1
|
Mizuno Y, Suebboonprathueng T, Onoe S, Akizawa H, Nishijima KI, Takahashi K, Kuge Y. Design of a Tetravalent RGD Peptide Capable of Simultaneous Binding with Multiple Integrin αvβ3 for Targeted Radionuclide Therapy. J Med Chem 2025; 68:6518-6533. [PMID: 40083181 DOI: 10.1021/acs.jmedchem.4c03007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
For targeted radionuclide therapy, radioligands that exhibit high and persistent tumor uptake are indispensable. We previously synthesized a 99mTc-labeled hexavalent RGD peptide (99mTc-(RGD)6) as a tumor imaging agent targeting integrin αvβ3. 99mTc-(RGD)6 showed high in vivo tumor uptake with long retention due to simultaneous binding to multiple integrin αvβ3 receptors. The purpose of this study was to apply this finding to the design of a multivalent RGD peptide labeled with 211At, a promising α-emitting radionuclide for radionuclide therapy. As a candidate compound, a tetravalent RGD peptide (H2N-(RGD)4) was synthesized and radiolabeled with 125I, a homologous element of At, for basic studies. As expected, 125I-(RGD)4 retained the capability of simultaneous binding and showed comparable in vivo tumor uptake to 99mTc-(RGD)6. Finally, 211At-(RGD)4 was synthesized with >95% radiochemical purity and exhibited an almost identical biodistribution pattern to 125I-(RGD)4. These results indicate that 211At-(RGD)4 might be a potential radioligand for integrin αvβ3-targeted radionuclide therapy.
Collapse
Affiliation(s)
- Yuki Mizuno
- Central Institute of Isotope Science, Hokkaido University, Sapporo 060-0815, Japan
- Global Center for Biomedical Science and Engineering, Hokkaido University, Sapporo 060-8638, Japan
| | | | - Satoru Onoe
- Laboratory of Physical Chemistry, Showa Pharmaceutical University, Machida 194-8543, Japan
| | - Hiromichi Akizawa
- Laboratory of Physical Chemistry, Showa Pharmaceutical University, Machida 194-8543, Japan
| | - Ken-Ichi Nishijima
- Advanced Clinical Research Center, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Kazuhiro Takahashi
- Advanced Clinical Research Center, Fukushima Medical University, Fukushima 960-1295, Japan
| | - Yuji Kuge
- Central Institute of Isotope Science, Hokkaido University, Sapporo 060-0815, Japan
- Global Center for Biomedical Science and Engineering, Hokkaido University, Sapporo 060-8638, Japan
| |
Collapse
|
2
|
Jin ZH, Degardin M, Furukawa T, Uehara T, Tsuji AB, Suzuki H, Wakizaka H, Sugyo A, Aung W, Suzuki H, Nagatsu K, Zhang MR, Dumy P, Boturyn D, Higashi T. Evaluation of the Gly-Phe-Lys Linker to Reduce the Renal Radioactivity of a [ 64Cu]Cu-Labeled Multimeric cRGD Peptide. ACS OMEGA 2025; 10:4102-4120. [PMID: 39926504 PMCID: PMC11799997 DOI: 10.1021/acsomega.4c10621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/02/2025] [Accepted: 01/08/2025] [Indexed: 02/11/2025]
Abstract
Radiometal-labeled peptide-based radiopharmaceuticals (RLPB-radiopharmaceuticals) are promising for cancer imaging and targeted radiotherapy; however, their effectiveness is often compromised by the high retention of nonspecific radioactivity in the kidneys due to renal excretion pathways. Current strategies to address this issue have limitations, highlighting the need for innovative approaches to improve targeting specificity and therapeutic efficacy. We aimed to evaluate the applicability of the Gly-Phe-Lys (GFK) tripeptide, a renal brush border (RBB) enzyme-cleavable linkage, to reduce renal radioactivity in RLPB-radiopharmaceuticals using the integrin-targeting radiopeptide [64Cu]Cu-cyclam-RAFT-c(-RGDfK-)4 ([64Cu]Cu-cyclam-RaftRGD). We designed and synthesized the model compound [64Cu]Cu-cyclam-GFK(benzoyl [Bz]), its predictive metabolites, and GFK-incorporated [64Cu]Cu-cyclam-RaftRGD derivatives [64Cu]Cu-cyclam-GFK-RaftRGD and [64Cu]Cu-cyclam-GFK(beta-alanine [βA])3-RaftRGD. In vitro studies showed that dual radiometabolites, namely, [64Cu]Cu-cyclam-G and [64Cu]Cu-cyclam-GF, were simultaneously released from [64Cu]Cu-cyclam-GFK(Bz) by different RBB enzymes, whereas both RaftRGD derivatives released only [64Cu]Cu-cyclam-GF. When injected into mice, [64Cu]Cu-cyclam-GFK(Bz) and the two RaftRGD derivatives led to the urinary excretion of [64Cu]Cu-cyclam-G and [64Cu]Cu-cyclam-GF, respectively. PET imaging and biodistribution studies showed the increased rates of reduction in renal radioactivity levels for the two RaftRGD derivatives compared to the parental [64Cu]Cu-cyclam-RaftRGD (e.g., PET: 1 to 24 h postinjection, 73.0 ± 2.3 and 75.6 ± 1.8 vs 43.0 ± 4.5%, p < 0.0001; biodistribution: 3 to 24 h, 61.1 and 74.4 vs 22.8%). Taken together, these results indicate that the designed renal cleavage occurred in vivo. We also noted the steric interference of the RaftRGD moiety on enzyme access, the spacer effect of the trimeric βA sequence (reduced steric hindrance), and the altered radiopharmacokinetics (e.g., initially increased renal accumulation) of the RaftRGD compounds upon linker incorporation. These findings provide important insights into the chemical design of RLPB-radiopharmaceuticals with reduced renal retention based on the RBB strategy.
Collapse
Affiliation(s)
- Zhao-Hui Jin
- Department
of Molecular Imaging and Theranostics, Institute for Quantum Medical
Science, National Institutes for Quantum
Science and Technology (QST), Chiba 263-8555, Japan
| | - Mélissa Degardin
- Département
de Chimie Moléculaire, CNRS, Université
Grenoble Alpes, cedex 9, Grenoble 38058, France
| | - Takako Furukawa
- Department
of Molecular Imaging and Theranostics, Institute for Quantum Medical
Science, National Institutes for Quantum
Science and Technology (QST), Chiba 263-8555, Japan
- Department
of Integrated Health Sciences, Graduate School of Medicine, Nagoya University, Nagoya 461-8673, Japan
| | - Tomoya Uehara
- Laboratory
of Molecular Imaging and Radiotherapy, Graduate School of Pharmaceutical
Sciences, Chiba University, Chiba 260-8675, Japan
| | - Atsushi B. Tsuji
- Department
of Molecular Imaging and Theranostics, Institute for Quantum Medical
Science, National Institutes for Quantum
Science and Technology (QST), Chiba 263-8555, Japan
| | - Hiroyuki Suzuki
- Laboratory
of Molecular Imaging and Radiotherapy, Graduate School of Pharmaceutical
Sciences, Chiba University, Chiba 260-8675, Japan
| | - Hidekatsu Wakizaka
- Department
of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical
Science, National Institutes for Quantum
Science and Technology (QST), Chiba 263-8555, Japan
| | - Aya Sugyo
- Department
of Molecular Imaging and Theranostics, Institute for Quantum Medical
Science, National Institutes for Quantum
Science and Technology (QST), Chiba 263-8555, Japan
| | - Winn Aung
- Department
of Molecular Imaging and Theranostics, Institute for Quantum Medical
Science, National Institutes for Quantum
Science and Technology (QST), Chiba 263-8555, Japan
| | - Hisashi Suzuki
- Department
of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical
Science, National Institutes for Quantum
Science and Technology (QST), Chiba 263-8555, Japan
| | - Kotaro Nagatsu
- Department
of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical
Science, National Institutes for Quantum
Science and Technology (QST), Chiba 263-8555, Japan
| | - Ming-Rong Zhang
- Department
of Advanced Nuclear Medicine Sciences, Institute for Quantum Medical
Science, National Institutes for Quantum
Science and Technology (QST), Chiba 263-8555, Japan
| | - Pascal Dumy
- Institut
des Biomolécules Max Mousseron, École Nationale Supérieure
de Chimie de Montpellier, Université
de Montpellier, cedex 5, Montpellier 34296, France
| | - Didier Boturyn
- Département
de Chimie Moléculaire, CNRS, Université
Grenoble Alpes, cedex 9, Grenoble 38058, France
| | - Tatsuya Higashi
- Department
of Molecular Imaging and Theranostics, Institute for Quantum Medical
Science, National Institutes for Quantum
Science and Technology (QST), Chiba 263-8555, Japan
| |
Collapse
|
3
|
Stangl S, Nguyen NT, Brosch-Lenz J, Šimeček J, Weber WA, Kossatz S, Notni J. Efficiency of succinylated gelatin and amino acid infusions for kidney uptake reduction of radiolabeled αvβ6-integrin targeting peptides: considerations on clinical safety profiles. Eur J Nucl Med Mol Imaging 2024; 51:3191-3201. [PMID: 38717591 PMCID: PMC11369040 DOI: 10.1007/s00259-024-06738-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 04/26/2024] [Indexed: 09/03/2024]
Abstract
PURPOSE 68Ga-Trivehexin is an investigational PET radiopharmaceutical (NCT05799274) targeting αvβ6-integrin for PET imaging of carcinomas. 177Lu-D0301 is a structurally related therapeutic peptide tetramer. However, it showed considerable kidney uptake in rodents, impeding clinical applicability. We therefore evaluated the impact of different kidney protection strategies on the biodistribution of both agents in normal and tumor-bearing mice. METHODS Ex-vivo biodistribution of 68Ga-Trivehexin (90 min p.i.) and 177Lu-D0301 (90 min and 24 h p.i.) was determined in healthy C57BL/6N and H2009 (human lung adenocarcinoma) xenografted CB17-SCID mice without and with co-infusion of 100 µL of solutions containing 2.5% arginine + 2.5% lysine (Arg/Lys), 4% succinylated gelatin (gelofusine, gelo), or combinations thereof. Arg/Lys was injected either i.p. 30 min before and after the radiopharmaceutical, or i.v. 2 min before the radiopharmaceutical. Gelo was administered either i.v. 2 min prior activity, or pre-mixed and injected together with the radiopharmaceutical (n = 5 per group). C57BL/6N mice were furthermore imaged by PET (90 min p.i.) and SPECT (24 h p.i.). RESULTS Kidney uptake of 68Ga-Trivehexin in C57BL/6N mice was reduced by 15% (Arg/Lys i.p.), 25% (Arg/Lys i.v.), and 70% (gelo i.v.), 90 min p.i., relative to control. 177Lu-D0301 kidney uptake was reduced by 2% (Arg/Lys i.p.), 41% (Arg/Lys i.v.), 61% (gelo i.v.) and 66% (gelo + Arg/Lys i.v.) 24 h p.i., compared to control. Combination of Arg/Lys and gelo provided no substantial benefit. Gelo furthermore reduced kidney uptake of 177Lu-D0301 by 76% (90 min p.i.) and 85% (24 h p.i.) in H2009 bearing SCID mice. Since tumor uptake was not (90 min p.i.) or only slightly reduced (15%, 24 h p.i.), the tumor/kidney ratio was improved by factors of 3.3 (90 min p.i.) and 2.6 (24 h p.i.). Reduction of kidney uptake was demonstrated by SPECT, which also showed that the remaining activity was located in the cortex. CONCLUSIONS The kidney uptake of both investigated radiopharmaceuticals was more efficiently reduced by gelofusine (61-85%) than Arg/Lys (25-41%). Gelofusine appears particularly suitable for reducing renal uptake of αvβ6-integrin targeted 177Lu-labeled peptide multimers because its application led to approximately three times higher tumor-to-kidney ratios. Since the incidence of severe adverse events (anaphylaxis) with succinylated gelatin products (reportedly 0.0062-0.038%) is comparable to that of gadolinium-based MRI or iodinated CT contrast agents (0.008% and 0.04%, respectively), clinical use of gelofusine during radioligand therapy appears feasible if similar risk management strategies as for contrast agents are applied.
Collapse
Affiliation(s)
- Stefan Stangl
- Department of Nuclear Medicine, University Hospital Klinikum Rechts Der Isar, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Nghia Trong Nguyen
- Department of Nuclear Medicine, University Hospital Klinikum Rechts Der Isar, School of Medicine and Health, Technical University of Munich, Munich, Germany
- Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Julia Brosch-Lenz
- Department of Nuclear Medicine, University Hospital Klinikum Rechts Der Isar, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | | | - Wolfgang A Weber
- Department of Nuclear Medicine, University Hospital Klinikum Rechts Der Isar, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Susanne Kossatz
- Department of Nuclear Medicine, University Hospital Klinikum Rechts Der Isar, School of Medicine and Health, Technical University of Munich, Munich, Germany.
- Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine and Health, Technical University of Munich, Munich, Germany.
- Department of Chemistry, School of Natural Sciences, Technical University of Munich, Munich, Germany.
| | - Johannes Notni
- TRIMT GmbH, Radeberg, Germany.
- Institute of Pathology, School of Medicine and Health, Technische Universität München, München, Germany.
| |
Collapse
|
4
|
Ollier C, Méndez-Ardoy A, Ortega-Caballero F, Jiménez-Blanco JL, Le Bris N, Tripier R. Extending the Scope of the C-Functionalization of Cyclam via Copper(I)-Catalyzed Alkyne-Azide Cycloaddition to Bifunctional Chelators of Interest. J Org Chem 2024; 89:5988-5999. [PMID: 38602478 DOI: 10.1021/acs.joc.3c02854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
Cyclam, known for its potent chelation properties, is explored for diverse applications through selective N-functionalization, offering versatile ligands for catalysis, medical research, and materials science. The challenges arising from N-alkylation, which could decrease the coordination properties, are addressed by introducing a robust C-functionalization method. The facile two-step synthesis proposed here involves the click chemistry-based C-functionalization of a hydroxyethyl cyclam derivative using Cu(I)-catalyzed alkyne-azide cycloaddition (CuAAC). Boc-protecting groups prevent undesired copper coordination, resulting in compounds with a wide range of functionalities. The optimized synthesis conditions enable C-functional cyclams to be obtained easily and advantageously, with high application potential in the previously cited fields. The methodology has been extended to trehalose-based Siamese twin amphiphiles, enabling efficient gene delivery applications.
Collapse
Affiliation(s)
- Cédric Ollier
- Univ Brest, UMR-CNRS 6521 CEMCA, 6 avenue Victor le Gorgeu, 29238 Brest, France
| | - Alejandro Méndez-Ardoy
- Department of Organic Chemistry, Faculty of Chemistry, University of Seville, c/Professor Garcia Gonzalez 1, 41012 Sevilla, Spain
| | - Fernando Ortega-Caballero
- Department of Organic Chemistry, Faculty of Chemistry, University of Seville, c/Professor Garcia Gonzalez 1, 41012 Sevilla, Spain
| | - José L Jiménez-Blanco
- Department of Organic Chemistry, Faculty of Chemistry, University of Seville, c/Professor Garcia Gonzalez 1, 41012 Sevilla, Spain
| | - Nathalie Le Bris
- Univ Brest, UMR-CNRS 6521 CEMCA, 6 avenue Victor le Gorgeu, 29238 Brest, France
| | - Raphaël Tripier
- Univ Brest, UMR-CNRS 6521 CEMCA, 6 avenue Victor le Gorgeu, 29238 Brest, France
| |
Collapse
|
5
|
de Roode KE, Joosten L, Behe M. Towards the Magic Radioactive Bullet: Improving Targeted Radionuclide Therapy by Reducing the Renal Retention of Radioligands. Pharmaceuticals (Basel) 2024; 17:256. [PMID: 38399470 PMCID: PMC10892921 DOI: 10.3390/ph17020256] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 02/07/2024] [Accepted: 02/09/2024] [Indexed: 02/25/2024] Open
Abstract
Targeted radionuclide therapy (TRT) is an emerging field and has the potential to become a major pillar in effective cancer treatment. Several pharmaceuticals are already in routine use for treating cancer, and there is still a high potential for new compounds for this application. But, a major issue for many radiolabeled low-to-moderate-molecular-weight molecules is their clearance via the kidneys and their subsequent reuptake. High renal accumulation of radioactive compounds may lead to nephrotoxicity, and therefore, the kidneys are often the dose-limiting organs in TRT with these radioligands. Over the years, different strategies have been developed aiming for reduced kidney retention and enhanced therapeutic efficacy of radioligands. In this review, we will give an overview of the efforts and achievements of the used strategies, with focus on the therapeutic potential of low-to-moderate-molecular-weight molecules. Among the strategies discussed here is coadministration of compounds that compete for binding to the endocytic receptors in the proximal tubuli. In addition, the influence of altering the molecular design of radiolabeled ligands on pharmacokinetics is discussed, which includes changes in their physicochemical properties and implementation of cleavable linkers or albumin-binding moieties. Furthermore, we discuss the influence of chelator and radionuclide choice on reabsorption of radioligands by the kidneys.
Collapse
Affiliation(s)
- Kim E. de Roode
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands;
- Tagworks Pharmaceuticals, Toernooiveld 1, 6525 ED Nijmegen, The Netherlands
| | - Lieke Joosten
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands;
| | - Martin Behe
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institut, 5232 Villigen, Switzerland
| |
Collapse
|
6
|
Laferriere-Holloway TS, Rios A, Lu Y, Okoro CC, van Dam RM. A rapid and systematic approach for the optimization of radio thin-layer chromatography resolution. J Chromatogr A 2023; 1687:463656. [PMID: 36463649 PMCID: PMC9894532 DOI: 10.1016/j.chroma.2022.463656] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022]
Abstract
Radiopharmaceutical analysis is limited by conventional methods. Radio-HPLC may be inaccurate for some compounds (e.g., 18F-radiopharmaceuticals) due to radionuclide sequester. Radio-TLC is simpler, faster, and detects all species but has limited resolution. Imaging-based readout of TLC plates (e.g., using Cerenkov luminescence imaging) can improve readout resolution, but the underlying chromatographic separation efficiency may be insufficient to resolve chemically similar species such as product and precursor-derived impurities. This study applies a systematic mobile phase optimization method, PRISMA, to improve radio-TLC resolution. The PRISMA method optimizes the mobile phase by selecting the correct solvent, optimizing solvent polarity, and optimizing composition. Without prior knowledge of impurities and by simply observing the separation resolution between a radiopharmaceutical and its nearest radioactive or non-radioactive impurities (observed via UV imaging) for different mobile phases, the PRISMA method enabled the development of high-resolution separation conditions for a wide range of 18F-radiopharmaceuticals ( [18F]PBR-06, [18F]FEPPA, [18F]Fallypride, [18F]FPEB, and [18F]FDOPA). Each optimization required a single batch of crude radiopharmaceutical and a few hours. Interestingly, the optimized TLC method provided greater accuracy (compared to other published TLC methods) in determining the product abundance of one radiopharmaceutical studied in more depth ( [18F]Fallypride) and was capable of resolving a comparable number of species as isocratic radio-HPLC. We used the PRISMA-optimized mobile phase for [18F]FPEB in combination with multi-lane radio-TLC techniques to evaluate reaction performance during high-throughput synthesis optimization of [18F]FPEB. The PRISMA methodology, in combination with high-resolution radio-TLC readout, enables a rapid and systematic approach to achieving high-resolution and accurate analysis of radiopharmaceuticals without the need for radio-HPLC.
Collapse
Affiliation(s)
- Travis S Laferriere-Holloway
- Department of Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA; Crump Institute for Molecular Imaging, UCLA, Los Angeles, CA, USA.
| | - Alejandra Rios
- Physics and Biology in Medicine Interdepartmental Graduate Program, UCLA, Los Angeles, CA, USA; Crump Institute for Molecular Imaging, UCLA, Los Angeles, CA, USA
| | - Yingqing Lu
- Physics and Biology in Medicine Interdepartmental Graduate Program, UCLA, Los Angeles, CA, USA; Crump Institute for Molecular Imaging, UCLA, Los Angeles, CA, USA
| | - Chelsea C Okoro
- Institute for Society and Genetics, UCLA, Los Angeles, CA, USA; Crump Institute for Molecular Imaging, UCLA, Los Angeles, CA, USA
| | - R Michael van Dam
- Department of Molecular & Medical Pharmacology, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA; Physics and Biology in Medicine Interdepartmental Graduate Program, UCLA, Los Angeles, CA, USA; Crump Institute for Molecular Imaging, UCLA, Los Angeles, CA, USA.
| |
Collapse
|
7
|
Multiplexed Imaging Reveals the Spatial Relationship of the Extracellular Acidity-Targeting pHLIP with Necrosis, Hypoxia, and the Integrin-Targeting cRGD Peptide. Cells 2022; 11:cells11213499. [DOI: 10.3390/cells11213499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 10/24/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
pH (low) insertion peptides (pHLIPs) have been developed for cancer imaging and therapy targeting the acidic extracellular microenvironment. However, the characteristics of intratumoral distribution (ITD) of pHLIPs are not yet fully understood. This study aimed to reveal the details of the ITD of pHLIPs and their spatial relationship with other tumor features of concern. The fluorescent dye-labeled pHLIPs were intravenously administered to subcutaneous xenograft mouse models of U87MG and IGR-OV1 expressing αVβ3 integrins (using large necrotic tumors). The αVβ3 integrin-targeting Cy5.5-RAFT-c(-RGDfK-)4 was used as a reference. In vivo and ex vivo fluorescence imaging, whole-tumor section imaging, fluorescence microscopy, and multiplexed fluorescence colocalization analysis were performed. The ITD of fluorescent dye-labeled pHLIPs was heterogeneous, having a high degree of colocalization with necrosis. A direct one-to-one comparison of highly magnified images revealed the cellular localization of pHLIP in pyknotic, karyorrhexis, and karyolytic necrotic cells. pHLIP and hypoxia were spatially contiguous but not overlapping cellularly. The hypoxic region was found between the ITDs of pHLIP and the cRGD peptide and the Ki-67 proliferative activity remained detectable in the pHLIP-accumulated regions. The results provide a better understanding of the characteristics of ITD of pHLIPs, leading to new insights into the theranostic applications of pHLIPs.
Collapse
|
8
|
Hiroyama S, Matsunaga K, Ito M, Iimori H, Tajiri M, Nakano Y, Shimosegawa E, Abe K. Usefulness of 18F-FPP-RGD2 PET in pathophysiological evaluation of lung fibrosis using a bleomycin-induced rat model. Eur J Nucl Med Mol Imaging 2022; 49:4358-4368. [DOI: 10.1007/s00259-022-05908-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 07/07/2022] [Indexed: 11/04/2022]
|
9
|
Special radionuclide production activities – recent developments at QST and throughout Japan. RADIOCHIM ACTA 2022. [DOI: 10.1515/ract-2021-1124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
National Institutes for Quantum Science and Technology (QST), formerly known as the National Institute of Radiological Sciences (NIRS), has been engaged in work on radiopharmaceutical science using cyclotrons since 1974. Eight pioneering researchers founded the basis of this field of research at NIRS, and to the present, many researchers and technicians have accumulated both scientific and technical achievements, as well as inherited the spirit of research. Besides, in recent years, we have developed production systems with AVF-930 cyclotron for various ‘non-standard’ radioisotopes applied in both diagnosis and therapy. Here, we review the past 50 years of our activities on radioisotope and radiopharmaceutical development, as well as more recent activities.
Collapse
|
10
|
Chigoho DM, Bridoux J, Hernot S. Reducing the renal retention of low- to moderate-molecular-weight radiopharmaceuticals. Curr Opin Chem Biol 2021; 63:219-228. [PMID: 34325089 DOI: 10.1016/j.cbpa.2021.06.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/18/2021] [Accepted: 06/23/2021] [Indexed: 02/06/2023]
Abstract
The field of nuclear imaging and therapy is rapidly progressing with the development of targeted radiopharmaceuticals that show rapid targeting and rapid clearance with minimal background. Unfortunately, they are often reabsorbed in the kidneys, leading to possible nephrotoxicity, limiting the therapeutic dose, and/or reducing imaging quality. The blocking of endocytic receptors has been extensively used as a strategy to reduce kidney radiation. Alternatively, the physicochemical properties of radiotracers can be modulated to either prevent their reuptake or promote the excretion of radiometabolites. Other interesting strategies focus on the insertion of a cleavable linker between the radiolabel and the targeting moiety or pretargeting approaches in which the targeting moiety and radiolabel are administered separately. In the context of this review, we will discuss the latest advances and insights on strategies used to reduce renal retention of low- to moderate-molecular-weight radiopharmaceuticals.
Collapse
Affiliation(s)
- Dora Mugoli Chigoho
- Laboratory for in Vivo Cellular and Molecular Imaging, ICMI-BEFY/MIMA, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Jessica Bridoux
- Laboratory for in Vivo Cellular and Molecular Imaging, ICMI-BEFY/MIMA, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium
| | - Sophie Hernot
- Laboratory for in Vivo Cellular and Molecular Imaging, ICMI-BEFY/MIMA, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium.
| |
Collapse
|
11
|
Zheng L, Wang M, Shen X, Wang C, Wan L, Tang L, Chen G, Chen G. Applicability of Monocyte Activation Test for Pyrogen Detection in Succinylated Gelatin Injection. CURR PHARM ANAL 2021. [DOI: 10.2174/1573412916666191230125803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background::
Pyrogens are fever-inducing substances and pyrogen detection is mandatory in
parenteral pharmaceuticals. Succinylated Gelatin Injection (SGI) is a biopharmaceutical product, containing
multi-component, and it is administered parenterally.
Objective:
The study aimed to assess pyrogen in SGI and to evaluate the feasibility of the Monocyte
Activation Test (MAT) for pyrogen detection in a multi-component pharmaceutical product.
Method:
In the present study, the Bacterial Endotoxin Test (BET) and the Monocyte Activation Test
(MAT) were employed to assess pyrogen in SGI. The MAT method was developed on the basis of the
HL-60/IL-6 assay. HL-60 cells were incubated with lipopolysaccharide (LPS) standards and sample
solutions. The endotoxin produced by the incubation, interleukin-6 (IL-6), was measured by ELISA.
The MAT method was validated and main parameters were investigated. Finally, the pyrogenicity of
SGIs from two different enterprises was determined by the developed MAT method.
Results:
The BET failed in the test for interfering factors and the MAT was proved suitable for the pyrogen
detection of SGI. All the products examined showed negative results in the pyrogen detection
test.
Conclusion:
The MAT method is feasible in pyrogen detection of SGI. It can be applied in pyrogen
detection for quality and safety control of multi-component biological products.
Collapse
Affiliation(s)
- Luxia Zheng
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 200040,China
| | - Mingren Wang
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 200040,China
| | - Xiong Shen
- Department of Pharmacy, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai 200032,China
| | - Can Wang
- Shanghai Institute for Food and Drug Control, 1500 Zhangheng Road, Shanghai 201203,China
| | - Liqing Wan
- Shanghai Institute for Food and Drug Control, 1500 Zhangheng Road, Shanghai 201203,China
| | - Liming Tang
- Shanghai Institute for Food and Drug Control, 1500 Zhangheng Road, Shanghai 201203,China
| | - Guiliang Chen
- Shanghai Institute of Pharmaceutical Industry, China State Institute of Pharmaceutical Industry, 285 Gebaini Road, Shanghai 200040,China
| | - Gang Chen
- Shanghai Institute for Food and Drug Control, 1500 Zhangheng Road, Shanghai 201203,China
| |
Collapse
|
12
|
Jin ZH, Tsuji AB, Degardin M, Sugyo A, Obara S, Wakizaka H, Nagatsu K, Hu K, Zhang MR, Dumy P, Boturyn D, Higashi T. Radiotheranostic Agent 64Cu-cyclam-RAFT-c(-RGDfK-) 4 for Management of Peritoneal Metastasis in Ovarian Cancer. Clin Cancer Res 2020; 26:6230-6241. [PMID: 32933998 DOI: 10.1158/1078-0432.ccr-20-1205] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 08/11/2020] [Accepted: 09/10/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE Ovarian cancer peritoneal metastases (OCPMs) are a pathophysiologically heterogeneous group of tumors that are rarely curable. αVβ3 integrin (αVβ3) is overexpressed on tumoral neovessels and frequently on ovarian cancer cells. Here, using two clinically relevant αVβ3-positive OCPM mouse models, we studied the theranostic potential of an αVβ3-specific radiopeptide, 64Cu-cyclam-RAFT-c(-RGDfK-)4 (64Cu-RaftRGD), and its intra- and intertumoral distribution in relation to the tumor microenvironment. EXPERIMENTAL DESIGN αVβ3-expressing peritoneal and subcutaneous models of ovarian carcinoma (IGR-OV1 and NIH:OVCAR-3) were established in nude mice. 64Cu-RaftRGD was administered either intravenously or intraperitoneally. We performed intratumoral distribution (ITD) studies, PET/CT imaging and quantification, biodistribution assay and radiation dosimetry, and therapeutic efficacy and toxicity studies. RESULTS Intraperitoneal administration was an efficient route for targeting 64Cu-RaftRGD to OCPMs with excellent tumor penetration. Using the fluorescence surrogate, Cy5.5-RaftRGD, in our unique high-resolution multifluorescence analysis, we found that the ITD of 64Cu-RaftRGD was spatially distinct from, but complementary to, that of hypoxia. 64Cu-RaftRGD-based PET enabled clear visualization of multiple OCPM deposits and ascites and biodistribution analysis demonstrated an inverse correlation between tumor uptake and tumor size (1.2-17.2 mm). 64Cu-RaftRGD at a radiotherapeutic dose (148 MBq/0.357 nmol) showed antitumor activities by inhibiting tumor cell proliferation and inducing apoptosis, with negligible toxicity. CONCLUSIONS Collectively, these results demonstrate the all-in-one potential of 64Cu-RaftRGD for imaging guided radiotherapy of OCPM by targeting both tumoral neovessels and cancerous cells. On the basis of the ITD finding, we propose that pairing αVβ3- and hypoxia-targeted radiotherapies could improve therapeutic efficacy by overcoming the heterogeneity of ITD encountered with single-agent treatments.
Collapse
Affiliation(s)
- Zhao-Hui Jin
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan.
| | - Atsushi B Tsuji
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan.
| | | | - Aya Sugyo
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Satoshi Obara
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Hidekatsu Wakizaka
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Kotaro Nagatsu
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Kuan Hu
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Ming-Rong Zhang
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | - Pascal Dumy
- Institut des Biomolécules Max Mousseron, École Nationale Supérieure de Chimie de Montpellier, Université de Montpellier, Montpellier, France
| | | | - Tatsuya Higashi
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| |
Collapse
|
13
|
Hiroyama S, Rokugawa T, Ito M, Iimori H, Morita I, Maeda H, Fujisawa K, Matsunaga K, Shimosegawa E, Abe K. Quantitative evaluation of hepatic integrin α vβ 3 expression by positron emission tomography imaging using 18F-FPP-RGD 2 in rats with non-alcoholic steatohepatitis. EJNMMI Res 2020; 10:118. [PMID: 33026561 PMCID: PMC7541810 DOI: 10.1186/s13550-020-00704-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022] Open
Abstract
Background Integrin αvβ3, which are expressed by activated hepatic stellate cells in non-alcoholic steatohepatitis (NASH), play an important role in the fibrosis. Recently, we reported that an RGD peptide positron emission tomography (PET) probe is useful as a predictor of hepatic fibrosis. Kinetic analysis of the RGD PET probe has been performed in tumours, but not in hepatic fibrosis. Therefore, we aimed to quantify hepatic integrin αvβ3 in a model of NASH by kinetic analysis using 18F-FPP-RGD2, an integrin αvβ3 PET probe. Methods 18F-FPP-RGD2 PET/CT scans were performed in control and NASH rats. Tissue kinetic analyses were performed using a one-tissue, two-compartment (1T2C) and a two-tissue, three-compartment (2T3C) model using an image-derived input function (IDIF) for the left ventricle. We then conducted correlation analysis between standard uptake values (SUVs) or volume of distribution (VT), evaluated using compartment kinetic analysis and integrin αv or β3 protein expression. Results Biochemical and histological evaluation confirmed the development of NASH rats. Integrin αvβ3 protein expression and hepatic SUV were higher in NASH- than normal rats. The hepatic activity of 18F-FPP-RGD2 peaked rapidly after administration and then gradually decreased, whereas left ventricular activity rapidly disappeared. The 2T3C model was found to be preferable for 18F-FPP-RGD2 kinetic analysis in the liver. The VT (IDIF) for 18F-FPP-RGD2, calculated using the 2T3C model, was significantly higher in NASH- than normal rats and correlated strongly with hepatic integrin αv and β3 protein expression. The strengths of these correlations were similar to those between SUV60–90 min and hepatic integrin αv or β3 protein expression. Conclusions We have demonstrated that the VT (IDIF) of 18F-FPP-RGD2, calculated using kinetic modelling, positively correlates with integrin αv and β3 protein in the liver of NASH rats. These findings suggest that hepatic VT (IDIF) provides a quantitative assessment of integrin αvβ3 protein in liver.
Collapse
Affiliation(s)
- Shuichi Hiroyama
- Translational Research Unit, Biomarker R&D Department, Shionogi & Co., Ltd., 3-1-1 Futaba-cho, Toyonaka, Osaka, 561-0825, Japan.
| | - Takemi Rokugawa
- Translational Research Unit, Biomarker R&D Department, Shionogi & Co., Ltd., 3-1-1 Futaba-cho, Toyonaka, Osaka, 561-0825, Japan
| | - Miwa Ito
- Translational Research Unit, Biomarker R&D Department, Shionogi & Co., Ltd., 3-1-1 Futaba-cho, Toyonaka, Osaka, 561-0825, Japan
| | - Hitoshi Iimori
- Research Laboratory for Development, Shionogi & Co., Ltd., 3-1-1 Futaba-cho, Toyonaka, Osaka, 561-0825, Japan
| | - Ippei Morita
- Laboratory for Advanced Medicine Research, Shionogi & Co., Ltd., 3-1-1 Futaba-cho, Toyonaka, Osaka, 561-0825, Japan
| | - Hiroki Maeda
- Laboratory for Innovative Therapy Research, Shionogi & Co., Ltd., 3-1-1 Futaba-cho, Toyonaka, Osaka, 561-0825, Japan
| | - Kae Fujisawa
- Research Laboratory for Development, Shionogi & Co., Ltd., 3-1-1 Futaba-cho, Toyonaka, Osaka, 561-0825, Japan
| | - Keiko Matsunaga
- Department of Molecular Imaging in Medicine, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Eku Shimosegawa
- Department of Molecular Imaging in Medicine, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kohji Abe
- Translational Research Unit, Biomarker R&D Department, Shionogi & Co., Ltd., 3-1-1 Futaba-cho, Toyonaka, Osaka, 561-0825, Japan
| |
Collapse
|
14
|
Avellini T, Soni N, Silvestri N, Fiorito S, De Donato F, De Mei C, Walther M, Cassani M, Ghosh S, Manna L, Stephan H, Pellegrino T. Cation Exchange Protocols to Radiolabel Aqueous Stabilized ZnS, ZnSe, and CuFeS 2 Nanocrystals with 64Cu for Dual Radio- and Photo-Thermal Therapy. ADVANCED FUNCTIONAL MATERIALS 2020; 30:2002362. [PMID: 32684910 PMCID: PMC7357593 DOI: 10.1002/adfm.202002362] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/03/2020] [Accepted: 04/08/2020] [Indexed: 05/04/2023]
Abstract
Here, cation exchange (CE) reactions are exploited to radiolabel ZnSe, ZnS, and CuFeS2 metal chalcogenide nanocrystals (NCs) with 64Cu. The CE protocol requires one simple step, to mix the water-soluble NCs with a 64Cu solution, in the presence of vitamin C used to reduce Cu(II) to Cu(I). Given the quantitative cation replacement on the NCs, a high radiochemical yield, up to 99%, is reached. Also, provided that there is no free 64Cu, no purification step is needed, making the protocol easily translatable to the clinic. A unique aspect of the approach is the achievement of an unprecedentedly high specific activity: by exploiting a volumetric CE, the strategy enables to concentrate a large dose of 64Cu (18.5 MBq) in a small NC dose (0.18 µg), reaching a specific activity of 103 TBq g-1. Finally, the characteristic dielectric resonance peak, still present for the radiolabeled 64Cu:CuFeS2 NCs after the partial-CE reaction, enables the generation of heat under clinical laser exposure (1 W cm-2). The synergic toxicity of photo-ablation and 64Cu ionization is here proven on glioblastoma and epidermoid carcinoma tumor cells, while no intrinsic cytotoxicity is seen from the NC dose employed for these dual experiments.
Collapse
Affiliation(s)
- Tommaso Avellini
- Istituto Italiano di Tecnologia (IIT)via Morego 30Genova16163Italy
| | - Nisarg Soni
- Istituto Italiano di Tecnologia (IIT)via Morego 30Genova16163Italy
| | | | - Sergio Fiorito
- Istituto Italiano di Tecnologia (IIT)via Morego 30Genova16163Italy
| | | | - Claudia De Mei
- Istituto Italiano di Tecnologia (IIT)via Morego 30Genova16163Italy
| | - Martin Walther
- Institut für Radiopharmazeutische KrebsforschungHelmholtz‐Zentrum Dresden‐RossendorfBautzner Landstraße 400Dresden01328Germany
| | - Marco Cassani
- Istituto Italiano di Tecnologia (IIT)via Morego 30Genova16163Italy
- Present address:
International Clinical Research Center (FNUSA‐ICRC)Center for Translational MedicineBrno62500Czech Republic
| | - Sandeep Ghosh
- Istituto Italiano di Tecnologia (IIT)via Morego 30Genova16163Italy
- Present address:
Epi Process TechnologyASM America Inc.3440 East University DrivePhoenixAZ85034‐7200USA
| | - Liberato Manna
- Istituto Italiano di Tecnologia (IIT)via Morego 30Genova16163Italy
| | - Holger Stephan
- Institut für Radiopharmazeutische KrebsforschungHelmholtz‐Zentrum Dresden‐RossendorfBautzner Landstraße 400Dresden01328Germany
| | | |
Collapse
|
15
|
Borbély A, Thoreau F, Figueras E, Kadri M, Coll J, Boturyn D, Sewald N. Synthesis and Biological Characterization of Monomeric and Tetrameric RGD-Cryptophycin Conjugates. Chemistry 2020; 26:2602-2605. [PMID: 31943410 PMCID: PMC7064988 DOI: 10.1002/chem.201905437] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Indexed: 01/28/2023]
Abstract
The effective delivery of cytotoxic agents to tumor cells is a key challenge in anticancer therapy. Multivalent integrinspecific ligands are considered a promising tool to increase the binding affinity, selectivity, and internalization efficiency of small-molecule drug conjugates. Herein, we report the synthesis and biological evaluation of a multimeric conjugate containing the high-affinity integrin αv β3 binding ligand RAFT-c(RGDfK)4 , a lysosomally cleavable Val-Cit linker, and cryptophycin-55 glycinate, a potent inhibitor of tubulin polymerization. In vitro cytotoxicity assays verified that the multimeric RGD-cryptophycin conjugate displays improved potency compared to the monomeric analogue in integrin αv β3 overexpressing tumor cell lines, while significantly reduced activity was observed in the integrin-negative cell line.
Collapse
Affiliation(s)
- Adina Borbély
- Organic and Bioorganic ChemistryDepartment of ChemistryBielefeld UniversityUniversitätsstraße 2533615BielefeldGermany
| | - Fabien Thoreau
- CNRS, Department of Molecular ChemistryUniversity Grenoble Alpes, UMR 525038000GrenobleFrance
| | - Eduard Figueras
- Organic and Bioorganic ChemistryDepartment of ChemistryBielefeld UniversityUniversitätsstraße 2533615BielefeldGermany
| | - Malika Kadri
- Institute for Advanced BiosciencesUniversity Grenoble Alpes, INSERM U1209—UMR CNRS 530938700GrenobleFrance
| | - Jean‐Luc Coll
- Institute for Advanced BiosciencesUniversity Grenoble Alpes, INSERM U1209—UMR CNRS 530938700GrenobleFrance
| | - Didier Boturyn
- CNRS, Department of Molecular ChemistryUniversity Grenoble Alpes, UMR 525038000GrenobleFrance
| | - Norbert Sewald
- Organic and Bioorganic ChemistryDepartment of ChemistryBielefeld UniversityUniversitätsstraße 2533615BielefeldGermany
| |
Collapse
|
16
|
Ohya T, Minegishi K, Suzuki H, Nagatsu K, Fukada M, Hanyu M, Zhang MR. Development of a remote purification apparatus with disposable evaporator for the routine production of high-quality 64Cu for clinical use. Appl Radiat Isot 2019; 146:127-132. [DOI: 10.1016/j.apradiso.2019.01.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 01/25/2019] [Accepted: 01/26/2019] [Indexed: 10/27/2022]
|
17
|
Zhou Z, Devoogdt N, Zalutsky MR, Vaidyanathan G. An Efficient Method for Labeling Single Domain Antibody Fragments with 18F Using Tetrazine- Trans-Cyclooctene Ligation and a Renal Brush Border Enzyme-Cleavable Linker. Bioconjug Chem 2018; 29:4090-4103. [PMID: 30384599 DOI: 10.1021/acs.bioconjchem.8b00699] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Single domain antibody fragments (sdAbs) labeled with 18F have shown promise for assessing the status of oncological targets such as the human epidermal growth factor receptor 2 (HER2) by positron emission tomography (PET). Earlier, we evaluated two residualizing prosthetic agents for 18F-labeling of anti-HER2 sdAbs; however, these methods resulted in poor labeling yields and high uptake of 18F activity in the kidneys. To potentially mitigate these limitations, we have now developed an 18F labeling method that utilizes the trans-cyclooctene (TCO)-tetrazine (Tz)-based inverse-electron demand Diels-Alder reaction (IEDDAR) in tandem with a renal brush border enzyme-cleavable glycine-lysine (GK) linker in the prosthetic moiety. The HER2-targeted sdAb 2Rs15d was derivatized with TCO-GK-PEG4-NHS or TCO-PEG4-NHS, which lacks the cleavable linker. As an additional control, the non HER2-specific sdAb R3B23 was derivatized with TCO-GK-PEG4-NHS. The resultant sdAb conjugates were labeled with 18F by IEDDAR using [18F]AlF-NOTA-PEG4-methyltetrazine. As a positive control, the 2Rs15d sdAb was radioiodinated using the well-characterized residualizing prosthetic agent, N-succinimidyl 4-guanidinomethyl-3-[125I]iodobenzoate ([125I]SGMIB). Synthesis of [18F]AlF-NOTA-Tz-TCO-GK-2Rs15d was achieved with an overall radiochemical yield (RCY) of 17.8 ± 1.5% ( n = 5) in 90 min, a significant improvement over prior methods (3-4% in 2-3 h). In vitro assays indicated that [18F]AlF-NOTA-Tz-TCO-GK-2Rs15d bound with high affinity and immunoreactivity to HER2. In normal mice, when normalized to coinjected [125I]SGMIB-2Rs15d, the kidney uptake of [18F]AlF-NOTA-Tz-TCO-GK-2Rs15d was 15- and 28-fold lower ( P < 0.001) than that seen for the noncleavable control ([18F]AlF-NOTA-Tz-TCO-2Rs15d) at 1 and 3 h, respectively. Uptake of [18F]AlF-NOTA-Tz-TCO-GK-2Rs15d in HER2-expressing SKOV-3 ovarian carcinoma xenografts implanted in athymic mice was about 80% of that seen for coinjected [125I]SGMIB-2Rs15d. On the other hand, kidney uptake was 5-6-fold lower, and as a result, tumor-to-kidney ratios were 4-fold higher for [18F]AlF-NOTA-Tz-TCO-GK-2Rs15d than those for [125I]SGMIB-2Rs15d. SKOV-3 xenografts were clearly delineated even at 1 h after administration of [18F]AlF-NOTA-Tz-TCO-GK-2Rs15d by Micro-PET/CT imaging with even higher contrast observed thereafter. In conclusion, this strategy warrants further evaluation for labeling small proteins such as sdAbs because it offers the benefits of good radiochemical yields and enhanced tumor-to-normal tissue ratios, particularly in the kidney.
Collapse
Affiliation(s)
- Zhengyuan Zhou
- Department of Radiology , Duke University Medical Center , Durham , North Carolina 27710 , United States
| | - Nick Devoogdt
- In vivo Cellular and Molecular Imaging laboratory , Vrije Universiteit Brussel , 1090 , Brussels , Belgium
| | - Michael R Zalutsky
- Department of Radiology , Duke University Medical Center , Durham , North Carolina 27710 , United States
| | - Ganesan Vaidyanathan
- Department of Radiology , Duke University Medical Center , Durham , North Carolina 27710 , United States
| |
Collapse
|
18
|
Jin ZH, Tsuji AB, Degardin M, Sugyo A, Yoshii Y, Nagatsu K, Zhang MR, Fujibayashi Y, Dumy P, Boturyn D, Higashi T. Uniform intratumoral distribution of radioactivity produced using two different radioagents, 64Cu-cyclam-RAFT-c(-RGDfK-) 4 and 64Cu-ATSM, improves therapeutic efficacy in a small animal tumor model. EJNMMI Res 2018; 8:54. [PMID: 29923139 PMCID: PMC6008272 DOI: 10.1186/s13550-018-0407-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 06/05/2018] [Indexed: 12/15/2022] Open
Abstract
Background The present study proposed a new concept for targeted radionuclide therapy (TRT) to improve the intratumoral distribution of radioactivity using two different radiopharmaceuticals. We examined the efficacy of a combination of a tetrameric cyclic Arg-Gly-Asp (cRGD) peptide-based radiopharmaceutical, 64Cu-cyclam-RAFT-c(-RGDfK-)4 (64Cu-RaftRGD, an αVβ3 integrin [αVβ3] tracer), and 64Cu-diacetyl-bis (N4-methylthiosemicarbazone) (64Cu-ATSM, a supposed tracer for hypoxic metabolism) in a small animal tumor model. Results Mice with subcutaneous αVβ3-positive U87MG glioblastoma xenografts were used. The intratumoral distribution of a near-infrared dye, Cy5.5-labeled RAFT-c(-RGDfK-)4 (Cy5.5-RaftRGD), 64Cu-RaftRGD, and 64Cu-ATSM was visualized by fluorescence imaging and autoradiography of the co-injected Cy5.5-RaftRGD with 64Cu-RaftRGD or 64Cu-ATSM at 3 h postinjection. Mice were treated with a single intravenous dose of the vehicle solution (control), 18.5 or 37 MBq of 64Cu-RaftRGD or 64Cu-ATSM, or a combination (18.5 MBq of each agent). The tumor volume, tumor cell proliferation, body weight, survival, and tumor and organ uptake of radiopharmaceuticals were assessed. It was shown that Cy5.5-RaftRGD colocalized with 64Cu-RaftRGD and could be used as a surrogate for the radioactive agent. The intratumoral distribution of Cy5.5-RaftRGD and 64Cu-ATSM was discordant and nearly complementary, indicating a more uniform distribution of radioactivity achievable with the combined use of 64Cu-RaftRGD and 64Cu-ATSM. Neither 64Cu-RaftRGD nor 64Cu-ATSM showed significant effects on tumor growth at 18.5 MBq. The combination of both (18.5 MBq each) showed sustained inhibitory effects against tumor growth and tumor cell proliferation and prolonged the survival of the mice, compared to that by either single agent at 37 MBq. Interestingly, the uptake of the combination by the tumor was higher than that of 64Cu-RaftRGD alone, but lower than that of 64Cu-ATSM alone. The kidneys showed the highest uptake of 64Cu-RaftRGD, whereas the liver exhibited the highest uptake of 64Cu-ATSM. No obvious adverse effects were observed in all treated mice. Conclusions The combination of 64Cu-RaftRGD and 64Cu-ATSM achieved an improved antitumor effect owing to the more uniform intratumoral distribution of radioactivity. Thus, combining different radiopharmaceuticals to improve the intratumoral distribution would be a promising concept for more effective and safer TRT. Electronic supplementary material The online version of this article (10.1186/s13550-018-0407-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhao-Hui Jin
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Anagawa 4-9-1, Inage, Chiba, 263-8555, Japan.
| | - Atsushi B Tsuji
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Anagawa 4-9-1, Inage, Chiba, 263-8555, Japan
| | - Mélissa Degardin
- Département de Chimie Moléculaire-UMR CNRS 5250, Université Grenoble Alpes, 38041, Grenoble Cedex 9, France
| | - Aya Sugyo
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Anagawa 4-9-1, Inage, Chiba, 263-8555, Japan
| | - Yukie Yoshii
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Anagawa 4-9-1, Inage, Chiba, 263-8555, Japan
| | - Kotaro Nagatsu
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Anagawa 4-9-1, Inage, Chiba, 263-8555, Japan
| | - Ming-Rong Zhang
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Anagawa 4-9-1, Inage, Chiba, 263-8555, Japan
| | - Yasuhisa Fujibayashi
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Anagawa 4-9-1, Inage, Chiba, 263-8555, Japan
| | - Pascal Dumy
- IBMM, UMR-5247, Université de Montpellier, CNRS, École Nationale Supérieure de Chimie de Montpellier, 34296, Montpellier Cedex 5, France
| | - Didier Boturyn
- Département de Chimie Moléculaire-UMR CNRS 5250, Université Grenoble Alpes, 38041, Grenoble Cedex 9, France
| | - Tatsuya Higashi
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Anagawa 4-9-1, Inage, Chiba, 263-8555, Japan
| |
Collapse
|
19
|
Kanno I, Seki C, Takuwa H, Jin ZH, Boturyn D, Dumy P, Furukawa T, Saga T, Ito H, Masamoto K. Positron emission tomography of cerebral angiogenesis and TSPO expression in a mouse model of chronic hypoxia. J Cereb Blood Flow Metab 2018; 38:687-696. [PMID: 28128020 PMCID: PMC5888851 DOI: 10.1177/0271678x16689800] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The present study aimed to examine whether positron emission tomography (PET) could evaluate cerebral angiogenesis. Mice were housed in a hypoxic chamber with 8-9% oxygen for 4, 7, and 14 days, and the angiogenic responses were evaluated with a radiotracer, 64Cu-cyclam-RAFT-c(-RGDfK-)4, which targeted αVβ3 integrin and was imaged with PET. The PET imaging results showed little uptake during all of the hypoxic periods. Immunofluorescence staining of the β3 integrin, CD61, revealed weak expression, while the microvessel density assessed by CD31 staining increased with the hypoxic duration. These observations suggest that the increased vascular density originated from other types of vascular remodeling, unlike angiogenic sprouting. We then searched for any signs of vascular remodeling that could be detected using PET. PET imaging of 11C-PK11195, a marker of the 18-kDa translocator protein (TSPO), revealed a transient increase at day 4 of hypoxia. Because the immunofluorescence of glial markers showed unchanged staining over the early phase of hypoxia, the observed upregulation of TSPO expression probably originated from non-glial cells (e.g. vascular cells). In conclusion, a transient increase in TSPO probe uptake was detected with PET at only the early phase of hypoxia, which indicates an early sign of vascular remodeling induced by hypoxia.
Collapse
Affiliation(s)
- Iwao Kanno
- 1 Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | - Chie Seki
- 1 Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | - Hiroyuki Takuwa
- 1 Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | - Zhao-Hui Jin
- 1 Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | - Didier Boturyn
- 2 Département de Chimie Moléculaire, Université Grenoble Alpes, Grenoble, France
| | - Pascal Dumy
- 3 Institut des Biomolécules Max Mousseron, École Nationale Supérieure de Chimie de Montpellier, Montpellier, France
| | - Takako Furukawa
- 1 Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | - Tsuneo Saga
- 1 Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | - Hiroshi Ito
- 1 Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | - Kazuto Masamoto
- 1 Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan.,4 Brain Science Inspired Life Support Research Center, University of Electro-Communications, Tokyo, Japan
| |
Collapse
|
20
|
Gelofusine Ameliorates Colistin-Induced Nephrotoxicity. Antimicrob Agents Chemother 2017; 61:AAC.00985-17. [PMID: 28923868 DOI: 10.1128/aac.00985-17] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Accepted: 09/09/2017] [Indexed: 01/06/2023] Open
Abstract
Colistin therapy is used as the last line of defense against life-threatening Gram-negative infections. Nephrotoxicity is the major dose-limiting side effect that impedes optimal dosing of patients. This study aims to examine the nephroprotective effect of the plasma volume expander gelofusine against colistin-induced nephrotoxicity. Renal protection was assessed in mice that were subcutaneously injected with colistin sulfate (14 mg/kg of body weight × 6 doses every 2 h; accumulated dose, 84 mg/kg) and simultaneously injected in the intraperitoneal region with gelofusine (75, 150, 300, or 600 mg/kg × 6). At 2 and 20 h after the last colistin dose, mice were euthanized, and the severity of renal alteration was examined histologically. Histological findings in mice revealed that colistin-induced nephrotoxicity was ameliorated by gelofusine in a dose-dependent manner, whereas significant histological abnormalities were detected in the kidneys of mice in the colistin-only group. The impact of coadministered gelofusine on colistin pharmacokinetics was investigated in rats. Rats were administered a single intravenous dose of gelofusine at 400 mg/kg 15 min prior to the intravenous administration of colistin (1 mg/kg). Gelofusine codosing did not alter the pharmacokinetics of colistin in rats; however, gelofusine did significantly lower the accumulation of colistin in the kidney tissue of mice. This is the first study demonstrating the protective effect of gelofusine against colistin-induced nephrotoxicity. These findings highlight the clinical potential of gelofusine as a safe adjunct for ameliorating the nephrotoxicity and increasing the therapeutic index of polymyxins.
Collapse
|
21
|
67Cu-Radiolabeling of a multimeric RGD peptide for αVβ3 integrin-targeted radionuclide therapy. Nucl Med Commun 2017; 38:347-355. [DOI: 10.1097/mnm.0000000000000646] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
22
|
Ohya T, Nagatsu K, Suzuki H, Fukada M, Minegishi K, Hanyu M, Fukumura T, Zhang MR. Efficient preparation of high-quality 64 Cu for routine use. Nucl Med Biol 2016; 43:685-691. [DOI: 10.1016/j.nucmedbio.2016.07.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 07/27/2016] [Indexed: 11/16/2022]
|
23
|
Jin ZH, Furukawa T, Degardin M, Sugyo A, Tsuji AB, Yamasaki T, Kawamura K, Fujibayashi Y, Zhang MR, Boturyn D, Dumy P, Saga T. αVβ3 Integrin-Targeted Radionuclide Therapy with 64Cu-cyclam-RAFT-c(-RGDfK-)4. Mol Cancer Ther 2016; 15:2076-85. [DOI: 10.1158/1535-7163.mct-16-0040] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 06/20/2016] [Indexed: 11/16/2022]
|
24
|
Litau S, Seibold U, Vall-Sagarra A, Fricker G, Wängler B, Wängler C. Comparative Assessment of Complex Stabilities of Radiocopper Chelating Agents by a Combination of Complex Challenge and in vivo Experiments. ChemMedChem 2015; 10:1200-8. [PMID: 26011290 DOI: 10.1002/cmdc.201500132] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Indexed: 12/12/2022]
Abstract
For (64) Cu radiolabeling of biomolecules to be used as in vivo positron emission tomography (PET) imaging agents, various chelators are commonly applied. It has not yet been determined which of the most potent chelators--NODA-GA ((1,4,7-triazacyclononane-4,7-diyl)diacetic acid-1-glutaric acid), CB-TE2A (2,2'-(1,4,8,11-tetraazabicyclo[6.6.2]hexadecane-4,11-diyl)diacetic acid), or CB-TE1A-GA (1,4,8,11-tetraazabicyclo[6.6.2]hexadecane-4,11-diyl-8-acetic acid-1-glutaric acid)--forms the most stable complexes resulting in PET images of highest quality. We determined the (64) Cu complex stabilities for these three chelators by a combination of complex challenge and an in vivo approach. For this purpose, bioconjugates of the chelating agents with the gastrin-releasing peptide receptor (GRPR)-affine peptide PESIN and an integrin αv β3 -affine c(RGDfC) tetramer were synthesized and radiolabeled with (64) Cu in excellent yields and specific activities. The (64) Cu-labeled biomolecules were evaluated for their complex stabilities in vitro by conducting a challenge experiment with the respective other chelators as challengers. The in vivo stabilities of the complexes were also determined, showing the highest stability for the (64) Cu-CB-TE1A-GA complex in both experimental setups. Therefore, CB-TE1A-GA is the most appropriate chelating agent for *Cu-labeled radiotracers and in vivo imaging applications.
Collapse
Affiliation(s)
- Shanna Litau
- Biomedical Chemistry, Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim (Germany).,Molecular Imaging and Radiochemistry, Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim (Germany)
| | - Uwe Seibold
- Biomedical Chemistry, Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim (Germany).,Molecular Imaging and Radiochemistry, Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim (Germany)
| | - Alicia Vall-Sagarra
- Biomedical Chemistry, Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim (Germany)
| | - Gert Fricker
- Institute of Pharmacy and Molecular Biotechnology, University of Heidelberg, Im Neuenheimer Feld 329, 69120 Heidelberg (Germany)
| | - Björn Wängler
- Molecular Imaging and Radiochemistry, Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim (Germany)
| | - Carmen Wängler
- Biomedical Chemistry, Department of Clinical Radiology and Nuclear Medicine, Medical Faculty Mannheim of Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim (Germany).
| |
Collapse
|
25
|
Jin ZH, Furukawa T, Kumata K, Xie L, Yui J, Wakizaka H, Fujibayashi Y, Zhang MR, Saga T. Development of the Fibronectin–Mimetic Peptide KSSPHSRN(SG) 5RGDSP as a Novel Radioprobe for Molecular Imaging of the Cancer Biomarker α 5β 1 Integrin. Biol Pharm Bull 2015; 38:1722-31. [DOI: 10.1248/bpb.b15-00344] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Zhao-Hui Jin
- Molecular Imaging Center, National Institute of Radiological Sciences
| | - Takako Furukawa
- Molecular Imaging Center, National Institute of Radiological Sciences
| | - Katsushi Kumata
- Molecular Imaging Center, National Institute of Radiological Sciences
| | - Lin Xie
- Molecular Imaging Center, National Institute of Radiological Sciences
| | - Joji Yui
- Molecular Imaging Center, National Institute of Radiological Sciences
| | | | | | - Ming-Rong Zhang
- Molecular Imaging Center, National Institute of Radiological Sciences
| | - Tsuneo Saga
- Molecular Imaging Center, National Institute of Radiological Sciences
| |
Collapse
|