collapse
|
39
|
Radiometal-theranostics: the first 20 years*. J Radioanal Nucl Chem 2022. [DOI: 10.1007/s10967-022-08624-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
AbstractThis review describes the basic principles of radiometal-theranostics and its dawn based on the development of the positron-emitting 86Y and 86Y-labeled radiopharmaceuticals to quantify biodistribution and dosimetry of 90Y-labeled analogue therapeutics. The nuclear and inorganic development of 86Y (including nuclear and cross section data, irradiation, radiochemical separation and recovery) led to preclinical and clinical evaluation of 86Y-labeled citrate and EDTMP complexes and yielded organ radiation doses in terms of mGy/MBq 90Y. The approach was extended to [86/90Y]Y-DOTA-TOC, yielding again yielded organ radiation doses in terms of mGy/MBq 90Y. The review further discusses the consequences of this early development in terms of further radiometals that were used (68Ga, 177Lu etc.), more chelators that were developed, new biological targets that were addressed (SSTR, PSMA, FAP, etc.) and subsequent generations of radiometal-theranostics that resulted out of that.
Collapse
|
40
|
Sun J, Huangfu Z, Yang J, Wang G, Hu K, Gao M, Zhong Z. Imaging-guided targeted radionuclide tumor therapy: From concept to clinical translation. Adv Drug Deliv Rev 2022; 190:114538. [PMID: 36162696 DOI: 10.1016/j.addr.2022.114538] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 09/03/2022] [Accepted: 09/11/2022] [Indexed: 01/24/2023]
Abstract
Since the first introduction of sodium iodide I-131 for use with thyroid patients almost 80 years ago, more than 50 radiopharmaceuticals have reached the markets for a wide range of diseases, especially cancers. The nuclear medicine paradigm also shifts from solely molecular imaging or radionuclide therapy to imaging-guided radionuclide therapy, which is deemed a vital component of precision cancer therapy and an emerging medical modality for personalized medicine. The imaging-guided radionuclide therapy highlights the systematic integration of targeted nuclear diagnostics and radionuclide therapeutics. Regarding this, nuclear imaging serves to "visualize" the lesions and guide the therapeutic strategy, followed by administration of a precise patient specific dose of radiotherapeutics for treatment according to the absorbed dose to different organs and tumors calculated by dosimetry tools, and finally repeated imaging to predict the prognosis. This strategy leads to significantly enhanced therapeutic efficacy, improved patient outcomes, and manageable adverse events. In this review, we provide an overview of imaging-guided targeted radionuclide therapy for different tumors such as advanced prostate cancer and neuroendocrine tumors, with a focus on development of new radioligands and their preclinical and clinical results, and further discuss about challenges and future perspectives.
Collapse
Affiliation(s)
- Juan Sun
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Zhenyuan Huangfu
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Jiangtao Yang
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China
| | - Guanglin Wang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China.
| | - Kuan Hu
- Department of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical Sciences, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, Chiba 263-8555, Japan.
| | - Mingyuan Gao
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, People's Republic of China
| | - Zhiyuan Zhong
- College of Pharmaceutical Sciences, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, People's Republic of China; Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, People's Republic of China.
| |
Collapse
|
41
|
Toyohara J, Al-Qahtani M, Huang YY, Cazzola E, Todde S, Furumoto S, Mikolajczak R, Decristoforo C, Gillings N, Yang M, Reilly R, Duatti A, Denkova A, Schirrmacher R, Carlucci G, Seimbille Y, Liu Z, Ellis B, Cornelissen BT, Kopka K, Bernardes E. Highlight selection of radiochemistry and radiopharmacy developments by editorial board. EJNMMI Radiopharm Chem 2022; 7:25. [PMID: 36182995 PMCID: PMC9526771 DOI: 10.1186/s41181-022-00177-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/05/2022] [Accepted: 09/07/2022] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND The Editorial Board of EJNMMI Radiopharmacy and Chemistry releases a biannual highlight commentary to update the readership on trends in the field of radiopharmaceutical development. MAIN BODY This commentary of highlights has resulted in 21 different topics selected by each coauthoring Editorial Board member addressing a variety of aspects ranging from novel radiochemistry to first in man application of novel radiopharmaceuticals. CONCLUSION Trends in radiochemistry and radiopharmacy are highlighted demonstrating the progress in the research field in various topics including new PET-labelling methods, FAPI-tracers and imaging, and radionuclide therapy being the scope of EJNMMI Radiopharmacy and Chemistry.
Collapse
Affiliation(s)
- Jun Toyohara
- PET Radiopharmaceutical Sciences, Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Mohammed Al-Qahtani
- Cyclotron and Radiopharmaceuticals Department, King Faisal Specialist Hospital and Research Center (KFSHRC), Riyadh, Saudi Arabia
| | - Ya-Yao Huang
- Molecular Imaging Center, National Taiwan University, Taipei, Taiwan
| | - Emiliano Cazzola
- Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Sacro Cuore-Don Calabria Hospital, Negrar (Vr), Italy
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Ospedale Sacro Cuore-Don Calabria, Negrar (Vr), Italia
| | - Sergio Todde
- University of Milano-Bicocca, Tecnomed Foundation, Monza, Italy
| | - Shozo Furumoto
- Cyclotron and Radioisotope Center (CYRIC), Tohoku University, Sendai, Japan
| | - Renata Mikolajczak
- Radioisotope Centre POLATOM, National Centre for Nuclear Research (NCBJ), Otwock, Poland
- Ośrodek Radioizotopów POLATOM, NARODOWE CENTRUM BADAŃ JĄDROWYCH (NCBJ), Otwock, Poland
| | - Clemens Decristoforo
- Department of Nuclear Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Nic Gillings
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Min Yang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu China
| | - Raymond Reilly
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON Canada
| | - Adriano Duatti
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
- Dipartimento di Scienze Chimiche, Farmaceutiche e Agrarie Università di Ferrara, Ferrara, Italia
| | - Antonia Denkova
- Department of Radiation Science and Technology, Delft University of Technology, Delft, The Netherlands
| | - Ralf Schirrmacher
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton, AB Canada
| | - Giuseppe Carlucci
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles (UCLA), Los Angeles, CA USA
| | - Yann Seimbille
- Department of Radiology and Nuclear Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Zhaofei Liu
- Medical Isotopes Research Center, Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Centre, Beijing, China
| | - Beverley Ellis
- Manchester University NHS Foundation Trust, Manchester, UK
| | - Bart T. Cornelissen
- Department of Nuclear Medicine and Molecular Imaging, Groningen (UMCG), University Medical Center, University of Groningen, Groningen, The Netherlands
| | - Klaus Kopka
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Emerson Bernardes
- Energy and Nuclear Research Institute (IPEN-CNEN/SP), Cidade Universitária, São Paulo, Brazil
- Instituto de Pesquisas Energéticas e Nucleares (IPEN-CNEN/SP), Cidade Universitária, São Paulo, Brasil
| |
Collapse
|
42
|
Spoormans K, Crabbé M, Struelens L, De Saint-Hubert M, Koole M. A Review on Tumor Control Probability (TCP) and Preclinical Dosimetry in Targeted Radionuclide Therapy (TRT). Pharmaceutics 2022; 14:2007. [PMID: 36297446 PMCID: PMC9608466 DOI: 10.3390/pharmaceutics14102007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 12/05/2022] Open
Abstract
Targeted radionuclide therapy (TRT) uses radiopharmaceuticals to specifically irradiate tumor cells while sparing healthy tissue. Response to this treatment highly depends on the absorbed dose. Tumor control probability (TCP) models aim to predict the tumor response based on the absorbed dose by taking into account the different characteristics of TRT. For instance, TRT employs radiation with a high linear energy transfer (LET), which results in an increased effectiveness. Furthermore, a heterogeneous radiopharmaceutical distribution could result in a heterogeneous dose distribution at a tissue, cellular as well as subcellular level, which will generally reduce the tumor response. Finally, the dose rate in TRT is protracted, relatively low, and variable over time. This allows cells to repair more DNA damage, which may reduce the effectiveness of TRT. Within this review, an overview is given on how these characteristics can be included in TCP models, while some experimental findings are also discussed. Many parameters in TCP models are preclinically determined and TCP models also play a role in the preclinical stage of radiopharmaceutical development; however, this all depends critically on the calculated absorbed dose. Accordingly, an overview of the existing preclinical dosimetry methods is given, together with their limitation and applications. It can be concluded that although the theoretical extension of TCP models from external beam radiotherapy towards TRT has been established quite well, the experimental confirmation is lacking. Thus, requiring additional comprehensive studies at the sub-cellular, cellular, and organ level, which should be provided with accurate preclinical dosimetry.
Collapse
Affiliation(s)
- Kaat Spoormans
- Research in Dosimetric Applications, Belgian Nuclear Research Center (SCK CEN), 2400 Mol, Belgium
- Unit of Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, Katholieke Universiteit Leuven (KUL), 3000 Leuven, Belgium
| | - Melissa Crabbé
- NURA Research Group, Belgian Nuclear Research Center (SCK CEN), 2400 Mol, Belgium
| | - Lara Struelens
- Research in Dosimetric Applications, Belgian Nuclear Research Center (SCK CEN), 2400 Mol, Belgium
| | - Marijke De Saint-Hubert
- Research in Dosimetric Applications, Belgian Nuclear Research Center (SCK CEN), 2400 Mol, Belgium
| | - Michel Koole
- Unit of Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, Katholieke Universiteit Leuven (KUL), 3000 Leuven, Belgium
| |
Collapse
|
43
|
Comparison of the Anti-Tumour Activity of the Somatostatin Receptor (SST) Antagonist [177Lu]Lu-Satoreotide Tetraxetan and the Agonist [177Lu]Lu-DOTA-TATE in Mice Bearing AR42J SST2-Positive Tumours. Pharmaceuticals (Basel) 2022; 15:ph15091085. [PMID: 36145306 PMCID: PMC9506113 DOI: 10.3390/ph15091085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 11/16/2022] Open
Abstract
Limited experiments have compared the treatment effects of repetitive cycles of radiolabelled somatostatin (SST) analogues. In vitro and in vivo experiments were conducted in an AR42J cancer cell model, comparing the antagonist [177Lu]Lu-satoreotide tetraxetan with the agonist [177Lu]Lu-DOTA-TATE in terms of their binding properties, biodistribution, anti-tumour activity and toxicity. Histopathological and immunohistochemical examinations were performed at different timepoints. In the in vitro assays, [177Lu]Lu-satoreotide tetraxetan recognised twice as many SST2 binding sites as [177Lu]Lu-DOTA-TATE. In mice treated once a week for four consecutive weeks, [177Lu]Lu-satoreotide tetraxetan (15 MBq) revealed a significantly greater median time taken to reach a tumour volume of 850 mm3 (68 days) compared to [177Lu]Lu-DOTA-TATE at 15 MBq (43 days) or 30 MBq (48 days). This was associated with a higher tumour uptake, enhanced DNA damage and no or mild effects on body weight, haematological toxicity, or renal toxicity with [177Lu]Lu-satoreotide tetraxetan (15 MBq). At the end of the study, complete tumour senescence was noted in 20% of animals treated with [177Lu]Lu-satoreotide tetraxetan, in 13% of those treated with [177Lu]Lu-DOTA-TATE at 30 MBq, and in none of those treated with [177Lu]Lu-DOTA-TATE at 15 MBq. In conclusion, repeated administrations of [177Lu]Lu-satoreotide tetraxetan were able to potentiate peptide receptor radionuclide therapy with a higher tumour uptake, longer median survival, and enhanced DNA damage, with a favourable efficacy/safety profile compared to [177Lu]Lu-DOTA-TATE.
Collapse
|
44
|
Pacak K, Taieb D, Jha A. Functional Imaging of Neuroendocrine Tumors: Stacking the Odds in a Patient's Favor. J Clin Endocrinol Metab 2022; 107:e3953-e3954. [PMID: 35536703 PMCID: PMC9387716 DOI: 10.1210/clinem/dgac298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Indexed: 01/18/2023]
Affiliation(s)
- Karel Pacak
- Correspondence: Karel Pacak, MD, PhD, DSc, Section on Medical Neuroendocrinology & Head, Developmental Endocrinology, Metabolism, Genetics and Endocrine Oncology Affinity Group, Eunice Kennedy Shriver NICHD, NIH, Bldg 10, CRC, Rm 1E-3140, 10 Center Dr, MSC-1109, Bethesda, MD 20892-1109, USA.
| | - David Taieb
- Department of Nuclear Medicine, La Timone University Hospital, European Center for Research in Medical Imaging, Aix-Marseille University, 13385 Marseille, France
| | - Abhishek Jha
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland 20892-1109, USA
| |
Collapse
|
45
|
Fani M, Mansi R, Nicolas GP, Wild D. Radiolabeled Somatostatin Analogs-A Continuously Evolving Class of Radiopharmaceuticals. Cancers (Basel) 2022; 14:cancers14051172. [PMID: 35267479 PMCID: PMC8909681 DOI: 10.3390/cancers14051172] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/21/2022] [Accepted: 02/22/2022] [Indexed: 11/16/2022] Open
Abstract
Somatostatin receptors (SSTs) are recognized as favorable molecular targets in neuroendocrine tumors (NETs) and neuroendocrine neoplasms (NENs), with subtype 2 (SST2) being the predominantly and most frequently expressed. PET/CT imaging with 68Ga-labeled SST agonists, e.g., 68Ga-DOTA-TOC (SomaKit TOC®) or 68Ga-DOTA-TATE (NETSPOT®), plays an important role in staging and restaging these tumors and can identify patients who qualify and would potentially benefit from peptide receptor radionuclide therapy (PRRT) with the therapeutic counterparts 177Lu-DOTA-TOC or 177Lu-DOTA-TATE (Lutathera®). This is an important feature of SST targeting, as it allows a personalized treatment approach (theranostic approach). Today, new developments hold promise for enhancing diagnostic accuracy and therapeutic efficacy. Among them, the use of SST2 antagonists, such as JR11 and LM3, has shown certain advantages in improving image sensitivity and tumor radiation dose, and there is evidence that they may find application in other oncological indications beyond NETs and NENs. In addition, PRRT performed with more cytotoxic α-emitters, such as 225Ac, or β- and Auger electrons, such as 161Tb, presents higher efficacy. It remains to be seen if any of these new developments will overpower the established radiolabeled SST analogs and PRRT with β--emitters.
Collapse
Affiliation(s)
- Melpomeni Fani
- Division of Radiopharmaceutical Chemistry, University Hospital Basel, 4031 Basel, Switzerland;
- Correspondence:
| | - Rosalba Mansi
- Division of Radiopharmaceutical Chemistry, University Hospital Basel, 4031 Basel, Switzerland;
| | - Guillaume P. Nicolas
- Division of Nuclear Medicine, University Hospital Basel, 4031 Basel, Switzerland; (G.P.N.); (D.W.)
- ENETS Center of Excellence for Neuroendocrine and Endocrine Tumors, University Hospital Basel, 4031 Basel, Switzerland
| | - Damian Wild
- Division of Nuclear Medicine, University Hospital Basel, 4031 Basel, Switzerland; (G.P.N.); (D.W.)
- ENETS Center of Excellence for Neuroendocrine and Endocrine Tumors, University Hospital Basel, 4031 Basel, Switzerland
| |
Collapse
|
46
|
Harris PE, Zhernosekov K. The evolution of PRRT for the treatment of neuroendocrine tumors; What comes next? Front Endocrinol (Lausanne) 2022; 13:941832. [PMID: 36387893 PMCID: PMC9659917 DOI: 10.3389/fendo.2022.941832] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 10/12/2022] [Indexed: 12/02/2022] Open
Abstract
Lu-177 has been developed for the treatment of patients with peptide receptor radionuclide therapy (PRRT). A second generation pure no-carrier-added Lu-177 has a high specific activity and has waste disposal advantages over the first generation carrier-added Lu-177. PRRT has recently been developed for the treatment of neuroendocrine tumors (NETs). The majority of pancreatic and gastroenteric NETs (GEP-NETs) express the somatostatin receptors (SSTRs) 2 and 5. These receptors can be specifically targeted with a somatostatin peptide analogue (DOTATOC/DOTATATE) which can be chelated to a positron emission tomography (PET) emitting radioisotope such as Ga-68 for imaging or to a β-emitting radioisotope Lu-177 for therapy. A key advantage of this approach is that the receptor expression can be demonstrated by PET imaging before the patient is treated. Clinical studies in G1 and G2 GEP-NETS have demonstrated that PRRT is extremely effective in terms of progression free survival (PFS), symptom control and quality of life, with a well-established safety profile. A beneficial effect on outcome survival awaits to be confirmed. The first commercially available product Lu-177-DOTATATE was approved following the NETTER-1 trial in G1 and G2 GE-NETS. Lu-177-DOTATATE 7,4 GBq every 8 weeks for 4 cycles, together with octreotide LAR 30 mg monthly, demonstrated a median PFS of 28,4 months compared to 8,5 months for octreotide LAR 60 mg monthly. A second pivotal study COMPETE is currently in progress, comparing no carrier-added (n.c.a.) Lu-177-DOTATOC to the m-TOR inhibitor Everolimus in both GE-NETs and PNETs. Two studies, NETTER-2 and COMPOSE are currently underway in patients with high grade G2 and G3 NETs. Novel SSTR antagonists are being developed as next generation targeting molecules for SSTR2-expressing tumors. Antagonists have a higher tumor binding to receptors than agonists, opening up the potential indications for SSTR2 targeting to tumors which have a relatively lower expression of SSTR2 compared to NET such as small cell lung cancer, hepatocellular carcinoma and breast cancer. In addition to Lu-177, radioisotopes with different radiation properties such as Tb-161 and the α-emitter Ac-225 are being developed which have the potential to improve treatment efficacy across the range of G1 to G3 NETs.
Collapse
|