1
|
Chatzopoulou E, Bousaidi N, Guilbert T, Rucher G, Rose J, Germain S, Rouzet F, Chaussain C, Muller L, Gorin C. Multiscale Imaging to Monitor Functional SHED-Supported Engineered Vessels. J Dent Res 2024; 103:1392-1402. [PMID: 39290146 DOI: 10.1177/00220345241271122] [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: 09/19/2024] Open
Abstract
Regeneration of orofacial tissues is hampered by the lack of adequate vascular supply. Implantation of in vitro engineered, prevascularized constructs has emerged as a strategy to allow the rapid vascularization of the entire graft. Given the angiogenic properties of dental pulp stem cells, we hereby established a preclinical model of prevascularized constructs loaded with stem cells from human exfoliating deciduous teeth (SHED) in a 3-dimensional-printed material and provided a functional analysis of their in vivo angiogenesis, vascular perfusion, and permeability. Three different cell-loaded collagen hydrogels (SHED-human umbilical vein endothelial cell [HUVEC], HUVEC with SHED-conditioned medium, and SHED alone) were cast in polylactic acid (PLA) grids and ectopically implanted in athymic mice. At day 10, in vivo positron emission tomography (PETscan) revealed a significantly increased uptake of radiotracer targeting activated endothelial cells in the SHED-HUVEC group compared to the other groups. At day 30, ex vivo micro-computed tomography imaging confirmed that SHED-HUVEC constructs had a significantly increased vascular volume compared to the other ones. Injection of species-specific lectins analyzed by 2-photon microscopy demonstrated blood perfusion of the engineered human vessels in both prevascularized groups. However, in vivo quantification showed increased vessel density in the SHED-HUVEC group. In addition, coinjection of fluorescent lectin and dextran revealed that prevascularization with SHED prevented vascular leakage, demonstrating the active role of SHED in the maturation of human-engineered microvascular networks. This preclinical study introduces a novel PLA prevascularized and implantable construct, along with an array of imaging techniques, to validate the ability of SHED to promote functional human-engineered vessels, further highlighting the interest of SHED for orofacial tissue engineering. Furthermore, this study validates the use of PETscan for the early detection of in vivo angiogenesis, which may be applied in the clinic to monitor the performance of prevascularized grafts.
Collapse
Affiliation(s)
- E Chatzopoulou
- Université Paris Cité, URP2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant, Montrouge, France
- AP-HP, Services de médecine bucco-dentaire, FHU DDS-Net, GH Paris Nord et Paris Est, France
| | - N Bousaidi
- Université Paris Cité, URP2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant, Montrouge, France
| | - T Guilbert
- Université Paris Cité, CNRS, INSERM U1016, Institut Cochin, Paris, France
| | - G Rucher
- Université Paris Cité, LVTS, INSERM U1148, France
- Université Paris Cité, UMS 34-FRIM, France
| | - J Rose
- AP-HP, Département de médecine nucléaire, Hôpital Bichat, Paris, France
| | - S Germain
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR7241, INSERM U1050, Université PSL, Paris, France
| | - F Rouzet
- Université Paris Cité, LVTS, INSERM U1148, France
- AP-HP, Département de médecine nucléaire, Hôpital Bichat, Paris, France
| | - C Chaussain
- Université Paris Cité, URP2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant, Montrouge, France
- AP-HP, Services de médecine bucco-dentaire, FHU DDS-Net, GH Paris Nord et Paris Est, France
| | - L Muller
- Center for Interdisciplinary Research in Biology (CIRB), Collège de France, CNRS UMR7241, INSERM U1050, Université PSL, Paris, France
| | - C Gorin
- Université Paris Cité, URP2496 Pathologies, Imagerie et Biothérapies Orofaciales et Plateforme Imagerie du Vivant, Montrouge, France
- AP-HP, Services de médecine bucco-dentaire, FHU DDS-Net, GH Paris Nord et Paris Est, France
| |
Collapse
|
2
|
Basaco Bernabeu T, Mansi R, Del Pozzo L, Zanger S, Gaonkar RH, McDougall L, De Rose F, Jaafar-Thiel L, Herz M, Eiber M, Ulaner GA, Weber WA, Fani M. 61Cu-PSMA-Targeted PET for Prostate Cancer: From Radiotracer Development to First-in-Human Imaging. J Nucl Med 2024; 65:1427-1434. [PMID: 39025646 PMCID: PMC11372264 DOI: 10.2967/jnumed.123.267126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 06/05/2024] [Indexed: 07/20/2024] Open
Abstract
The demand for PET tracers that target prostate-specific membrane antigen (PSMA) continues to increase. Meeting this demand with approved 68Ga- and 18F-labeled PSMA tracers is challenging outside of major urban centers. This is because the short physical half-life of these radionuclides makes it necessary to produce them near their sites of usage. To overcome this challenge, we propose cyclotron-produced 61Cu for labeling PSMA PET tracers. 61Cu can be produced on a large scale, and its 3.33-h half-life allows shipping over considerably longer distances than possible for 68Ga and 18F. Production of true theranostic twins using 61Cu and the β--emitter 67Cu is also feasible. Methods: PSMA-I&T (DOTAGA-(l-y)fk(sub-KuE)) and its derivative in which the DOTAGA chelator was replaced by NODAGA (NODAGA-(l-y)fk(sub-KuE)), herein reported as DOTAGA-PSMA-I&T and NODAGA-PSMA-I&T, respectively, were labeled with 61Cu and compared with [68Ga]Ga-DOTAGA-PSMA-I&T, [68Ga]Ga-NODAGA-PSMA-I&T, [68Ga]Ga-PSMA-11, and [18F]PSMA-1007. In vitro (lipophilicity, affinity, cellular uptake, and distribution) and in vivo (PET/CT, biodistribution, and stability) studies were performed in LNCaP cells and xenografts. Human dosimetry estimates were calculated for [61Cu]Cu-NODAGA-PSMA-I&T. First-in-human imaging with [61Cu]Cu-NODAGA-PSMA-I&T was performed in a patient with metastatic prostate cancer. Results: [61Cu]Cu-DOTAGA-PSMA-I&T and [61Cu]Cu-NODAGA-PSMA-I&T were synthesized with radiochemical purity of more than 97%, at an apparent molar activity of 24 MBq/nmol, without purification after labeling. In vitro, natural Cu (natCu)-DOTAGA-PSMA-I&T and natCu-NODAGA-PSMA-I&T showed high affinity for PSMA (inhibitory concentration of 50%, 11.2 ± 2.3 and 9.3 ± 1.8 nM, respectively), although lower than the reference natGa-PSMA-11 (inhibitory concentration of 50%, 2.4 ± 0.4 nM). Their cellular uptake and distribution were comparable to those of [68Ga]Ga-PSMA-11. In vivo, [61Cu]Cu-NODAGA-PSMA-I&T showed significantly lower uptake in nontargeted tissues than [61Cu]Cu-DOTAGA-PSMA-I&T and higher tumor uptake (14.0 ± 5.0 percentage injected activity per gram of tissue [%IA/g]) than [61Cu]Cu-DOTAGA-PSMA-I&T (6.06 ± 0.25 %IA/g, P = 0.0059), [68Ga]Ga-PSMA-11 (10.2 ± 1.5 %IA/g, P = 0.0972), and [18F]PSMA-1007 (9.70 ± 2.57 %IA/g, P = 0.080) at 1 h after injection. Tumor uptake was also higher for [61Cu]Cu-NODAGA-PSMA-I&T at 4 h after injection (10.7 ± 3.3 %IA/g) than for [61Cu]Cu-DOTAGA-PSMA-I&T (4.88 ± 0.63 %IA/g, P = 0.0014) and [18F]PSMA-1007 (6.28 ± 2.19 %IA/g, P = 0.0145). Tumor-to-nontumor ratios of [61Cu]Cu-NODAGA-PSMA-I&T were superior to those of [61Cu]Cu-DOTAGA-PSMA-I&T and comparable to those of [68Ga]Ga-PSMA-11 and [18F]PSMA-1007 at 1 h after injection and increased significantly between 1 and 4 h after injection in most cases. Human dosimetry estimates for [61Cu]Cu-NODAGA-PSMA-I&T were similar to the ones reported for 18F-PSMA ligands. First-in-human imaging demonstrated multifocal osseous and hepatic metastases. Conclusion: [61Cu]Cu-NODAGA-PSMA-I&T is a promising PSMA radiotracer that compares favorably with [68Ga]Ga-PSMA-11 and [18F]PSMA-1007, while allowing delayed imaging.
Collapse
Affiliation(s)
- Tais Basaco Bernabeu
- Division of Radiopharmaceutical Chemistry, University Hospital Basel, Basel, Switzerland
| | - Rosalba Mansi
- Division of Radiopharmaceutical Chemistry, University Hospital Basel, Basel, Switzerland
| | - Luigi Del Pozzo
- Division of Radiopharmaceutical Chemistry, University Hospital Basel, Basel, Switzerland
| | - Sandra Zanger
- Division of Radiopharmaceutical Chemistry, University Hospital Basel, Basel, Switzerland
| | - Raghuvir H Gaonkar
- Division of Radiopharmaceutical Chemistry, University Hospital Basel, Basel, Switzerland
| | - Lisa McDougall
- Division of Radiopharmaceutical Chemistry, University Hospital Basel, Basel, Switzerland
| | | | | | - Michael Herz
- Department of Nuclear Medicine, Technical University of Munich, Munich, Germany
- Bavarian Cancer Research Center, Munich, Germany
| | - Matthias Eiber
- Department of Nuclear Medicine, Technical University of Munich, Munich, Germany
- Bavarian Cancer Research Center, Munich, Germany
| | - Gary A Ulaner
- Molecular Imaging and Therapy, Hoag Family Cancer Institute, Irvine, California; and
- Departments of Radiology and Translational Genomics, University of Southern California, Los Angeles, California
| | - Wolfgang A Weber
- Department of Nuclear Medicine, Technical University of Munich, Munich, Germany
- Bavarian Cancer Research Center, Munich, Germany
| | - Melpomeni Fani
- Division of Radiopharmaceutical Chemistry, University Hospital Basel, Basel, Switzerland;
| |
Collapse
|
3
|
Zhang T, Ma X, Xu M, Cai J, Cai J, Cao Y, Zhang Z, Ji X, He J, Cabrera GOF, Wu X, Zhao W, Wu Z, Xie J, Li Z. Chelator boosted tumor-retention and pharmacokinetic properties: development of 64Cu labeled radiopharmaceuticals targeting neurotensin receptor. Eur J Nucl Med Mol Imaging 2024; 51:3322-3333. [PMID: 38771516 PMCID: PMC11368631 DOI: 10.1007/s00259-024-06754-2] [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: 12/18/2023] [Accepted: 05/05/2024] [Indexed: 05/22/2024]
Abstract
PURPOSE Accumulating evidence suggests that neurotensin (NTS) and neurotensin receptors (NTSRs) play key roles in lung cancer progression by triggering multiple oncogenic signaling pathways. This study aims to develop Cu-labeled neurotensin receptor 1 (NTSR1)-targeting agents with the potential for both imaging and therapeutic applications. METHOD A series of neurotensin receptor antagonists (NRAs) with variable propylamine (PA) linker length and different chelators were synthesized, including [64Cu]Cu-CB-TE2A-iPA-NRA ([64Cu]Cu-4a-c, i = 1, 2, 3), [64Cu]Cu-NOTA-2PA-NRA ([64Cu]Cu-4d), [64Cu]Cu-DOTA-2PA-NRA ([64Cu]Cu-4e, also known as [64Cu]Cu-3BP-227), and [64Cu]Cu-DOTA-VS-2PA-NRA ([64Cu]Cu-4f). The series of small animal PET/CT were conducted in H1299 lung cancer model. The expression profile of NTSR1 was also confirmed by IHC using patient tissue samples. RESULTS For most of the compounds studied, PET/CT showed prominent tumor uptake and high tumor-to-background contrast, but the tumor retention was strongly influenced by the chelators used. For previously reported 4e, [64Cu]Cu-labeled derivative showed initial high tumor uptake accompanied by rapid tumor washout at 24 h. The newly developed [64Cu]Cu-4d and [64Cu]Cu-4f demonstrated good tumor uptake and tumor-to-background contrast at early time points, but were less promising in tumor retention. In contrast, our lead compound [64Cu]Cu-4b demonstrated 9.57 ± 1.35, 9.44 ± 2.38 and 9.72 ± 4.89%ID/g tumor uptake at 4, 24, and 48 h p.i., respectively. Moderate liver uptake (11.97 ± 3.85, 9.80 ± 3.63, and 7.72 ± 4.68%ID/g at 4, 24, and 48 h p.i.) was observed with low uptake in most other organs. The PA linker was found to have a significant effect on drug distribution. Compared to [64Cu]Cu-4b, [64Cu]Cu-4a had a lower background, including a greatly reduced liver uptake, while the tumor uptake was only moderately reduced. Meanwhile, [64Cu]Cu-4c showed increased uptake in both the tumor and the liver. The clinical relevance of NTSR1 was also demonstrated by the elevated tumor expression in patient tissue samples. CONCLUSIONS Through the side-by-side comparison, [64Cu]Cu-4b was identified as the lead agent for further evaluation based on its high and sustained tumor uptake and moderate liver uptake. It can not only be used to efficiently detect NTSR1 expression in lung cancer (for diagnosis, patient screening, and treatment monitoring), but also has the great potential to treat NTSR-positive lesions once chelating to the beta emitter 67Cu.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Radiology, Biomedical Research Imaging Center, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina , 27599, USA.
- Department of Radiopharmaceuticals, Nuclear Medicine Clinical Transformation Center, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China.
| | - Xinrui Ma
- Department of Radiology, Biomedical Research Imaging Center, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina , 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, Raleigh, NC, North Carolina State University, NC 27599, USA
| | - Muyun Xu
- Department of Radiology, Biomedical Research Imaging Center, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina , 27599, USA
| | - Jinghua Cai
- Department of Radiology, Biomedical Research Imaging Center, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina , 27599, USA
| | - Jianhua Cai
- Department of Radiology, Biomedical Research Imaging Center, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina , 27599, USA
| | - Yanguang Cao
- Division of Pharmacotherapy and Experimental Therapeutics, School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Zhihao Zhang
- Department of Radiopharmaceuticals, Nuclear Medicine Clinical Transformation Center, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Xin Ji
- Department of Radiopharmaceuticals, Nuclear Medicine Clinical Transformation Center, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Jian He
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - German Oscar Fonseca Cabrera
- Department of Radiology, Biomedical Research Imaging Center, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina , 27599, USA
| | - Xuedan Wu
- Department of Radiology, Biomedical Research Imaging Center, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina , 27599, USA
| | - Weiling Zhao
- Department of Radiology, Biomedical Research Imaging Center, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina , 27599, USA
| | - Zhanhong Wu
- Department of Radiology, Biomedical Research Imaging Center, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina , 27599, USA
| | - Jin Xie
- Department of Chemistry, University of Georgia, 302 East Campus Road, Athens, GA, 30602, USA.
| | - Zibo Li
- Department of Radiology, Biomedical Research Imaging Center, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina , 27599, USA.
| |
Collapse
|
4
|
Shi G, Liu X, Du Y, Tian J. RGD targeted magnetic ferrite nanoparticles enhance antitumor immunotherapeutic efficacy by activating STING signaling pathway. iScience 2024; 27:109062. [PMID: 38660408 PMCID: PMC11039334 DOI: 10.1016/j.isci.2024.109062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/06/2023] [Accepted: 01/25/2024] [Indexed: 04/26/2024] Open
Abstract
Manganese has been used in tumor imaging for their ability to provide T1-weighted MRI signal. Recent research find Mn2+ can induce activation of the stimulator of interferon gene (STING) pathway to create an active and favorable tumor immune microenvironment. However, the direct injection of Mn2+ often results in toxicity. In this study, we developed an RGD targeted magnetic ferrite nanoparticle (RGD-MnFe2O4) to facilitate tumor targeted imaging and improve tumor immunotherapy. Magnetic resonance imaging and fluorescence molecular imaging were performed to monitor its in vivo biodistribution. We found that RGD-MnFe2O4 showed active tumor targeting and longer accumulation at tumor sites. Moreover, RGD-MnFe2O4 can activate STING pathway with low toxicity to enhance the PD-L1 expression. Furthermore, combining RGD-MnFe2O4 and anti-PD-L1 antibody (aPD-L1) can treat several types of immunogenic tumors through promoting the accumulation of tumor-infiltrating cytotoxic T cells. In general, our study provides a promising new strategy for the targeted and multifunctional theranostic nanoparticle for the improvement of tumor immunotherapy.
Collapse
Affiliation(s)
- Guangyuan Shi
- University of Science and Technology of China, Hefei 230026, China
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Xiaoli Liu
- Northwest University, Xi’an 710127, China
| | - Yang Du
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100080, China
| | - Jie Tian
- Science and Engineering, Beihang University, Beijing 100191, China
| |
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
|
Price T, Wagner L, Rosecker V, Havlíčková J, Prior TJ, Kubíček V, Hermann P, Stasiuk GJ. Inorganic Chemistry of the Tripodal Picolinate Ligand Tpaa with Gallium(III) and Radiolabeling with Gallium-68. Inorg Chem 2023; 62:20769-20776. [PMID: 37793007 PMCID: PMC10731648 DOI: 10.1021/acs.inorgchem.3c02459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Indexed: 10/06/2023]
Abstract
We report here the improved synthesis of the tripodal picolinate chelator Tpaa, with an overall yield of 41% over five steps, in comparison to the previously reported 6% yield. Tpaa was investigated for its coordination chemistry with Ga(III) and radiolabeling properties with gallium-68 (68Ga). The obtained crystal structure for [Ga(Tpaa)] shows that the three picolinate arms coordinate to the Ga(III) ion, fully occupying the octahedral coordination geometry. This is supported by 1H NMR which shows that the three arms are symmetrical when coordinated to Ga(III). Assessment of the thermodynamic stability through potentiometry gives log KGa-Tpaa = 21.32, with a single species being produced across the range of pH 3.5-7.5. Tpaa achieved >99% radiochemical conversion with 68Ga under mild conditions ([Tpaa] = 6.6 μM, pH 7.4, 37 °C) with a molar activity of 3.1 GBq μmol-1. The resulting complex, [68Ga][Ga(Tpaa)], showed improved stability over the previously reported [68Ga][Ga(Dpaa)(H2O)] in a serum challenge, with 32% of [68Ga][Ga(Tpaa)] remaining intact after 30 min of incubation with fetal bovine serum.
Collapse
Affiliation(s)
- Thomas
W. Price
- Department
of Imaging Chemistry and Biology, School of Biomedical Engineering
and Imaging Sciences, King’s College
London, London SE1 7EH, United
Kingdom
| | - Laurène Wagner
- Department
of Imaging Chemistry and Biology, School of Biomedical Engineering
and Imaging Sciences, King’s College
London, London SE1 7EH, United
Kingdom
| | - Veronika Rosecker
- Department
of Imaging Chemistry and Biology, School of Biomedical Engineering
and Imaging Sciences, King’s College
London, London SE1 7EH, United
Kingdom
| | - Jana Havlíčková
- Department
of Inorganic Chemistry, Faculty of Science, Charles University, Hlavova 8, 128 40 Prague 2, Czech Republic
| | - Timothy J. Prior
- Chemistry,
School of Natural Sciences, University of
Hull, Cottingham Road, Hull HU6 7RX, United Kingdom
| | - Vojtěch Kubíček
- Department
of Inorganic Chemistry, Faculty of Science, Charles University, Hlavova 8, 128 40 Prague 2, Czech Republic
| | - Petr Hermann
- Department
of Inorganic Chemistry, Faculty of Science, Charles University, Hlavova 8, 128 40 Prague 2, Czech Republic
| | - Graeme J. Stasiuk
- Department
of Imaging Chemistry and Biology, School of Biomedical Engineering
and Imaging Sciences, King’s College
London, London SE1 7EH, United
Kingdom
| |
Collapse
|
7
|
Wang Y, Yuan H, Tang S, Liu Y, Cai P, Liu N, Chen Y, Zhou Z. The effects of novel macrocyclic chelates on the targeting properties of the 68Ga-labeled Gastrin releasing peptide receptor antagonist RM2. EJNMMI Res 2023; 13:56. [PMID: 37285007 PMCID: PMC10247930 DOI: 10.1186/s13550-023-01005-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 06/01/2023] [Indexed: 06/08/2023] Open
Abstract
BACKGROUND The gastrin-releasing peptide receptor (GRPr) is a molecular target for the visualization of prostate cancer. Bombesin (BN) analogs are short peptides with a high affinity for GRPr. RM2 is a bombesin-based antagonist. It has been demonstrated that RM2 have superior in vivo biodistribution and targeting properties than high-affinity receptor agonists. This study developed new RM2-like antagonists by introducing the novel bifunctional chelators AAZTA5 and DATA5m to RM2. RESULTS The effects of different macrocyclic chelating groups on drug targeting properties and the possibility of preparing 68Ga-radiopharmaceuticals in a kit-based protocol were investigated using 68Ga-labeled entities. Both new RM2 variants were labelled with 68Ga3+ resulting in high yields, stability, and low molarity of the ligand. DATA5m-RM2 and AAZTA5-RM2 incorporated 68Ga3+ nearly quantitatively at room temperature within 3-5 min, and the labelling yield for 68Ga-DOTA-RM2 was approximately 10% under the same conditions. 68Ga-AAZTA5-RM2 showed stronger hydrophilicity according to partition coefficient. Although the maximal cellular uptake values of the three compounds were similar, 68Ga-AAZTA5-RM2 and 68Ga-DATA5m-RM2 peaked more rapidly. Biodistribution studies showed high and specific tumor uptake, with a maximum of 9.12 ± 0.81 percentage injected activity per gram of tissue (%ID/g) for 68Ga-DATA5m-RM2 and 7.82 ± 0.61%ID/g for 68Ga-AAZTA5-RM2 at 30 min after injection. CONCLUSIONS The conditions for complexation of DATA5m-RM2 and AAZTA5-RM2 with gallium-68 are milder, faster and require less amount of precursors than DOTA-RM2. Chelators had an evident influence on the pharmacokinetics and targeting properties of 68Ga-X-RM2 derivatives. Positively charged 68Ga-DATA5m-RM2 provided a high tumor uptake, high image contrast and good capability of targeting GRPr.
Collapse
Affiliation(s)
- Yinwen Wang
- The Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Jiangyang District, Luzhou, Sichuan, China
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
| | - Hongmei Yuan
- The Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Jiangyang District, Luzhou, Sichuan, China
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
| | - Sufan Tang
- The Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Jiangyang District, Luzhou, Sichuan, China
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
| | - Yang Liu
- The Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Jiangyang District, Luzhou, Sichuan, China
- Institute of Nuclear Medicine, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
| | - Ping Cai
- The Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Jiangyang District, Luzhou, Sichuan, China
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China
| | - Nan Liu
- Department of Nuclear Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Yue Chen
- The Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China.
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Jiangyang District, Luzhou, Sichuan, China.
- Institute of Nuclear Medicine, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China.
| | - Zhijun Zhou
- The Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China.
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Jiangyang District, Luzhou, Sichuan, China.
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China.
- Institute of Nuclear Medicine, Southwest Medical University, Jiangyang District, Luzhou, Sichuan, China.
| |
Collapse
|
8
|
Failla M, Floresta G, Abbate V. Peptide-based positron emission tomography probes: current strategies for synthesis and radiolabelling. RSC Med Chem 2023; 14:592-623. [PMID: 37122545 PMCID: PMC10131587 DOI: 10.1039/d2md00397j] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/05/2023] [Indexed: 01/09/2023] Open
Abstract
In medical imaging, techniques such as magnetic resonance imaging, contrast-enhanced computerized tomography, and positron emission tomography (PET) are extensively available and routinely used for disease diagnosis and treatment. Peptide-based targeting PET probes are usually small peptides with high affinity and specificity to specific cellular and tissue targets opportunely engineered for acting as PET probes. For instance, either the radioisotope (e.g., 18F, 11C) can be covalently linked to the peptide-probe or another ligand that strongly complexes the radioisotope (e.g., 64Cu, 68Ga) through multiple coordinative bonds can be chemically conjugated to the peptide delivery moiety. The main advantages of these probes are that they are cheaper than classical antibody-based PET tracers and can be efficiently chemically modified to be radiolabelled with virtually any radionuclide making them very attractive for clinical use. The goal of this review is to report and summarize recent technologies in peptide PET-based molecular probes synthesis and radiolabelling with the most used radioisotopes in 2022.
Collapse
Affiliation(s)
- Mariacristina Failla
- Department of Drug Science and Technology, University of Turin Via P. Giuria 9 10125 Turin Italy
| | - Giuseppe Floresta
- King's College London, Institute of Pharmaceutical Science Franklin Wilkins Building London SE1 9NH UK
- Department of Drug and Health Sciences, University of Catania Catania Italy
| | - Vincenzo Abbate
- King's College London, Institute of Pharmaceutical Science Franklin Wilkins Building London SE1 9NH UK
| |
Collapse
|
9
|
Tokgöz S, Boss M, Prasad S, Shah P, Laverman P, van Riel M, Gotthardt M. Protocol for Clinical GLP-1 Receptor PET/CT Imaging with [ 68Ga]Ga-NODAGA-Exendin-4. Methods Mol Biol 2022; 2592:143-153. [PMID: 36507990 DOI: 10.1007/978-1-0716-2807-2_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Imaging with radiolabeled exendin enables detection and characterization of glucagon-like peptide 1 receptors (GLP-1Rs) in vivo with high specificity. The novel radiotracer [68Ga]Ga-NODAGA-exendin-4 forms a stable complex after a simple and fast labeling procedure. Beta-cell mass in the islets of Langerhans can be visualized using [68Ga]Ga-NODAGA-exendin-4, which is promising for research into diabetes mellitus (DM) pathophysiology. Furthermore, this radiotracer enables very sensitive detection of insulinomas, resulting from vast overexpression of GLP-1Rs, and seems promising for the detection of focal lesions in congenital hyperinsulinism (CHI). Here, we describe the procedures involved in [68Ga]Ga-NODAGA-exendin-4 positron emission tomography (PET)/computed tomography (CT) imaging including the radiolabeling of the NODAGA-exendin conjugate with 68Ga, quality controls, and PET/CT.
Collapse
Affiliation(s)
- S Tokgöz
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - M Boss
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - S Prasad
- Department of Nuclear Medicine, Radiopharmacy, Berlin Experimental Radionuclide Imaging Center (BERIC), Berlin, Germany
| | - P Shah
- Department of Pediatric Endocrinology, Barts Health NHS Trust (The Royal London Childrens Hospital), London, UK
| | - P Laverman
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - M van Riel
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - M Gotthardt
- Department of Medical Imaging, Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
10
|
Uzal-Varela R, Patinec V, Tripier R, Valencia L, Maneiro M, Canle M, Platas-Iglesias C, Esteban-Gómez D, Iglesias E. On the dissociation pathways of copper complexes relevant as PET imaging agents. J Inorg Biochem 2022; 236:111951. [PMID: 35963110 DOI: 10.1016/j.jinorgbio.2022.111951] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/15/2022] [Accepted: 07/27/2022] [Indexed: 02/07/2023]
Abstract
Several bifunctional chelators have been synthesized in the last years for the development of new 64Cu-based PET agents for in vivo imaging. When designing a metal-based PET probe, it is important to achieve high stability and kinetic inertness once the radioisotope is coordinated. Different competitive assays are commonly used to evaluate the possible dissociation mechanisms that may induce Cu(II) release in the body. Among them, acid-assisted dissociation tests or transchelation challenges employing EDTA or SOD are frequently used to evaluate both solution thermodynamics and the kinetic behavior of potential metal-based systems. Despite of this, the Cu(II)/Cu(I) bioreduction pathway that could be promoted by the presence of bioreductants still remains little explored. To fill this gap we present here a detailed spectroscopic study of the kinetic behavior of different macrocyclic Cu(II) complexes. The complexes investigated include the cross-bridge cyclam derivative [Cu(CB-TE1A)]+, whose structure was determined using single-crystal X-ray diffraction. The acid-assisted dissociation mechanism was investigated using HClO4 and HCl to analyse the effect of the counterion on the rate constants. The complexes were selected so that the effects of complex charge and coordination polyhedron could be assessed. Cyclic voltammetry experiments were conducted to investigate whether the reduction to Cu(I) falls within the window of common bioreducing agents. The most striking behavior concerns the [Cu(NO2Th)]2+ complex, a 1,4,7-triazacyclononane derivative containing two methylthiazolyl pendant arms. This complex is extremely inert with respect to dissociation following the acid-catalyzed mechanism, but dissociates rather quickly in the presence of a bioreductant like ascorbic acid.
Collapse
Affiliation(s)
- Rocío Uzal-Varela
- Centro de Investigacións Científicas Avanzadas (CICA) and Departamento de Química, Facultade de Ciencias, Universidade da Coruña, 15071 A Coruña, Galicia, Spain
| | - Véronique Patinec
- 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
| | - Laura Valencia
- Departamento de Química Inorgánica, Universidade de Vigo, Facultad de Ciencias, 36310 Pontevedra, Spain
| | - Marcelino Maneiro
- Departamento de Química Inorgánica, Universidade de Santiago de Compostela, Facultade de Ciencias, 27002 Lugo, Spain
| | - Moisés Canle
- Centro de Investigacións Científicas Avanzadas (CICA) and Departamento de Química, Facultade de Ciencias, Universidade da Coruña, 15071 A Coruña, Galicia, Spain
| | - Carlos Platas-Iglesias
- Centro de Investigacións Científicas Avanzadas (CICA) and Departamento de Química, Facultade de Ciencias, Universidade da Coruña, 15071 A Coruña, Galicia, Spain
| | - David Esteban-Gómez
- Centro de Investigacións Científicas Avanzadas (CICA) and Departamento de Química, Facultade de Ciencias, Universidade da Coruña, 15071 A Coruña, Galicia, Spain.
| | - Emilia Iglesias
- Centro de Investigacións Científicas Avanzadas (CICA) and Departamento de Química, Facultade de Ciencias, Universidade da Coruña, 15071 A Coruña, Galicia, Spain.
| |
Collapse
|
11
|
Price TW, Renard I, Prior TJ, Kubíček V, Benoit DM, Archibald SJ, Seymour AM, Hermann P, Stasiuk GJ. Bn2DT3A, a Chelator for 68Ga Positron Emission Tomography: Hydroxide Coordination Increases Biological Stability of [ 68Ga][Ga(Bn 2DT3A)(OH)] . Inorg Chem 2022; 61:17059-17067. [PMID: 36251390 DOI: 10.1021/acs.inorgchem.2c01992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The chelator Bn2DT3A was used to produce a novel 68Ga complex for positron emission tomography (PET). Unusually, this system is stabilized by a coordinated hydroxide in aqueous solutions above pH 5, which confers sufficient stability for it to be used for PET. Bn2DT3A complexes Ga3+ in a hexadentate manner, forming a mer-mer complex with log K([Ga(Bn2DT3A)]) = 18.25. Above pH 5, the hydroxide ion coordinates the Ga3+ ion following dissociation of a coordinated amine. Bn2DT3A radiolabeling displayed a pH-dependent speciation, with [68Ga][Ga(Bn2DT3A)(OH)]- being formed above pH 5 and efficiently radiolabeled at pH 7.4. Surprisingly, [68Ga][Ga(Bn2DT3A)(OH)]- was found to show an increased stability in vitro (for over 2 h in fetal bovine serum) compared to [68Ga][Ga(Bn2DT3A)]. The biodistribution of [68Ga][Ga(Bn2DT3A)(OH)]- in healthy rats showed rapid clearance and excretion via the kidneys, with no uptake seen in the lungs or bones.
Collapse
Affiliation(s)
- Thomas W Price
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, U.K.,Department of Biomedical Sciences, University of Hull, Cottingham Road, Hull HU6 7RX, U.K.,Positron Emission Tomography Research Center, University of Hull, Cottingham Road, Hull HU6 7RX, U.K
| | - Isaline Renard
- Department of Biomedical Sciences, University of Hull, Cottingham Road, Hull HU6 7RX, U.K.,Positron Emission Tomography Research Center, University of Hull, Cottingham Road, Hull HU6 7RX, U.K
| | - Timothy J Prior
- Chemistry, University of Hull, Cottingham Road, Hull HU6 7RX, U.K
| | - Vojtěch Kubíček
- Department of Inorganic Chemistry, Faculty of Science, Charles University, Hlavova 2030, Prague 2, Czech Republic
| | - David M Benoit
- E.A. Milne Centre for Astrophysics, Department of Physics and Mathematics, University of Hull, Cottingham Road, Hull HU6 7RX, U.K
| | - Stephen J Archibald
- Department of Biomedical Sciences, University of Hull, Cottingham Road, Hull HU6 7RX, U.K.,Positron Emission Tomography Research Center, University of Hull, Cottingham Road, Hull HU6 7RX, U.K
| | - Anne-Marie Seymour
- Department of Biomedical Sciences, University of Hull, Cottingham Road, Hull HU6 7RX, U.K
| | - Petr Hermann
- Department of Inorganic Chemistry, Faculty of Science, Charles University, Hlavova 2030, Prague 2, Czech Republic
| | - Graeme J Stasiuk
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, U.K
| |
Collapse
|
12
|
Chelation of Theranostic Copper Radioisotopes with S-Rich Macrocycles: From Radiolabelling of Copper-64 to In Vivo Investigation. Molecules 2022; 27:molecules27134158. [PMID: 35807404 PMCID: PMC9268100 DOI: 10.3390/molecules27134158] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 06/21/2022] [Accepted: 06/22/2022] [Indexed: 12/10/2022] Open
Abstract
Copper radioisotopes are generally employed for cancer imaging and therapy when firmly coordinated via a chelating agent coupled to a tumor-seeking vector. However, the biologically triggered Cu2+-Cu+ redox switching may constrain the in vivo integrity of the resulting complex, leading to demetallation processes. This unsought pathway is expected to be hindered by chelators bearing N, O, and S donors which appropriately complements the borderline-hard and soft nature of Cu2+ and Cu+. In this work, the labelling performances of a series of S-rich polyazamacrocyclic chelators with [64Cu]Cu2+ and the stability of the [64Cu]Cu-complexes thereof were evaluated. Among the chelators considered, the best results were obtained with 1,7-bis [2-(methylsulfanyl)ethyl]-4,10,diacetic acid-1,4,7,10-tetraazacyclododecane (DO2A2S). DO2A2S was labelled at high molar activities in mild reaction conditions, and its [64Cu]Cu2+ complex showed excellent integrity in human serum over 24 h. Biodistribution studies in BALB/c nude mice performed with [64Cu][Cu(DO2A2S)] revealed a behavior similar to other [64Cu]Cu-labelled cyclen derivatives characterized by high liver and kidney uptake, which could either be ascribed to transchelation phenomena or metabolic processing of the intact complex.
Collapse
|
13
|
Gao X, Xu J, Yao T, Liu X, Zhang H, Zhan C. Peptide-decorated nanocarriers penetrating the blood-brain barrier for imaging and therapy of brain diseases. Adv Drug Deliv Rev 2022; 187:114362. [PMID: 35654215 DOI: 10.1016/j.addr.2022.114362] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 05/11/2022] [Accepted: 05/25/2022] [Indexed: 12/12/2022]
Abstract
Blood-Brain Barrier (BBB) is one of the most important physiological barriers strictly restricting the substance exchange between blood and brain tissues. While the BBB protects the brain from infections and toxins and maintains brain homeostasis, it is also recognized as the main obstacle to the penetration of therapeutics and imaging agents into the brain. Due to high specificity and affinity, peptides are frequently exploited to decorate nanocarriers across the BBB for diagnosis and/or therapy purposes. However, there are still some challenges that restrict their clinical application, such as stability, safety and immunocompatibility. In this review, we summarize the biological and pathophysiological characteristics of the BBB, strategies across the BBB, and recent progress on peptide decorated nanocarriers for brain diseases diagnosis and therapy. The challenges and opportunities for their translation are also discussed.
Collapse
|
14
|
Brandt F, Ullrich M, Laube M, Kopka K, Bachmann M, Löser R, Pietzsch J, Pietzsch HJ, van den Hoff J, Wodtke R. "Clickable" Albumin Binders for Modulating the Tumor Uptake of Targeted Radiopharmaceuticals. J Med Chem 2021; 65:710-733. [PMID: 34939412 DOI: 10.1021/acs.jmedchem.1c01791] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The intentional binding of radioligands to albumin gains increasing attention in the context of radiopharmaceutical cancer therapy as it can lead to an enhanced radioactivity uptake into the tumor lesions and, thus, to a potentially improved therapeutic outcome. However, the influence of the radioligand's albumin-binding affinity on the time profile of tumor uptake has been only partly addressed so far. Based on the previously identified Nε-4-(4-iodophenyl)butanoyl-lysine scaffold, we designed "clickable" lysine-derived albumin binders (cLABs) and determined their dissociation constants toward albumin by novel assay methods. Structure-activity relationships were derived, and selected cLABs were applied for the modification of the somatostatin receptor subtype 2 ligand (Tyr3)octreotate. These novel conjugates were radiolabeled with copper-64 and subjected to a detailed in vitro and in vivo radiopharmacological characterization. Overall, the results of this study provide an incentive for further investigations of albumin binders for applications in endoradionuclide therapies.
Collapse
Affiliation(s)
- Florian Brandt
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany.,Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, 01069 Dresden, Germany
| | - Martin Ullrich
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Markus Laube
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Klaus Kopka
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany.,Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, 01069 Dresden, Germany
| | - Michael Bachmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany.,National Center for Tumor Diseases (NCT) Dresden, University Hospital Carl Gustav Carus, Fetscherstraße 74, 01307 Dresden, Germany
| | - Reik Löser
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany.,Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, 01069 Dresden, Germany
| | - Jens Pietzsch
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany.,Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, 01069 Dresden, Germany
| | - Hans-Jürgen Pietzsch
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany.,Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, 01069 Dresden, Germany
| | - Jörg van den Hoff
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany.,Technische Universität Dresden, Department of Nuclear Medicine, University Hospital Carl Gustav Carus, Fetscherstraße 74, 01307 Dresden, Germany
| | - Robert Wodtke
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany
| |
Collapse
|
15
|
Steiger K, Quigley NG, Groll T, Richter F, Zierke MA, Beer AJ, Weichert W, Schwaiger M, Kossatz S, Notni J. There is a world beyond αvβ3-integrin: Multimeric ligands for imaging of the integrin subtypes αvβ6, αvβ8, αvβ3, and α5β1 by positron emission tomography. EJNMMI Res 2021; 11:106. [PMID: 34636990 PMCID: PMC8506476 DOI: 10.1186/s13550-021-00842-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/27/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND In the context of nuclear medicine and theranostics, integrin-related research and development was, for most of the time, focused predominantly on 'RGD peptides' and the subtype αvβ3-integrin. However, there are no less than 24 known integrins, and peptides without the RGD sequence as well as non-peptidic ligands play an equally important role as selective integrin ligands. On the other hand, multimerization is a well-established method to increase the avidity of binding structures, but multimeric radiopharmaceuticals have not made their way into clinics yet. In this review, we describe how these aspects have been interwoven in the framework of the German Research Foundation's multi-group interdisciplinary funding scheme CRC 824, yielding a series of potent PET imaging agents for selective imaging of various integrin subtypes. RESULTS The gallium-68 chelator TRAP was utilized to elaborate symmetrical trimers of various peptidic and non-peptidic integrin ligands. Preclinical data suggested a high potential of the resulting Ga-68-tracers for PET-imaging of the integrins α5β1, αvβ8, αvβ6, and αvβ3. For the first three, we provide some additional immunohistochemistry data in human cancers, which suggest several future clinical applications. Finally, application of αvβ3- and αvβ6-integrin tracers in pancreatic carcinoma patients revealed that unlike αvβ3-targeted PET, αvβ6-integrin PET is not characterized by off-target uptake and thus, enables a substantially improved imaging of this type of cancer. CONCLUSIONS Novel radiopharmaceuticals targeting a number of different integrins, above all, αvβ6, have proven their clinical potential and will play an increasingly important role in future theranostics.
Collapse
Affiliation(s)
- Katja Steiger
- Institut Für Pathologie Und Pathologische Anatomie, Technische Universität München, Munich, Germany
| | - Neil Gerard Quigley
- Institut Für Pathologie Und Pathologische Anatomie, Technische Universität München, Munich, Germany
| | - Tanja Groll
- Institut Für Pathologie Und Pathologische Anatomie, Technische Universität München, Munich, Germany
| | - Frauke Richter
- Institut Für Pathologie Und Pathologische Anatomie, Technische Universität München, Munich, Germany
| | | | | | - Wilko Weichert
- Institut Für Pathologie Und Pathologische Anatomie, Technische Universität München, Munich, Germany
| | - Markus Schwaiger
- Klinik Für Nuklearmedizin Und Zentralinstitut Für Translationale Krebsforschung (TranslaTUM), Klinikum Rechts Der Isar der Technischen Universität München, Munich, Germany
| | - Susanne Kossatz
- Klinik Für Nuklearmedizin Und Zentralinstitut Für Translationale Krebsforschung (TranslaTUM), Klinikum Rechts Der Isar der Technischen Universität München, Munich, Germany
| | - Johannes Notni
- Institut Für Pathologie Und Pathologische Anatomie, Technische Universität München, Munich, Germany. .,Experimental Radiopharmacy, Clinic for Nuclear Medicine, University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany.
| |
Collapse
|
16
|
Peng T, Wang X, Li Z, Bi L, Gao J, Yang M, Wang Y, Yao X, Shan H, Jin H. Preclinical Evaluation of [ 64Cu]NOTA-CP01 as a PET Imaging Agent for Metastatic Esophageal Squamous Cell Carcinoma. Mol Pharm 2021; 18:3638-3648. [PMID: 34424706 DOI: 10.1021/acs.molpharmaceut.1c00600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Targeting metastatic esophageal squamous cell carcinoma (ESCC) has been a challenge in clinical practice. Emerging evidence demonstrates that C-X-C chemokine receptor 4 (CXCR4) highly expresses in ESCC and plays a pivotal role in the process of tumor metastasis. We developed a copper-64 (t1/2 = 12.7 h, 19% beta+) labeling route of NOTA-CP01 derived from LY2510924, a cyclopeptide-based CXCR4 potent antagonist, in an attempt to noninvasively visualize CXCR4 expression in metastatic ESCC. Precursor NOTA-CP01 was designed by modifying the C-terminus of LY2510925 with bis-t-butyl NOTA via a butane-1,4-diamine linker. The radiolabeling process was finished within 15 min with high radiochemical yield (>95%), radiochemical purity (>99%), and specific activity (10.5-21 GBq/μmol) (non-decay-corrected). The in vitro solubility and stability tests revealed that [64Cu]NOTA-CP01 had a high water solubility (log P = -3.44 ± 0.12, n = 5) and high stability in saline and fetal bovine serum. [64Cu]NOTA-CP01 exhibited CXCR4-specific binding with a nanomolar affinity (IC50 = 1.61 ± 0.96 nM, Kd = 0.272 ± 0.14 nM) similar to that of the parental LY2510924. The in vitro cell uptake assay indicated that the [64Cu]NOTA-CP01-selective accumulation in EC109 cells was CXCR4-specific. Molecular docking of the CXCR4/NOTA-CP01 complex suggested that the Lys, Arg, and NOTA of this ligand have a strong polar interaction with the key residues of CXCR4, which explains the tight affinity of [64Cu]NOTA-CP01 for CXCR4. To test the target engagement in vivo, prolonged-time positron emission computed tomography (PET) imaging was performed at 0.5, 4, 6, 8, 12, 16, and 24 h postinjection of [64Cu]NOTA-CP01 to the EC109 tumor-bearing mice. The EC109 tumors were most visible with high contrast to the contralateral background at 6 h postinjection. The tracer revealed receptor-specific tumor accumulation, which was illustrated by effective blocking via coinjection with a blocking dose of LY2510924. Quantification analysis of the prolonged-time images showed that there was obvious radioactivity accumulation in the tumor (1.27 ± 0.19%ID/g) with the best tumor-to-blood ratio (4.79 ± 0.06) and tumor-to-muscle ratio (15.44 ± 2.94) at 6 h postinjection of the probe. The immunofluorescence and immunohistochemistry confirmed the positive expression of CXCR4 in the EC109 tumor and ESCC and metastatic lymph nodes of patients, respectively. We concluded that [64Cu]NOTA-CP01 possessed a very high target engagement for CXCR4-positive ESCC and could be a potential candidate in the clinical detection of metastatic ESCC.
Collapse
Affiliation(s)
- Tukang Peng
- Guangdong Provincial Key Laboratory of Biomedical Imaging, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Xiaohui Wang
- Guangdong Provincial Key Laboratory of Biomedical Imaging, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Zhijun Li
- Guangdong Provincial Key Laboratory of Biomedical Imaging, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Lei Bi
- Guangdong Provincial Key Laboratory of Biomedical Imaging, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Jiebing Gao
- Department of Radiology, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Min Yang
- Guangdong Provincial Key Laboratory of Biomedical Imaging, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Yuwei Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau 999078, China
- College of Pharmacy, Shaanxi University of Chinese Medicine, Shiji Ave., Xi'an-Xianyang New Economic Zone, Xianyang, Shaanxi Province 712046, China
| | - Xiaojun Yao
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Taipa, Macau 999078, China
| | - Hong Shan
- Guangdong Provincial Key Laboratory of Biomedical Imaging, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
- Department of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| | - Hongjun Jin
- Guangdong Provincial Key Laboratory of Biomedical Imaging, Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province 519000, China
| |
Collapse
|
17
|
Martin S, Maus S, Stemler T, Rosar F, Khreish F, Holland JP, Ezziddin S, Bartholomä MD. Proof-of-Concept Study of the NOTI Chelating Platform: Preclinical Evaluation of 64Cu-Labeled Mono- and Trimeric c(RGDfK) Conjugates. Mol Imaging Biol 2021; 23:95-108. [PMID: 32856224 PMCID: PMC7782405 DOI: 10.1007/s11307-020-01530-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/14/2020] [Accepted: 08/09/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE We recently developed a chelating platform based on the macrocycle 1,4,7-triazacyclononane with up to three five-membered azaheterocyclic arms for the preparation of 68Ga- and 64Cu-based radiopharmaceuticals. Based on this platform, the chelator scaffold NOTI-TVA with three additional carboxylic acid groups for bioconjugation was synthesized and characterized. The primary aims of this proof-of-concept study were (1) to evaluate if trimeric radiotracers on the basis of the NOTI-TVA 6 scaffold can be developed, (2) to determine if the additional substituents for bioconjugation at the non-coordinating NH atoms of the imidazole residues of the building block NOTI influence the metal binding properties, and (3) what influence multiple targeting vectors have on the biological performance of the radiotracer. The cyclic RGDfK peptide that specifically binds to the αvß3 integrin receptor was selected as the biological model system. PROCEDURES Two different synthetic routes for the preparation of NOTI-TVA 6 were explored. Three c(RGDfK) peptide residues were conjugated to the NOTI-TVA 6 building block by standard peptide chemistry providing the trimeric bioconjugate NOTI-TVA-c(RGDfK)3 9. Labeling of 9 with [64Cu]CuCl2 was performed manually at pH 8.2 at ambient temperature. Binding affinities of Cu-8, the Cu2+ complex of the previously described monomer NODIA-Me-c(RGDfK) 8, and the trimer Cu-9 to integrin αvß3 were determined in competitive cell binding experiments in the U-87MG cell line. The pharmacokinetics of both 64Cu-labeled conjugates [64Cu]Cu-8 and [64Cu]Cu-9 were determined by small-animal PET imaging and ex vivo biodistribution studies in mice bearing U-87MG xenografts. RESULTS Depending on the synthetic route, NOTI-TVA 6 was obtained with an overall yield up to 58 %. The bioconjugate 9 was prepared in 41 % yield. Both conjugates [64Cu]Cu-8 and [64Cu]Cu-9 were radiolabeled quantitatively at ambient temperature in high molar activities of Am ~ 20 MBq nmol-1 in less than 5 min. Competitive inhibitory constants IC50 of c(RDGfK) 7, Cu-8, and Cu-9 were determined to be 159.5 ± 1.3 nM, 256.1 ± 2.1 nM, and 99.5 ± 1.1 nM, respectively. In small-animal experiments, both radiotracers specifically delineated αvß3 integrin-positive U-87MG tumors with low uptake in non-target organs and rapid blood clearance. The trimer [64Cu]Cu-9 showed a ~ 2.5-fold higher tumor uptake compared with the monomer [64Cu]Cu-8. CONCLUSIONS Functionalization of NOTI at the non-coordinating NH atoms of the imidazole residues for bioconjugation was straightforward and allowed the preparation of a homotrimeric RGD conjugate. After optimization of the synthesis, required building blocks to make NOTI-TVA 6 are now available on multi-gram scale. Modifications at the imidazole groups had no measurable impact on metal binding properties in vitro and in vivo suggesting that the NOTI scaffold is a promising candidate for the development of 64Cu-labeled multimeric/multifunctional radiotracers.
Collapse
Affiliation(s)
- Sebastian Martin
- Department of Nuclear Medicine, Saarland University - Medical Center, Kirrbergerstrasse, D-66421, Homburg, Germany
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital, Rue de Bugnon 25A, CH-1011, Lausanne, Switzerland
| | - Stephan Maus
- Department of Nuclear Medicine, Saarland University - Medical Center, Kirrbergerstrasse, D-66421, Homburg, Germany
| | - Tobias Stemler
- Department of Nuclear Medicine, Saarland University - Medical Center, Kirrbergerstrasse, D-66421, Homburg, Germany
| | - Florian Rosar
- Department of Nuclear Medicine, Saarland University - Medical Center, Kirrbergerstrasse, D-66421, Homburg, Germany
| | - Fadi Khreish
- Department of Nuclear Medicine, Saarland University - Medical Center, Kirrbergerstrasse, D-66421, Homburg, Germany
| | - Jason P Holland
- Department of Chemistry, University of Zurich, Winterthurerstrasse 190, CH-8057, Zurich, Switzerland
| | - Samer Ezziddin
- Department of Nuclear Medicine, Saarland University - Medical Center, Kirrbergerstrasse, D-66421, Homburg, Germany
| | - Mark D Bartholomä
- Department of Nuclear Medicine, Saarland University - Medical Center, Kirrbergerstrasse, D-66421, Homburg, Germany.
- Department of Nuclear Medicine, University of Freiburg - Medical Center, Hugstetterstrasse 55, 79106, Freiburg, Germany.
| |
Collapse
|
18
|
Rangger C, Haubner R. Radiolabelled Peptides for Positron Emission Tomography and Endoradiotherapy in Oncology. Pharmaceuticals (Basel) 2020; 13:E22. [PMID: 32019275 PMCID: PMC7169460 DOI: 10.3390/ph13020022] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 02/07/2023] Open
Abstract
This review deals with the development of peptide-based radiopharmaceuticals for the use with positron emission tomography and peptide receptor radiotherapy. It discusses the pros and cons of this class of radiopharmaceuticals as well as the different labelling strategies, and summarises approaches to optimise metabolic stability. Additionally, it presents different target structures and addresses corresponding tracers, which are already used in clinical routine or are being investigated in clinical trials.
Collapse
Affiliation(s)
| | - Roland Haubner
- Department of Nuclear Medicine, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria;
| |
Collapse
|
19
|
Zaibaq NG, Pollard AC, Collins MJ, Pisaneschi F, Pagel MD, Wilson LJ. Evaluation of the Biodistribution of Serinolamide-Derivatized C 60 Fullerene. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E143. [PMID: 31941058 PMCID: PMC7023239 DOI: 10.3390/nano10010143] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 12/31/2019] [Accepted: 01/08/2020] [Indexed: 12/12/2022]
Abstract
Carbon nanoparticles have consistently been of great interest in medicine. However, there are currently no clinical materials based on carbon nanoparticles, due to inconsistent biodistribution and excretion data. In this work, we have synthesized a novel C60 derivative with a metal chelating agent (1,4,7-Triazacyclononane-1,4,7-triacetic acid; NOTA) covalently bound to the C60 cage and radiolabeled with copper-64 (t1/2 = 12.7 h). Biodistribution of the material was assessed in vivo using positron emission tomography (PET). Bingel-Hirsch chemistry was employed to functionalize the fullerene cage with highly water-soluble serinolamide groups allowing this new C60 conjugate to clear quickly from mice almost exclusively through the kidneys. Comparing the present results to the larger context of reports of biocompatible fullerene derivatives, this work offers an important evaluation of the in vivo biodistribution, using experimental evidence to establish functionalization guidelines for future C60-based biomedical platforms.
Collapse
Affiliation(s)
- Nicholas G. Zaibaq
- Department of Chemistry and Smalley-Curl Institute, Rice University, 6100 Main St, Houston, TX 77005, USA; (N.G.Z.); (A.C.P.); (M.J.C.)
| | - Alyssa C. Pollard
- Department of Chemistry and Smalley-Curl Institute, Rice University, 6100 Main St, Houston, TX 77005, USA; (N.G.Z.); (A.C.P.); (M.J.C.)
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, 1881 East Rd, Houston, TX 77054, USA;
| | - Michael J. Collins
- Department of Chemistry and Smalley-Curl Institute, Rice University, 6100 Main St, Houston, TX 77005, USA; (N.G.Z.); (A.C.P.); (M.J.C.)
| | - Federica Pisaneschi
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, 1881 East Rd, Houston, TX 77054, USA;
| | - Mark D. Pagel
- Department of Chemistry and Smalley-Curl Institute, Rice University, 6100 Main St, Houston, TX 77005, USA; (N.G.Z.); (A.C.P.); (M.J.C.)
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, 1881 East Rd, Houston, TX 77054, USA;
| | - Lon J. Wilson
- Department of Chemistry and Smalley-Curl Institute, Rice University, 6100 Main St, Houston, TX 77005, USA; (N.G.Z.); (A.C.P.); (M.J.C.)
| |
Collapse
|
20
|
Comparison of DOTA and NODAGA as chelates for 68Ga-labelled CDP1 as novel infection PET imaging agents. J Radioanal Nucl Chem 2019. [DOI: 10.1007/s10967-019-06693-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
21
|
Kaeopookum P, Petrik M, Summer D, Klinger M, Zhai C, Rangger C, Haubner R, Haas H, Hajduch M, Decristoforo C. Comparison of 68Ga-labeled RGD mono- and multimers based on a clickable siderophore-based scaffold. Nucl Med Biol 2019; 78-79:1-10. [PMID: 31678781 DOI: 10.1016/j.nucmedbio.2019.09.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 09/23/2019] [Accepted: 09/30/2019] [Indexed: 02/06/2023]
Abstract
Cyclic pentapeptides containing the amino acid sequence arginine-glycine-aspartic (RGD) have been widely applied to target αvβ3 integrin, which is upregulated in various tumors during tumor-induced angiogenesis. Multimeric cyclic RGD peptides have been reported to be advantageous over monomeric counterparts for angiogenesis imaging. Here, we prepared mono-, di-, and trimeric cyclic arginine-glycine-aspartic-D-phenylalanine-lysine (c (RGDfK)) derivatives by conjugation with the natural chelator fusarinine C (FSC) using click chemistry based on copper (I)-catalyzed azide-alkyne cycloaddition (CuAAC). The αvβ3 binding properties of 68Ga-labeled mono-, di-, and trimeric c(RGDfK) peptides were evaluated in vitro as well as in vivo and compared with the references monomeric [68Ga]GaNODAGA-c(RGDfK) and trimeric [68Ga]GaFSC(suc-c(RGDfK))3. All 68Ga-labeled c(RGDfK) peptides displayed hydrophilicity (logD = -2.96 to -3.80), low protein binding and were stable in phosphate buffered-saline (PBS) and serum up to 2 h. In vitro internalization assays with human melanoma M21 (αvβ3-positive) and M21-L (αvβ3-negative) cell lines showed specific uptake of all derivatives and increased in the series: mono- < di- < trimeric peptide. The highest tumor uptake, tumor-to-background ratios, and image contrast were found for the dimeric [68Ga]GaMAFC(c(RGDfK)aza)2. In conclusion, we developed a novel strategy for direct, straight forward preparation of mono-, di-, and trimeric c(RGDfK) conjugates based on the FSC scaffold. Interestingly, the best αvβ3 imaging properties were found for the dimeric [68Ga]GaMAFC(c(RGDfK)aza)2.
Collapse
Affiliation(s)
- Piriya Kaeopookum
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria; Research and Development Division, Thailand Institute of Nuclear Technology, Nakhon Nayok, Thailand
| | - Milos Petrik
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Dominik Summer
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Maximilian Klinger
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Chuangyan Zhai
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria; School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Christine Rangger
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Roland Haubner
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria
| | - Hubertus Haas
- Division of Molecular Biology, Biocenter, Medical University Innsbruck, Innsbruck, Austria
| | - Marian Hajduch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacky University, Olomouc, Czech Republic
| | - Clemens Decristoforo
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria.
| |
Collapse
|
22
|
Vatsa R, Shukla J, Kumar S, Chakraboarty S, Dash A, Singh G, Mittal BR. Effect of Macro-Cyclic Bifunctional Chelators DOTA and NODAGA on Radiolabeling and In Vivo Biodistribution of Ga-68 Cyclic RGD Dimer. Cancer Biother Radiopharm 2019; 34:427-435. [DOI: 10.1089/cbr.2019.2811] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Rakhee Vatsa
- Department of Nuclear Medicine and PET, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Jaya Shukla
- Department of Nuclear Medicine and PET, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Sunil Kumar
- Department of Nuclear Medicine and PET, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | | | - Ashutosh Dash
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Gurpreet Singh
- Department of General Surgery, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Bhagwant Rai Mittal
- Department of Nuclear Medicine and PET, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
23
|
Mansi R, Fani M. Design and development of the theranostic pair 177 Lu-OPS201/ 68 Ga-OPS202 for targeting somatostatin receptor expressing tumors. J Labelled Comp Radiopharm 2019; 62:635-645. [PMID: 31112618 DOI: 10.1002/jlcr.3755] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/03/2019] [Accepted: 05/13/2019] [Indexed: 12/26/2022]
Abstract
Radiolabeled somatostatin receptor (sstr) antagonists have shown superiority in different preclinical and clinical settings compared with the well-established and clinically used agonists for targeting sstr-expressing tumors, with regard to pharmacokinetics, tumor uptake, and retention. The theranostic pair 177 Lu-OPS201/68 Ga-OPS202, based on the sstr2 antagonist JR11 (Cpa-c[d-Cys-Aph(Hor)-d-Aph(Cbm)-Lys-Thr-Cys]-d-Tyr-NH2 ), is the most advanced pair of the antagonist family in terms of preclinical development and is currently under clinical evaluation. OPS201 and OPS202 share the same amino acid sequence (JR11) but feature different conjugated chelators needed for radiolabeling, DOTA for OPS201 and NODAGA for OPS202. In this review, the design and development of the peptidic analog, JR11, and the selection of chelators and radiometals that led to 177 Lu-OPS201/68 Ga-OPS202 are discussed. Furthermore, the preclinical evaluation of both radiolabeled analogs from bench to bedside and the clinical trials involving the theranostic pair are presented.
Collapse
Affiliation(s)
- Rosalba Mansi
- Division of Radiopharmaceutical Chemistry, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Melpomeni Fani
- Division of Radiopharmaceutical Chemistry, University Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
24
|
Thomas G, Boudon J, Maurizi L, Moreau M, Walker P, Severin I, Oudot A, Goze C, Poty S, Vrigneaud JM, Demoisson F, Denat F, Brunotte F, Millot N. Innovative Magnetic Nanoparticles for PET/MRI Bimodal Imaging. ACS OMEGA 2019; 4:2637-2648. [PMID: 31459499 PMCID: PMC6648431 DOI: 10.1021/acsomega.8b03283] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 01/14/2019] [Indexed: 05/21/2023]
Abstract
Superparamagnetic iron oxide nanoparticles were developed as positron emission tomography (PET) and magnetic resonance imaging (MRI) bimodal imaging agents. These nanoparticles (NPs), with a specific nanoflower morphology, were first synthesized and simultaneously functionalized with 3,4-dihydroxy-l-phenylalanine (LDOPA) under continuous hydrothermal conditions. The resulting NPs exhibited a low hydrodynamic size of 90 ± 2 nm. The functional groups of LDOPA (-NH2 and -COOH) were successfully used for the grafting of molecules of interest in a second step. The nanostructures were modified by poly(ethylene glycol) (PEG) and a new macrocyclic chelator MANOTA for further 64Cu radiolabeling for PET imaging. The functionalized NPs showed promising bimodal (PET and MRI) imaging capability with high r 2 and r 2* (T 2 and T 2* relaxivities) values and good stability. They were mainly uptaken from liver and kidneys. No cytotoxicity effect was observed. These NPs appear as a good candidate for bimodal tracers in PET/MRI.
Collapse
Affiliation(s)
- Guillaume Thomas
- ICB
UMR 6303 CNRS-Université Bourgogne Franche-Comté, 21000 Dijon, France
| | - Julien Boudon
- ICB
UMR 6303 CNRS-Université Bourgogne Franche-Comté, 21000 Dijon, France
| | - Lionel Maurizi
- ICB
UMR 6303 CNRS-Université Bourgogne Franche-Comté, 21000 Dijon, France
| | - Mathieu Moreau
- ICMUB
UMR 6302 CNRS-Université Bourgogne Franche-Comté, 21000 Dijon, France
| | - Paul Walker
- Département
de Spectroscopie par Résonance Magnétique, CHU Dijon, 21000 Dijon, France
| | - Isabelle Severin
- UBFC-AgrosupDijon-INSERM
U 1231, 1 Esplanade Erasme, 21000 Dijon, France
| | - Alexandra Oudot
- Plateforme
d’Imagerie Préclinique, Service de Médecine Nucléaire, Centre Georges François Leclerc, 21000 Dijon, France
| | - Christine Goze
- ICMUB
UMR 6302 CNRS-Université Bourgogne Franche-Comté, 21000 Dijon, France
| | - Sophie Poty
- ICMUB
UMR 6302 CNRS-Université Bourgogne Franche-Comté, 21000 Dijon, France
| | - Jean-Marc Vrigneaud
- Plateforme
d’Imagerie Préclinique, Service de Médecine Nucléaire, Centre Georges François Leclerc, 21000 Dijon, France
| | - Fréderic Demoisson
- ICB
UMR 6303 CNRS-Université Bourgogne Franche-Comté, 21000 Dijon, France
| | - Franck Denat
- ICMUB
UMR 6302 CNRS-Université Bourgogne Franche-Comté, 21000 Dijon, France
| | - François Brunotte
- Plateforme
d’Imagerie Préclinique, Service de Médecine Nucléaire, Centre Georges François Leclerc, 21000 Dijon, France
| | - Nadine Millot
- ICB
UMR 6303 CNRS-Université Bourgogne Franche-Comté, 21000 Dijon, France
- E-mail:
| |
Collapse
|
25
|
Jung S, Anusha JR, Park S, Yu KH, Raj CJ, Kim BC. HER2 inhibition efficiency of 6-amino-2-methyl-2-phenethyl-2H-benzopyran and feasibility of the 64Cu-labeled benzopyran derivative in cancer diagnosis. NEW J CHEM 2019. [DOI: 10.1039/c9nj02893e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The benzopyran derivative 6-amino-2-methyl-2-phenethyl-2H-benzopyran inhibits the overexpression of the protein HER2, and the 64Cu-labeled compound is promising for cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Soonjae Jung
- Division of Applied RI
- Korea Institute of Radiological & Medical Sciences
- Seoul-01812
- Republic of Korea
| | - J. R. Anusha
- Department of Chemistry
- Dongguk University
- Seoul-04620
- Republic of Korea
- Department of Advanced Zoology and Biotechnology
| | - Seungil Park
- Department of Chemistry
- Dongguk University
- Seoul-04620
- Republic of Korea
| | - Kook Hyun Yu
- Department of Chemistry
- Dongguk University
- Seoul-04620
- Republic of Korea
| | - C. Justin Raj
- Department of Chemistry
- Dongguk University
- Seoul-04620
- Republic of Korea
| | - Byung Chul Kim
- Department of Printed Electronics Engineering
- Sunchon National University
- 255, Jungang-ro
- Suncheon-si
- Republic of Korea
| |
Collapse
|
26
|
Collignon AM, Lesieur J, Anizan N, Azzouna RB, Poliard A, Gorin C, Letourneur D, Chaussain C, Rouzet F, Rochefort GY. Early angiogenesis detected by PET imaging with 64Cu-NODAGA-RGD is predictive of bone critical defect repair. Acta Biomater 2018; 82:111-121. [PMID: 30312778 DOI: 10.1016/j.actbio.2018.10.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 10/04/2018] [Accepted: 10/07/2018] [Indexed: 12/15/2022]
Abstract
Therapies using stem cells may be applicable to all fields of regenerative medicine, including craniomaxillofacial surgery. Dental pulp stem cells (DPSCs) have demonstrated in vitro and in vivo osteogenic and proangiogenic properties. The aim of the study was to evaluate whether early angiogenesis investigated by nuclear imaging can predict bone formation within a mouse critical bone defect. Two symmetrical calvarial critical-sized defects were created. Defects were left empty or filled with i) DPSC-containing dense collagen scaffold, ii) 5% hypoxia-primed DPSC-containing dense collagen scaffold, iii) acellular dense collagen scaffold, or iv) left empty. Early angiogenesis assessed by PET using 64Cu-NODAGA-RGD as a tracer was found to be correlated with bone formation determined by micro-CT within the defects from day 30, and to be correlated to the late calcium apposition observed at day 90 using 18F-Na PET. These results suggest that nuclear imaging of angiogenesis, a technique applicable in clinical practice, is a promising approach for early prediction of bone grafting outcome, thus potentially allowing to anticipate alternative regenerative strategies. STATEMENT OF SIGNIFICANCE: Bone defects are a major concern in medicine. As life expectancy increases, the number of bone lesions grows, and occurring complications lead to a delay or even lack of consolidation. Therefore, to be able to predict healing or the absence of scarring at early times would be very interesting. This would not "waste time" for the patient. We report here that early nuclear imaging of angiogenesis, using 64Cu-NODAGA-RGD as a tracer, associated with nuclear imaging of mineralization, using 18F-Na as a tracer, is correlated to late bone healing objectivized by classical histology and microtomography. This nuclear imaging represents a promising approach for early prediction of bone grafting outcome in clinical practice, thus potentially allowing to anticipate alternative regenerative strategies.
Collapse
Affiliation(s)
- Anne-Margaux Collignon
- EA 2496 Orofacial Pathologies, Imagery and Biotherapies, Dental School Faculty, University Paris Descartes and Life Imaging Platform (PIV), Montrouge, France; University Hospitals, AP-HP, Paris, France
| | - Julie Lesieur
- EA 2496 Orofacial Pathologies, Imagery and Biotherapies, Dental School Faculty, University Paris Descartes and Life Imaging Platform (PIV), Montrouge, France
| | - Nadège Anizan
- Fédération de Recherche en Imagerie Multimodale (FRIM), Inserm UMS-34, Université Paris Diderot, Paris, France
| | - Rana Ben Azzouna
- University Hospitals, AP-HP, Paris, France; Fédération de Recherche en Imagerie Multimodale (FRIM), Inserm UMS-34, Université Paris Diderot, Paris, France; INSERM U1148, Laboratory of Vascular Translational Science, University Paris Diderot, University Paris 13, X Bichat Hospital, and Département Hospitalo-Universitaire (DHU) FIRE, F-75018 Paris, France
| | - Anne Poliard
- EA 2496 Orofacial Pathologies, Imagery and Biotherapies, Dental School Faculty, University Paris Descartes and Life Imaging Platform (PIV), Montrouge, France
| | - Caroline Gorin
- EA 2496 Orofacial Pathologies, Imagery and Biotherapies, Dental School Faculty, University Paris Descartes and Life Imaging Platform (PIV), Montrouge, France; University Hospitals, AP-HP, Paris, France
| | - Didier Letourneur
- INSERM U1148, Laboratory of Vascular Translational Science, University Paris Diderot, University Paris 13, X Bichat Hospital, and Département Hospitalo-Universitaire (DHU) FIRE, F-75018 Paris, France
| | - Catherine Chaussain
- EA 2496 Orofacial Pathologies, Imagery and Biotherapies, Dental School Faculty, University Paris Descartes and Life Imaging Platform (PIV), Montrouge, France; University Hospitals, AP-HP, Paris, France
| | - Francois Rouzet
- University Hospitals, AP-HP, Paris, France; Fédération de Recherche en Imagerie Multimodale (FRIM), Inserm UMS-34, Université Paris Diderot, Paris, France; INSERM U1148, Laboratory of Vascular Translational Science, University Paris Diderot, University Paris 13, X Bichat Hospital, and Département Hospitalo-Universitaire (DHU) FIRE, F-75018 Paris, France.
| | - Gael Y Rochefort
- EA 2496 Orofacial Pathologies, Imagery and Biotherapies, Dental School Faculty, University Paris Descartes and Life Imaging Platform (PIV), Montrouge, France.
| |
Collapse
|
27
|
Ocak M, Beaino W, White A, Zeng D, Cai Z, Anderson CJ. 64Cu-Labeled Phosphonate Cross-Bridged Chelator Conjugates of c(RGDyK) for PET/CT Imaging of Osteolytic Bone Metastases. Cancer Biother Radiopharm 2018; 33:74-83. [PMID: 29634417 DOI: 10.1089/cbr.2017.2419] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVE The goal of this research was to evaluate c(RGDyK) conjugated to phosphonate-based cross-bridged chelators using Cu-free click chemistry in the 4T1 mouse mammary tumor bone metastasis model in comparison with 64Cu-CB-TE2A-c(RGDyK), which previously showed selective binding to integrin αvβ3 on osteoclasts. EXPERIMENTAL Two phosphonate-based cross-bridged chelators (CB-TE1A1P and CB-TE1K1P) were conjugated to c(RGDyK) through bio-orthogonal strain-promoted alkyne-azide cycloaddition. In vitro and in vivo evaluation of the 64Cu-labeled TE1A1P-DBCO-c(RGDyK) (AP-c(RGDyK)), TE1K1P-PEG4-DBCO-c(RGDyK) (KP-c(RGDyK)), and CB-TE2A-c(RGDyK) were compared in the 4T1 mouse model of bone metastasis. The affinities of the unconjugated and chelator-c(RGDyK) analogs for αvβ3 integrin were determined using a competitive-binding assay. For in vivo evaluation, BALB/c mice were injected with 1 × 105 4T1/Luc cells in the left ventricle. Formation of metastases was monitored by bioluminescence imaging (BLI) followed by small-animal PET/CT 2 h postinjection of radiotracers. RESULTS The chelator-peptide conjugates showed similar affinity to integrin αvβ3, in the low nM range. PET imaging demonstrated a higher uptake in bones having metastases for all 64Cu-labeled c(RGDyK) analogs compared with bones in nontumor-bearing mice. The correlation between uptake of 64Cu-AP-c(RGDyK) and 64Cu-KP-c(RGDyK) in bones with metastases based on PET/CT imaging, and osteoclast number based on histomorphometry, was improved over the previously investigated 64Cu-CB-TE2A-c(RGDyK). CONCLUSION These data suggest that the phosphonate chelator conjugates of c(RDGyK) peptides are promising PET tracers suitable for imaging tumor-associated osteoclasts in bone metastases.
Collapse
Affiliation(s)
- Meltem Ocak
- 1 Department of Radiology, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Wissam Beaino
- 1 Department of Radiology, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Alexander White
- 1 Department of Radiology, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Dexing Zeng
- 1 Department of Radiology, University of Pittsburgh , Pittsburgh, Pennsylvania.,2 Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Zhengxin Cai
- 1 Department of Radiology, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Carolyn J Anderson
- 1 Department of Radiology, University of Pittsburgh , Pittsburgh, Pennsylvania.,2 Department of Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania.,3 Department of Pharmacology and Chemical Biology, University of Pittsburgh , Pittsburgh, Pennsylvania.,4 Department of Bioengineering, University of Pittsburgh , Pittsburgh, Pennsylvania
| |
Collapse
|
28
|
Debordeaux F, Chansel-Debordeaux L, Pinaquy JB, Fernandez P, Schulz J. What about αvβ3 integrins in molecular imaging in oncology? Nucl Med Biol 2018; 62-63:31-46. [DOI: 10.1016/j.nucmedbio.2018.04.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 03/19/2018] [Accepted: 04/30/2018] [Indexed: 10/17/2022]
|
29
|
Läppchen T, Holland JP, Kiefer Y, Bartholomä MD. Preparation and preclinical evaluation of a 68Ga-labelled c(RGDfK) conjugate comprising the bifunctional chelator NODIA-Me. EJNMMI Radiopharm Chem 2018; 3:6. [PMID: 29756024 PMCID: PMC5932101 DOI: 10.1186/s41181-018-0043-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 04/10/2018] [Indexed: 11/23/2022] Open
Abstract
Background We recently developed a chelating platform based on the macrocycle 1,4,7-triazacyclononane with up to three, five-membered azaheterocyclic arms for the development of 68Ga- and 64Cu-based radiopharmaceuticals. Here, a 68Ga-labelled conjugate comprising the bifunctional chelator NODIA-Me in combination with the αvß3-targeting peptide c(RGDfK) has been synthesized and characterized. The primary aim was to evaluate further the potential of our NODIA-Me chelating system for the development of 68Ga-labelled radiotracers. Results The BFC NODIA-Me was conjugated to c(RGDfK) by standard peptide chemistry to obtain the final bioconjugate NODIA-Me-c(RGDfK) 3 in 72% yield. Labelling with [68Ga]GaCl3 was accomplished in a fully automated, cGMP compliant process to give [68Ga]3 in high radiochemical yield (98%) and moderate specific activity (~ 8 MBq nmol− 1). Incorporation of the Ga-NODIA-Me chelate to c(RGDfK) 2 had only minimal influence on the affinity to integrin αvß3 (IC50 values [natGa]3 = 205.1 ± 1.4 nM, c(RGDfK) 2 = 159.5 ± 1.3 nM) as determined in competitive cell binding experiments in U-87 MG cell line. In small-animal PET imaging and ex vivo biodistribution studies, the radiotracer [68Ga]3 showed low uptake in non-target organs and specific tumor uptake in U-87 MG tumors. Conclusion The results suggest that the bifunctional chelator NODIA-Me is an interesting alternative to existing ligands for the development of 68Ga-labelled radiopharmaceuticals.
Collapse
Affiliation(s)
- Tilman Läppchen
- Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetterstrasse 55, D-79106 Freiburg, Germany.,2Department of Nuclear Medicine, Inselspital, Bern University Hospital and University of Bern, Freiburgstrasse, CH-3010 Bern, Switzerland
| | - Jason P Holland
- Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetterstrasse 55, D-79106 Freiburg, Germany.,3Department of Chemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Yvonne Kiefer
- Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetterstrasse 55, D-79106 Freiburg, Germany
| | - Mark D Bartholomä
- Department of Nuclear Medicine, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetterstrasse 55, D-79106 Freiburg, Germany
| |
Collapse
|
30
|
Läppchen T, Kiefer Y, Holland JP, Bartholomä MD. In vitro and in vivo evaluation of the bifunctional chelator NODIA-Me in combination with a prostate-specific membrane antigen targeting vector. Nucl Med Biol 2018; 60:45-54. [DOI: 10.1016/j.nucmedbio.2018.03.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 02/14/2018] [Accepted: 03/07/2018] [Indexed: 01/21/2023]
|
31
|
Hedhli J, Slania SLL, Płoska A, Czerwinski A, Konopka CJ, Wozniak M, Banach M, Dobrucki IT, Kalinowski L, Dobrucki LW. Evaluation of a dimeric-cRGD peptide for targeted PET-CT imaging of peripheral angiogenesis in diabetic mice. Sci Rep 2018; 8:5401. [PMID: 29599497 PMCID: PMC5876368 DOI: 10.1038/s41598-018-23372-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 03/06/2018] [Indexed: 11/09/2022] Open
Abstract
The α V β3 integrin plays an important role in many physiological functions and pathological disorders. α V β3 is minimally expressed in normal quiescent endothelial cells, but significantly upregulated during neovascularization. In this study, we evaluated a 64Cu-labeled dimeric cRGD tracer targeted at α V β3 integrin and report its applicability to assess peripheral angiogenesis in diabetes mellitus (DM). We established a murine model of type-1 DM characterized by elevated glucose, glycated serum protein (GSP), and glycated hemoglobin A1c (HbA1c). We demonstrated that our imaging probe is specific to α V β3 integrin under both normo- and hyperglycemic conditions. We found that the analysis of in vivo PET-CT images correlated well with gamma well counting (GWC). Both GWC and PET-CT imaging demonstrated increased uptake of 64Cu-NOTA-PEG4-cRGD2 in the ischemic hindlimb in contrast to non-ischemic control. GWC of the distal ischemic tissue from DM mice showed significantly lower probe accumulation than in non-DM mice. The immunofluorescence staining of the ischemic tissues showed a 3-fold reduction in CD31 and 4-fold reduction in the α V β3 expression in DM vs. non-DM animals. In conclusion, we successfully demonstrated that diabetes-associated reductions in peripheral angiogenesis can be non-invasively detected with PET-CT imaging using targeted dimeric-cRGD probe.
Collapse
Affiliation(s)
- Jamila Hedhli
- Beckman Institute for Advanced Science and Technology, Urbana, IL, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Stephanie L L Slania
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Agata Płoska
- Beckman Institute for Advanced Science and Technology, Urbana, IL, USA
- Department of Medical Laboratory Diagnostics and Central Bank of Frozen Tissues & Genetic Specimens, Medical University of Gdansk, Gdansk, Poland
| | | | - Christian J Konopka
- Beckman Institute for Advanced Science and Technology, Urbana, IL, USA
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Marcin Wozniak
- Beckman Institute for Advanced Science and Technology, Urbana, IL, USA
- Department of Medical Laboratory Diagnostics and Central Bank of Frozen Tissues & Genetic Specimens, Medical University of Gdansk, Gdansk, Poland
| | - Maciej Banach
- Department of Hypertension, Medical University of Lodz, Lodz, Poland
| | - Iwona T Dobrucki
- Beckman Institute for Advanced Science and Technology, Urbana, IL, USA
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics and Central Bank of Frozen Tissues & Genetic Specimens, Medical University of Gdansk, Gdansk, Poland
- Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.PL), Gdansk, Poland
| | - Lawrence W Dobrucki
- Beckman Institute for Advanced Science and Technology, Urbana, IL, USA.
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA.
- Department of Medical Laboratory Diagnostics and Central Bank of Frozen Tissues & Genetic Specimens, Medical University of Gdansk, Gdansk, Poland.
- Biobanking and Biomolecular Resources Research Infrastructure Poland (BBMRI.PL), Gdansk, Poland.
| |
Collapse
|
32
|
Boros E, Pinkhasov OR, Caravan P. Metabolite profiling with HPLC-ICP-MS as a tool for in vivo characterization of imaging probes. EJNMMI Radiopharm Chem 2018; 3:2. [PMID: 29503859 PMCID: PMC5824709 DOI: 10.1186/s41181-017-0037-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 12/26/2017] [Indexed: 11/10/2022] Open
Abstract
Background Current analytical methods for characterizing pharmacokinetic and metabolic properties of positron emission tomography (PET) and single photon emission computed tomography (SPECT) probes are limited. Alternative methods to study tracer metabolism are needed. The study objective was to assess the potential of high performance liquid chromatography - inductively coupled plasma - mass spectrometry (HPLC-ICP-MS) for quantification of molecular probe metabolism and pharmacokinetics using stable isotopes. Methods Two known peptide-DOTA conjugates were chelated with natGa and natIn. Limit of detection of HPLC-ICP-MS for 69Ga and 115In was determined. Rats were administered 50-150 nmol of Ga- and/or In-labeled probes, blood was serially sampled, and plasma analyzed by HPLC-ICP-MS using both reverse phase and size exclusion chromatography. Results The limits of detection were 0.16 pmol for 115In and 0.53 pmol for 69Ga. Metabolites as low as 0.001 %ID/g could be detected and transchelation products identified. Simultaneous administration of Ga- and In-labeled probes allowed the determination of pharmacokinetics and metabolism of both probes in a single animal. Conclusions HPLC-ICP-MS is a robust, sensitive and radiation-free technique to characterize the pharmacokinetics and metabolism of imaging probes.
Collapse
Affiliation(s)
- Eszter Boros
- A. A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Suite 2301, Charlestown, MA 02129 USA.,3Present address: Department of Chemistry, Stony Brook University, 100 Nicolls road, Stony Brook, New York, NY 11790 USA
| | - Omar R Pinkhasov
- A. A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Suite 2301, Charlestown, MA 02129 USA
| | - Peter Caravan
- A. A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, 149 13th Street, Suite 2301, Charlestown, MA 02129 USA.,2Institute for Innovation in Imaging, Department of Radiology, Massachusetts General Hospital, Building 149, Room 2301, 13th Street, Charlestown, Boston, MA 02129 USA
| |
Collapse
|
33
|
Price TW, Greenman J, Stasiuk GJ. Current advances in ligand design for inorganic positron emission tomography tracers 68Ga, 64Cu, 89Zr and 44Sc. Dalton Trans 2018; 45:15702-15724. [PMID: 26865360 DOI: 10.1039/c5dt04706d] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
A key part of the development of metal based Positron Emission Tomography probes is the chelation of the radiometal. In this review the recent developments in the chelation of four positron emitting radiometals, 68Ga, 64Cu, 89Zr and 44Sc, are explored. The factors that effect the chelation of each radio metal and the ideal ligand system will be discussed with regards to high in vivo stability, complexation conditions, conjugation to targeting motifs and complexation kinetics. A series of cyclic, cross-bridged and acyclic ligands will be discussed, such as CP256 which forms stable complexes with 68Ga under mild conditions and PCB-TE2A which has been shown to form a highly stable complex with 64Cu. 89Zr and 44Sc have seen significant development in recent years with a number of chelates being applied to each metal - eight coordinate di-macrocyclic terephthalamide ligands were found to rapidly produce more stable complexes with 89Zr than the widely used DFO.
Collapse
Affiliation(s)
- Thomas W Price
- School of Biological, Biomedical and Environmental Sciences, The University of Hull, HU6 7RX, UK. and Positron Emission Tomography Research Centre, The University of Hull, HU6 7RX, UK
| | - John Greenman
- School of Biological, Biomedical and Environmental Sciences, The University of Hull, HU6 7RX, UK.
| | - Graeme J Stasiuk
- School of Biological, Biomedical and Environmental Sciences, The University of Hull, HU6 7RX, UK. and Positron Emission Tomography Research Centre, The University of Hull, HU6 7RX, UK
| |
Collapse
|
34
|
Sarkar S, Bhatt N, Ha YS, Huynh PT, Soni N, Lee W, Lee YJ, Kim JY, Pandya DN, An GI, Lee KC, Chang Y, Yoo J. High in Vivo Stability of 64Cu-Labeled Cross-Bridged Chelators Is a Crucial Factor in Improved Tumor Imaging of RGD Peptide Conjugates. J Med Chem 2018; 61:385-395. [DOI: 10.1021/acs.jmedchem.7b01671] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Swarbhanu Sarkar
- Department
of Molecular Medicine, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu 41944, South Korea
| | - Nikunj Bhatt
- Department
of Molecular Medicine, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu 41944, South Korea
| | - Yeong Su Ha
- Department
of Molecular Medicine, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu 41944, South Korea
| | - Phuong Tu Huynh
- Department
of Molecular Medicine, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu 41944, South Korea
| | - Nisarg Soni
- Department
of Molecular Medicine, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu 41944, South Korea
| | - Woonghee Lee
- Department
of Molecular Medicine, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu 41944, South Korea
| | - Yong Jin Lee
- Department
of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, South Korea
| | - Jung Young Kim
- Department
of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, South Korea
| | - Darpan N. Pandya
- Department
of Molecular Medicine, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu 41944, South Korea
| | - Gwang Il An
- Department
of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, South Korea
| | - Kyo Chul Lee
- Department
of RI-Convergence Research, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, South Korea
| | - Yongmin Chang
- Department
of Molecular Medicine, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu 41944, South Korea
| | - Jeongsoo Yoo
- Department
of Molecular Medicine, BK21 Plus KNU Biomedical Convergence Program, Kyungpook National University, Daegu 41944, South Korea
| |
Collapse
|
35
|
Mansour N, Paquette M, Ait-Mohand S, Dumulon-Perreault V, Guérin B. Evaluation of a novel GRPR antagonist for prostate cancer PET imaging: [ 64 Cu]-DOTHA 2 -PEG-RM26. Nucl Med Biol 2018; 56:31-38. [DOI: 10.1016/j.nucmedbio.2017.10.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 10/17/2017] [Accepted: 10/18/2017] [Indexed: 02/08/2023]
|
36
|
Synthesis and Preliminary Evaluation of68Ga-NOTA-Biphenyl-c(RGDyK) for the Quantification of Integrin αvβ3. B KOREAN CHEM SOC 2017. [DOI: 10.1002/bkcs.11316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
37
|
Tsionou MI, Knapp CE, Foley CA, Munteanu CR, Cakebread A, Imberti C, Eykyn TR, Young JD, Paterson BM, Blower PJ, Ma MT. Comparison of macrocyclic and acyclic chelators for gallium-68 radiolabelling. RSC Adv 2017; 7:49586-49599. [PMID: 29308192 PMCID: PMC5708347 DOI: 10.1039/c7ra09076e] [Citation(s) in RCA: 106] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 10/06/2017] [Indexed: 01/12/2023] Open
Abstract
Gallium-68 (68Ga) is a positron-emitting isotope used for clinical PET imaging of peptide receptor expression. 68Ga radiopharmaceuticals used in molecular PET imaging consist of disease-targeting biomolecules tethered to chelators that complex 68Ga3+. Ideally, the chelator will rapidly, quantitatively and stably coordinate 68Ga3+ at room temperature, near neutral pH and low chelator concentration, allowing for simple routine radiopharmaceutical formulation. Identification of chelators that fulfil these requirements will facilitate development of kit-based 68Ga radiopharmaceuticals. Herein the reaction of a range of widely used macrocyclic and acyclic chelators with 68Ga3+ is reported. Radiochemical yields have been measured under conditions of varying chelator concentrations, pH (3.5 and 6.5) and temperature (25 and 90 °C). These chelators are: 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA), 1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA), 1,4,7-triazacyclononane macrocycles substituted with phosphonic (NOTP) and phosphinic (TRAP) groups at the amine, bis(2-hydroxybenzyl)ethylenediaminediacetic acid (HBED), a tris(hydroxypyridinone) containing three 1,6-dimethyl-3-hydroxypyridin-4-one groups (THP) and the hexadentate tris(hydroxamate) siderophore desferrioxamine-B (DFO). Competition studies have also been undertaken to assess relative complexation efficiencies of each chelator for 68Ga3+ under different pH and temperature conditions. Performing radiolabelling reactions at pH 6.5, 25 °C and 5-50 μM chelator concentration resulted in near quantitative radiochemical yields for all chelators, except DOTA. Radiochemical yields either decreased or were not substantially improved when the reactions were undertaken at lower pH or at higher temperature, except in the case of DOTA. THP and DFO were the most effective 68Ga3+ chelators at near-neutral pH and 25 °C, rapidly providing near-quantitative radiochemical yields at very low chelator concentrations. NOTP and HBED were only slightly less effective under these conditions. In competition studies with all other chelators, THP demonstrated highest reactivity for 68Ga3+ complexation under all conditions. These data point to THP possessing ideal properties for rapid, one-step kit-based syntheses of 68Ga-biomolecules for molecular PET imaging. LC-MS and 1H, 13C{1H} and 71Ga NMR studies of HBED complexes of Ga3+ showed that under the analytical conditions employed in this study, multiple HBED-bound Ga complexes exist. X-ray diffraction data indicated that crystals isolated from these solutions contained octahedral [Ga(HBED)(H2O)], with HBED coordinated in a pentadentate N2O3 mode, with only one phenolic group coordinated to Ga3+, and the remaining coordination site occupied by a water molecule.
Collapse
Affiliation(s)
- Maria Iris Tsionou
- King's College London, Division of Imaging Sciences and Biomedical Engineering, St Thomas' Hospital, London SE1 7EH, UK.
| | - Caroline E Knapp
- Department of Chemistry, University College London, 20 Gordon Street, London WC1H 0AJ, UK
| | - Calum A Foley
- King's College London, Division of Imaging Sciences and Biomedical Engineering, St Thomas' Hospital, London SE1 7EH, UK.
| | - Catherine R Munteanu
- King's College London, Division of Imaging Sciences and Biomedical Engineering, St Thomas' Hospital, London SE1 7EH, UK.
| | - Andrew Cakebread
- Division of Analytical and Environmental Sciences, King's College London, Franklin Wilkin's Building, London SE1 9NH, UK
| | - Cinzia Imberti
- King's College London, Division of Imaging Sciences and Biomedical Engineering, St Thomas' Hospital, London SE1 7EH, UK.
| | - Thomas R Eykyn
- King's College London, Division of Imaging Sciences and Biomedical Engineering, St Thomas' Hospital, London SE1 7EH, UK.
| | - Jennifer D Young
- King's College London, Division of Imaging Sciences and Biomedical Engineering, St Thomas' Hospital, London SE1 7EH, UK.
| | - Brett M Paterson
- School of Chemistry, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, 3010, Victoria, Australia
| | - Philip J Blower
- King's College London, Division of Imaging Sciences and Biomedical Engineering, St Thomas' Hospital, London SE1 7EH, UK.
| | - Michelle T Ma
- King's College London, Division of Imaging Sciences and Biomedical Engineering, St Thomas' Hospital, London SE1 7EH, UK.
| |
Collapse
|
38
|
Garousi J, Andersson KG, Dam JH, Olsen BB, Mitran B, Orlova A, Buijs J, Ståhl S, Löfblom J, Thisgaard H, Tolmachev V. The use of radiocobalt as a label improves imaging of EGFR using DOTA-conjugated Affibody molecule. Sci Rep 2017; 7:5961. [PMID: 28729680 PMCID: PMC5519605 DOI: 10.1038/s41598-017-05700-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 06/01/2017] [Indexed: 02/04/2023] Open
Abstract
Several anti-cancer therapies target the epidermal growth factor receptor (EGFR). Radionuclide imaging of EGFR expression in tumours may aid in selection of optimal cancer therapy. The 111In-labelled DOTA-conjugated ZEGFR:2377 Affibody molecule was successfully used for imaging of EGFR-expressing xenografts in mice. An optimal combination of radionuclide, chelator and targeting protein may further improve the contrast of radionuclide imaging. The aim of this study was to evaluate the targeting properties of radiocobalt-labelled DOTA-ZEGFR:2377. DOTA-ZEGFR:2377 was labelled with 57Co (T1/2 = 271.8 d), 55Co (T1/2 = 17.5 h), and, for comparison, with the positron-emitting radionuclide 68Ga (T1/2 = 67.6 min) with preserved specificity of binding to EGFR-expressing A431 cells. The long-lived cobalt radioisotope 57Co was used in animal studies. Both 57Co-DOTA-ZEGFR:2377 and 68Ga-DOTA-ZEGFR:2377 demonstrated EGFR-specific accumulation in A431 xenografts and EGFR-expressing tissues in mice. Tumour-to-organ ratios for the radiocobalt-labelled DOTA-ZEGFR:2377 were significantly higher than for the gallium-labelled counterpart already at 3 h after injection. Importantly, 57Co-DOTA-ZEGFR:2377 demonstrated a tumour-to-liver ratio of 3, which is 7-fold higher than the tumour-to-liver ratio for 68Ga-DOTA-ZEGFR:2377. The results of this study suggest that the positron-emitting cobalt isotope 55Co would be an optimal label for DOTA-ZEGFR:2377 and further development should concentrate on this radionuclide as a label.
Collapse
Affiliation(s)
- Javad Garousi
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Ken G Andersson
- Department of Protein Technology, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Johan H Dam
- Department of Nuclear Medicine, Odense University Hospital, Sdr. Boulevard 29, 5000, Odense, Denmark
| | - Birgitte B Olsen
- Department of Nuclear Medicine, Odense University Hospital, Sdr. Boulevard 29, 5000, Odense, Denmark
| | - Bogdan Mitran
- Division of Molecular Imaging, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Anna Orlova
- Division of Molecular Imaging, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Jos Buijs
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Stefan Ståhl
- Department of Protein Technology, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - John Löfblom
- Department of Protein Technology, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Helge Thisgaard
- Department of Nuclear Medicine, Odense University Hospital, Sdr. Boulevard 29, 5000, Odense, Denmark
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
39
|
Abstract
Background Integrin-targeting radiopharmaceuticals have potential broad applications, spanning from cancer theranostics to cardiovascular diseases. We have previously reported preclinical dosimetry results of 68Ga-NODAGA-RGDyK in mice. This study presents the first human dosimetry of 68Ga-NODAGA-RGDyK in the five consecutive patients included in a clinical imaging protocol of carotid atherosclerotic plaques. Five male patients underwent whole-body time-of-flight (TOF) PET/CT scans 10, 60 and 120 min after tracer injection (200 MBq). Quantification of 68Ga activity concentration was first validated by a phantom study. To be used as input in OLINDA/EXM, time-activity curves were derived from manually drawn regions of interest over the following organs: brain, thyroid, lungs, heart, liver, spleen, stomach, kidneys, red marrow, pancreas, small intestine, colon, urinary bladder and whole body. A separate dosimetric analysis was performed for the choroid plexuses. Female dosimetry was extrapolated from male data. Effective doses (EDs) were estimated according to both ICRP60 and ICRP103 assuming 30-min and 1-h voiding cycles. Results The body regions receiving the highest dose were urinary bladder, kidneys and choroid plexuses. For a 30-min voiding cycle, the EDs were 15.7 and 16.5 μSv/MBq according to ICRP60 and ICRP103, respectively. The extrapolation to female dosimetry resulted in organ absorbed doses 17% higher than those of male patients, on average. The 1-h voiding cycle extrapolation resulted in EDs of 19.3 and 19.8 μSv/MBq according to ICRP60 and ICRP103, respectively. A comparison is made with previous mouse dosimetry and with other human studies employing different RGD-based radiopharmaceuticals. Conclusions According to ICRP60/ICRP103 recommendations, an injection of 200 MBq 68Ga-NODAGA-RGDyK leads to an ED in man of 3.86/3.92 mSv. For future therapeutic applications, specific attention should be directed to delivered dose to kidneys and potentially also to the choroid plexuses. Trial registration Clinical trial.gov, NCT01608516
Collapse
|
40
|
Abstract
Angiogenesis imaging is important for diagnostic and therapeutic treatment of various malignant and nonmalignant diseases. The Arg-Gly-Asp (RGD) sequence has been known to bind with the αvβ3 integrin that is expressed on the surface of angiogenic blood vessels or tumor cells. Thus, various radiolabeled derivatives of RGD peptides have been developed for angiogenesis imaging. Among the various radionuclides, (68)Ga was the most widely studied for RGD peptide imaging because of its excellent nuclear physical properties, easy-to-label chemical properties, and cost-effectiveness owing to the availability of a (68)Ge-(68)Ga generator. Thus, various (68)Ga-labeled RGD derivatives have been developed and applied for preclinical and clinical studies. Clinical trials were performed for both malignant and nonmalignant diseases. Breast cancer, glioma, and lung cancer were malignant, and myocardial infarction, atherosclerosis, and moyamoya disease were nonmalignant among the investigated diseases. Further, these (68)Ga-labeled RGD derivatives could be applied to assess the effects of antiangiogenic treatment or theragnosis or both, of cancers. In conclusion, the angiogenesis imaging technology using (68)Ga-labeled RGD derivatives might be useful for the development of new therapeutic assessments, and for diagnostic and theragnostic applications.
Collapse
Affiliation(s)
- Jae Seon Eo
- Department of Nuclear Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Jae Min Jeong
- Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
41
|
Pirisedigh A, Blais V, Ait-Mohand S, Abdallah K, Holleran BJ, Leduc R, Dory YL, Gendron L, Guérin B. Synthesis and Evaluation of a 64Cu-Conjugate, a Selective δ-Opioid Receptor Positron Emission Tomography Imaging Agent. Org Lett 2017; 19:2018-2021. [DOI: 10.1021/acs.orglett.7b00575] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Azadeh Pirisedigh
- Department
of Nuclear Medicine and Radiobiology, ‡Department of Pharmacology and
Physiology, Faculty of Medicine and Health Sciences,
and §Laboratoire de Synthèse
Supramoléculaire, Department of Chemistry, Faculty of Sciences,
Institut de Pharmacologie, Université de Sherbrooke, Centre
de recherche du CHUS, 3001, 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Véronique Blais
- Department
of Nuclear Medicine and Radiobiology, ‡Department of Pharmacology and
Physiology, Faculty of Medicine and Health Sciences,
and §Laboratoire de Synthèse
Supramoléculaire, Department of Chemistry, Faculty of Sciences,
Institut de Pharmacologie, Université de Sherbrooke, Centre
de recherche du CHUS, 3001, 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Samia Ait-Mohand
- Department
of Nuclear Medicine and Radiobiology, ‡Department of Pharmacology and
Physiology, Faculty of Medicine and Health Sciences,
and §Laboratoire de Synthèse
Supramoléculaire, Department of Chemistry, Faculty of Sciences,
Institut de Pharmacologie, Université de Sherbrooke, Centre
de recherche du CHUS, 3001, 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Khaled Abdallah
- Department
of Nuclear Medicine and Radiobiology, ‡Department of Pharmacology and
Physiology, Faculty of Medicine and Health Sciences,
and §Laboratoire de Synthèse
Supramoléculaire, Department of Chemistry, Faculty of Sciences,
Institut de Pharmacologie, Université de Sherbrooke, Centre
de recherche du CHUS, 3001, 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Brian J. Holleran
- Department
of Nuclear Medicine and Radiobiology, ‡Department of Pharmacology and
Physiology, Faculty of Medicine and Health Sciences,
and §Laboratoire de Synthèse
Supramoléculaire, Department of Chemistry, Faculty of Sciences,
Institut de Pharmacologie, Université de Sherbrooke, Centre
de recherche du CHUS, 3001, 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Richard Leduc
- Department
of Nuclear Medicine and Radiobiology, ‡Department of Pharmacology and
Physiology, Faculty of Medicine and Health Sciences,
and §Laboratoire de Synthèse
Supramoléculaire, Department of Chemistry, Faculty of Sciences,
Institut de Pharmacologie, Université de Sherbrooke, Centre
de recherche du CHUS, 3001, 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Yves L. Dory
- Department
of Nuclear Medicine and Radiobiology, ‡Department of Pharmacology and
Physiology, Faculty of Medicine and Health Sciences,
and §Laboratoire de Synthèse
Supramoléculaire, Department of Chemistry, Faculty of Sciences,
Institut de Pharmacologie, Université de Sherbrooke, Centre
de recherche du CHUS, 3001, 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Louis Gendron
- Department
of Nuclear Medicine and Radiobiology, ‡Department of Pharmacology and
Physiology, Faculty of Medicine and Health Sciences,
and §Laboratoire de Synthèse
Supramoléculaire, Department of Chemistry, Faculty of Sciences,
Institut de Pharmacologie, Université de Sherbrooke, Centre
de recherche du CHUS, 3001, 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| | - Brigitte Guérin
- Department
of Nuclear Medicine and Radiobiology, ‡Department of Pharmacology and
Physiology, Faculty of Medicine and Health Sciences,
and §Laboratoire de Synthèse
Supramoléculaire, Department of Chemistry, Faculty of Sciences,
Institut de Pharmacologie, Université de Sherbrooke, Centre
de recherche du CHUS, 3001, 12e Avenue Nord, Sherbrooke, Québec J1H 5N4, Canada
| |
Collapse
|
42
|
Moreau M, Poty S, Vrigneaud JM, Walker P, Guillemin M, Raguin O, Oudot A, Bernhard C, Goze C, Boschetti F, Collin B, Brunotte F, Denat F. MANOTA: a promising bifunctional chelating agent for copper-64 immunoPET. Dalton Trans 2017; 46:14659-14668. [DOI: 10.1039/c7dt01772c] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A comparison of four bifunctional chelating agents showed superior behaviour of a new NOTA derivative for 64Cu labelling of antibody fragments.
Collapse
|
43
|
Colin DJ, Inkster JAH, Germain S, Seimbille Y. Preclinical validations of [ 18F]FPyPEGCBT- c(RGDfK): a 18F-labelled RGD peptide prepared by ligation of 2-cyanobenzothiazole and 1,2-aminothiol to image angiogenesis. EJNMMI Radiopharm Chem 2017; 1:16. [PMID: 29564392 PMCID: PMC5843817 DOI: 10.1186/s41181-016-0019-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 10/11/2016] [Indexed: 12/13/2022] Open
Abstract
Background αVβ3, αVβ5 and α5β1 integrins are known to be involved in carcinogenesis and are overexpressed in many types of tumours compared to healthy tissues; thereby they have been selected as promising therapeutic targets. Positron emission tomography (PET) is providing a unique non-invasive screening assay to discriminate which patient is more prone to benefit from antiangiogenic therapies, and extensive research has been carried out to develop a clinical radiopharmaceutical that binds specifically to integrin receptors. We recently reported the synthesis of a new 18F-labelled RGD peptide prepared by 2-cyanobenzothiazole (CBT)/1,2-aminothiol conjugation. This study aims at characterising the preclinical biologic properties of this new tumour-targeting ligand, named [18F]FPyPEGCBT-c(RGDfK). The in vitro binding properties of [18F]FPyPEGCBT-c(RGDfK) were analysed by standard binding assay in U-87 MG and SKOV-3 cancer models and its selectivity towards integrins by siRNA depletions. Its preclinical potential was studied in mice bearing subcutaneous tumours by ex vivo biodistribution studies and in vivo microPET/CT imaging. Results In vitro, FPyPEGCBT-c(RGDfK) efficiently bound RGD-recognising integrins as compared to a control c(RGDfV) peptide (IC50 = 30.8 × 10−7 M vs. 6.0 × 10−7 M). [18F]FPyPEGCBT-c(RGDfK) cell uptake was mediated by an active transport through binding to αV, β3 and β5 but not to β1 subunits. In vivo, this new tracer demonstrated specific tumour uptake with %ID/g of 2.9 and 2.4 in U-87 MG and SKOV-3 tumours 1 h post injection. Tumour-to-muscle ratios of 4 were obtained 1 h after intravenous administration of the tracer allowing good visualisation of the tumours. However, unfavourable background accumulation and high hepatobiliary excretion were observed. Conclusion [18F]FPyPEGCBT-c(RGDfK) specifically detects tumours expressing RGD-recognising integrin receptors in preclinical studies. Further optimisation of this radioligand may yield candidates with improved imaging properties and would warrant the further use of this efficient labelling technique for alternative targeting vectors. Electronic supplementary material The online version of this article (doi:10.1186/s41181-016-0019-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Didier J Colin
- MicroPET/SPECT/CT Imaging Laboratory, Centre for BioMedical Imaging (CIBM), University Hospital of Geneva, 1211 Geneva, Switzerland
| | - James A H Inkster
- Cyclotron Unit, University Hospital of Geneva, 1211 Geneva, Switzerland
| | - Stéphane Germain
- MicroPET/SPECT/CT Imaging Laboratory, Centre for BioMedical Imaging (CIBM), University Hospital of Geneva, 1211 Geneva, Switzerland
| | - Yann Seimbille
- Cyclotron Unit, University Hospital of Geneva, 1211 Geneva, Switzerland.,TRIUMF, Life Sciences Division, 4004 Wesbrook Mall, Vancouver, BC V6T 2A3 Canada
| |
Collapse
|
44
|
Lee JW, Lee YJ, Shin UC, Kim SW, Kim BI, Lee KC, Kim JY, Park JA. Improved Pharmacokinetics Following PEGylation and Dimerization of a c(RGD-ACH-K) Conjugate Used for Tumor Positron Emission Tomography Imaging. Cancer Biother Radiopharm 2016; 31:295-301. [PMID: 27754748 DOI: 10.1089/cbr.2016.2036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Improving the in vivo pharmacokinetics (PK) of positron emission tomography (PET) radiotracers is of critical importance to tumor diagnosis and therapy. In the case of peptide-based radiotracers, the modification and addition of a linker or spacer functional group often offer faster in vivo pharmacokinetic behavior. In this study, the authors introduced two new PEGlyated dimeric c(RGD-ACH-K) conjugates, in which an aminocyclohexane carboxylic acid (ACH) is inserted into the ring chain of the cyclic RGD peptides, with a common bifunctional chelator (DOTA or NOTA) used for labeling with radiometals (including 68Ga and 64Cu). The addition of polyethylene glycol (PEG) and dimerization of c(RGD-ACH-K) affected the PK of the renal system and the tumor-targeting ability, relative to unmodified molecule. As a result, both 64Cu-DOTA-E[c(RGD-ACH-K)]2 (complex 1) and 64Cu-NOTA-E[c(RGD-ACH-K)]2 (complex 2) exhibited specific tumor-targeting properties relative to tumor-blocking control group, most likely resulting from improved in vivo tumor imaging. The in vivo tumor-to-blood ratio of the 64Cu(NOTA) complex shows better PET imaging than that of the 64Cu(DOTA) complex, which should lead to improved dosimetry and increased suitability for noninvasive monitoring of tumor growth or tumor-targeted radionuclide therapy.
Collapse
Affiliation(s)
- Ji Woong Lee
- 1 Molecular Imaging Research Center, Korea Institute of Radiological & Medical Sciences , Seoul, Republic of Korea.,2 Department of Integrated Biomedical and Life Science, Korea University , Seoul, Republic of Korea
| | - Yong Jin Lee
- 1 Molecular Imaging Research Center, Korea Institute of Radiological & Medical Sciences , Seoul, Republic of Korea
| | - Un Chol Shin
- 1 Molecular Imaging Research Center, Korea Institute of Radiological & Medical Sciences , Seoul, Republic of Korea
| | - Suhng Wook Kim
- 2 Department of Integrated Biomedical and Life Science, Korea University , Seoul, Republic of Korea
| | - Byung Il Kim
- 3 Department of Nuclear Medicine, Korea Cancer Center Hospital , Seoul, Republic of Korea
| | - Kyo Chul Lee
- 1 Molecular Imaging Research Center, Korea Institute of Radiological & Medical Sciences , Seoul, Republic of Korea
| | - Jung Young Kim
- 1 Molecular Imaging Research Center, Korea Institute of Radiological & Medical Sciences , Seoul, Republic of Korea
| | - Ji-Ae Park
- 1 Molecular Imaging Research Center, Korea Institute of Radiological & Medical Sciences , Seoul, Republic of Korea
| |
Collapse
|
45
|
Greiser J, Niksch T, Weigand W, Freesmeyer M. Investigations on the Ga(III) Complex of EOB-DTPA and Its 68Ga Radiolabeled Analogue. J Vis Exp 2016. [PMID: 27584545 DOI: 10.3791/54334] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
We demonstrate a method for the isolation of EOB-DTPA (3,6,9-triaza-3,6,9-tris(carboxymethyl)-4-(ethoxybenzyl)-undecanedioic acid) from its Gd(III) complex and protocols for the preparation of its novel non-radioactive, i.e., natural Ga(III) as well as radioactive (68)Ga complex. The ligand as well as the Ga(III) complex were characterized by nuclear magnetic resonance (NMR) spectroscopy, mass spectrometry and elemental analysis. (68)Ga was obtained by a standard elution method from a (68)Ge/(68)Ga generator. Experiments to evaluate the (68)Ga-labeling efficiency of EOB-DTPA at pH 3.8-4.0 were performed. Established analysis techniques radio TLC (thin layer chromatography) and radio HPLC (high performance liquid chromatography) were used to determine the radiochemical purity of the tracer. As a first investigation of the (68)Ga tracers' lipophilicity the n-octanol/water distribution coefficient of (68)Ga species present in a pH 7.4 solution was determined by an extraction method. In vitro stability measurements of the tracer in various media at physiological pH were performed, revealing different rates of decomposition.
Collapse
Affiliation(s)
- Julia Greiser
- Clinic of Nuclear Medicine, University Hospital Jena
| | - Tobias Niksch
- Clinic of Nuclear Medicine, University Hospital Jena
| | | | | |
Collapse
|
46
|
Lee JW, Park JA, Lee YJ, Shin UC, Kim SW, Kim BI, Lim SM, An GI, Kim JY, Lee KC. New Glucocyclic RGD Dimers for Positron Emission Tomography Imaging of Tumor Integrin Receptors. Cancer Biother Radiopharm 2016; 31:209-16. [PMID: 27403677 DOI: 10.1089/cbr.2016.2015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Most studies of radiolabeled arginine-glycine-aspartic acid (RGD) peptides have shown in vitro affinity for integrin ανβ3, allowing for the targeting of receptor-positive tumors in vivo. However, major differences have been found in the pharmacokinetic profiles of different radiolabeled RGD peptide analogs. The purposes of this study were to prepare (64)Cu-DOTA-gluco-E[c(RGDfK)]2 (R8), (64)Cu-NOTA-gluco-E[c(RGDfK)]2 (R9), and (64)Cu-NODAGA-gluco-E[c(RGDfK)]2 (R10) and compare their pharmacokinetics and tumor imaging properties using small-animal positron emission tomography (PET). All three compounds were produced with high specific activity within 10 minutes. The IC50 values were similar for all the substances, and their affinities were greater than that of c(RGDyK). R8, R9, and R10 were stable for 24 hours in human and mouse serums and showed high uptake in U87MG tumors with high tumor-to-blood ratios. Compared to the control, a cyclic RGD peptide dimer without glucosamine, R10, showed low uptake in the liver. Because of their good imaging qualities and improved pharmacokinetics, (64)Cu-labeled dimer RGD conjugates (R8, R9, and R10) may have potential applications as PET radiotracers. R9 (NOTA) with highly in vivo stability consequentially showed an improved PET tumor uptake than R8 (DOTA) or R10 (NODAGA).
Collapse
Affiliation(s)
- Ji Woong Lee
- 1 Molecular Imaging Research Center, Korea Institute of Radiological & Medical Sciences , Seoul, Republic of Korea.,2 Department of Integrated Biomedical and Life Science, Korea University , Seoul, Republic of Korea
| | - Ji-Ae Park
- 1 Molecular Imaging Research Center, Korea Institute of Radiological & Medical Sciences , Seoul, Republic of Korea
| | - Yong Jin Lee
- 1 Molecular Imaging Research Center, Korea Institute of Radiological & Medical Sciences , Seoul, Republic of Korea
| | - Un Chol Shin
- 1 Molecular Imaging Research Center, Korea Institute of Radiological & Medical Sciences , Seoul, Republic of Korea
| | - Suhng Wook Kim
- 2 Department of Integrated Biomedical and Life Science, Korea University , Seoul, Republic of Korea
| | - Byung Il Kim
- 3 Department of Nuclear Medicine, Korea Cancer Center Hospital , Seoul, Republic of Korea
| | - Sang Moo Lim
- 3 Department of Nuclear Medicine, Korea Cancer Center Hospital , Seoul, Republic of Korea
| | - Gwang Il An
- 1 Molecular Imaging Research Center, Korea Institute of Radiological & Medical Sciences , Seoul, Republic of Korea
| | - Jung Young Kim
- 1 Molecular Imaging Research Center, Korea Institute of Radiological & Medical Sciences , Seoul, Republic of Korea
| | - Kyo Chul Lee
- 1 Molecular Imaging Research Center, Korea Institute of Radiological & Medical Sciences , Seoul, Republic of Korea
| |
Collapse
|
47
|
Gai Y, Sun L, Hui W, Ouyang Q, Anderson CJ, Xiang G, Ma X, Zeng D. New Bifunctional Chelator p-SCN-PhPr-NE3TA for Copper-64: Synthesis, Peptidomimetic Conjugation, Radiolabeling, and Evaluation for PET Imaging. Inorg Chem 2016; 55:6892-901. [PMID: 27347690 DOI: 10.1021/acs.inorgchem.6b00395] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Bifunctional chelators play an important role in developing metallic radionuclide-based radiopharmaceuticals. In this study, a new bifunctional ligand, p-SCN-PhPr-NE3TA, was synthesized and conjugated to a very late antigen-4 targeting peptidomimetic, LLP2A, for evaluating its application in (64)Cu-based positron emission tomography (PET) imaging. The new ligand exhibited strong selective coordination of Cu(II), leading to a robust Cu complex, even in the presence of 10-fold Fe(III). The LLP2A conjugate of p-SCN-PhPr-NE3TA was prepared and successfully labeled with (64)Cu under mild conditions. The conjugate (64)Cu-NE3TA-PEG4-LLP2A showed significantly higher specific activity, compared with (64)Cu-NOTA-PEG4-LLP2A, while maintaining comparable serum stability. Subsequent biodistribution studies and PET imaging in mice bearing B16F10 xenografts confirmed its favorable in vivo performance and high tumor uptake with low background, rendering p-SCN-PhPr-NE3TA a promising bifunctional chelator for (64)Cu-based radiopharmaceuticals.
Collapse
Affiliation(s)
- Yongkang Gai
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology , 13 Hangkong Road, Wuhan 430030, China.,Department of Radiology, University of Pittsburgh , Pittsburgh, Pennsylvania 15219, United States
| | - Lingyi Sun
- Department of Radiology, University of Pittsburgh , Pittsburgh, Pennsylvania 15219, United States
| | - Wenqi Hui
- College of Pharmacy, The Third Military Medical University , Chongqing 400038, China
| | - Qin Ouyang
- College of Pharmacy, The Third Military Medical University , Chongqing 400038, China
| | - Carolyn J Anderson
- Department of Radiology, University of Pittsburgh , Pittsburgh, Pennsylvania 15219, United States.,Departments of Pharmacology & Chemical Biology and Bioengineering, University of Pittsburgh , Pittsburgh, Pennsylvania 15219, United States
| | - Guangya Xiang
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology , 13 Hangkong Road, Wuhan 430030, China
| | - Xiang Ma
- School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology , 13 Hangkong Road, Wuhan 430030, China
| | - Dexing Zeng
- Department of Radiology, University of Pittsburgh , Pittsburgh, Pennsylvania 15219, United States
| |
Collapse
|
48
|
Banerjee SR, Chen Z, Pullambhatla M, Lisok A, Chen J, Mease RC, Pomper MG. Preclinical Comparative Study of (68)Ga-Labeled DOTA, NOTA, and HBED-CC Chelated Radiotracers for Targeting PSMA. Bioconjug Chem 2016; 27:1447-55. [PMID: 27076393 PMCID: PMC5290997 DOI: 10.1021/acs.bioconjchem.5b00679] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
(68)Ga-labeled, low-molecular-weight imaging agents that target the prostate-specific membrane antigen (PSMA) are increasingly used clinically to detect prostate and other cancers with positron emission tomography (PET). The goal of this study was to compare the pharmacokinetics of three PSMA-targeted radiotracers: (68)Ga-1, using DOTA-monoamide as the chelating agent; (68)Ga-2, containing the macrocyclic chelating agent p-SCN-Bn-NOTA; and (68)Ga-DKFZ-PSMA-11, currently in clinical trials, which uses the acyclic chelating agent, HBED-CC. The PSMA-targeting scaffold for all three agents utilized a similar Glu-urea-Lys-linker construct. Each radiotracer enabled visualization of PSMA+ PC3 PIP tumor, kidney, and urinary bladder as early as 15 min post-injection using small animal PET/computed tomography (PET/CT). (68)Ga-2 demonstrated the fastest rate of clearance from all tissues in this series and displayed higher uptake in PSMA+ PC3 PIP tumor compared to (68)Ga-1 at 1 h post-injection. There was no significant difference in PSMA+ PC3 PIP tumor uptake for the three agents at 2 and 3 h post-injection. (68)Ga-DKFZ-PSMA-11 demonstrated the highest uptake and retention in normal tissues, including kidney, blood, spleen, and salivary glands and PSMA-negative PC3 flu tumors up to 3 h post-injection. In this preclinical evaluation (68)Ga-2 had the most advantageous characteristics for PSMA-targeted PET imaging.
Collapse
Affiliation(s)
- Sangeeta Ray Banerjee
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, Maryland 21287, United States
| | - Zhengping Chen
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, Maryland 21287, United States
| | - Mrudula Pullambhatla
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, Maryland 21287, United States
| | - Ala Lisok
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, Maryland 21287, United States
| | - Jian Chen
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, Maryland 21287, United States
| | - Ronnie C. Mease
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, Maryland 21287, United States
| | - Martin G. Pomper
- Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, Maryland 21287, United States
| |
Collapse
|
49
|
Notni J, Steiger K, Hoffmann F, Reich D, Schwaiger M, Kessler H, Wester HJ. Variation of Specific Activities of 68Ga-Aquibeprin and 68Ga-Avebetrin Enables Selective PET Imaging of Different Expression Levels of Integrins α5β1 and αvβ3. J Nucl Med 2016; 57:1618-1624. [DOI: 10.2967/jnumed.116.173948] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 03/29/2016] [Indexed: 12/22/2022] Open
|
50
|
Farkas R, Siwowska K, Ametamey SM, Schibli R, van der Meulen NP, Müller C. (64)Cu- and (68)Ga-Based PET Imaging of Folate Receptor-Positive Tumors: Development and Evaluation of an Albumin-Binding NODAGA-Folate. Mol Pharm 2016; 13:1979-87. [PMID: 27145400 DOI: 10.1021/acs.molpharmaceut.6b00143] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
A number of folate-based radioconjugates have been synthesized and evaluated for nuclear imaging purposes of folate receptor (FR)-positive tumors and potential therapeutic application. A common shortcoming of radiofolates is, however, a significant accumulation of radioactivity in the kidneys. This situation has been faced by modifying the folate conjugate with an albumin-binding entity to increase the circulation time of the radiofolate, which led to significantly improved tumor-to-kidney ratios. The aim of this study was to develop an albumin-binding folate conjugate with a NODAGA-chelator (rf42) for labeling with (64)Cu and (68)Ga, allowing application for PET imaging. The folate conjugate rf42 was synthesized in 8 steps, with an overall yield of 5%. Radiolabeling with (64)Cu and (68)Ga was carried out at room temperature within 10 min resulting in (64)Cu-rf42 and (68)Ga-rf42 with >95% radiochemical purity. (64)Cu-rf42 and (68)Ga-rf42 were stable (>95% intact) in phosphate-buffered saline over more than 4 half-lives of the corresponding radionuclide. In vitro, the plasma protein-bound fraction of (64)Cu-rf42 and (68)Ga-rf42 was determined to be >96%. Cell experiments proved FR-specific uptake of both radiofolates, as it was reduced to <1% when KB tumor cells were coincubated with excess folic acid. In vivo, high accumulation of (64)Cu-rf42 and (68)Ga-rf42 was found in KB tumors of mice (14.52 ± 0.99% IA/g and 11.92 ± 1.68% IA/g, respectively) at 4 h after injection. The tumor-to-kidney ratios were in the range of 0.43-0.55 over the first 4 h of investigation. At later time points (up to 72 h p.i. of (64)Cu-rf42) the tumor-to-kidney ratio increased to 0.73. High-quality PET/CT images were obtained 2 h after injection of (64)Cu-rf42 and (68)Ga-rf42, respectively, allowing distinct visualization of tumors and kidneys. Comparison of PET/CT images obtained with (64)Cu-rf42 and a (64)Cu-labeled DOTA-folate conjugate (cm10) clearly proved the superiority of NODAGA for stable coordination of (64)Cu. (64)Cu-cm10 showed high liver uptake, most probably as a consequence of released (64)Cu(2+). The data reported in this study clearly proved the promising features of (64)Cu-rf42, particularly in terms of favorable tumor-to-kidney ratios. The relatively long half-life of (64)Cu (T1/2 = 12.7 h) matches well with the enhanced circulation time of the albumin-binding NODAGA-folate, allowing PET imaging at longer time points after injection than is possible when using (68)Ga (T1/2 = 68 min).
Collapse
Affiliation(s)
- Renáta Farkas
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institut , Villigen-PSI, Switzerland
| | - Klaudia Siwowska
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institut , Villigen-PSI, Switzerland
| | - Simon M Ametamey
- Department of Chemistry and Applied Biosciences, ETH Zurich , Zurich, Switzerland
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institut , Villigen-PSI, Switzerland.,Department of Chemistry and Applied Biosciences, ETH Zurich , Zurich, Switzerland
| | - Nicholas P van der Meulen
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institut , Villigen-PSI, Switzerland.,Laboratory of Radiochemistry, Paul Scherrer Institut , Villigen-PSI, Switzerland
| | - Cristina Müller
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Paul Scherrer Institut , Villigen-PSI, Switzerland.,Department of Chemistry and Applied Biosciences, ETH Zurich , Zurich, Switzerland
| |
Collapse
|