1
|
Lundgren Mortensen AC, Berglund H, Jha P, Stenman A, Selvaraju RK, Lundqvist H, Hofström C, Persson H, Juhlin CC, Zedenius J, Frejd FY, Nestor M. Dual-Nuclide Biodistribution and Therapeutic Evaluation of a Novel Antibody-Based Radiopharmaceutical in Anaplastic Thyroid Cancer Xenografts. Mol Cancer Ther 2025; 24:753-762. [PMID: 39976160 DOI: 10.1158/1535-7163.mct-24-0524] [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: 06/29/2024] [Revised: 09/27/2024] [Accepted: 02/18/2025] [Indexed: 02/21/2025]
Abstract
Anaplastic thyroid cancer (ATC) is a rare but severe form of thyroid cancer responsible for approximately 50% of thyroid cancer deaths. Consequently, the identification of innovative therapies remains crucial for the effective treatment of ATC. Molecular radiotherapy is a rapidly growing field within oncology, and the cell surface antigen CD44v6, which is overexpressed in several cancers, is a plausible target for molecular radiotherapy of ATC. IHC of 39 patient samples with ATC was evaluated for CD44v6 expression. Biodistribution and dosimetry of iodine-125 (125I)-/lutetium-177 (177Lu)-labeled UU-40, a CD44v6-specific antibody, followed by in vivo efficacy in two ATC xenograft models with varying target expression levels (ACT-1 and BHT-101), accompanied by single-photon emission computed tomography (SPECT) imaging, evaluated radiolabeled UU-40 for therapeutic efficiency in ATC xenografts. The IHC revealed CD44v6 immunoreactivity in 46% of patient samples with ATC. The biodistribution favored 177Lu-labeled UU-40 over the 125I-labeled antibody and confirmed the in vivo specificity of both radioconjugates. The in vivo efficacy and accompanied SPECT imaging of a moderate CD44v6-expressing xenograft model (BHT-101) verified the tumor specificity, as well as the target-specific effect of 177Lu-labeled UU-40 on tumor growth and survival. A 100% complete response rate was demonstrated as a result of therapy using a single dose of 16 MBq 177Lu-labeled UU-40 in a high CD44v6-expressing xenograft model (ACT-1), and SPECT imaging revealed excellent tumor uptake of the radioconjugate at 14 days after injection. This study verifies the expression of CD44v6 in ATC and strengthens the superiority and promise of 177Lu-labeled UU-40 over 131I-labeled UU-40 for antibody-based molecular radiotherapy of CD44v6-positive ATC.
Collapse
Affiliation(s)
- Anja Charlotte Lundgren Mortensen
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory (SciLifeLab), Uppsala University, Uppsala, Sweden
| | - Hanna Berglund
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory (SciLifeLab), Uppsala University, Uppsala, Sweden
| | - Preeti Jha
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory (SciLifeLab), Uppsala University, Uppsala, Sweden
- Department of Radiology, University of Texas Southwestern Medical Centre, Dallas, Texas
| | - Adam Stenman
- Department of Oncology-Pathology, Karolinska Institutet, Solna, Sweden
- Department of Pathology and Cancer Diagnostics, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Ram Kumar Selvaraju
- Department of Medicinal Chemistry, Preclinical PET-MRI Platform, Uppsala University, Uppsala, Sweden
| | - Hans Lundqvist
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory (SciLifeLab), Uppsala University, Uppsala, Sweden
| | - Camilla Hofström
- Drug Discovery and Development Platform, Science for Life Laboratory (SciLifeLab), Stockholm, Sweden
| | - Helena Persson
- Drug Discovery and Development Platform, Science for Life Laboratory (SciLifeLab), Stockholm, Sweden
| | - C Christofer Juhlin
- Department of Oncology-Pathology, Karolinska Institutet, Solna, Sweden
- Department of Pathology and Cancer Diagnostics, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Jan Zedenius
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Department of Breast, Endocrine Tumors and Sarcoma, Karolinska University Hospital, Stockholm, Sweden
| | - Fredrik Y Frejd
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory (SciLifeLab), Uppsala University, Uppsala, Sweden
| | - Marika Nestor
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory (SciLifeLab), Uppsala University, Uppsala, Sweden
| |
Collapse
|
2
|
Tosato M, Favaretto C, Kleynhans J, Burgoyne AR, Gestin JF, van der Meulen NP, Jalilian A, Köster U, Asti M, Radchenko V. Alpha Atlas: Mapping global production of α-emitting radionuclides for targeted alpha therapy. Nucl Med Biol 2025; 142-143:108990. [PMID: 39809026 DOI: 10.1016/j.nucmedbio.2024.108990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 12/06/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025]
Abstract
Targeted Alpha Therapy has shown great promise in cancer treatment, sparking significant interest over recent decades. However, its broad adoption has been impeded by the scarcity of alpha-emitters and the complexities related to their use. The availability of these radionuclides is often constrained by the intricate production processes and purification, as well as regulatory and logistical challenges. Moreover, the high cost and technical difficulties associated with handling and applying alpha-emitting radionuclides pose additional barriers to their clinical implementation. This Alpha Atlas provides an in-depth overview of the leading alpha-particle emitting radionuclide candidates for clinical use, focusing on their production processes and supply chains. By mapping the current facilities that produce and supply these radionuclides, this atlas aims to assist researchers, clinicians, and industries in initiating or scaling up the applications of alpha-emitters. The Alpha Atlas aspires to act as a strategic guide, facilitating collaboration and driving forward the integration of these potent therapeutic agents into cancer treatment practices.
Collapse
Affiliation(s)
- Marianna Tosato
- Radiopharmaceutical Chemistry Laboratory (RACHEL), Nuclear Medicine Unit, AUSL-IRCCS Reggio Emilia, 42123 Reggio Emilia, Italy.
| | - Chiara Favaretto
- Radiopharmacy and Cyclotron Department, IRCCS Sacro Cuore Don Calabria, Negrar 37024, Verona, Italy
| | - Janke Kleynhans
- Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Andrew R Burgoyne
- Oak Ridge National Laboratory, 1 Bethel Valley Road, Oak Ridge, TN 37830, United States
| | - Jean-François Gestin
- Nantes Université, Inserm, CNRS, Université d'Angers, CRCI2NA, 44000 Nantes, France
| | - Nicholas P van der Meulen
- PSI Center for Life Sciences, 5232 Villigen-PSI, Switzerland; PSI Center for Nuclear Engineering and Sciences, 5232 Villigen-PSI, Switzerland
| | - Amirreza Jalilian
- Department of Nuclear Safety and Security, International Atomic Energy Agency, 1220 Vienna, Austria
| | - Ulli Köster
- Institut Laue-Langevin, 38042 Grenoble, France
| | - Mattia Asti
- Radiopharmaceutical Chemistry Laboratory (RACHEL), Nuclear Medicine Unit, AUSL-IRCCS Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Valery Radchenko
- Life Sciences Division, TRIUMF, BC V6T 2A3 Vancouver, British Columbia, Canada; Department of Chemistry, University of British Columbia, V6T 1Z1 Vancouver, British Columbia, Canada
| |
Collapse
|
3
|
Shchukina AA, Zubenko AD, Tarasenko OV, Larenkov AA, Bubenshchikov VB, Chernikova EY, Fedorov YV, Fedorova OA. Evaluation of chelating agents based on pyridine-azacrown compounds H 4PATA, PATAM, and H 4PATPA for 68Ga and 177Lu. Nucl Med Biol 2025; 140-141:108972. [PMID: 39550886 DOI: 10.1016/j.nucmedbio.2024.108972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 11/19/2024]
Abstract
In this article, we present the synthesis and characterization of three macrocyclic chelators, H4PATA, PATAM, and H4PATPA, based on a pyridine-azacrown compound. Their complexation with 68Ga and 177Lu has been thoroughly investigated using MALDI TOF MS, 1H NMR spectroscopy, radiolabeling studies, and experiments in vitro with fetal bovine serum and a 1000-fold molar excess of H4EDTA. Our studies have shown that the chelators H4PATA and H4PATPA form complexes at room temperature with both radionuclides (RCY > 80 % and > 90 % for complexes with H4PATA and H4PATPA after 30 min, respectively). The chelator PATAM requires high temperature (95 °C) for complexation. In vitro stability assays in fetal bovine serum as well as H4EDTA-challenge revealed that transchelation occurs for all complexes with 68Ga. However, complexes of the ligands H4PATA and PATAM with 177Lu were found stable. Thus, taking into account the radiolabeling at room temperature and in vitro stability of the complex [177Lu]Lu·PATA, our investigations revealed the chelator H4PATA is a candidate for radiopharmaceutical use with 177Lu.
Collapse
Affiliation(s)
- Anna A Shchukina
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, Vavilova st., 28, 119334 Moscow, Russian Federation.
| | - Anastasia D Zubenko
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, Vavilova st., 28, 119334 Moscow, Russian Federation
| | - Oksana V Tarasenko
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, Vavilova st., 28, 119334 Moscow, Russian Federation; D. I. Mendeleev University of Chemical Technology of Russia, Miusskaya Sq. 9, 125047 Moscow, Russian Federation
| | - Anton A Larenkov
- State Research Center-Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Zhivopisnaya Str., 46, 123098 Moscow, Russian Federation
| | - Viktor B Bubenshchikov
- State Research Center-Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Zhivopisnaya Str., 46, 123098 Moscow, Russian Federation
| | - Ekaterina Y Chernikova
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, Vavilova st., 28, 119334 Moscow, Russian Federation
| | - Yury V Fedorov
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, Vavilova st., 28, 119334 Moscow, Russian Federation
| | - Olga A Fedorova
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, Vavilova st., 28, 119334 Moscow, Russian Federation; D. I. Mendeleev University of Chemical Technology of Russia, Miusskaya Sq. 9, 125047 Moscow, Russian Federation
| |
Collapse
|
4
|
Moreno-Alcántar G, Drexler M, Casini A. Assembling a new generation of radiopharmaceuticals with supramolecular theranostics. Nat Rev Chem 2024; 8:893-914. [PMID: 39468298 DOI: 10.1038/s41570-024-00657-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2024] [Indexed: 10/30/2024]
Abstract
Supramolecular chemistry has been used to tackle some of the major challenges in modern science, including cancer therapy and diagnosis. Supramolecular platforms provide synthetic flexibility, rapid generation through self-assembly, facile labelling, unique topologies, tunable reversibility of the enabling noncovalent interactions, and opportunities for host-guest chemistry and mechanical bonding. In this Review, we summarize recent advances in the design and radiopharmaceutical application of discrete self-assembled coordination complexes and mechanically interlocked molecules - namely, metallacages and rotaxanes, respectively - as well as in situ-forming supramolecular aggregates, specifically pinpointing their potential as next-generation radiotheranostic agents. The outlook of such supramolecular constructs for potential applications in the clinic is discussed.
Collapse
Affiliation(s)
- Guillermo Moreno-Alcántar
- Department of Chemistry, School of Natural Sciences, Technical University of Munich, Garching bei München, Germany
| | - Marike Drexler
- Department of Chemistry, School of Natural Sciences, Technical University of Munich, Garching bei München, Germany
| | - Angela Casini
- Department of Chemistry, School of Natural Sciences, Technical University of Munich, Garching bei München, Germany.
- Munich Data Science Institute (MDSI), Technical University of Munich, Garching bei München, Germany.
| |
Collapse
|
5
|
Larenkov A, Mitrofanov I, Rakhimov M. Improvement of End-of-Synthesis Radiochemical Purity of 177Lu-DOTA-PSMA-Ligands with Alternative Synthesis Approaches: Conversion Upswing and Side-Products Minimization. Pharmaceutics 2024; 16:1535. [PMID: 39771514 PMCID: PMC11677419 DOI: 10.3390/pharmaceutics16121535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 11/22/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Radiochemical purity is a key criterion for the quality of radiopharmaceuticals used in clinical practice. The joint improvement of analytical methods capable of identifying related radiochemical impurities and determining the actual radiochemical purity, as well as the improvement of synthesis methods to minimize the formation of possible radiochemical impurities, is integral to the implementation of high-tech nuclear medicine procedures. PSMA-targeted radionuclide therapy with lutetium-177 has emerged as an effective treatment option for prostate cancer, and [177Lu]Lu-PSMA-617 and [177Lu]Lu-PSMAI&T have achieved global recognition as viable radiopharmaceuticals. Recently, it was shown that specific radiochemical impurities can form during the synthesis of [177Lu]Lu-PSMA-617 because of a spontaneous, thermally mediated condensation of the Glu-C(O)-Lys fragment, resulting in the formation of three different cyclic forms (with no affinity for PSMA). During this study, we identified another impurity, a product of detachment of the Glu-CO fragment from PSMA-617, caused by heating. The total content of all four thermally mediated degradation products may reach 9-11% during classical incubation for 30 min at 95 °C, reducing the radiochemical purity to an unacceptable level (albeit with high levels of radiochemical conversion). It is reasonable to assume that the formation of similar impurities is characteristic of all PSMA-specific vectors that contain Glu-C(O)-Lys pharmacophores. Because the formation of these impurities directly depends on the temperature and incubation time, to reduce their content in the reaction mixture at the end of the synthesis, it is necessary to select conditions to achieve a high level of radiochemical conversion for the minimum possible time and/or at the minimum sufficient temperature. METHODS In this study, using [177Lu]Lu-PSMA-617 as an example, we evaluated the efficiency of alternative methods of synthesis with microwave heating and co-solvent (ethanol) addition to ensure radiochemical yield and radiochemical purity in the shortest possible time and at the minimum necessary and sufficient synthesis temperature. RESULTS Both approaches achieved a significant reduction in the impurities content, while achieving satisfactory synthesis yields in a short time. In addition to improving the synthesis parameters and radiochemical purity, the use of microwave heating and the addition of ethanol reduces the negative influence of other auxiliaries on labeling kinetics. Notably, the addition of ethanol under certain conditions allowed [177Lu]Lu-PSMA-617 to be synthesized at room temperature for only 10 min. This makes it possible to achieve exceptionally high real radiochemical purity of the preparations, determined only by the quality of the original precursor. The approaches considered in this study can be successfully applied to improve the synthesis process and quality parameters of the finished product, both for known radiopharmaceuticals and for those under development.
Collapse
|
6
|
Kanagasundaram T, Sun Y, Lee KK, MacMillan SN, Brugarolas P, Wilson JJ. Fluorine-18 incorporation and radiometal coordination in macropa ligands for PET imaging and targeted alpha therapy. Chem Commun (Camb) 2024; 60:11940-11943. [PMID: 39352495 DOI: 10.1039/d4cc04165h] [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] [Indexed: 10/06/2024]
Abstract
The development of theranostic agents for radiopharmaceuticals based on therapeutic alpha emitters marks an important clinical need. We describe a strategy for the development of theranostic agents of this type via the functionalization of the ligand with the diagnostic radionuclide fluorine-18. An analogue of macropa, an 18-membered macrocyclic chelator with high affinity for alpha therapeutic radiometals, was synthesized and its complexation properties with metal ions were determined. The new macropa-F ligand was used for quantitative radiometal complexation with lead-203 and bismuth-207, as surrogates for their alpha-emitting radioisotopes. As a diagnostic partner, a radiofluorinated macropa ligand was used for quantitative bismuth(III) and lead(II) complexation. All fluorine-18 and radiometal complexes are highly stable in human serum over several days. This study presents a new proof-of-principle approach for developing theranostic agents based on alpha-emitting radionuclides and fluorine-18.
Collapse
Affiliation(s)
- Thines Kanagasundaram
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY-14853, USA
| | - Yang Sun
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA-02114, USA.
| | - Kevin K Lee
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY-14853, USA
- Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, CA-93106, USA.
| | - Samantha N MacMillan
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY-14853, USA
| | - Pedro Brugarolas
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA-02114, USA.
| | - Justin J Wilson
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY-14853, USA
- Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, CA-93106, USA.
| |
Collapse
|
7
|
Wang J, Wang L, Zhang Y, Pan S, Lin Y, Wu J, Bu M. Design, Synthesis, and Anticancer Activity of Novel Enmein-Type Diterpenoid Derivatives Targeting the PI3K/Akt/mTOR Signaling Pathway. Molecules 2024; 29:4066. [PMID: 39274913 PMCID: PMC11396751 DOI: 10.3390/molecules29174066] [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: 07/27/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/16/2024] Open
Abstract
The enmein-type diterpenoids are a class of anticancer ent-Kaurane diterpnoids that have received much attention in recent years. Herein, a novel 1,14-epoxy enmein-type diterpenoid 4, was reported in this project for the first time. A series of novel enmein-type diterpenoid derivatives were also synthesized and tested for anticancer activities. Among all the derivatives, compound 7h exhibited the most significant inhibitory effect against A549 cells (IC50 = 2.16 µM), being 11.03-folds better than its parental compound 4. Additionally, 7h exhibited relatively weak anti-proliferative activity (IC50 > 100 µM) against human normal L-02 cells, suggesting that it had excellent anti-proliferative selectivity for cancer cells. Mechanism studies suggested that 7h induced G0/G1 arrest and apoptosis in A549 cells by inhibiting the PI3K/AKT/mTOR pathway. This process was associated with elevated intracellular ROS levels and collapsed MMP. In summary, these data identified 7h as a promising lead compound that warrants further investigation of its anticancer properties.
Collapse
Affiliation(s)
- Jiafeng Wang
- College of Pathology, Qiqihar Medical University, Qiqihar 161006, China; (J.W.); (Y.Z.); (S.P.)
| | - Lu Wang
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, China; (L.W.); (Y.L.)
| | - Yingbo Zhang
- College of Pathology, Qiqihar Medical University, Qiqihar 161006, China; (J.W.); (Y.Z.); (S.P.)
| | - Siwen Pan
- College of Pathology, Qiqihar Medical University, Qiqihar 161006, China; (J.W.); (Y.Z.); (S.P.)
| | - Yu Lin
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, China; (L.W.); (Y.L.)
| | - Jiale Wu
- College of Life and Health, Hainan University, Haikou 570228, China;
| | - Ming Bu
- College of Pharmacy, Qiqihar Medical University, Qiqihar 161006, China; (L.W.); (Y.L.)
| |
Collapse
|
8
|
Deiser S, Fenzl S, König V, Drexler M, Smith LM, George ME, Beck R, Witney TH, Inoue S, Casini A. (SiFA)SeFe: A Hydrophilic Silicon-Based Fluoride Acceptor Enabling Versatile Peptidic Radiohybrid Tracers. J Med Chem 2024; 67:14077-14094. [PMID: 39115131 PMCID: PMC11345769 DOI: 10.1021/acs.jmedchem.4c00924] [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: 04/18/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 08/23/2024]
Abstract
The radiohybrid (rh) concept to design targeted (and chemically identical) radiotracers for imaging or radionuclide therapy of tumors has gained momentum. For this strategy, a new bifunctional Silicon-based Fluoride Acceptor (SiFA) moiety (SiFA)SeFe was synthesized, endowed with improved hydrophilicity and high versatility of integration into rh-compounds. Preliminary radiolabeling and stability studies under different conditions were conducted using model bioconjugate peptides. Further, three somatostatin receptor 2 (sstR2)-targeted rh-compounds ((SiFA)SeFe-rhTATE1-3, TATE = (Tyr3)-octreotate) were developed. Compound (SiFA)SeFe-rhTATE3, enables labeling with 18F for PET imaging or chelation of 177Lu for therapy. The rh-compounds possess comparable receptor binding affinity and in vitro performance as good as the clinically proven gold standards. SstR2-specificity was further shown for (SiFA)SeFe-rhTATE2 using the chicken chorioallantoic membrane (CAM) model. The biodistribution of two compounds in mice showed high accumulation in tumors and excretion via the kidneys, demonstrating the clinical applicability of the (SiFA)SeFe moiety.
Collapse
Affiliation(s)
- Sandra Deiser
- Chair
of Medicinal and Bioinorganic Chemistry, Department of Chemistry,
School of Natural Sciences, Technical University
of Munich, Lichtenbergstr. 4, 85748 Garching b. München, Germany
- Chair
of Pharmaceutical Radiochemistry, Department of Chemistry, School
of Natural Sciences, Technical University
of Munich, Walther-Meißner-Str.
3, 85748 Garching
b. München, Germany
| | - Sebastian Fenzl
- Chair
of Pharmaceutical Radiochemistry, Department of Chemistry, School
of Natural Sciences, Technical University
of Munich, Walther-Meißner-Str.
3, 85748 Garching
b. München, Germany
- Institute
of Silicon Chemistry, Department of Chemistry, School of Natural Sciences, Technical University of Munich, Lichtenbergstr. 4, 85748 Garching b. München, Germany
| | - Victor König
- Chair
of Pharmaceutical Radiochemistry, Department of Chemistry, School
of Natural Sciences, Technical University
of Munich, Walther-Meißner-Str.
3, 85748 Garching
b. München, Germany
| | - Marike Drexler
- Chair
of Medicinal and Bioinorganic Chemistry, Department of Chemistry,
School of Natural Sciences, Technical University
of Munich, Lichtenbergstr. 4, 85748 Garching b. München, Germany
- Chair
of Pharmaceutical Radiochemistry, Department of Chemistry, School
of Natural Sciences, Technical University
of Munich, Walther-Meißner-Str.
3, 85748 Garching
b. München, Germany
| | - Lydia M. Smith
- School
of Biomedical Engineering and Imaging Sciences King’s College London St Thomas’ Hospital, London SE1 7EH, U.K.
| | - Madeleine E. George
- School
of Biomedical Engineering and Imaging Sciences King’s College London St Thomas’ Hospital, London SE1 7EH, U.K.
| | - Roswitha Beck
- Chair
of Pharmaceutical Radiochemistry, Department of Chemistry, School
of Natural Sciences, Technical University
of Munich, Walther-Meißner-Str.
3, 85748 Garching
b. München, Germany
| | - Timothy H. Witney
- School
of Biomedical Engineering and Imaging Sciences King’s College London St Thomas’ Hospital, London SE1 7EH, U.K.
| | - Shigeyoshi Inoue
- Institute
of Silicon Chemistry, Department of Chemistry, School of Natural Sciences, Technical University of Munich, Lichtenbergstr. 4, 85748 Garching b. München, Germany
| | - Angela Casini
- Chair
of Medicinal and Bioinorganic Chemistry, Department of Chemistry,
School of Natural Sciences, Technical University
of Munich, Lichtenbergstr. 4, 85748 Garching b. München, Germany
- Chair
of Pharmaceutical Radiochemistry, Department of Chemistry, School
of Natural Sciences, Technical University
of Munich, Walther-Meißner-Str.
3, 85748 Garching
b. München, Germany
| |
Collapse
|
9
|
Garaulet G, Báez BB, Medrano G, Rivas-Sánchez M, Sánchez-Alonso D, Martinez-Torrecuadrada JL, Mulero F. Radioimmunotheragnosis in Cancer Research. Cancers (Basel) 2024; 16:2896. [PMID: 39199666 PMCID: PMC11352548 DOI: 10.3390/cancers16162896] [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: 06/13/2024] [Revised: 08/02/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
The combination of immunoPET-where an antibody (Ab) is labeled with an isotope for PET imaging-and radioimmunotherapy (RIT), using the same antibody with a therapeutic isotope, offers significant advantages in cancer management. ImmunoPET allows non-invasive imaging of antigen expression, which aids in patient selection for subsequent radioimmunotherapy. It also facilitates the assessment of tumor response to therapy, allowing for treatment adjustments if necessary. In addition, immunoPET provides critical pharmacokinetic data, including antibody biodistribution and clearance rates, which are essential for dosimetry calculations and treatment protocol optimization. There are still challenges to overcome. Identifying appropriate target antigens that are selectively expressed on cancer cells while minimally expressed on normal tissues remains a major hurdle to reduce off-target toxicity. In addition, it is critical to optimize the pharmacokinetics of radiolabeled antibodies to maximize tumor uptake and minimize normal tissue uptake, particularly in vital organs such as the liver and kidney. This approach offers the potential for targeted and personalized cancer therapy with reduced systemic toxicity by exploiting the specificity of monoclonal antibodies and the cytotoxic effects of radiation. However, further research is needed to address remaining challenges and to optimize these technologies for clinical use.
Collapse
Affiliation(s)
- Guillermo Garaulet
- Molecular Imaging Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (G.G.); (B.B.B.); (G.M.)
| | - Bárbara Beatriz Báez
- Molecular Imaging Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (G.G.); (B.B.B.); (G.M.)
| | - Guillermo Medrano
- Molecular Imaging Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (G.G.); (B.B.B.); (G.M.)
| | - María Rivas-Sánchez
- Protein Production Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (M.R.-S.); (D.S.-A.)
| | - David Sánchez-Alonso
- Protein Production Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (M.R.-S.); (D.S.-A.)
| | | | - Francisca Mulero
- Molecular Imaging Unit, Spanish National Cancer Center—CNIO, 28029 Madrid, Spain; (G.G.); (B.B.B.); (G.M.)
| |
Collapse
|
10
|
Tosato M, Randhawa P, Asti M, Hemmingsen LBS, O'Shea CA, Thaveenrasingam P, Sauer SPA, Chen S, Graiff C, Menegazzo I, Baron M, Radchenko V, Ramogida CF, Di Marco V. Capturing Mercury-197m/g for Auger Electron Therapy and Cancer Theranostic with Sulfur-Containing Cyclen-Based Macrocycles. Inorg Chem 2024; 63:14241-14255. [PMID: 39024562 DOI: 10.1021/acs.inorgchem.4c02418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
The interest in mercury radioisotopes, 197mHg (t1/2 = 23.8 h) and 197gHg (t1/2 = 64.14 h), has recently been reignited by the dual diagnostic and therapeutic nature of their nuclear decays. These isotopes emit γ-rays suitable for single photon emission computed tomography imaging and Auger electrons which can be exploited for treating small and metastatic tumors. However, the clinical utilization of 197m/gHg radionuclides is obstructed by the lack of chelators capable of securely binding them to tumor-seeking vectors. This work aims to address this challenge by investigating a series of chemically tailored macrocyclic platforms with sulfur-containing side arms, namely, 1,4,7,10-tetrakis[2-(methylsulfanyl)ethyl]-1,4,7,10-tetraazacyclododecane (DO4S), 1,4,7-tris[2-(methylsulfanyl)ethyl]-1,4,7,10-tetraazacyclododecane (DO3S), and 1,7-bis[2-(methylsulfanyl)ethyl]-1,4,7,10-tetraazacyclododecane-4,10-diacetic acid (DO2A2S). 1,4,7,10-Tetrazacyclododecane-1,4,7,10-tetracetic acid (DOTA), the widest explored chelator in nuclear medicine, and the nonfunctionalized backbone 1,4,7,10-tetrazacyclododecane (cyclen) were considered as well to shed light on the role of the sulfanyl arms in the metal coordination. To this purpose, a comprehensive experimental and theoretical study encompassing aqueous coordination chemistry investigations through potentiometry, nuclear magnetic resonance (NMR) spectroscopy, X-ray crystallography, and density functional theory (DFT) calculations, as well as concentration- and temperature-dependent [197m/gHg]Hg2+ radiolabeling and in vitro stability assays in human serum was conducted. The obtained results reveal that the investigated chelators rapidly complex Hg2+ in aqueous media, forming extremely thermodynamically stable 1:1 metal-to-ligand complexes with superior stabilities compared to those of DOTA or cyclen. These complexes exhibited 6- to 8-fold coordination environments, with donors statically bound to the metal center, as evidenced by the presence of 1H-199Hg spin-spin coupling via NMR. A similar octacoordinated environment was also found for DOTA in both solution and solid state, but in this case, multiple slowly exchanging conformers were detected at ambient temperature. The sulfur-rich ligands quantitatively incorporate cyclotron-produced [197m/gHg]Hg2+ under relatively mild reaction conditions (pH = 7 and T = 50 °C), with the resulting radioactive complexes exhibiting decent stability in human serum (up to 75% after 24 h). By developing viable chelators and understanding the impact of structural modifications, our research addresses the scarcity of suitable chelating agents for 197m/gHg, offering promise for its future in vivo application as a theranostic Auger-emitter radiometal.
Collapse
Affiliation(s)
- Marianna Tosato
- Department of Chemical Sciences, University of Padova, 35131 Padova, Italy
- Department of Chemistry, Simon Fraser University, BC V5A 0A7 Burnaby, British Columbia, Canada
- Life Sciences Division, TRIUMF, BC V6T 2A3 Vancouver, British Columbia, Canada
- Radiopharmaceutical Chemistry Section, Nuclear Medicine Unit, AUSL-IRCCS Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Parmissa Randhawa
- Department of Chemistry, Simon Fraser University, BC V5A 0A7 Burnaby, British Columbia, Canada
- Life Sciences Division, TRIUMF, BC V6T 2A3 Vancouver, British Columbia, Canada
| | - Mattia Asti
- Radiopharmaceutical Chemistry Section, Nuclear Medicine Unit, AUSL-IRCCS Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Lars B S Hemmingsen
- Department of Chemistry, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Catriona Ann O'Shea
- Department of Chemistry, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | | | - Stephan P A Sauer
- Department of Chemistry, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Shaohuang Chen
- Department of Chemistry, Simon Fraser University, BC V5A 0A7 Burnaby, British Columbia, Canada
- Life Sciences Division, TRIUMF, BC V6T 2A3 Vancouver, British Columbia, Canada
| | - Claudia Graiff
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, 43124 Parma, Italy
| | - Ileana Menegazzo
- Department of Chemical Sciences, University of Padova, 35131 Padova, Italy
| | - Marco Baron
- Department of Chemical Sciences, University of Padova, 35131 Padova, Italy
| | - Valery Radchenko
- Life Sciences Division, TRIUMF, BC V6T 2A3 Vancouver, British Columbia, Canada
- Department of Chemistry, University of British Columbia, BC V6T 1Z1 Vancouver, British Columbia, Canada
| | - Caterina F Ramogida
- Department of Chemistry, Simon Fraser University, BC V5A 0A7 Burnaby, British Columbia, Canada
- Life Sciences Division, TRIUMF, BC V6T 2A3 Vancouver, British Columbia, Canada
| | - Valerio Di Marco
- Department of Chemical Sciences, University of Padova, 35131 Padova, Italy
| |
Collapse
|
11
|
Simms ME, Li Z, Sibley MM, Ivanov AS, Lara CM, Johnstone TC, Kertesz V, Fears A, White FD, Thorek DLJ, Thiele NA. PYTA: a universal chelator for advancing the theranostic palette of nuclear medicine. Chem Sci 2024; 15:11279-11286. [PMID: 39055008 PMCID: PMC11268510 DOI: 10.1039/d3sc06854d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 06/05/2024] [Indexed: 07/27/2024] Open
Abstract
To clinically advance the growing arsenal of radiometals available to image and treat cancer, chelators with versatile binding properties are needed. Herein, we evaluated the ability of the py2[18]dieneN6 macrocycle PYTA to interchangeably bind and stabilize 225Ac3+, [177Lu]Lu3+, [111In]In3+ and [44Sc]Sc3+, a chemically diverse set of radionuclides that can be used complementarily for targeted alpha therapy, beta therapy, single-photon emission computed tomography (SPECT) imaging, and positron emission tomography (PET) imaging, respectively. Through NMR spectroscopy and X-ray diffraction, we show that PYTA possesses an unusual degree of flexibility for a macrocyclic chelator, undergoing dramatic conformational changes that enable it to optimally satisfy the disparate coordination properties of each metal ion. Subsequent radiolabeling studies revealed that PYTA quantitatively binds all 4 radiometals at room temperature in just minutes at pH 6. Furthermore, these complexes were found to be stable in human serum over 2 half-lives. These results surpass those obtained for 2 state-of-the-art chelators for nuclear medicine, DOTA and macropa. The stability of 225Ac-PYTA and [44Sc]Sc-PYTA, the complexes having the most disparity with respect to metal-ion size, was further probed in mice. The resulting PET images (44Sc) and ex vivo biodistribution profiles (44Sc and 225Ac) of the PYTA complexes differed dramatically from those of unchelated [44Sc]Sc3+ and 225Ac3+. These differences provide evidence that PYTA retains this size-divergent pair of radionuclides in vivo. Collectively, these studies establish PYTA as a new workhorse chelator for nuclear medicine and warrant its further investigation in targeted constructs.
Collapse
Affiliation(s)
- Megan E Simms
- Chemical Sciences Division, Oak Ridge National Laboratory Oak Ridge TN 37831 USA
| | - Zhiyao Li
- Department of Radiology, Washington University in St. Louis School of Medicine St. Louis MO 63110 USA
- Program in Quantitative Molecular Therapeutics, Washington University in St. Louis School of Medicine St. Louis MO 63110 USA
| | - Megan M Sibley
- Chemical Sciences Division, Oak Ridge National Laboratory Oak Ridge TN 37831 USA
| | - Alexander S Ivanov
- Chemical Sciences Division, Oak Ridge National Laboratory Oak Ridge TN 37831 USA
| | - Caroline M Lara
- Department of Biological Sciences, University of Notre Dame Notre Dame IN 46556 USA
| | - Timothy C Johnstone
- Department of Chemistry and Biochemistry, University of California Santa Cruz Santa Cruz CA 95064 USA
| | - Vilmos Kertesz
- Biosciences Division, Oak Ridge National Laboratory Oak Ridge TN 37831 USA
| | - Amanda Fears
- Department of Radiology, Washington University in St. Louis School of Medicine St. Louis MO 63110 USA
- Program in Quantitative Molecular Therapeutics, Washington University in St. Louis School of Medicine St. Louis MO 63110 USA
| | - Frankie D White
- Radioisotope Science and Technology Division, Oak Ridge National Laboratory Oak Ridge TN 37831 USA
| | - Daniel L J Thorek
- Department of Radiology, Washington University in St. Louis School of Medicine St. Louis MO 63110 USA
- Program in Quantitative Molecular Therapeutics, Washington University in St. Louis School of Medicine St. Louis MO 63110 USA
- Department of Biomedical Engineering, Washington University in St. Louis St. Louis MO 63110 USA
- Oncologic Imaging Program, Siteman Cancer Center, Washington University in St. Louis School of Medicine St. Louis MO 63110 USA
| | - Nikki A Thiele
- Chemical Sciences Division, Oak Ridge National Laboratory Oak Ridge TN 37831 USA
| |
Collapse
|
12
|
Roy T, Pogorilyy E, Kumarananthan CP, Kvitastein UA, Foscato M, Törnroos KW, Adamsen TCH, Le Roux E. Synthesis and stability of the [ 45Ti]Ti-DOTA complex: en route towards aza-macrocyclic 45Ti-based radiopharmaceuticals. Chem Commun (Camb) 2024; 60:7148-7151. [PMID: 38860653 DOI: 10.1039/d4cc01800a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2024]
Abstract
We report the use of DOTA as a chelator for titanium. The resulting complex is fully characterised and in vitro stability studies reveal its high kinetic inertness against transmetallation and transchelation. The radiolabeling of DOTA with 45Ti, via a guaiacol-based liquid-liquid extraction method, leads to a high radiochemical conversion up to 98%.
Collapse
Affiliation(s)
- Tamal Roy
- Department of Chemistry, University of Bergen, Allégaten 41, Bergen, Norway.
| | - Eduard Pogorilyy
- Department of Chemistry, University of Bergen, Allégaten 41, Bergen, Norway.
| | - Chubina P Kumarananthan
- Department of Radiology, Haukeland University Hospital, Centre for Nuclear Medicine and PET, Jonas Lies vei 65, Bergen, Norway
| | - Unni A Kvitastein
- Department of Radiology, Haukeland University Hospital, Centre for Nuclear Medicine and PET, Jonas Lies vei 65, Bergen, Norway
| | - Marco Foscato
- Department of Chemistry, University of Bergen, Allégaten 41, Bergen, Norway.
| | - Karl W Törnroos
- Department of Chemistry, University of Bergen, Allégaten 41, Bergen, Norway.
| | - Tom C H Adamsen
- Department of Chemistry, University of Bergen, Allégaten 41, Bergen, Norway.
- Department of Radiology, Haukeland University Hospital, Centre for Nuclear Medicine and PET, Jonas Lies vei 65, Bergen, Norway
| | - Erwan Le Roux
- Department of Chemistry, University of Bergen, Allégaten 41, Bergen, Norway.
| |
Collapse
|
13
|
Franchi S, Madabeni A, Tosato M, Gentile S, Asti M, Orian L, Di Marco V. Navigating through the coordination preferences of heavy alkaline earth metals: Laying the foundations for 223Ra- and 131/135mBa-based targeted alpha therapy and theranostics of cancer. J Inorg Biochem 2024; 256:112569. [PMID: 38701687 DOI: 10.1016/j.jinorgbio.2024.112569] [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: 02/26/2024] [Revised: 04/04/2024] [Accepted: 04/18/2024] [Indexed: 05/05/2024]
Abstract
The clinical success of [223Ra]RaCl2 (Xofigo®) for the palliative treatment of bone metastases in patients with prostate cancer has highlighted the therapeutic potential of α-particle emission. Expanding the applicability of radium-223 in Targeted Alpha Therapy of non-osseous tumors is followed up with significant interest, as it holds the potential to unveil novel treatment options in the comprehensive management of cancer. Moreover, the use of barium radionuclides, like barium-131 and -135m, is still unfamiliar in nuclear medicine applications, although they can be considered as radium-223 surrogates for imaging purposes. Enabling these applications requires the establishment of chelators able to form stable complexes with radium and barium radionuclides. Until now, only a limited number of ligands have been suggested and these molecules have been primarily inspired by existing structures known for their ability to complex large metal cations. However, a systematic inspection of chelators specifically tailored to Ra2+ and Ba2+ has yet to be conducted. This work delves into a comprehensive investigation of a series of small organic ligands, aiming to unveil the coordination preferences of both radium-223 and barium-131/135m. Electronic binding energies of both metal cations to each ligand were theoretically computed via Density Functional Theory calculations (COSMO-ZORA-PBE-D3/TZ2P), while thermodynamic stability constants were experimentally determined for Ba2+-ligand complexes by potentiometry, NMR and UV-Vis spectroscopies. The outcomes revealed malonate, 2-hydroxypyridine 1-oxide and picolinate as the most favorable building blocks to design multidentate chelators. These findings serve as foundation guidelines, propelling the development of cutting-edge radium-223- and barium-131/135m-based radiopharmaceuticals for Targeted Alpha Therapy and theranostics of cancer.
Collapse
Affiliation(s)
- Sara Franchi
- Department of Chemical Sciences, University of Padova, 35131 Padova, Italy.
| | - Andrea Madabeni
- Department of Chemical Sciences, University of Padova, 35131 Padova, Italy.
| | - Marianna Tosato
- Radiopharmaceutical Chemistry Section, Nuclear Medicine Unit, AUSL-IRCCS Reggio Emilia, 42122 Reggio Emilia, Italy.
| | - Silvia Gentile
- Department of Chemical Sciences, University of Padova, 35131 Padova, Italy.
| | - Mattia Asti
- Radiopharmaceutical Chemistry Section, Nuclear Medicine Unit, AUSL-IRCCS Reggio Emilia, 42122 Reggio Emilia, Italy.
| | - Laura Orian
- Department of Chemical Sciences, University of Padova, 35131 Padova, Italy; National Institute of Nuclear Physics, National Laboratories of Legnaro (INFN-LNL), 35020 Legnaro, Padova, Italy.
| | - Valerio Di Marco
- Department of Chemical Sciences, University of Padova, 35131 Padova, Italy.
| |
Collapse
|
14
|
Wu J, Chen J, Zhao Y, Yuan M, Chen X, He X, Zhang J, Shao G, Sun Q. Molecular SPECT/CT Profiling of Claudin18.2 Expression In Vivo: Implication for Patients with Gastric Cancer. Mol Pharm 2024; 21:3447-3458. [PMID: 38843446 DOI: 10.1021/acs.molpharmaceut.4c00155] [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: 07/02/2024]
Abstract
Zolbetuximab (IMAB362), a monoclonal antibody targeting Claudin18.2 (CLDN 18.2), demonstrates a significant clinical benefit in patients with advanced gastroesophageal cancers. The noninvasive assessment of CLDN18.2 expression through molecular imaging offers a potential avenue for expedited monitoring and the stratification of patients into risk groups. This study elucidates that CLDN18.2 is expressed at a noteworthy frequency in primary gastric cancers and their metastases. The iodogen method was employed to label IMAB362 with 123I/131I. The results demonstrated the efficient and reproducible synthesis of 123I-IMAB362, with a specific binding affinity to CLDN18.2. Immuno-single-photon emission computed tomography (SPECT) imaging revealed the rapid accumulation of 123I-IMAB362 in gastric cancer xenografts at 12 h, remaining stable for 3 days in patient-derived tumor xenograft models. Additionally, tracer uptake of 123I-IMAB362 in MKN45 cells surpassed that in MKN28 cells at each time point, with tumor uptake correlating significantly with CLDN18.2 expression levels. Positron emission tomography/computed tomography imaging indicated that tumor uptake of 18F-FDG and the functional/viable tumor volume in the 131I-IMAB362 group were significantly lower than those in the 123I-IMAB362 group on day 7. In conclusion, 123I-IMAB362 immuno-SPECT imaging offers an effective method for direct, noninvasive, and whole-body quantitative assessment of tumor CLDN18.2 expression in vivo. This approach holds promise for accelerating the monitoring and stratification of patients with gastric cancer.
Collapse
Affiliation(s)
- Jian Wu
- Department of Science and Technology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| | - Jun Chen
- Department of Nuclear Medicine, The Affiliated Taizhou People's Hospital of Nanjing University of Chinese Medicine, Taizhou 225300, China
- Department of Nuclear Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, China
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| | - Yuetong Zhao
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| | - Mengyun Yuan
- Department of Science and Technology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| | - Xu Chen
- Department of Science and Technology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| | - Xiangdong He
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| | - Jun Zhang
- Department of Nuclear Medicine, The Affiliated Taizhou People's Hospital of Nanjing University of Chinese Medicine, Taizhou 225300, China
- Department of Nuclear Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, China
| | - Guoqiang Shao
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| | - Qingmin Sun
- Department of Science and Technology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, China
| |
Collapse
|
15
|
Harriswangler C, McNeil BL, Brandariz I, Valencia L, Esteban-Gómez D, Ramogida CF, Platas-Iglesias C. Incorporation of Carboxylate Pendant Arms into 18-Membered Macrocycles: Effects on [ nat/203Pb]Pb(II) Complexation. Chemistry 2024; 30:e202400434. [PMID: 38466910 DOI: 10.1002/chem.202400434] [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: 01/31/2024] [Revised: 03/04/2024] [Accepted: 03/11/2024] [Indexed: 03/13/2024]
Abstract
We present a detailed investigation on the coordination chemistry of [nat/203Pb]Pb(II) with chelators H4PYTA and H4CHX-PYTA. These chelators belong to the family of ligands derived from the 18-membered macrocyclic backbone PYAN and present varying degrees of rigidity due to the presence of either ethyl or cyclohexyl spacers. A complete study of the stable Pb(II) complexes is carried out via NMR, X-Ray crystallography, stability constant determination and computational studies. While these studies indicated that Pb(II) complexation is achieved, and the thermodynamic stability of the resulting complexes is very high, a certain degree of fluxionality does exist in both cases. Nevertheless, radiolabeling studies were carried out using SPECT (single photon emission computed tomography) compatible isotope lead-203 (203Pb, t1/2=51.9 h), and while both chelators complex the radioisotope, the incorporation of carboxylate pendant arms appears to be detrimental towards the stability of the complexes when compared to the previously described amide analogues. Additionally, incorporation of a cyclohexyl spacer does not improve the kinetic inertness of the system.
Collapse
Affiliation(s)
- Charlene Harriswangler
- Centro Interdisciplinar de Química e Bioloxía (CICA) and Departamento de Química, Universidade da Coruña, Campus da Zapateira-Rúa da Fraga 10, 15001, A Coruña, Spain
| | - Brooke L McNeil
- Department of Chemistry, Simon Fraser University, 8888 University Drive, V5A 1S6, Burnaby, British Columbia, Canada
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, V6T 2A3, Vancouver, British Columbia, Canada
| | - Isabel Brandariz
- Centro Interdisciplinar de Química e Bioloxía (CICA) and Departamento de Química, Universidade da Coruña, Campus da Zapateira-Rúa da Fraga 10, 15001, A Coruña, Spain
| | - Laura Valencia
- Departamento de Química Inorgánica, Facultad de Ciencias, Universidade de Vigo, As Lagoas, Marcosende, 36310, Pontevedra, Spain
| | - David Esteban-Gómez
- Centro Interdisciplinar de Química e Bioloxía (CICA) and Departamento de Química, Universidade da Coruña, Campus da Zapateira-Rúa da Fraga 10, 15001, A Coruña, Spain
| | - Caterina F Ramogida
- Department of Chemistry, Simon Fraser University, 8888 University Drive, V5A 1S6, Burnaby, British Columbia, Canada
- Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, V6T 2A3, Vancouver, British Columbia, Canada
| | - Carlos Platas-Iglesias
- Centro Interdisciplinar de Química e Bioloxía (CICA) and Departamento de Química, Universidade da Coruña, Campus da Zapateira-Rúa da Fraga 10, 15001, A Coruña, Spain
| |
Collapse
|
16
|
Binmujlli MA. Radiological and Molecular Analysis of Radioiodinated Anastrozole and Epirubicin as Innovative Radiopharmaceuticals Targeting Methylenetetrahydrofolate Dehydrogenase 2 in Solid Tumors. Pharmaceutics 2024; 16:616. [PMID: 38794278 PMCID: PMC11126143 DOI: 10.3390/pharmaceutics16050616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/13/2024] [Accepted: 04/18/2024] [Indexed: 05/26/2024] Open
Abstract
In the dynamic field of radiopharmaceuticals, innovating targeted agents for cancer diagnosis and therapy is crucial. Our study enriches this evolving landscape by evaluating the potential of radioiodinated anastrozole ([125I]anastrozole) and radioiodinated epirubicin ([125I]epirubicin) as targeting agents against MTHFD2-driven tumors. MTHFD2, which is pivotal in one-carbon metabolism, is notably upregulated in various cancers, presenting a novel target for radiopharmaceutical application. Through molecular docking and 200 ns molecular dynamics (MD) simulations, we assess the binding efficiency and stability of [125I]anastrozole and [125I]epirubicin with MTHFD2. Molecular docking illustrates that [125I]epirubicin has a superior binding free energy (∆Gbind) of -41.25 kJ/mol compared to -39.07 kJ/mol for [125I]anastrozole and -38.53 kJ/mol for the control ligand, suggesting that it has a higher affinity for MTHFD2. MD simulations reinforce this, showing stable binding, as evidenced by root mean square deviation (RMSD) values within a narrow range, underscoring the structural integrity of the enzyme-ligand complexes. The root mean square fluctuation (RMSF) analysis indicates consistent dynamic behavior of the MTHFD2 complex upon binding with [125I]anastrozole and [125I]epirubicin akin to the control. The radius of gyration (RG) measurements of 16.90 Å for MTHFD2-[125I]anastrozole and 16.84 Å for MTHFD2-[125I]epirubicin confirm minimal structural disruption upon binding. The hydrogen bond analysis reveals averages of two and three stable hydrogen bonds for [125I]anastrozole and [125I]epirubicin complexes, respectively, highlighting crucial stabilizing interactions. The MM-PBSA calculations further endorse the thermodynamic favorability of these interactions, with binding free energies of -48.49 ± 0.11 kJ/mol for [125I]anastrozole and -43.8 kJ/mol for MTHFD2-. The significant contribution of Van der Waals and electrostatic interactions to the binding affinities of [125I]anastrozole and [125I]epirubicin, respectively, underscores their potential efficacy for targeted tumor imaging and therapy. These computational findings lay the groundwork for the future experimental validation of [125I]anastrozole and [125I]epirubicin as MTHFD2 inhibitors, heralding a notable advancement in precision oncology tools. The data necessitate subsequent in vitro and in vivo assays to corroborate these results.
Collapse
Affiliation(s)
- Mazen Abdulrahman Binmujlli
- Department of Internal Medicine, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), P.O. Box 90950, Riyadh 11623, Saudi Arabia
| |
Collapse
|
17
|
Baun C, Naghavi-Behzad M, Hildebrandt MG, Gerke O, Thisgaard H. Gastrin-releasing peptide receptor as a theranostic target in breast cancer: a systematic scoping review. Semin Nucl Med 2024; 54:256-269. [PMID: 38342656 DOI: 10.1053/j.semnuclmed.2024.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 02/13/2024]
Abstract
The gastrin-releasing peptide receptor (GRPR) is known to be overexpressed in breast cancer, making it a promising target for both imaging and therapy within a theranostic framework. Various radioligands targeting GRPR have undergone investigation in preclinical and clinical studies related to breast cancer. This systematic scoping review aimed to assess the current evidence on GRPR-targeted radioligands for diagnostic and therapeutic applications in breast cancer. The methodology followed the PRISMA-ScR protocol. The literature search was conducted in September 2023 and encompassed MEDLINE, Embase, Cochrane, and Scopus databases. We included original peer-reviewed studies focused on breast cancer patients or in vivo breast cancer models. Two reviewers performed the study selection process independently. Data were extracted, synthesized, and categorized into preclinical and clinical studies, further subdivided based on radioligand properties. A total of 35 original studies were included in the review, with three of them evaluating therapeutic outcomes. The results indicated that GRPR-radioantagonists are superior to GRPR-agonists, exhibiting preferable in vivo stability, rapid, specific tumor targeting, and enhanced retention. Both preclinical and clinical evaluations demonstrated renal excretion and high uptake in normal GRPR-expressing tissue, primarily the pancreas. A significant positive correlation was observed between GRPR and estrogen-receptor expression. In the clinical setting, GRPR-radioligands effectively detected primary tumors and, to a lesser extent, lymph node metastases. Moreover, GRPR-targeted radioantagonists successfully identified distant metastases originating from various sites in advanced metastatic disease, strongly correlated with positive estrogen receptor expression. Preclinical therapeutic evaluation of GRPR-radioligands labeled with lutetium-177 showed promising tumor responses, and none of the studies reported any observed or measured side effects, indicating a safe profile. In conclusion, the evidence presented in this review indicates a preference for GRPR-targeted antagonists over agonists, owing to their superior kinetics and promising diagnostic potential. Clinical assessments suggested diagnostic value for GRPR-targeted theranostics in breast cancer patients, particularly those with high estrogen receptor expression. Nevertheless, in the therapeutic clinical context, paying attention to the radiation dose administered to the pancreas and kidneys is crucial.
Collapse
Affiliation(s)
- Christina Baun
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, Odense, Denmark.
| | - Mohammad Naghavi-Behzad
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Malene Grubbe Hildebrandt
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Center for Personalized Response Monitoring in Oncology (PREMIO), Odense University Hospital, Odense, Denmark; Centre for Innovative Medical Technology, Odense University Hospital, Odense, Denmark
| | - Oke Gerke
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Helge Thisgaard
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark; Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
18
|
Zubenko AD, Shchukina AA, Chernikova EY, Egorova BV, Ikonnikova IS, Priselkova AB, Larenkov AA, Bubenshchikov VB, Mitrofanov AA, Fedorov YV, Fedorova OA. Synthesis of new acyclic chelators H4aPyta and H6aPyha and their complexes with Cu 2+, Ga 3+, Y 3+, and Bi 3. Dalton Trans 2024; 53:1141-1155. [PMID: 38105658 DOI: 10.1039/d3dt03451h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
In this article, we present the synthesis and characterization of new acyclic pyridine-containing polyaminocarboxylate ligands H4aPyta and H6aPyha, which differ in structural rigidity and the number of chelating groups. Their abilities to form complexes with Cu2+, Ga3+, Y3+, and Bi3+ cations, as well as the stability of the complexes, were evaluated by potentiometric titration method, radiolabeling with the corresponding radionuclides, in vitro studies, mass spectrometry, and HPLC. The structures of the resulting complexes were determined using NMR spectroscopy and DFT calculations. The results obtained made it possible to evaluate the influence of the structural features of the complexes on their stability. The developed chelators H4aPyta and H6aPyha were proved to be promising for further research in the field of radiopharmaceuticals.
Collapse
Affiliation(s)
- Anastasia D Zubenko
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 St. Vavilova, 28, GSP-1, Moscow, Russian Federation.
| | - Anna A Shchukina
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 St. Vavilova, 28, GSP-1, Moscow, Russian Federation.
| | - Ekaterina Y Chernikova
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 St. Vavilova, 28, GSP-1, Moscow, Russian Federation.
| | - Bayirta V Egorova
- Lomonosov Moscow State University, 119991 Leninskie Gory, 1/3, Moscow, Russian Federation
| | - Irina S Ikonnikova
- Lomonosov Moscow State University, 119991 Leninskie Gory, 1/3, Moscow, Russian Federation
| | - Anna B Priselkova
- Lomonosov Moscow State University, 119991 Leninskie Gory, 1/3, Moscow, Russian Federation
| | - Anton A Larenkov
- State Research Center-Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Zhivopisnaya Str., Bld. 46, 123098 Moscow, Russian Federation
| | - Viktor B Bubenshchikov
- State Research Center-Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Zhivopisnaya Str., Bld. 46, 123098 Moscow, Russian Federation
| | - Artem A Mitrofanov
- Lomonosov Moscow State University, 119991 Leninskie Gory, 1/3, Moscow, Russian Federation
| | - Yury V Fedorov
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 St. Vavilova, 28, GSP-1, Moscow, Russian Federation.
| | - Olga A Fedorova
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, 119991 St. Vavilova, 28, GSP-1, Moscow, Russian Federation.
| |
Collapse
|
19
|
Tosato M, Gandini A, Happel S, Bas M, Donzella A, Zenoni A, Salvini A, Andrighetto A, Di Marco V, Asti M. Chromatographic separation of silver-111 from neutron-irradiated palladium target: toward direct labeling of radiotracers. EJNMMI Radiopharm Chem 2023; 8:43. [PMID: 38123869 PMCID: PMC10733254 DOI: 10.1186/s41181-023-00232-0] [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: 10/27/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Silver-111 is a promising β--emitting radioisotope with ideal characteristics for targeted radionuclide therapy and associated single photon emission tomography imaging. Its decay properties closely resemble the clinically established lutetium-177, making it an attractive candidate for therapeutic applications. In addition, the clinical value of silver-111 is further enhanced by the existence of the positron-emitting counterpart silver-103, thus imparting a truly theranostic potential to this element. A so-fitting matching pair could potentially overcome the current limitations associated with the forced use of chemically different isotopes as imaging surrogates of lutetium-177, leading to more accurate and efficient diagnosis and treatment. However, the use of silver-111-based radiopharmaceuticals in vivo has faced obstacles due to the challenges related to its production and radiochemical separation from the target material. To address these issues, this study aims to implement a chromatographic separation methodology for the purification of reactor-produced silver-111. The ultimate goal is to achieve a ready-to-use formulation for the direct radiolabeling of tumour-seeking biomolecules. RESULTS A two-step sequence chromatographic process was validated for cold Ag-Pd separation and then translated to the radioactive counterpart. Silver-111 was produced via the 110Pd(n,γ)111Pd nuclear reaction on a natural palladium target and the subsequent β--decay of palladium-111. Silver-111 was chemically separated from the metallic target via the implemented chromatographic process by using commercially available LN and TK200 resins. The effectiveness of the separations was assessed by inductively coupled plasma optical emission spectroscopy and γ-spectrometry, respectively, and the Ag+ retrieval was afforded in pure water. Recovery of silver-111 was > 90% with a radionuclidic purity > 99% and a separation factor of around 4.21·10-4. CONCLUSIONS The developed separation method was suitable to obtain silver-111 with high molar activity in a ready-to-use water-based formulation that can be directly employed for the labeling of radiotracers. By successfully establishing a robust and efficient production and purification method for silver-111, this research paves the way for its wider application in targeted radionuclide therapy and precision imaging.
Collapse
Affiliation(s)
- Marianna Tosato
- Radiopharmaceutical Chemistry Section, Nuclear Medicine Unit, AUSL di Reggio Emilia: Azienda Unità Sanitaria Locale - IRCCS Tecnologie Avanzate e Modelli Assistenziali in Oncologia di Reggio Emilia, Via Amendola 2, 42122, Reggio Emilia, Italy
- Department of Chemical Sciences, University of Padova, 35131, Padua, Italy
| | - Andrea Gandini
- Laboratory of Applied Nuclear Energy, 27100, Pavia, Italy
| | | | - Marine Bas
- TrisKem International SAS, 35170, Brittany, France
| | - Antonietta Donzella
- Department of Mechanical and Industrial Engineering, University of Brescia, 25123, Brescia, Italy
- Italian Institute of Nuclear Physics, Pavia Section, 27100, Pavia, Italy
| | - Aldo Zenoni
- Department of Mechanical and Industrial Engineering, University of Brescia, 25123, Brescia, Italy
- Italian Institute of Nuclear Physics, Pavia Section, 27100, Pavia, Italy
| | - Andrea Salvini
- Laboratory of Applied Nuclear Energy, 27100, Pavia, Italy
| | - Alberto Andrighetto
- Italian Institute of Nuclear Physics, Legnaro National Laboratories, 35020, Legnaro, (Padova), Italy
| | - Valerio Di Marco
- Department of Chemical Sciences, University of Padova, 35131, Padua, Italy
| | - Mattia Asti
- Radiopharmaceutical Chemistry Section, Nuclear Medicine Unit, AUSL di Reggio Emilia: Azienda Unità Sanitaria Locale - IRCCS Tecnologie Avanzate e Modelli Assistenziali in Oncologia di Reggio Emilia, Via Amendola 2, 42122, Reggio Emilia, Italy.
| |
Collapse
|
20
|
Kopp I, Cieslik P, Anger K, Josephy T, Neupert L, Velmurugan G, Gast M, Wadepohl H, Brühlmann SA, Walther M, Kopka K, Bachmann M, Stephan H, Kubeil M, Comba P. Bispidine Chelators for Radiopharmaceutical Applications with Lanthanide, Actinide, and Main Group Metal Ions. Inorg Chem 2023; 62:20754-20768. [PMID: 37707798 DOI: 10.1021/acs.inorgchem.3c02340] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Octadentate and specifically nonadentate ligands with a bispidine scaffold (3,7-diazabicyclo[3.3.1]nonane) are known to be efficiently coordinated to a range of metal ions of interest in radiopharmaceutical chemistry and lead to exceedingly stable and inert complexes. Nonadentate bispidine L2 (with a tridentate bipyridine acetate appended to N3 and a picolinate at N7) has been shown before to be an ideal chelator for 111In3+, 177Lu3+, and 225Ac3+, nuclides of interest for diagnosis and therapy, and a proof-of-principle study with an SSTR2-specific octreotate has shown potential for theranostic applications. We now have extended these studies in two directions. First, we present ligand derivative L3, in which the bipyridine acetate is substituted with terpyridine, a softer donor for metal ions with a preference for more covalency. L3 did not fulfill the hopes because complexation is much less efficient. While for Bi3+ and Pb2+ the ligand is an excellent chelator with properties similar to those of L2, Lu3+ and La3+ show very slow and inefficient complexation with L3 in contrast to L2, and 225Ac3+ is not fully coordinated, even at an increased temperature (92% radiochemical yield at 80 °C, 60 min, [L3] = 10-4 M). These observations have led to a hypothesis for the complexation pathway that is in line with all of the experimental data and supported by a preliminary density functional theory analysis, which is important for the design of further optimized bispidine chelators. Second, the coordination chemistry of L2 has been extended to Bi3+, La3+, and Pb2+, including solid state and solution structural work, complex stabilities, radiolabeling, and radiostability studies. All complexes of this ligand (La3+, Ac3+, Lu3+, Bi3+, In3+, and Pb2+), including nuclides for targeted α therapy (TAT), single-photon emission computed tomography, and positron emission tomography, are formed efficiently under physiological conditions, i.e., suitable for the labeling of delicate biological vectors such as antibodies, and the complexes are very stable and inert. Importantly, for TAT with 225Ac, the daughter nuclides 213Bi and 209Pb also form stable complexes, and this is important for reducing damage to healthy tissue.
Collapse
Affiliation(s)
- Ina Kopp
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Patrick Cieslik
- Universität Heidelberg, Anorganisch-Chemisches Institut, INF 270, 69120 Heidelberg, Germany
| | - Karl Anger
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Thomas Josephy
- Universität Heidelberg, Anorganisch-Chemisches Institut, INF 270, 69120 Heidelberg, Germany
| | - Lucca Neupert
- Universität Heidelberg, Anorganisch-Chemisches Institut, INF 270, 69120 Heidelberg, Germany
| | - Gunasekaran Velmurugan
- Universität Heidelberg, Anorganisch-Chemisches Institut, INF 270, 69120 Heidelberg, Germany
| | - Michael Gast
- Universität Heidelberg, Anorganisch-Chemisches Institut, INF 270, 69120 Heidelberg, Germany
| | - Hubert Wadepohl
- Universität Heidelberg, Anorganisch-Chemisches Institut, INF 270, 69120 Heidelberg, Germany
| | - Santiago Andrés Brühlmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Martin Walther
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Klaus Kopka
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany
- Technische Universität Dresden, Faculty of Chemistry and Food Chemistry, School of Science, 01069 Dresden, Germany
- National Center for Tumor Diseases (NCT) Dresden, University Hospital Carl Gustav Carus, Fetscherstraße 74, 01307 Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, 01307 Dresden, Germany
| | - Michael Bachmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany
- National Center for Tumor Diseases (NCT) Dresden, University Hospital Carl Gustav Carus, Fetscherstraße 74, 01307 Dresden, Germany
- Technische Universität Dresden, Medical Faculty Carl Gustav Carus, 01069 Dresden, Germany
| | - Holger Stephan
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Manja Kubeil
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Peter Comba
- Universität Heidelberg, Anorganisch-Chemisches Institut, INF 270, 69120 Heidelberg, Germany
- Universität Heidelberg, Interdisciplinary Center for Scientific Computing, INF 205, 69120 Heidelberg, Germany
| |
Collapse
|
21
|
Kelderman CAA, Maclean RC, Hungnes IN, Davey PRWJ, Salimova E, de Veer M, Patel N, Ma MT, Paterson BM. Technetium Nitrido Complexes of Tetradentate Thiosemicarbazones: Kit-Based Radiolabeling, Characterization, and In Vivo Evaluation. Inorg Chem 2023; 62:20791-20805. [PMID: 37855107 DOI: 10.1021/acs.inorgchem.3c02650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2023]
Abstract
Bis(thiosemicarbazone) and pyridylhydrazone-thiosemicarbazone chelators have demonstrated utility in nuclear medicine. In particular, the 64Cu2+ complexes have been extensively developed for hypoxia imaging and molecular imaging of peptide and protein markers of disease. However, the chemistry and application of bis(thiosemicarbazone) and pyridylhydrazone-thiosemicarbazone chelators in combination with 99mTc, the most widely used radionuclide in nuclear medicine, is underexplored. Herein, a series of bis(thiosemicarbazone) and pyridylhydrazone-thiosemicarbazone chelators were radiolabeled with nitrido-technetium-99m in an optimized one-pot synthesis from [99mTc]TcO4-. Optimization of the radiochemical syntheses allowed for production of the complexes in >90% radiochemical conversion with apparent molar activities of 3.3-5 GBq/μmol. Competition experiments demonstrated the excellent stability of the complexes. The nitrido-technetium-99 complexes were synthesized, and the chemical identities were investigated using mass spectrometry, spectroscopy, and density functional theory calculations. Complexation of nitrido-rhenium(V) was achieved with the N4-dialkylated bis(thiosemicarbazones). Planar imaging and ex vivo biodistribution studies of the five 99mTc complexes were conducted on healthy BALB/c mice to determine in vivo behavior. The lipophilic nature of the complexes resulted in uptake of 1.6-5.7% ID g-1 in the brain at 2 min postinjection and retention of 0.4-1.7% ID g-1 at 15 min postinjection. The stability of the complexes and the biodistribution data demonstrate that these chelators are ideal platforms for future production of radiopharmaceutical candidates.
Collapse
Affiliation(s)
| | - Rachel C Maclean
- School of Chemistry, Monash University, Clayton, Victoria 3800, Australia
- Centre for Advanced Imaging, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Ingebjørg N Hungnes
- School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, London SE1 7EH, United Kingdom
| | - Patrick R W J Davey
- School of Chemistry, Monash University, Clayton, Victoria 3800, Australia
- Department of Chemistry, Simon Fraser University, Burnaby, British Columbia V5A 1S6, Canada
| | - Ekaterina Salimova
- Monash Biomedical Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - Michael de Veer
- Monash Biomedical Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - Natasha Patel
- School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, London SE1 7EH, United Kingdom
| | - Michelle T Ma
- School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, London SE1 7EH, United Kingdom
| | - Brett M Paterson
- School of Chemistry, Monash University, Clayton, Victoria 3800, Australia
- Centre for Advanced Imaging, The University of Queensland, St. Lucia, Queensland 4072, Australia
- Monash Biomedical Imaging, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
22
|
Marlin A, Koller A, Madarasi E, Cordier M, Esteban-Gómez D, Platas-Iglesias C, Tircsó G, Boros E, Patinec V, Tripier R. H 3nota Derivatives Possessing Picolyl and Picolinate Pendants for Ga 3+ Coordination and 67Ga 3+ Radiolabeling. Inorg Chem 2023; 62:20634-20645. [PMID: 37552617 DOI: 10.1021/acs.inorgchem.3c01417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2023]
Abstract
We synthesized, thanks to the regiospecific N-functionalization using an orthoamide intermediate, two 1,4,7-triazacyclononane derivatives containing an acetate arm and either a methylpyridine or a picolinic acid group, respectively, Hnoapy and H2noapa, as new Ga3+ chelators for potential use in nuclear medicine. The corresponding Ga3+ complexes were synthesized and structurally characterized in solution by 1H and 13C NMR. The [Ga(noapy)]2+ complex appears to exist in solution as two diasteroisomeric pairs of enantiomers, as confirmed by density functional theory (DFT) calculations, while for [Ga(noapa)]+, a single species is present in solution. Solid-state investigations were possible for the [Ga(noapa)]+ complex, which crystallized from water as a pair of enantiomers. The average length of the N-Ga bonds of 2.090 Å is identical with that found for the [Ga(nota)] complex, showing that the presence of the picolinate arm does not hinder the coordination of the ligand to the metal ion. Protonation constants of noapy- and noapa2- were determined by potentiometric titrations, providing an overall basicity ∑log KiH (i = 1-4) that increases in the order noapy- < noapa2- < nota3- with increases in the negative charge of the ligand. Stability constants determined by pH-potentiometric titrations supplemented with 71Ga NMR data show that the stabilities of [Ga(noapy)]2+ and [Ga(noapa)]+ are lower compared to that of [Ga(nota)] but higher than those of other standards such as [Ga(aazta)]-. 67Ga radiolabeling studies were performed in order to demonstrate the potential of these chelators for 67/68Ga-based radiopharmaceuticals. The labelings of Hnoapy and H2noapa were nearly identical, outperforming H3nota. Stability studies were conducted in phosphate-buffered saline and in the presence of human serum transferrin, revealing no significant decomplexation of [67Ga][Ga(noapy)]2+ and [67Ga][Ga(noapa)]+ compared to [67Ga][Ga(nota)]. Finally, all complexes were found to be highly hydrophilic, with calculated log D7.4 values of -3.42 ± 0.05, -3.34 ± 0.04, and -3.00 ± 0.23 for Hnoapy, H2noapa, and H3nota, respectively, correlating with the charge of each complex and the electrostatic potentials obtained with DFT.
Collapse
Affiliation(s)
- Axia Marlin
- CEMCA, Université Brest, UMR 6521, CNRS, 6 avenue Victor le Gorgeu, 29238 Brest, France
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, New York 11794, United States
| | - Angus Koller
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, New York 11794, United States
| | - Enikö Madarasi
- Department of Physical Chemistry, Faculty of Science and Technology, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
- Doctoral School of Chemistry at the University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Marie Cordier
- Institut des Sciences Chimiques de Rennes, Université Rennes, UMR 6226, CNRS, F-35000 Rennes, France
| | - David Esteban-Gómez
- Centro de Investigacións Científicas Avanzadas and Departamento de Química, Universidade da Coruña, Campus da Zapateira, rúa da Fraga 10, 15008A Coruña, Spain
| | - Carlos Platas-Iglesias
- Centro de Investigacións Científicas Avanzadas and Departamento de Química, Universidade da Coruña, Campus da Zapateira, rúa da Fraga 10, 15008A Coruña, Spain
| | - Gyula Tircsó
- Department of Physical Chemistry, Faculty of Science and Technology, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Eszter Boros
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, New York 11794, United States
| | - Véronique Patinec
- CEMCA, Université Brest, UMR 6521, CNRS, 6 avenue Victor le Gorgeu, 29238 Brest, France
| | - Raphaël Tripier
- CEMCA, Université Brest, UMR 6521, CNRS, 6 avenue Victor le Gorgeu, 29238 Brest, France
| |
Collapse
|
23
|
Gawne PJ, Ferreira M, Papaluca M, Grimm J, Decuzzi P. New Opportunities and Old Challenges in the Clinical translation of Nanotheranostics. NATURE REVIEWS. MATERIALS 2023; 8:783-798. [PMID: 39022623 PMCID: PMC11251001 DOI: 10.1038/s41578-023-00581-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 06/27/2023] [Indexed: 07/20/2024]
Abstract
Nanoparticle-based systems imbued with both diagnostic and therapeutic functions, known as nanotheranostics, have enabled remarkable progress in guiding focal therapy, inducing active responses to endogenous and exogenous biophysical stimuli, and stratifying patients for optimal treatment. However, although in recent years more nanotechnological platforms and techniques have been implemented in the clinic, several important challenges remain that are specific to nanotheranostics. In this Review, we first discuss some of the many ways of 'constructing' nanotheranostics, focusing on the different imaging modalities and therapeutic strategies. We then outline nanotheranostics that are currently used in humans at different stages of clinical development, identifying specific advantages and opportunities. Finally, we define critical steps along the winding road of preclinical and clinical development and suggest actions to overcome technical, manufacturing, regulatory and economical challenges for the safe and effective clinical translation of nanotheranostics.
Collapse
Affiliation(s)
- Peter J. Gawne
- UCL Cancer Institute, University College London, London, UK
- Centre for Cancer Biomarkers and Biotherapeutics, Barts Cancer Institute, Queen Mary, University of London, London, UK
- School of Biomedical Engineering and Imaging Sciences, King’s College London, London, UK
| | - Miguel Ferreira
- Department of Radiology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Marisa Papaluca
- School of Public Health, Imperial College of London, South Kensington CampusLondon, UK
| | - Jan Grimm
- Molecular Pharmacology Program and Department of Radiology, Memorial Sloan-Kettering Cancer, Center, New York, NY, USA
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via, Morego 30, 16163, Genoa, IT
| |
Collapse
|
24
|
Franchi S, Asti M, Di Marco V, Tosato M. The Curies' element: state of the art and perspectives on the use of radium in nuclear medicine. EJNMMI Radiopharm Chem 2023; 8:38. [PMID: 37947909 PMCID: PMC10638329 DOI: 10.1186/s41181-023-00220-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 10/19/2023] [Indexed: 11/12/2023] Open
Abstract
BACKGROUND The alpha-emitter radium-223 (223Ra) is presently used in nuclear medicine for the palliative treatment of bone metastases from castration-resistant prostate cancer. This application arises from its advantageous decay properties and its intrinsic ability to accumulate in regions of high bone turnover when injected as a simple chloride salt. The commercial availability of [223Ra]RaCl2 as a registered drug (Xofigo®) is a further additional asset. MAIN BODY The prospect of extending the utility of 223Ra to targeted α-therapy of non-osseous cancers has garnered significant interest. Different methods, such as the use of bifunctional chelators and nanoparticles, have been explored to incorporate 223Ra in proper carriers designed to precisely target tumor sites. Nevertheless, the search for a suitable scaffold remains an ongoing challenge, impeding the diffusion of 223Ra-based radiopharmaceuticals. CONCLUSION This review offers a comprehensive overview of the current role of radium radioisotopes in nuclear medicine, with a specific focus on 223Ra. It also critically examines the endeavors conducted so far to develop constructs capable of incorporating 223Ra into cancer-targeting drugs. Particular emphasis is given to the chemical aspects aimed at providing molecular scaffolds for the bifunctional chelator approach.
Collapse
Affiliation(s)
- Sara Franchi
- Department of Chemical Sciences, University of Padova, Via Marzolo 1, 35131, Padua, Italy
| | - Mattia Asti
- Radiopharmaceutical Chemistry Section, Nuclear Medicine Unit, AUSL di Reggio Emilia: Azienda Unità Sanitaria Locale - IRCCS Tecnologie Avanzate e Modelli Assistenziali in Oncologia di Reggio Emilia, Via Amendola 2, 42122, Reggio Emilia, Italy
| | - Valerio Di Marco
- Department of Chemical Sciences, University of Padova, Via Marzolo 1, 35131, Padua, Italy
| | - Marianna Tosato
- Radiopharmaceutical Chemistry Section, Nuclear Medicine Unit, AUSL di Reggio Emilia: Azienda Unità Sanitaria Locale - IRCCS Tecnologie Avanzate e Modelli Assistenziali in Oncologia di Reggio Emilia, Via Amendola 2, 42122, Reggio Emilia, Italy.
| |
Collapse
|
25
|
Baun C, Dam JH, Hildebrandt MG, Ewald JD, Kristensen BW, Gammelsrød VS, Olsen BB, Thisgaard H. Preclinical evaluation of [ 58mCo]Co-DOTA-PSMA-617 for Auger electron therapy of prostate cancer. Sci Rep 2023; 13:18837. [PMID: 37914790 PMCID: PMC10620164 DOI: 10.1038/s41598-023-43429-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/23/2023] [Indexed: 11/03/2023] Open
Abstract
Prostate-specific membrane antigen (PSMA), highly expressed in prostate cancer, is a promising target for radionuclide therapy. Auger electron-emitting radionuclides are well suited for targeted radionuclide therapy if they can be delivered close to the DNA of the targeted cells. This preclinical study evaluated the theranostic pair [55/58mCo]Co-DOTA-PSMA-617 for PET imaging and Auger electron therapy of prostate cancer. [58mCo]Co-DOTA-PSMA-617 was successfully prepared with > 99% radiochemical yield and purity. In vitro, uptake and subcellular distribution assays in PSMA-positive prostate cancer cells showed PSMA-specific uptake with high cell-associated activity in the nucleus. Incubation with [58mCo]Co-DOTA-PSMA-617 reduced cell viability and clonogenic survival in a significant dose-dependent manner (p < 0.05). Biodistribution of xenografted mice showed high specific tumor uptake of the cobalt-labeled PSMA ligand for all time points with rapid clearance from normal tissues, which PET imaging confirmed. In vivo, therapy with [58mCo]Co-DOTA-PSMA-617 in tumor-bearing mice demonstrated significantly increased median survival for treated mice compared to control animals (p = 0.0014). In conclusion, [55/58mCo]Co-DOTA-PSMA-617 displayed excellent in vitro and in vivo properties, offering significant survival benefits in mice with no observed toxicities.
Collapse
Affiliation(s)
- Christina Baun
- Department of Nuclear Medicine, Odense University Hospital, Kløvervænget 47, 5000, Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Center for Personalized Response Monitoring in Oncology (PREMIO), Odense University Hospital, Odense, Denmark
| | - Johan Hygum Dam
- Department of Nuclear Medicine, Odense University Hospital, Kløvervænget 47, 5000, Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Malene Grubbe Hildebrandt
- Department of Nuclear Medicine, Odense University Hospital, Kløvervænget 47, 5000, Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Center for Personalized Response Monitoring in Oncology (PREMIO), Odense University Hospital, Odense, Denmark
- Centre for Innovative Medical Technology, Odense University Hospital, Odense, Denmark
| | - Jesper Dupont Ewald
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Bjarne Winther Kristensen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Vigga Sand Gammelsrød
- Department of Nuclear Medicine, Odense University Hospital, Kløvervænget 47, 5000, Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Birgitte Brinkmann Olsen
- Department of Nuclear Medicine, Odense University Hospital, Kløvervænget 47, 5000, Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Surgical Pathology, Zealand University Hospital, Roskilde, Denmark
| | - Helge Thisgaard
- Department of Nuclear Medicine, Odense University Hospital, Kløvervænget 47, 5000, Odense C, Denmark.
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
26
|
Sharma S, Pandey MK. Radiometals in Imaging and Therapy: Highlighting Two Decades of Research. Pharmaceuticals (Basel) 2023; 16:1460. [PMID: 37895931 PMCID: PMC10610335 DOI: 10.3390/ph16101460] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/03/2023] [Accepted: 10/05/2023] [Indexed: 10/29/2023] Open
Abstract
The present article highlights the important progress made in the last two decades in the fields of molecular imaging and radionuclide therapy. Advancements in radiometal-based positron emission tomography, single photon emission computerized tomography, and radionuclide therapy are illustrated in terms of their production routes and ease of radiolabeling. Applications in clinical diagnostic and radionuclide therapy are considered, including human studies under clinical trials; their current stages of clinical translations and findings are summarized. Because the metalloid astatine is used for imaging and radionuclide therapy, it is included in this review. In regard to radionuclide therapy, both beta-minus (β-) and alpha (α)-emitting radionuclides are discussed by highlighting their production routes, targeted radiopharmaceuticals, and current clinical translation stage.
Collapse
Affiliation(s)
| | - Mukesh K. Pandey
- Division of Nuclear Medicine, Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA;
| |
Collapse
|
27
|
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
|
28
|
Bobba KN, Bidkar AP, Meher N, Fong C, Wadhwa A, Dhrona S, Sorlin A, Bidlingmaier S, Shuere B, He J, Wilson DM, Liu B, Seo Y, VanBrocklin HF, Flavell RR. Evaluation of 134Ce/ 134La as a PET Imaging Theranostic Pair for 225Ac α-Radiotherapeutics. J Nucl Med 2023; 64:1076-1082. [PMID: 37201957 PMCID: PMC10315697 DOI: 10.2967/jnumed.122.265355] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/07/2023] [Indexed: 05/20/2023] Open
Abstract
225Ac-targeted α-radiotherapy is a promising approach to treating malignancies, including prostate cancer. However, α-emitting isotopes are difficult to image because of low administered activities and a low fraction of suitable γ-emissions. The in vivo generator 134Ce/134La has been proposed as a potential PET imaging surrogate for the therapeutic nuclides 225Ac and 227Th. In this report, we detail efficient radiolabeling methods using the 225Ac-chelators DOTA and MACROPA. These methods were applied to radiolabeling of prostate cancer imaging agents, including PSMA-617 and MACROPA-PEG4-YS5, for evaluation of their in vivo pharmacokinetic characteristics and comparison to the corresponding 225Ac analogs. Methods: Radiolabeling was performed by mixing DOTA/MACROPA chelates with 134Ce/134La in NH4OAc, pH 8.0, at room temperature, and radiochemical yields were monitored by radio-thin-layer chromatography. In vivo biodistributions of 134Ce-DOTA/MACROPA.NH2 complexes were assayed through dynamic small-animal PET/CT imaging and ex vivo biodistribution studies over 1 h in healthy C57BL/6 mice, compared with free 134CeCl3 In vivo, preclinical imaging of 134Ce-PSMA-617 and 134Ce-MACROPA-PEG4-YS5 was performed on 22Rv1 tumor-bearing male nu/nu-mice. Ex vivo biodistribution was performed for 134Ce/225Ac-MACROPA-PEG4-YS5 conjugates. Results: 134Ce-MACROPA.NH2 demonstrated near-quantitative labeling with 1:1 ligand-to-metal ratios at room temperature, whereas a 10:1 ligand-to-metal ratio and elevated temperatures were required for DOTA. Rapid urinary excretion and low liver and bone uptake were seen for 134Ce/225Ac-DOTA/MACROPA. NH2 conjugates in comparison to free 134CeCl3 confirmed high in vivo stability. An interesting observation during the radiolabeling of tumor-targeting vectors PSMA-617 and MACROPA-PEG4-YS5-that the daughter 134La was expelled from the chelate after the decay of parent 134Ce-was confirmed through radio-thin-layer chromatography and reverse-phase high-performance liquid chromatography. Both conjugates, 134Ce-PSMA-617 and 134Ce-MACROPA-PEG4-YS5, displayed tumor uptake in 22Rv1 tumor-bearing mice. The ex vivo biodistribution of 134Ce-MACROPA.NH2, 134Ce-DOTA and 134Ce-MACROPA-PEG4-YS5 corroborated well with the respective 225Ac-conjugates. Conclusion: These results demonstrate the PET imaging potential for 134Ce/134La-labeled small-molecule and antibody agents. The similar 225Ac and 134Ce/134La-chemical and pharmacokinetic characteristics suggest that the 134Ce/134La pair may act as a PET imaging surrogate for 225Ac-based radioligand therapies.
Collapse
Affiliation(s)
- Kondapa Naidu Bobba
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Anil P Bidkar
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Niranjan Meher
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Cyril Fong
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Anju Wadhwa
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Suchi Dhrona
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Alex Sorlin
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Scott Bidlingmaier
- Department of Anesthesia, University of California, San Francisco, San Francisco, California
| | - Becka Shuere
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Jiang He
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia;
| | - David M Wilson
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Bin Liu
- Department of Anesthesia, University of California, San Francisco, San Francisco, California
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California; and
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Henry F VanBrocklin
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California;
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California; and
| | - Robert R Flavell
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California; and
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California
| |
Collapse
|
29
|
Benloucif A, Meyer D, Balasse L, Goubard A, Danner L, Bouhlel A, Castellano R, Guillet B, Chames P, Kerfelec B. Rapid nanobody-based imaging of mesothelin expressing malignancies compatible with blocking therapeutic antibodies. Front Immunol 2023; 14:1200652. [PMID: 37388728 PMCID: PMC10303918 DOI: 10.3389/fimmu.2023.1200652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/25/2023] [Indexed: 07/01/2023] Open
Abstract
Introduction Mesothelin (MSLN) is overexpressed in a wide variety of cancers with few therapeutic options and has recently emerged as an attractive target for cancer therapy, with a large number of approaches currently under preclinical and clinical investigation. In this respect, developing mesothelin specific tracers as molecular companion tools for predicting patient eligibility, monitoring then response to mesothelin-targeting therapies, and tracking the evolution of the disease or for real-time visualisation of tumours during surgery is of growing importance. Methods We generated by phage display a nanobody (Nb S1) and used enzymatic approaches were used to site-directed conjugate Nb S1 with either ATTO 647N fluorochrome or NODAGA chelator for fluorescence and positron emission tomography imaging (PET) respectively. Results We demonstrated that Nb S1 displays a high apparent affinity and specificity for human mesothelin and demonstrated that the binding, although located in the membrane distal domain of mesothelin, is not impeded by the presence of MUC16, the only known ligand of mesothelin, nor by the therapeutic antibody amatuximab. In vivo experiments showed that both ATTO 647N and [68Ga]Ga-NODAGA-S1 rapidly and specifically accumulated in mesothelin positive tumours compared to mesothelin negative tumours or irrelevant Nb with a high tumour/background ratio. The ex vivo biodistribution profile analysis also confirmed a significantly higher uptake of Nb S1 in MSLN-positive tumours than in MSLNlow tumours. Conclusion We demonstrated for the first time the use of an anti-MSLN nanobody as PET radiotracer for same day imaging of MSLN+ tumours, targeting an epitope compatible with the monitoring of amatuximab-based therapies and current SS1-derived-drug conjugates.
Collapse
Affiliation(s)
- Abdennour Benloucif
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Damien Meyer
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Laure Balasse
- Aix Marseille Univ, CNRS, Centre Européen de Recherche en Imagerie Medicale (CERIMED), Marseille, France
- Aix-marseille University, INSERM, INRAE, Centre de recherche en Cardiovasculaire et Nutrition (C2VN), Marseille, France
| | - Armelle Goubard
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, TrGET Preclinical Platform, Marseille, France
| | - Lucile Danner
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Ahlem Bouhlel
- Aix Marseille Univ, CNRS, Centre Européen de Recherche en Imagerie Medicale (CERIMED), Marseille, France
- Aix-marseille University, INSERM, INRAE, Centre de recherche en Cardiovasculaire et Nutrition (C2VN), Marseille, France
| | - Rémy Castellano
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, TrGET Preclinical Platform, Marseille, France
| | - Benjamin Guillet
- Aix Marseille Univ, CNRS, Centre Européen de Recherche en Imagerie Medicale (CERIMED), Marseille, France
- Aix-marseille University, INSERM, INRAE, Centre de recherche en Cardiovasculaire et Nutrition (C2VN), Marseille, France
| | - Patrick Chames
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| | - Brigitte Kerfelec
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, Marseille, France
| |
Collapse
|
30
|
Funeh CN, Bridoux J, Ertveldt T, De Groof TWM, Chigoho DM, Asiabi P, Covens P, D'Huyvetter M, Devoogdt N. Optimizing the Safety and Efficacy of Bio-Radiopharmaceuticals for Cancer Therapy. Pharmaceutics 2023; 15:pharmaceutics15051378. [PMID: 37242621 DOI: 10.3390/pharmaceutics15051378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/20/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
The precise delivery of cytotoxic radiation to cancer cells through the combination of a specific targeting vector with a radionuclide for targeted radionuclide therapy (TRT) has proven valuable for cancer care. TRT is increasingly being considered a relevant treatment method in fighting micro-metastases in the case of relapsed and disseminated disease. While antibodies were the first vectors applied in TRT, increasing research data has cited antibody fragments and peptides with superior properties and thus a growing interest in application. As further studies are completed and the need for novel radiopharmaceuticals nurtures, rigorous considerations in the design, laboratory analysis, pre-clinical evaluation, and clinical translation must be considered to ensure improved safety and effectiveness. Here, we assess the status and recent development of biological-based radiopharmaceuticals, with a focus on peptides and antibody fragments. Challenges in radiopharmaceutical design range from target selection, vector design, choice of radionuclides and associated radiochemistry. Dosimetry estimation, and the assessment of mechanisms to increase tumor uptake while reducing off-target exposure are discussed.
Collapse
Affiliation(s)
- Cyprine Neba Funeh
- Laboratory for In Vivo Cellular and Molecular Imaging, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K.001, 1090 Brussels, Belgium
| | - Jessica Bridoux
- Laboratory for In Vivo Cellular and Molecular Imaging, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K.001, 1090 Brussels, Belgium
| | - Thomas Ertveldt
- Laboratory for Molecular and Cellular Therapy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Timo W M De Groof
- Laboratory for In Vivo Cellular and Molecular Imaging, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K.001, 1090 Brussels, Belgium
| | - Dora Mugoli Chigoho
- Laboratory for In Vivo Cellular and Molecular Imaging, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K.001, 1090 Brussels, Belgium
| | - Parinaz Asiabi
- Laboratory for In Vivo Cellular and Molecular Imaging, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K.001, 1090 Brussels, Belgium
| | - Peter Covens
- Laboratory for In Vivo Cellular and Molecular Imaging, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K.001, 1090 Brussels, Belgium
| | - Matthias D'Huyvetter
- Laboratory for In Vivo Cellular and Molecular Imaging, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K.001, 1090 Brussels, Belgium
| | - Nick Devoogdt
- Laboratory for In Vivo Cellular and Molecular Imaging, Department of Medical Imaging, Vrije Universiteit Brussel, Laarbeeklaan 103/K.001, 1090 Brussels, Belgium
| |
Collapse
|
31
|
Fan Y, Pan D, Yang M, Wang X. Radiolabelling and in vivo radionuclide imaging tracking of emerging pollutants in environmental toxicology: A review. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 866:161412. [PMID: 36621508 DOI: 10.1016/j.scitotenv.2023.161412] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 12/27/2022] [Accepted: 01/02/2023] [Indexed: 06/17/2023]
Abstract
Emerging pollutants (EPs) have become a global concern, attracting tremendous attention because of serious threats to human and animal health. EP diversity emanates from their behaviour and ability to enter the body via multiple pathways and exhibit completely different distribution, transport, and excretion. To better understand the in vivo behaviour of EPs, we reviewed radiolabelling and in vivo radionuclide imaging tracking of various EPs, including micro- and nano-plastics, perfluoroalkyl substances, metal oxides, pharmaceutical and personal care products, and so on. Because this accurate and quantitative imaging approach requires the labelling of radionuclides onto EPs, the main strategies for radiolabelling were reviewed, such as synthesis with radioactive precursors, element exchange, proton beam activation, and modification. Spatial and temporal biodistribution of various EPs was summarised in a heat map, revealing that the absorption, transport, and excretion of EPs are markedly related to their type, size, and pathway into the body. These findings implicate the potential toxicity of diverse EPs in organs and tissues. Finally, we discussed the potential and challenges of radionuclide imaging tracking of EPs, which can be considered in future EPs studies.
Collapse
Affiliation(s)
- Yeli Fan
- School of Environmental Engineering, Wuxi University, Wuxi 214105, PR China
| | - Donghui Pan
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, PR China
| | - Min Yang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, PR China
| | - Xinyu Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, PR China.
| |
Collapse
|
32
|
Benfante V, Stefano A, Ali M, Laudicella R, Arancio W, Cucchiara A, Caruso F, Cammarata FP, Coronnello C, Russo G, Miele M, Vieni A, Tuttolomondo A, Yezzi A, Comelli A. An Overview of In Vitro Assays of 64Cu-, 68Ga-, 125I-, and 99mTc-Labelled Radiopharmaceuticals Using Radiometric Counters in the Era of Radiotheranostics. Diagnostics (Basel) 2023; 13:diagnostics13071210. [PMID: 37046428 PMCID: PMC10093267 DOI: 10.3390/diagnostics13071210] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Revised: 03/11/2023] [Accepted: 03/17/2023] [Indexed: 04/14/2023] Open
Abstract
Radionuclides are unstable isotopes that mainly emit alpha (α), beta (β) or gamma (γ) radiation through radiation decay. Therefore, they are used in the biomedical field to label biomolecules or drugs for diagnostic imaging applications, such as positron emission tomography (PET) and/or single-photon emission computed tomography (SPECT). A growing field of research is the development of new radiopharmaceuticals for use in cancer treatments. Preclinical studies are the gold standard for translational research. Specifically, in vitro radiopharmaceutical studies are based on the use of radiopharmaceuticals directly on cells. To date, radiometric β- and γ-counters are the only tools able to assess a preclinical in vitro assay with the aim of estimating uptake, retention, and release parameters, including time- and dose-dependent cytotoxicity and kinetic parameters. This review has been designed for researchers, such as biologists and biotechnologists, who would like to approach the radiobiology field and conduct in vitro assays for cellular radioactivity evaluations using radiometric counters. To demonstrate the importance of in vitro radiopharmaceutical assays using radiometric counters with a view to radiogenomics, many studies based on 64Cu-, 68Ga-, 125I-, and 99mTc-labeled radiopharmaceuticals have been revised and summarized in this manuscript.
Collapse
Affiliation(s)
- Viviana Benfante
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), 90015 Cefalù, Italy
| | - Alessandro Stefano
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), 90015 Cefalù, Italy
| | - Muhammad Ali
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy
| | | | - Walter Arancio
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy
| | - Antonino Cucchiara
- Department of Diagnostic and Therapeutic Services, IRCCS-ISMETT (Mediterranean Institute for Transplantation and Advanced Specialized Therapies), Via Tricomi 5, 90127 Palermo, Italy
| | - Fabio Caruso
- Department of Diagnostic and Therapeutic Services, IRCCS-ISMETT (Mediterranean Institute for Transplantation and Advanced Specialized Therapies), Via Tricomi 5, 90127 Palermo, Italy
| | - Francesco Paolo Cammarata
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), 90015 Cefalù, Italy
| | - Claudia Coronnello
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Giorgio Russo
- Institute of Molecular Bioimaging and Physiology, National Research Council (IBFM-CNR), 90015 Cefalù, Italy
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| | - Monica Miele
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy
| | - Alessandra Vieni
- Department of Diagnostic and Therapeutic Services, IRCCS-ISMETT (Mediterranean Institute for Transplantation and Advanced Specialized Therapies), Via Tricomi 5, 90127 Palermo, Italy
| | - Antonino Tuttolomondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy
| | - Anthony Yezzi
- Department of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Albert Comelli
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy
- National Biodiversity Future Center (NBFC), 90133 Palermo, Italy
| |
Collapse
|
33
|
Chakraborty K, Mondal J, An JM, Park J, Lee YK. Advances in Radionuclides and Radiolabelled Peptides for Cancer Therapeutics. Pharmaceutics 2023; 15:pharmaceutics15030971. [PMID: 36986832 PMCID: PMC10054444 DOI: 10.3390/pharmaceutics15030971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023] Open
Abstract
Radiopharmaceutical therapy, which can detect and treat tumours simultaneously, was introduced more than 80 years ago, and it has changed medical strategies with respect to cancer. Many radioactive radionuclides have been developed, and functional, molecularly modified radiolabelled peptides have been used to produce biomolecules and therapeutics that are vastly utilised in the field of radio medicine. Since the 1990s, they have smoothly transitioned into clinical application, and as of today, a wide variety of radiolabelled radionuclide derivatives have been examined and evaluated in various studies. Advanced technologies, such as conjugation of functional peptides or incorporation of radionuclides into chelating ligands, have been developed for advanced radiopharmaceutical cancer therapy. New radiolabelled conjugates for targeted radiotherapy have been designed to deliver radiation directly to cancer cells with improved specificity and minimal damage to the surrounding normal tissue. The development of new theragnostic radionuclides, which can be used for both imaging and therapy purposes, allows for more precise targeting and monitoring of the treatment response. The increased use of peptide receptor radionuclide therapy (PRRT) is also important in the targeting of specific receptors which are overexpressed in cancer cells. In this review, we provide insights into the development of radionuclides and functional radiolabelled peptides, give a brief background, and describe their transition into clinical application.
Collapse
Affiliation(s)
- Kushal Chakraborty
- Department of IT and Energy Convergence (BK21 FOUR), Korea National University of Transportation, Chungju 27469, Republic of Korea
| | - Jagannath Mondal
- Department of Green Bio Engineering, Graduate School, Korea National University of Transportation, Chungju 27469, Republic of Korea
- 4D Convergence Technology Institute, Korea National University of Transportation, Jeungpyeong 27909, Republic of Korea
| | - Jeong Man An
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Jooho Park
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Republic of Korea
- Research Institute for Biomedical & Health Science, Konkuk University, Chungju 27478, Republic of Korea
- Correspondence: (J.P.); (Y.-K.L.); Tel.: +82-43-841-5224 (Y.-K.L.)
| | - Yong-Kyu Lee
- Department of Green Bio Engineering, Graduate School, Korea National University of Transportation, Chungju 27469, Republic of Korea
- 4D Convergence Technology Institute, Korea National University of Transportation, Jeungpyeong 27909, Republic of Korea
- Correspondence: (J.P.); (Y.-K.L.); Tel.: +82-43-841-5224 (Y.-K.L.)
| |
Collapse
|
34
|
Mixdorf JC, Hoffman SLV, Aluicio-Sarduy E, Barnhart TE, Engle JW, Ellison PA. Copper-Mediated Radiobromination of (Hetero)Aryl Boronic Pinacol Esters. J Org Chem 2023; 88:2089-2094. [PMID: 36745853 PMCID: PMC9957949 DOI: 10.1021/acs.joc.2c02420] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A copper-mediated radiobromination of (hetero)aryl boronic pinacol esters is described. Cyclotron-produced [76/77Br]bromide was isolated using an anion exchange cartridge, wherein the pre-equilibration and elution solutions played a critical role in downstream deboro-bromination. The bromination tolerates a broad range of functional groups, labeling molecules with ranging electronic and steric effects. Bologically active radiopharmaceuticals were synthesized, including two radiobrominated inhibitors of poly ADP ribose polymerase, a clinically relevant chemotherapeutic target for ovarian, breast, and prostate cancers.
Collapse
Affiliation(s)
- Jason C. Mixdorf
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, 1111 Highland Avenue Madison, WI 53705
| | - Sabrina L. V. Hoffman
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, 1111 Highland Avenue Madison, WI 53705
| | - Eduardo Aluicio-Sarduy
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, 1111 Highland Avenue Madison, WI 53705
| | - Todd E. Barnhart
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, 1111 Highland Avenue Madison, WI 53705
| | - Jonathan W. Engle
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, 1111 Highland Avenue Madison, WI 53705
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, 1111 Highland Avenue Madison, WI 53705
| | - Paul A. Ellison
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, 1111 Highland Avenue Madison, WI 53705
| |
Collapse
|
35
|
Larenkov A, Mitrofanov I, Pavlenko E, Rakhimov M. Radiolysis-Associated Decrease in Radiochemical Purity of 177Lu-Radiopharmaceuticals and Comparison of the Effectiveness of Selected Quenchers against This Process. Molecules 2023; 28:molecules28041884. [PMID: 36838872 PMCID: PMC9967390 DOI: 10.3390/molecules28041884] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
The radiolytic degradation of vector molecules is a major factor affecting the shelf life of therapeutic radiopharmaceuticals. The development of time-stable dosage forms of radiopharmaceuticals is the key to their successful implementation in clinical practice. Using [177Lu]Lu-PSMA-617 molecule as an example, the time dependence of the change in radiochemical purity (RCP, %) under radiolysis conditions was studied. The dependence of [177Lu]Lu-PSMA-617 radiolysis on parameters such as time, radionuclide activity, buffer agent concentration, precursor amount, and preparation volume was evaluated. It was shown that the absorbed dose was the dominant factor influencing the RCP. The RCP value is inversely proportional to the absorbed dose in the [177Lu]Lu-PSMA-617 preparation and has an exponential dependence. The lutetium-177 dose factor ψ (Gy·mL·MBq-1) and PSMA-617 concentration-dependent dose constant κ (Gy-1) were evaluated for absorbed dose estimation via computer modeling, chemical dosimetry, and radiochemical purity monitoring under various conditions. The further refinement and application of the dependencies found can be useful for predicting the RCP value at the stage of optimizing the composition of the finished dosage form of therapeutic radiopharmaceuticals. The influence of the buffer agent (sodium acetate) concentration on [177Lu]Lu-PSMA-617 radiolytic degradation was shown and should be considered both when developing a dosage form, and when comparing the results of independent studies. The effectiveness of the addition of various stabilizing agents, such as DMSA, cysteine, gentisic acid, vanillin, methionine, adenine, dobesilic acid, thymine, uracil, nicotinamide, meglumine, and mannitol, in suppressing the effects of radiolysis was evaluated.
Collapse
|
36
|
Pedersen KS, Deville C, Søndergaard U, Jensen M, Jensen AI. Improved procedures for production and purification of 135La from enriched [ 135Ba]BaCO 3 on a 16.5 MeV cyclotron. Appl Radiat Isot 2023; 192:110612. [PMID: 36521259 DOI: 10.1016/j.apradiso.2022.110612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/19/2022] [Accepted: 12/08/2022] [Indexed: 12/13/2022]
Abstract
Lanthanum-135 (135La) is a favorable Auger electron emitter with a high Auger electron yield and low gamma emission, making it promising for Auger electron radiotherapy. However, successful application requires reliable and scalable 135La production. Up to now, metallic natural barium (natBa) is a commonly used target material, but this material is sensitive to moisture and oxidation. BaCO3 has also been tested, due to its higher chemical stability. However, BaCO3 has poor thermal conductivity, limiting the applicable current and making high yield production challenging. In this study, we pressed a mixture of enriched [135Ba]BaCO3 and fine aluminum (Al) powder to provide a stable target with improved thermal conductivity compared to pure BaCO3. After 4 h of irradiation with a 16.5 MeV proton beam at 20 μA current, 1.62 ± 0.18 GBq was produced from a 200 mg [135Ba]BaCO3:Al (1:2, w/w) target. This corresponded to a saturation yield of 11.91 ± 1.31 GBq (or 596 ± 66 MBq/μA). A purification procedure involving initial precipitation, followed by a single composite column containing a layer of TK200 resin and a second layer of branched DGA resin was developed, with 97.1 ± 3.6 % decay corrected 135La recovery. [135La]LaCl3 was obtained in an effective molar activity of 79.6 ± 25.3 MBq/nmol (DOTA titration), 104.0 ± 40.4 MBq/nmol (DTPA titration) and 186.5 ± 83.8 MBq/nmol (CHX-A″-DTPA titration), and a radionuclidic purity (RNP) of >99.9 % at end of purification, hereby demonstrating a purity suitable for radiopharmaceutical use.
Collapse
Affiliation(s)
- Kristina Søborg Pedersen
- The Hevesy Laboratory, Department of Health Technology, Technical University of Denmark, Frederiksborgvej 399, Building 202, 4000, Roskilde, Denmark
| | - Claire Deville
- The Hevesy Laboratory, Department of Health Technology, Technical University of Denmark, Frederiksborgvej 399, Building 202, 4000, Roskilde, Denmark
| | - Ursula Søndergaard
- University Hospital of North Norway, Sykehusvegen 38, 9019, Tromsø, Norway; Arctic University of Norway, Hansine Hansens veg 18, 9019, Tromsø, Norway
| | - Mikael Jensen
- The Hevesy Laboratory, Department of Health Technology, Technical University of Denmark, Frederiksborgvej 399, Building 202, 4000, Roskilde, Denmark.
| | - Andreas I Jensen
- The Hevesy Laboratory, Department of Health Technology, Technical University of Denmark, Frederiksborgvej 399, Building 202, 4000, Roskilde, Denmark.
| |
Collapse
|
37
|
Microfluidic Solvent Extraction of No-Carrier-Added 64Cu from Irradiated Zn target for Radiopharmaceutical Preparation. CHEMICAL ENGINEERING JOURNAL ADVANCES 2022. [DOI: 10.1016/j.ceja.2022.100433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
38
|
Galbiati A, Zana A, Bocci M, Millul J, Elsayed A, Mock J, Neri D, Cazzamalli S. A Dimeric FAP-Targeting Small-Molecule Radioconjugate with High and Prolonged Tumor Uptake. J Nucl Med 2022; 63:1852-1858. [PMID: 35589404 PMCID: PMC9730928 DOI: 10.2967/jnumed.122.264036] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 05/10/2022] [Indexed: 01/11/2023] Open
Abstract
Imaging procedures based on small-molecule radioconjugates targeting fibroblast activation protein (FAP) have recently emerged as a powerful tool for the diagnosis of a wide variety of tumors. However, the therapeutic potential of radiolabeled FAP-targeting agents is limited by their short residence time in neoplastic lesions. In this work, we present the development and in vivo characterization of BiOncoFAP, a new dimeric FAP-binding motif with an extended tumor residence time and favorable tumor-to-organ ratio. Methods: The binding properties of BiOncoFAP and its monovalent OncoFAP analog were assayed against recombinant human FAP. Preclinical experiments with 177Lu-OncoFAP-DOTAGA (177Lu-OncoFAP) and 177Lu-BiOncoFAP-DOTAGA (177Lu-BiOncoFAP) were performed on mice bearing FAP-positive HT-1080 tumors. Results: OncoFAP and BiOncoFAP displayed comparable subnanomolar dissociation constants toward recombinant human FAP in solution, but the bivalent BiOncoFAP bound more avidly to the target immobilized on solid supports. In a comparative biodistribution study, 177Lu-BiOncoFAP exhibited a more stable and prolonged tumor uptake than 177Lu-OncoFAP (∼20 vs. ∼4 percentage injected dose/g, respectively, at 24 h after injection). Notably, 177Lu-BiOncoFAP showed favorable tumor-to-organ ratios with low kidney uptake. Both 177Lu-OncoFAP and 177Lu-BiOncoFAP displayed potent antitumor efficacy when administered at therapeutic doses to tumor-bearing mice. Conclusion: 177Lu-BiOncoFAP is a promising candidate for radioligand therapy of cancer, with favorable in vivo tumor-to-organ ratios, a long tumor residence time, and potent anticancer efficacy.
Collapse
Affiliation(s)
- Andrea Galbiati
- Research and Development Department, Philochem AG, Otelfingen, Switzerland
| | - Aureliano Zana
- Research and Development Department, Philochem AG, Otelfingen, Switzerland
| | - Matilde Bocci
- Research and Development Department, Philochem AG, Otelfingen, Switzerland
| | - Jacopo Millul
- Research and Development Department, Philochem AG, Otelfingen, Switzerland
| | - Abdullah Elsayed
- Research and Development Department, Philochem AG, Otelfingen, Switzerland;,Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, Zurich, Switzerland; and
| | - Jacqueline Mock
- Research and Development Department, Philochem AG, Otelfingen, Switzerland
| | - Dario Neri
- Department of Chemistry and Applied Biosciences, Swiss Federal Institute of Technology, Zurich, Switzerland; and,Philogen S.p.A., Siena, Italy
| | - Samuele Cazzamalli
- Research and Development Department, Philochem AG, Otelfingen, Switzerland
| |
Collapse
|
39
|
Zha Z, Choi SR, Li L, Zhao R, Ploessl K, Yao X, Alexoff D, Zhu L, Kung HF. New PSMA-Targeting Ligands: Transformation from Diagnosis (Ga-68) to Radionuclide Therapy (Lu-177). J Med Chem 2022; 65:13001-13012. [DOI: 10.1021/acs.jmedchem.2c00852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Zhihao Zha
- Five Eleven Pharma Inc., Philadelphia, Pennsylvania 19104, United States
| | - Seok Rye Choi
- Five Eleven Pharma Inc., Philadelphia, Pennsylvania 19104, United States
| | - Linlin Li
- College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Ruiyue Zhao
- College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Karl Ploessl
- Five Eleven Pharma Inc., Philadelphia, Pennsylvania 19104, United States
| | - Xinyue Yao
- College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - David Alexoff
- Five Eleven Pharma Inc., Philadelphia, Pennsylvania 19104, United States
| | - Lin Zhu
- College of Chemistry, Beijing Normal University, Beijing 100875, P. R. China
| | - Hank F. Kung
- Five Eleven Pharma Inc., Philadelphia, Pennsylvania 19104, United States
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
40
|
Kelderman CAA, Davey PRWJ, Ma MT, de Veer M, Salimova E, Donnelly PS, Paterson BM. Hexadentate technetium-99m bis(thiosemicarbazonato) complexes: synthesis, characterisation and biodistribution. Dalton Trans 2022; 51:14064-14078. [PMID: 35822662 DOI: 10.1039/d2dt01264b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The syntheses of non-oxido/non-nitrido bis(thiosemicarbazonato)technetium(V) complexes featuring a series of alkyl and ether substituents is presented. The bis(thiosemicarbazones) were radiolabelled with technetium-99m using an optimised one-pot synthesis from [99mTc][TcO4]-. Mass spectrometry and computational chemistry data suggested a distorted trigonal prismatic coordination environment for the bis(thiosemicarbazonato)technetium(V) complexes by way of a bis(thiosemicarbazone)technetium(V)-oxido intermediate complex. The lipophilicities of the complexes were estimated using distribution ratios and three of the new complexes were investigated in mice using kinetic planar imaging and ex vivo biodistribution experiments and were compared to [99mTc][TcO4]-. Modification of the technetium complexes with various lipophilic functional groups altered the biodistributions of the complexes in mice despite evidence suggesting limited stability of the complexes to biologically relevant conditions. The most hydrophilic complex had higher uptake in the kidneys compared to the most lipophilic, which had higher liver uptake, suggesting modification of the excretion pathways.
Collapse
Affiliation(s)
| | | | - Michelle T Ma
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London SE1 7EH, UK
| | - Michael de Veer
- Monash Biomedical Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - Ekaterina Salimova
- Monash Biomedical Imaging, Monash University, Clayton, Victoria 3800, Australia
| | - Paul S Donnelly
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Brett M Paterson
- School of Chemistry, Monash University, Clayton, Victoria 3800, Australia. .,Monash Biomedical Imaging, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
41
|
Marlin A, Hierlmeier I, Guillou A, Bartholomä M, Tripier R, Patinec V. Bioconjugated chelates based on (methylpyridinyl)tacn: synthesis, 64Cu labeling and in vitro evaluation for prostate cancer targeting. Metallomics 2022; 14:6596882. [PMID: 35648482 DOI: 10.1093/mtomcs/mfac036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/02/2022] [Indexed: 11/13/2022]
Abstract
Three new bifunctional copper chelators based on the 1,4,7-triazacyclononane (tacn) platform have been synthesized and conjugated to peptide. The first one is constituted of the tacn with two methylpyridinyl and one methylthiazolyl carboxylic acid pendant arms, while, in the second and third ones, the macrocycle is functionalized by three methylpyridinyl groups, with an additional hexynoic acid chain on a carbon of one or two pyridine rings. These three bifunctional chelators have been conjugated to the antagonist JMV594 peptide for targeting the gastrin releasing peptide receptor (GRP-r), which is overexpressed in prostate cancer. The resulting monomeric bioconjugates have shown their efficiency to be radiolabeled with β+ emitter 64Cu, and the hydrophilicity and PC-3 cell internalisation properties of these radiolabeled conjugates have been studied. PC-3 cell binding affinity of mono- and dimeric metal-free and natCu metallated conjugates have been evaluated by IC50 measurements. The results demonstrate the potential of these methylpyridinyl tacn derivatives for radiopharmaceutical applications.
Collapse
Affiliation(s)
- Axia Marlin
- Univ Brest, UMR-CNRS 6521 CEMCA, 6 avenue Victor le Gorgeu, 29238 Brest, France
| | - Ina Hierlmeier
- Department of Nuclear Medicine, Saarland University-Medical Center, Kirrbergerstrasse, 66421 Homburg, Germany
| | - Amaury Guillou
- Univ Brest, UMR-CNRS 6521 CEMCA, 6 avenue Victor le Gorgeu, 29238 Brest, France
| | - Mark Bartholomä
- Department of Nuclear Medicine, Saarland University-Medical Center, Kirrbergerstrasse, 66421 Homburg, Germany
| | - Raphaël Tripier
- Univ Brest, UMR-CNRS 6521 CEMCA, 6 avenue Victor le Gorgeu, 29238 Brest, France
| | - Véronique Patinec
- Univ Brest, UMR-CNRS 6521 CEMCA, 6 avenue Victor le Gorgeu, 29238 Brest, France
| |
Collapse
|
42
|
Rösner L, Konken CP, Depke DA, Rentmeister A, Schäfers M. Covalent labeling of immune cells. Curr Opin Chem Biol 2022; 68:102144. [DOI: 10.1016/j.cbpa.2022.102144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/06/2022] [Accepted: 03/11/2022] [Indexed: 12/15/2022]
|
43
|
Holik HA, Ibrahim FM, Elaine AA, Putra BD, Achmad A, Kartamihardja AHS. The Chemical Scaffold of Theranostic Radiopharmaceuticals: Radionuclide, Bifunctional Chelator, and Pharmacokinetics Modifying Linker. Molecules 2022; 27:3062. [PMID: 35630536 PMCID: PMC9143622 DOI: 10.3390/molecules27103062] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/27/2022] [Accepted: 05/05/2022] [Indexed: 11/16/2022] Open
Abstract
Therapeutic radiopharmaceuticals have been researched extensively in the last decade as a result of the growing research interest in personalized medicine to improve diagnostic accuracy and intensify intensive therapy while limiting side effects. Radiometal-based drugs are of substantial interest because of their greater versatility for clinical translation compared to non-metal radionuclides. This paper comprehensively discusses various components commonly used as chemical scaffolds to build radiopharmaceutical agents, i.e., radionuclides, pharmacokinetic-modifying linkers, and chelators, whose characteristics are explained and can be used as a guide for the researcher.
Collapse
Affiliation(s)
- Holis Abdul Holik
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia; (F.M.I.); (A.A.E.); (B.D.P.)
| | - Faisal Maulana Ibrahim
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia; (F.M.I.); (A.A.E.); (B.D.P.)
| | - Angela Alysia Elaine
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia; (F.M.I.); (A.A.E.); (B.D.P.)
| | - Bernap Dwi Putra
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia; (F.M.I.); (A.A.E.); (B.D.P.)
| | - Arifudin Achmad
- Department of Nuclear Medicine and Molecular Theranostics, Faculty of Medicine, Universitas Padjadjaran/Hasan Sadikin General Hospital, Bandung 40161, Indonesia; (A.A.); (A.H.S.K.)
- Oncology and Stem Cell Working Group, Faculty of Medicine, Universitas Padjadjaran, Bandung 40161, Indonesia
| | - Achmad Hussein Sundawa Kartamihardja
- Department of Nuclear Medicine and Molecular Theranostics, Faculty of Medicine, Universitas Padjadjaran/Hasan Sadikin General Hospital, Bandung 40161, Indonesia; (A.A.); (A.H.S.K.)
| |
Collapse
|
44
|
Parrilha GL, dos Santos RG, Beraldo H. Applications of radiocomplexes with thiosemicarbazones and bis(thiosemicarbazones) in diagnostic and therapeutic nuclear medicine. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214418] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
45
|
Usama SM, Marker SC, Hernandez Vargas S, AghaAmiri S, Ghosh SC, Ikoma N, Tran Cao HS, Schnermann MJ, Azhdarinia A. Targeted Dual-Modal PET/SPECT-NIR Imaging: From Building Blocks and Construction Strategies to Applications. Cancers (Basel) 2022; 14:1619. [PMID: 35406390 PMCID: PMC8996983 DOI: 10.3390/cancers14071619] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 02/04/2023] Open
Abstract
Molecular imaging is an emerging non-invasive method to qualitatively and quantitively visualize and characterize biological processes. Among the imaging modalities, PET/SPECT and near-infrared (NIR) imaging provide synergistic properties that result in deep tissue penetration and up to cell-level resolution. Dual-modal PET/SPECT-NIR agents are commonly combined with a targeting ligand (e.g., antibody or small molecule) to engage biomolecules overexpressed in cancer, thereby enabling selective multimodal visualization of primary and metastatic tumors. The use of such agents for (i) preoperative patient selection and surgical planning and (ii) intraoperative FGS could improve surgical workflow and patient outcomes. However, the development of targeted dual-modal agents is a chemical challenge and a topic of ongoing research. In this review, we define key design considerations of targeted dual-modal imaging from a topological perspective, list targeted dual-modal probes disclosed in the last decade, review recent progress in the field of NIR fluorescent probe development, and highlight future directions in this rapidly developing field.
Collapse
Affiliation(s)
- Syed Muhammad Usama
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA; (S.M.U.); (S.C.M.)
| | - Sierra C. Marker
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA; (S.M.U.); (S.C.M.)
| | - Servando Hernandez Vargas
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA; (S.H.V.); (S.A.); (S.C.G.)
| | - Solmaz AghaAmiri
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA; (S.H.V.); (S.A.); (S.C.G.)
| | - Sukhen C. Ghosh
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA; (S.H.V.); (S.A.); (S.C.G.)
| | - Naruhiko Ikoma
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; (N.I.); (H.S.T.C.)
| | - Hop S. Tran Cao
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030, USA; (N.I.); (H.S.T.C.)
| | - Martin J. Schnermann
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA; (S.M.U.); (S.C.M.)
| | - Ali Azhdarinia
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA; (S.H.V.); (S.A.); (S.C.G.)
| |
Collapse
|
46
|
Chakravarty R, Patra S, Jagadeesan K, Thakare S, Chakraborty S. Electrochemical separation of 132/135La theranostic pair from proton irradiated Ba target. Sep Purif Technol 2022. [DOI: 10.1016/j.seppur.2021.119908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
47
|
Brachytherapy Approach Using 177Lu Conjugated Gold Nanostars and Evaluation of Biodistribution, Tumor Retention, Dosimetry and Therapeutic Efficacy in Head and Neck Tumor Model. Pharmaceutics 2021; 13:pharmaceutics13111903. [PMID: 34834318 PMCID: PMC8623985 DOI: 10.3390/pharmaceutics13111903] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/02/2021] [Accepted: 11/05/2021] [Indexed: 12/14/2022] Open
Abstract
Brachytherapy can provide sufficient doses to head and neck squamous cell carcinoma (HNSCC) with minimal damage to nearby normal tissues. In this study, the β--emitter 177Lu was conjugated to DTPA-polyethylene glycol (PEG) decorated gold nanostars (177Lu-DTPA-pAuNS) used in surface-enhanced Raman scattering and photothermal therapy (PTT). The accumulation and therapeutic efficacy of 177Lu-DTPA-pAuNS were compared with those of 177Lu-DTPA on an orthotopic HNSCC tumor model. The SPECT/CT imaging and biodistribution studies showed that 177Lu-DTPA-pAuNS can be accumulated in the tumor up to 15 days, but 177Lu-DTPA could not be detected at 24 h after injection. The tumor viability and growth were suppressed by injected 177Lu-DTPA-pAuNS but not nonconjugated 177Lu-DTPA, as evaluated by bioluminescent imaging. The radiation-absorbed dose of the normal organ was the highest in the liver (0.33 mSv/MBq) estimated in a 73 kg adult, but that of tumorsphere (0.5 g) was 3.55 mGy/MBq, while intravenous injection of 177Lu-DTPA-pAuNS resulted in 1.97 mSv/MBq and 0.13 mGy/MBq for liver and tumorsphere, respectively. We also observed further enhancement of tumor-suppressive effects by a combination of 177Lu-DTPA-pAuNS and PTT compared to 177Lu-DTPA-pAuNS alone. In conclusion, 177Lu-DTPA-pAuNS may be considered as a potential radiopharmaceutical agent for HNSCC brachytherapy.
Collapse
|
48
|
Rondon A, Rouanet J, Degoul F. Radioimmunotherapy in Oncology: Overview of the Last Decade Clinical Trials. Cancers (Basel) 2021; 13:cancers13215570. [PMID: 34771732 PMCID: PMC8583425 DOI: 10.3390/cancers13215570] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/26/2021] [Accepted: 11/05/2021] [Indexed: 12/17/2022] Open
Abstract
Simple Summary Monoclonal antibody-bearing radionuclides have been under clinical investigation over the last two decades for their use in theranostic (diagnostic and therapeutic) applications in cancer. However, despite the numerous trials that have been conducted, only two radioimmunotherapies (RIT) have been approved by the FDA for the targeted therapy of hematologic tumors expressing CD20 antigens. Moreover, RIT applications for solid cancers faced major issues—such as radiotoxicity due to low antibodies penetrance requiring substantial curative dose—where new discoveries concerning antibody engineering or radionuclides are trying to overcome. Here, we performed an overview of the last 11-year clinical trials involving RIT for solid and non-solid cancers conducted either with full antibodies or antibody fragments. We discussed the low-to-moderate efficiency of RIT compared to conventional therapies and described the last advances in clinic for antibodies carriers (F(ab′)2, Fab′, ScFv). Finally, we discussed about the complexity of RIT as a therapy and depicted both the issues and the prospects of such a strategy. Abstract The specific irradiation of tumors with selective radiolabeled antibodies constitutes an attractive therapeutic approach. Consequent preclinical research has been conducted by both biologists to identify pertinent targets and to select corresponding antibodies (mAb) and by radiochemists to radiolabel mAbs. These numerous preclinical investigations have ascertained the therapeutic interest of radioimmunotherapy (RIT) protocols in mice models. Here, we summarize the clinical studies that have been performed the last decade, including clinical trials (phases I, II, and III), prospective and retrospective studies, and cases series. We thereby reported 92 clinical studies. Among them, 62 concern the treatment of hematological malignancies, and 30 concern solid tumors. For hematologic diseases, the analysis was complex due to the high discrepancy of therapeutic strategies (first-line therapy, consolidation, stem cell transplantation conditioning) as well as the high variety of malignancies that were treated. The clinical studies from the last decade failed to expand anti-CD20 RIT indications but confirmed that RIT using radiolabeled anti-CD20 remains a pertinent choice for patients with relapse follicular lymphomas. For solid tumors, the positive benefit of RIT is more mitigated, apart for few malignancies that can be treated locally. Clinical trials also demonstrated the potential of some antibody formats, such as F(ab′)2, which has already been approved by the China State FDA under the trend name Licartin®. Despite disparate results, mAb fragments are an interesting prospect for the improvement of RIT efficiency as well as for pretargeted strategies that delay the injection of radioactive treatments from the mAb ones.
Collapse
Affiliation(s)
- Aurélie Rondon
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, UCLouvain, BE-1200 Brussels, Belgium
- Correspondence: (A.R.); (F.D.)
| | - Jacques Rouanet
- Imagerie Moléculaire et Stratégies Théranostiques, Inserm UMR1240, Université Clermont-Auvergne, F-63000 Clermont-Ferrand, France;
- Service de Dermatologie et d’Oncologie Cutanée, CHU Estaing, F-63011 Clermont-Ferrand, France
| | - Françoise Degoul
- CNRS 6293, INSERM U1103, GReD, Centre de Recherche et de Biologie Clinique, Université Clermont-Auvergne, F-63000 Clermont-Ferrand, France
- Correspondence: (A.R.); (F.D.)
| |
Collapse
|