1
|
Shchukina AA, Zubenko AD, Tarasenko OV, Larenkov AA, Bubenshchikov VB, Chernikova EY, Fedorov YV, Fedorova OA. Evaluation of chelating agents based on pyridine-azacrown compounds H 4PATA, PATAM, and H 4PATPA for 68Ga and 177Lu. Nucl Med Biol 2024; 140-141:108972. [PMID: 39550886 DOI: 10.1016/j.nucmedbio.2024.108972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 11/19/2024]
Abstract
In this article, we present the synthesis and characterization of three macrocyclic chelators, H4PATA, PATAM, and H4PATPA, based on a pyridine-azacrown compound. Their complexation with 68Ga and 177Lu has been thoroughly investigated using MALDI TOF MS, 1H NMR spectroscopy, radiolabeling studies, and experiments in vitro with fetal bovine serum and a 1000-fold molar excess of H4EDTA. Our studies have shown that the chelators H4PATA and H4PATPA form complexes at room temperature with both radionuclides (RCY > 80 % and > 90 % for complexes with H4PATA and H4PATPA after 30 min, respectively). The chelator PATAM requires high temperature (95 °C) for complexation. In vitro stability assays in fetal bovine serum as well as H4EDTA-challenge revealed that transchelation occurs for all complexes with 68Ga. However, complexes of the ligands H4PATA and PATAM with 177Lu were found stable. Thus, taking into account the radiolabeling at room temperature and in vitro stability of the complex [177Lu]Lu·PATA, our investigations revealed the chelator H4PATA is a candidate for radiopharmaceutical use with 177Lu.
Collapse
Affiliation(s)
- Anna A Shchukina
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, Vavilova st., 28, 119334 Moscow, Russian Federation.
| | - Anastasia D Zubenko
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, Vavilova st., 28, 119334 Moscow, Russian Federation
| | - Oksana V Tarasenko
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, Vavilova st., 28, 119334 Moscow, Russian Federation; D. I. Mendeleev University of Chemical Technology of Russia, Miusskaya Sq. 9, 125047 Moscow, Russian Federation
| | - Anton A Larenkov
- State Research Center-Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Zhivopisnaya Str., 46, 123098 Moscow, Russian Federation
| | - Viktor B Bubenshchikov
- State Research Center-Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency, Zhivopisnaya Str., 46, 123098 Moscow, Russian Federation
| | - Ekaterina Y Chernikova
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, Vavilova st., 28, 119334 Moscow, Russian Federation
| | - Yury V Fedorov
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, Vavilova st., 28, 119334 Moscow, Russian Federation
| | - Olga A Fedorova
- A. N. Nesmeyanov Institute of Organoelement Compounds of Russian Academy of Sciences, Vavilova st., 28, 119334 Moscow, Russian Federation; D. I. Mendeleev University of Chemical Technology of Russia, Miusskaya Sq. 9, 125047 Moscow, Russian Federation
| |
Collapse
|
2
|
Nazar AK, Basu S. Radiolabeled Somatostatin Analogs for Cancer Imaging. Semin Nucl Med 2024; 54:914-940. [PMID: 39122608 DOI: 10.1053/j.semnuclmed.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 07/01/2024] [Indexed: 08/12/2024]
Abstract
Somatostatin receptors (SSTR) are expressed by many tumours especially those related to neuro-endocrine origin and molecular functional imaging of SSTR expression using radiolabelled somatostatin analogs have revolutionized imaging of patients with these group of malignancies. Coming a long way from the first radiolabelled somatostatin analog 123I-Tyr-3-octreotide, there has been significant developments in terms of radionuclides used, the ligands and somatostatin derivatives. 111In-Pentetreotide extensively employed for imaging NETs at the beginning has now been replaced by 68Ga-SSA based PET-CT. SSA-PET/CT performs superior to conventional imaging modalities and has evolved in the mainframe for NET imaging. The advantages were multiple: (i) superior spatial resolution of PET versus SPECT, (ii) quantitative capabilities of PET aiding in disease activity and treatment response monitoring with better precision, (iii) shorter scan time and (iv) less patient exposure to radiation. The modality is indicated for staging, detecting the primary in CUP-NETs, restaging, treatment planning (along with FDG: the concept of dual-tracer PET-CT) as well as treatment response evaluation and follow-up of NETs. SSA PET/CT has also been incorporated in the guidelines for imaging of Pheochromocytoma-Paraganglioma, Medullary carcinoma thyroid, Meningioma and Tumor induced osteomalacia. At present, there is rising interest on (a) 18F-labelled SSA, (b) 64Cu-labelled SSA, and (c) somatostatin antagonists. 18F offers excellent imaging properties, 64Cu makes delayed imaging feasible which has implications in dosimetry and SSTR antagonists bind with the SST receptors with high affinity and specificity, providing high contrast images with less background, which can be translated to theranostics effectively. SSTR have been demonstrated in non-neuroendocrine tumours as well in the peer-reviewed literature, with studies demonstrating the potential of SSA PET/CT in Neuroblastoma, Nasopharyngeal carcinoma, carcinoma prostate (neuroendocrine differentiation) and lymphoma. This review will focus on the currently available SSAs and their history, different SPECT/PET agents, SSTR antagonists, comparison between the various imaging tracers, and their utility in both neuroendocrine and non-neuroendocrine tumors.
Collapse
Affiliation(s)
- Aamir K Nazar
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Centre Annexe, Mumbai; Homi Bhabha National Institute, Mumbai
| | - Sandip Basu
- Radiation Medicine Centre, Bhabha Atomic Research Centre, Tata Memorial Centre Annexe, Mumbai; Homi Bhabha National Institute, Mumbai.
| |
Collapse
|
3
|
Kleynhans J, Ebenhan T, Sathekge MM. Expanding Role for Gallium-68 PET Imaging in Oncology. Semin Nucl Med 2024; 54:778-791. [PMID: 38964934 DOI: 10.1053/j.semnuclmed.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 07/06/2024]
Abstract
Gallium-68 has gained substantial momentum since 2003 as a versatile radiometal that is extremely useful for application in the development of novel oncology targeting diagnostic radiopharmaceuticals. It is available through both generator produced radioactivity and via cyclotron production methods and can therefore be implemented in either small- or large-scale production facilities. It can also be implemented within different spectrum of infrastructure settings with relative ease. Whilst many of the radiopharmaceuticals are being development and investigated, which is summarized in this manuscript, [68Ga]Ga-SSTR2 and [68Ga]Ga-PSMA has prominence in current clinical guidelines. The novel tracer [68Ga]Ga-FAPi has also gained significant interest in the clinical context. A comparison of the labelling strategies followed to incorporate gallium-68 and fluorine-18 into the same molecular targeting constructs clearly demonstrate that gallium-68 complexation is the most convenient approach. Recently, cold kit based starting products are available to make the small-scale production of gallium-68 radiopharmaceuticals even more efficient when combined with generator produced gallium-68. The regulatory aspects is currently changing to support the implementation of gallium-68 and other diagnostic radiopharmaceuticals, simplifying the translation towards clinical use. Overall, the development of gallium-68 based radiopharmaceuticals is not only rapidly changing the landscape of diagnosis in oncology, but this growth also promotes innovation and progress in new applications of therapeutic radiometals such as lutetium-177 and actinium-225.
Collapse
Affiliation(s)
- Janke Kleynhans
- Department of Pharmaceutical and Pharmacological Sciences, Radiopharmaceutical Research, Katholieke Universiteit Leuven, Leuven, Belgium.
| | - Thomas Ebenhan
- Department of Nuclear Medicine, University of Pretoria, Pretoria, South Africa; Preclinical Imaging Facility, Nuclear Medicine Research Infrastructure, Pretoria, South Africa
| | - Mike Machaba Sathekge
- Preclinical Imaging Facility, Nuclear Medicine Research Infrastructure, Pretoria, South Africa; Department of Nuclear Medicine, University of Pretoria and Steve Biko Academic Hospital, Pretoria, South Africa
| |
Collapse
|
4
|
Ernst MJ, Abdulkader A, Hagenbach A, Claude G, Roca Jungfer M, Abram U. [Tc(NO)(Cp)(PPh 3)Cl] and [Tc(NO)(Cp)(PPh 3)(NCCH 3)](PF 6), and Their Reactions with Pyridine and Chalcogen Donors. Molecules 2024; 29:1114. [PMID: 38474627 DOI: 10.3390/molecules29051114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/25/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Reactions of the technetium(I) nitrosyl complex [Tc(NO)(Cp)(PPh3)Cl] with triphenylphosphine chalcogenides EPPh3 (E = O, S, Se), and Ag(PF6) in a CH2Cl2/MeOH mixture (v/v, 2/1) result in an exchange of the chlorido ligand and the formation of [Tc(NO)(Cp)(PPh3)(EPPh3)](PF6) compounds. The cationic acetonitrile complex [Tc(NO)(Cp)(PPh3)(NCCH3)]+ is formed when the reaction is conducted in NCCH3 without additional ligands. During the isolation of the corresponding PF6- salt a gradual decomposition of the anion was detected in the solvent mixture applied. The yields and the purity of the product increase when the BF4- salt is used instead. The acetonitrile ligand is bound remarkably strongly to technetium and exchange reactions readily proceed only with strong donors, such as pyridine or ligands with 'soft' donor atoms, such as the thioether thioxane. Substitutions on the cyclopentadienyl ring do not significantly influence the ligand exchange behavior of the starting material. 99Tc NMR spectroscopy is a valuable tool for the evaluation of reactions of the complexes of the present study. The extremely large chemical shift range of this method allows the ready detection of corresponding ligand exchange reactions. The observed 99Tc chemical shifts depend on the donor properties of the ligands. DFT calculations support the discussions about the experimental results and provide explanations for some of the unusual findings.
Collapse
Affiliation(s)
- Moritz Johannes Ernst
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Fabeckstr. 34/36, D-14195 Berlin, Germany
| | - Abdullah Abdulkader
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Fabeckstr. 34/36, D-14195 Berlin, Germany
| | - Adelheid Hagenbach
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Fabeckstr. 34/36, D-14195 Berlin, Germany
| | - Guilhem Claude
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Fabeckstr. 34/36, D-14195 Berlin, Germany
| | | | - Ulrich Abram
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Fabeckstr. 34/36, D-14195 Berlin, Germany
| |
Collapse
|
5
|
Pijeira MSO, Nunes PSG, Chaviano SL, Diaz AMA, DaSilva JN, Ricci-Junior E, Alencar LMR, Chen X, Santos-Oliveira R. Medicinal (Radio) Chemistry: Building Radiopharmaceuticals for the Future. Curr Med Chem 2024; 31:5481-5534. [PMID: 37594105 DOI: 10.2174/0929867331666230818092634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/30/2023] [Accepted: 07/13/2023] [Indexed: 08/19/2023]
Abstract
Radiopharmaceuticals are increasingly playing a leading role in diagnosing, monitoring, and treating disease. In comparison with conventional pharmaceuticals, the development of radiopharmaceuticals does follow the principles of medicinal chemistry in the context of imaging-altered physiological processes. The design of a novel radiopharmaceutical has several steps similar to conventional drug discovery and some particularity. In the present work, we revisited the insights of medicinal chemistry in the current radiopharmaceutical development giving examples in oncology, neurology, and cardiology. In this regard, we overviewed the literature on radiopharmaceutical development to study overexpressed targets such as prostate-specific membrane antigen and fibroblast activation protein in cancer; β-amyloid plaques and tau protein in brain disorders; and angiotensin II type 1 receptor in cardiac disease. The work addresses concepts in the field of radiopharmacy with a special focus on the potential use of radiopharmaceuticals for nuclear imaging and theranostics.
Collapse
Affiliation(s)
- Martha Sahylí Ortega Pijeira
- Laboratory of Nanoradiopharmaceuticals and Synthesis of Novel Radiopharmaceuticals, Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Rio de Janeiro 21941906, Brazil
| | - Paulo Sérgio Gonçalves Nunes
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas SP13083-970, Brazil
| | - Samila Leon Chaviano
- Laboratoire de Biomatériaux pour l'Imagerie Médicale, Axe Médicine Régénératrice, Centre de Recherche du Centre Hospitalier Universitaire de Québec - Université Laval, Québec, QC, Canada
| | - Aida M Abreu Diaz
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
- Institute de Génie Biomédical, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Jean N DaSilva
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal, Montréal, Québec, Canada
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
- Institute de Génie Biomédical, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Eduardo Ricci-Junior
- Laboratório de Desenvolvimento Galênico, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil
| | - Luciana Magalhães Rebelo Alencar
- Laboratory of Biophysics and Nanosystems, Federal University of Maranhão, Av. dos Portugueses, 1966, Vila Bacanga, São Luís MA65080-805, Brazil
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore 117597, Singapore
| | - Ralph Santos-Oliveira
- Laboratory of Nanoradiopharmaceuticals and Synthesis of Novel Radiopharmaceuticals, Brazilian Nuclear Energy Commission, Nuclear Engineering Institute, Rio de Janeiro 21941906, Brazil
- Laboratory of Radiopharmacy and Nanoradiopharmaceuticals, Rio de Janeiro State University, Rio de Janeiro 23070200, Brazil
| |
Collapse
|
6
|
Marlin A, Koller A, Madarasi E, Cordier M, Esteban-Gómez D, Platas-Iglesias C, Tircsó G, Boros E, Patinec V, Tripier R. H 3nota Derivatives Possessing Picolyl and Picolinate Pendants for Ga 3+ Coordination and 67Ga 3+ Radiolabeling. Inorg Chem 2023; 62:20634-20645. [PMID: 37552617 DOI: 10.1021/acs.inorgchem.3c01417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2023]
Abstract
We synthesized, thanks to the regiospecific N-functionalization using an orthoamide intermediate, two 1,4,7-triazacyclononane derivatives containing an acetate arm and either a methylpyridine or a picolinic acid group, respectively, Hnoapy and H2noapa, as new Ga3+ chelators for potential use in nuclear medicine. The corresponding Ga3+ complexes were synthesized and structurally characterized in solution by 1H and 13C NMR. The [Ga(noapy)]2+ complex appears to exist in solution as two diasteroisomeric pairs of enantiomers, as confirmed by density functional theory (DFT) calculations, while for [Ga(noapa)]+, a single species is present in solution. Solid-state investigations were possible for the [Ga(noapa)]+ complex, which crystallized from water as a pair of enantiomers. The average length of the N-Ga bonds of 2.090 Å is identical with that found for the [Ga(nota)] complex, showing that the presence of the picolinate arm does not hinder the coordination of the ligand to the metal ion. Protonation constants of noapy- and noapa2- were determined by potentiometric titrations, providing an overall basicity ∑log KiH (i = 1-4) that increases in the order noapy- < noapa2- < nota3- with increases in the negative charge of the ligand. Stability constants determined by pH-potentiometric titrations supplemented with 71Ga NMR data show that the stabilities of [Ga(noapy)]2+ and [Ga(noapa)]+ are lower compared to that of [Ga(nota)] but higher than those of other standards such as [Ga(aazta)]-. 67Ga radiolabeling studies were performed in order to demonstrate the potential of these chelators for 67/68Ga-based radiopharmaceuticals. The labelings of Hnoapy and H2noapa were nearly identical, outperforming H3nota. Stability studies were conducted in phosphate-buffered saline and in the presence of human serum transferrin, revealing no significant decomplexation of [67Ga][Ga(noapy)]2+ and [67Ga][Ga(noapa)]+ compared to [67Ga][Ga(nota)]. Finally, all complexes were found to be highly hydrophilic, with calculated log D7.4 values of -3.42 ± 0.05, -3.34 ± 0.04, and -3.00 ± 0.23 for Hnoapy, H2noapa, and H3nota, respectively, correlating with the charge of each complex and the electrostatic potentials obtained with DFT.
Collapse
Affiliation(s)
- Axia Marlin
- CEMCA, Université Brest, UMR 6521, CNRS, 6 avenue Victor le Gorgeu, 29238 Brest, France
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, New York 11794, United States
| | - Angus Koller
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, New York 11794, United States
| | - Enikö Madarasi
- Department of Physical Chemistry, Faculty of Science and Technology, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
- Doctoral School of Chemistry at the University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Marie Cordier
- Institut des Sciences Chimiques de Rennes, Université Rennes, UMR 6226, CNRS, F-35000 Rennes, France
| | - David Esteban-Gómez
- Centro de Investigacións Científicas Avanzadas and Departamento de Química, Universidade da Coruña, Campus da Zapateira, rúa da Fraga 10, 15008A Coruña, Spain
| | - Carlos Platas-Iglesias
- Centro de Investigacións Científicas Avanzadas and Departamento de Química, Universidade da Coruña, Campus da Zapateira, rúa da Fraga 10, 15008A Coruña, Spain
| | - Gyula Tircsó
- Department of Physical Chemistry, Faculty of Science and Technology, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Eszter Boros
- Department of Chemistry, Stony Brook University, 100 Nicolls Road, Stony Brook, New York 11794, United States
| | - Véronique Patinec
- CEMCA, Université Brest, UMR 6521, CNRS, 6 avenue Victor le Gorgeu, 29238 Brest, France
| | - Raphaël Tripier
- CEMCA, Université Brest, UMR 6521, CNRS, 6 avenue Victor le Gorgeu, 29238 Brest, France
| |
Collapse
|
7
|
Mitra JB, Mukherjee A, Kumar A, Chandak A, Rakshit S, Yadav HD, Pandey BN, Sarma HD. Imaging of bacterial infection: Harnessing positron emission tomography and Cherenkov luminescence imaging with UBI-derived octapeptide. Drug Dev Res 2023; 84:1513-1521. [PMID: 37571805 DOI: 10.1002/ddr.22103] [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: 05/22/2023] [Revised: 07/07/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023]
Abstract
Noninvasive imaging techniques for the early detection of infections are in high demand. In this study, we present the development of an infection imaging agent consisting of the antimicrobial peptide fragment UBI (31-38) conjugated to the chelator 1,4,7-triazacyclononane,1-glutaric acid-4,7-acetic acid (NODAGA), which allows for labeling with the positron emitter Ga-68. The preclinical evaluation of [68 Ga]Ga-NODAGA-UBI (31-38) was conducted to investigate its potential for imaging bacterial infections caused by Staphylococcus aureus. The octapeptide derived from ubiquicidin, UBI (31-38), was synthesized and conjugated with the chelator NODAGA. The conjugate was then radiolabeled with Ga-68. The radiolabeling process and the stability of the radio formulation were confirmed through chromatography. The study included both in vitro evaluations using S. aureus and in vivo evaluations in an animal model of infection and inflammation. Positron emission tomography (PET) and Cherenkov luminescence imaging (CLI) were performed to visualize the targeted localization of the radio formulation at the site of infection. Ex vivo biodistribution studies were carried out to quantify the uptake of the radio formulation in different organs and tissues. Additionally, the uptake of [18 F]Fluorodeoxyglucose ([18 F] FDG) in the animal model was also studied for comparison. The [68 Ga]Ga-NODAGA-UBI (31-38) complex consistently exhibited high radiochemical purity (>90%) after formulation. The complex demonstrated stability in saline, phosphate-buffered saline, and human serum for up to 3 h. Notably, the complex displayed significantly higher uptake in S. aureus, which was inhibited in the presence of unconjugated UBI (29-41) peptide, confirming the specificity of the formulation for bacterial membranes. Bacterial imaging capability was also observed in PET and CLI images. Biodistribution results indicated a substantial target-to-nontarget ratio of approximately 4 at 1 h postinjection of the radio formulation. Conversely, the uptake of [18 F]FDG in the animal model did not allow for the discrimination of infected and inflamed sites. Our studies have demonstrated that [68 Ga]Ga-NODAGA-UBI (31-38) holds promise as a radiotracer for imaging bacterial infections caused by S. aureus.
Collapse
Affiliation(s)
- Jyotsna Bhatt Mitra
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre (BARC), Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Archana Mukherjee
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre (BARC), Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Anuj Kumar
- Radiopharmaceuticals Division, Bhabha Atomic Research Centre (BARC), Mumbai, India
| | - Ashok Chandak
- Board of Radiation & Isotope Technology, Navi Mumbai, India
| | - Sutapa Rakshit
- Radiation Medicine Centre, Bhabha Atomic Research Centre (BARC), Mumbai, India
| | - Hansa D Yadav
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre (BARC), Mumbai, India
| | - Badri Narain Pandey
- Homi Bhabha National Institute, Mumbai, India
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre (BARC), Mumbai, India
| | - Haladhar Dev Sarma
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre (BARC), Mumbai, India
| |
Collapse
|
8
|
Vollett KDW, Szulc DA, Cheng HLM. A Manganese Porphyrin Platform for the Design and Synthesis of Molecular and Targeted MRI Contrast Agents. Int J Mol Sci 2023; 24:ijms24119532. [PMID: 37298480 DOI: 10.3390/ijms24119532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/24/2023] [Accepted: 05/29/2023] [Indexed: 06/12/2023] Open
Abstract
Magnetic resonance imaging (MRI) contrast agents, in contrast to the plethora of fluorescent agents available to target disease biomarkers or exogenous implants, have remained predominantly non-specific. That is, they do not preferentially accumulate in specific locations in vivo because doing so necessitates longer contrast retention, which is contraindicated for current gadolinium (Gd) agents. This double-edge sword implies that Gd agents can offer either rapid elimination (but lack specificity) or targeted accumulation (but with toxicity risks). For this reason, MRI contrast agent innovation has been severely constrained. Gd-free alternatives based on manganese (Mn) chelates have been largely ineffective, as they are inherently unstable. In this study, we present a Mn(III) porphyrin (MnP) platform for bioconjugation, offering the highest stability and chemical versatility compared to any other T1 contrast agent. We exploit the inherent metal stability conferred by porphyrins and the absence of pendant bases (found in Gd or Mn chelates) that limit versatile functionalization. As proof-of-principle, we demonstrate labeling of human serum albumin, a model protein, and collagen hydrogels for applications in in-vivo targeted imaging and material tracking, respectively. In-vitro and in-vivo results confirm unprecedented metal stability, ease of functionalization, and high T1 relaxivity. This new platform opens the door to ex-vivo validation by fluorescent imaging and multipurpose molecular imaging in vivo.
Collapse
Affiliation(s)
- Kyle D W Vollett
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
| | - Daniel A Szulc
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
| | - Hai-Ling Margaret Cheng
- Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
- Translational Biology & Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering, University of Toronto, Toronto, ON M5S 3G8, Canada
| |
Collapse
|
9
|
Polyak A, Képes Z, Trencsényi G. Implant Imaging: Perspectives of Nuclear Imaging in Implant, Biomaterial, and Stem Cell Research. Bioengineering (Basel) 2023; 10:bioengineering10050521. [PMID: 37237591 DOI: 10.3390/bioengineering10050521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/17/2023] [Accepted: 04/23/2023] [Indexed: 05/28/2023] Open
Abstract
Until now, very few efforts have been made to specifically trace, monitor, and visualize implantations, artificial organs, and bioengineered scaffolds for tissue engineering in vivo. While mainly X-Ray, CT, and MRI methods have been used for this purpose, the applications of more sensitive, quantitative, specific, radiotracer-based nuclear imaging techniques remain a challenge. As the need for biomaterials increases, so does the need for research tools to evaluate host responses. PET (positron emission tomography) and SPECT (single photon emission computer tomography) techniques are promising tools for the clinical translation of such regenerative medicine and tissue engineering efforts. These tracer-based methods offer unique and inevitable support, providing specific, quantitative, visual, non-invasive feedback on implanted biomaterials, devices, or transplanted cells. PET and SPECT can improve and accelerate these studies through biocompatibility, inertivity, and immune-response evaluations over long investigational periods at high sensitivities with low limits of detection. The wide range of radiopharmaceuticals, the newly developed specific bacteria, and the inflammation of specific or fibrosis-specific tracers as well as labeled individual nanomaterials can represent new, valuable tools for implant research. This review aims to summarize the opportunities of nuclear-imaging-supported implant research, including bone, fibrosis, bacteria, nanoparticle, and cell imaging, as well as the latest cutting-edge pretargeting methods.
Collapse
Affiliation(s)
- Andras Polyak
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - Zita Képes
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| | - György Trencsényi
- Division of Nuclear Medicine and Translational Imaging, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, Nagyerdei St. 98, H-4032 Debrecen, Hungary
| |
Collapse
|
10
|
Capponi PC, Mari M, Ferrari E, Asti M. Radiolabeled Chalcone Derivatives as Potential Radiotracers for β-Amyloid Plaques Imaging. Molecules 2023; 28:3233. [PMID: 37049995 PMCID: PMC10096019 DOI: 10.3390/molecules28073233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/30/2023] [Accepted: 04/03/2023] [Indexed: 04/09/2023] Open
Abstract
Natural products often provide a pool of pharmacologically relevant precursors for the development of various drug-related molecules. In this review, the research performed on some radiolabeled chalcone derivatives characterized by the presence of the α-β unsaturated carbonyl functional group as potential radiotracers for the imaging of β-amyloids plaques will be summarized. Chalcones' structural modifications and chemical approaches which allow their radiolabeling with the most common SPECT (Single Photon Emission Computed Tomography) and PET (Positron Emission Tomography) radionuclides will be described, as well as the state of the art regarding their in vitro binding affinity and in vivo biodistribution and pharmacokinetics in preclinical studies. Moreover, an explanation of the rationale behind their potential utilization as probes for Alzheimer's disease in nuclear medicine applications will be provided.
Collapse
Affiliation(s)
- Pier Cesare Capponi
- Radiopharmaceutical Chemistry Section, Nuclear Medicine Unit, AUSL-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42122 Reggio Emilia, Italy
| | - Matteo Mari
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia, Via G. Campi 103, 41125 Modena, Italy
| | - Erika Ferrari
- Department of Chemical and Geological Sciences, University of Modena and Reggio Emilia, Via G. Campi 103, 41125 Modena, Italy
| | - Mattia Asti
- Radiopharmaceutical Chemistry Section, Nuclear Medicine Unit, AUSL-IRCCS di Reggio Emilia, Viale Risorgimento 80, 42122 Reggio Emilia, Italy
| |
Collapse
|
11
|
[111In]In/[177Lu]Lu-AAZTA5-LM4 SST2R-Antagonists in Cancer Theranostics: From Preclinical Testing to First Patient Results. Pharmaceutics 2023; 15:pharmaceutics15030776. [PMID: 36986637 PMCID: PMC10053881 DOI: 10.3390/pharmaceutics15030776] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/03/2023] Open
Abstract
Aiming to expand the application of the SST2R-antagonist LM4 (DPhe-c[DCys-4Pal-DAph(Cbm)-Lys-Thr-Cys]-DTyr-NH2) beyond [68Ga]Ga-DATA5m-LM4 PET/CT (DATA5m, (6-pentanoic acid)-6-(amino)methy-1,4-diazepinetriacetate), we now introduce AAZTA5-LM4 (AAZTA5, 1,4-bis(carboxymethyl)-6-[bis(carboxymethyl)]amino-6-[pentanoic-acid]perhydro-1,4-diazepine), allowing for the convenient coordination of trivalent radiometals of clinical interest, such as In-111 (for SPECT/CT) or Lu-177 (for radionuclide therapy). After labeling, the preclinical profiles of [111In]In-AAZTA5-LM4 and [177Lu]Lu-AAZTA5-LM4 were compared in HEK293-SST2R cells and double HEK293-SST2R/wtHEK293 tumor-bearing mice using [111In]In-DOTA-LM3 and [177Lu]Lu-DOTA-LM3 as references. The biodistribution of [177Lu]Lu-AAZTA5-LM4 was additionally studied for the first time in a NET patient. Both [111In]In-AAZTA5-LM4 and [177Lu]Lu-AAZTA5-LM4 displayed high and selective targeting of the HEK293-SST2R tumors in mice and fast background clearance via the kidneys and the urinary system. This pattern was reproduced for [177Lu]Lu-AAZTA5-LM4 in the patient according to SPECT/CT results in a monitoring time span of 4–72 h pi. In view of the above, we may conclude that [177Lu]Lu-AAZTA5-LM4 shows promise as a therapeutic radiopharmaceutical candidate for SST2R-expressing human NETs, based on previous [68Ga]Ga-DATA5m-LM4 PET/CT, but further studies are needed to fully assess its clinical value. Furthermore, [111In]In-AAZTA5-LM4 SPECT/CT may represent a legitimate alternative diagnostic option in cases where PET/CT is not available.
Collapse
|
12
|
Braun D, Judmann B, Cheng X, Wängler B, Schirrmacher R, Fricker G, Wängler C. Synthesis, Radiolabeling, and In Vitro and In Vivo Characterization of Heterobivalent Peptidic Agents for Bispecific EGFR and Integrin α vβ 3 Targeting. ACS OMEGA 2023; 8:2793-2807. [PMID: 36687076 PMCID: PMC9850772 DOI: 10.1021/acsomega.2c07484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 12/15/2022] [Indexed: 06/17/2023]
Abstract
Radiolabeled heterobivalent peptidic ligands (HBPLs) are a highly promising compound class for the sensitive and specific visualization of tumors as they often exhibit superior properties compared to their monospecific counterparts and are able to concomitantly or complementarily address different receptor types. The combination of two receptor-specific agents targeting the epidermal growth factor receptor (EGFR) and the integrin αvβ3 in one HBPL would constitute a synergistic combination of binding motifs as these two receptor types are concurrently overexpressed on several human tumor types and are closely associated with disease progression and metastasis. Here, we designed and synthesized two heterobivalent radioligands consisting of the EGFR-specific peptide GE11 and αvβ3-specific cyclic RGD peptides, bearing a (1,4,7-triazacyclononane-4,7-diyl)diacetic acid-1-glutaric acid chelator for efficient radiolabeling and linkers of different lengths between both peptides. Both HBPLs were radiolabeled with 68Ga3+ in high radiochemical yields, purities of 96-99%, and molar activities of 36-88 GBq/μmol. [68Ga]Ga-1 and [68Ga]Ga-2 were evaluated for their log D(7.4) and stability toward degradation by human serum peptidases, showing a high hydrophilicity for both agents of -3.07 ± 0.01 and -3.44 ± 0.08 as well as a high stability toward peptidase degradation in human serum with half-lives of 272 and 237 min, respectively. Further on, the in vitro receptor binding profiles of both HBPLs to the target EGF and integrin αvβ3 receptors were assessed on EGFR-positive A431 and αvβ3-positive U87MG cells. Finally, we investigated the in vivo pharmacokinetics of HBPL [68Ga]Ga-1 by positron emission tomography/computed tomography imaging in A431 tumor-bearing xenograft mice to assess its potential for the receptor-specific visualization of EGFR- and/or αvβ3-expressing tumors. In these experiments, [68Ga]Ga-1 demonstrated a tumor uptake of 2.79 ± 1.66% ID/g, being higher than in all other organs and tissues apart from kidneys and blood at 2 h p.i. Receptor blocking studies revealed the observed tumor uptake to be solely mediated by integrin αvβ3, whereas no contribution of the GE11 peptide sequence to tumor uptake via the EGFR could be determined. Thus, the approach to develop radiolabeled EGFR- and integrin αvβ3-bispecific HBPLs is in general feasible although another peptide lead structure than GE11 should be used as the basis for the EGFR-specific part of the agents.
Collapse
Affiliation(s)
- Diana Braun
- Biomedical
Chemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty
Mannheim, Heidelberg University, 68167 Mannheim, Germany
- Molecular
Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine,
Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Benedikt Judmann
- Biomedical
Chemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty
Mannheim, Heidelberg University, 68167 Mannheim, Germany
- Molecular
Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine,
Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Xia Cheng
- Molecular
Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine,
Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Björn Wängler
- Molecular
Imaging and Radiochemistry, Clinic of Radiology and Nuclear Medicine,
Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Ralf Schirrmacher
- Department
of Oncology, Division of Oncological Imaging, University of Alberta, Edmonton, Alberta T6G 1Z2, Canada
| | - Gert Fricker
- Institute
of Pharmacy and Molecular Biotechnology, University of Heidelberg, 69120 Heidelberg, Germany
| | - Carmen Wängler
- Biomedical
Chemistry, Clinic of Radiology and Nuclear Medicine, Medical Faculty
Mannheim, Heidelberg University, 68167 Mannheim, Germany
| |
Collapse
|
13
|
Damerow H, Wängler B, Schirrmacher R, Fricker G, Wängler C. Synthesis of a Bifunctional Cross-Bridged Chelating Agent, Peptide Conjugation, and Comparison of 68 Ga Labeling and Complex Stability Characteristics with Established Chelators. ChemMedChem 2023; 18:e202200495. [PMID: 36259364 PMCID: PMC10100262 DOI: 10.1002/cmdc.202200495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/17/2022] [Indexed: 01/24/2023]
Abstract
[68 Ga]Ga3+ can be introduced into receptor-specific peptidic carriers via different chelators to obtain radiotracers for Positron Emission Tomography imaging and the chosen chelating agent considerably influences the in vivo pharmacokinetics of the corresponding radiopeptides. A chelator that should be a valuable alternative to established chelating agents for 68 Ga-radiolabeling of peptides would be a backbone-functionalized variant of the chelator CB-DO2A. Here, the bifunctional cross-bridged chelating agent CB-DO2A-GA was developed and compared to the established chelators DOTA, NODA-GA and DOTA-GA. For this purpose, CB-DO2A-GA(tBu)2 was introduced into the peptide Tyr3 -octreotate (TATE) and in direct comparison to the corresponding DOTA-, NODA-GA-, and DOTA-GA-modified TATE analogs, CB-DO2A-GA-TATE required harsher reaction conditions for 68 Ga-incorporation. Regarding the hydrophilicity profile of the resulting radiopeptides, a decrease in hydrophilicity from [68 Ga]Ga-DOTA-GA-TATE (logD(7.4) of -4.11±0.11) to [68 Ga]Ga-CB-DO2A-GA-TATE (-3.02±0.08) was observed. Assessing the stability against metabolic degradation and complex challenge, [68 Ga]Ga-CB-DO2A-GA demonstrated a very high kinetic inertness, exceeding that of [68 Ga]Ga-DOTA-GA. Therefore, CB-DO2A-GA is a valuable alternative to established chelating agents for 68 Ga-radiolabeling of peptides, especially when the formation of a very stable, positively charged 68 Ga-complex is pursued.
Collapse
Affiliation(s)
- Helen Damerow
- Clinic of Radiology and Nuclear Medicine, Biomedical ChemistryMedical Faculty Mannheim of Heidelberg University68167MannheimGermany
| | - Björn Wängler
- Clinic of Radiology and Nuclear Medicine, Molecular Imaging and RadiochemistryMedical Faculty Mannheim of Heidelberg University68167MannheimGermany
| | - Ralf Schirrmacher
- Department of Oncology, Division of Oncological ImagingUniversity of AlbertaEdmontonABT6G 1Z2Canada
| | - Gert Fricker
- Institute of Pharmacy and Molecular BiotechnologyUniversity of Heidelberg69120HeidelbergGermany
| | - Carmen Wängler
- Clinic of Radiology and Nuclear Medicine, Biomedical ChemistryMedical Faculty Mannheim of Heidelberg University68167MannheimGermany
| |
Collapse
|
14
|
Marjanovic-Painter B, Kleynhans J, Zeevaart JR, Rohwer E, Ebenhan T. A decade of ubiquicidin development for PET imaging of infection: A systematic review. Nucl Med Biol 2023; 116-117:108307. [PMID: 36435145 DOI: 10.1016/j.nucmedbio.2022.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/25/2022] [Accepted: 11/07/2022] [Indexed: 11/15/2022]
Abstract
BACKGROUND Ubiquicidin is a peptide fragment with selective binding to negatively charged bacterial cell membranes. Besides its earlier labelling with gamma emitting radionuclides, it has been labelled with Positron Emission Tomography (PET) radionuclides in the last decade for imaging infection and distinguishing infectious disease from sterile inflammation. This systematic review aims to evaluate the technology readiness level of PET based ubiquicidin radiopharmaceuticals. METHODS Two independent researchers reviewed all articles and abstracts pertaining ubiquicidin and PET imaging that are currently available. Scopus, Google Scholar and PubMed/Medline were used in the search. Upon completion of the literature search all articles and abstracts were evaluated and duplicates were excluded. All non-PET articles as well as review articles without new data were deemed ineligible. RESULTS From a total of 17 papers and 10 abstracts the studies were grouped into development, preclinical and clinical studies. Development was published in 15/17 (88%) publications and 6/10 (60%) abstracts, preclinical applications in 9/17 (53%) publications and 1/10 (10%) of abstracts. Finally, clinical studies made up 6/17 (35%) of full publications and 4/10 (40%) of the available abstracts. Development results were the most abundant. All the findings in the different areas of development of ubiquicidin as PET radiopharmaceutical are summarized in this paper. CONCLUSION Labelling procedures are generally uncomplicated and relatively fast and there are indications of adequate product stability. The production of PET radiopharmaceuticals based on UBI will therefore not be a barrier for clinical introduction of this technology. Systematization and unification of criteria for preclinical imaging and larger clinical trials are needed to ensure the translation of this radiopharmaceutical into the clinic. Therefore a conclusion with regards to the clinical relevance of ubiquicidin based PET is not yet possible.
Collapse
Affiliation(s)
| | - Janke Kleynhans
- Nuclear Medicine Research Infrastructure NPC, Pretoria, South Africa
| | - Jan Rijn Zeevaart
- Radiochemistry, The South African Nuclear Energy Corporation, Pelindaba, South Africa; Nuclear Medicine Research Infrastructure NPC, Pretoria, South Africa
| | - Egmont Rohwer
- Department of Chemistry, University of Pretoria, Pretoria, South Africa
| | - Thomas Ebenhan
- Radiochemistry, The South African Nuclear Energy Corporation, Pelindaba, South Africa; Department of Nuclear Medicine, University of Pretoria, Pretoria, South Africa; Nuclear Medicine Research Infrastructure NPC, Pretoria, South Africa.
| |
Collapse
|
15
|
Kanellopoulos P, Nock BA, Greifenstein L, Baum RP, Roesch F, Maina T. [ 68Ga]Ga-DATA 5m-LM4, a PET Radiotracer in the Diagnosis of SST 2R-Positive Tumors: Preclinical and First Clinical Results. Int J Mol Sci 2022; 23:ijms232314590. [PMID: 36498918 PMCID: PMC9740503 DOI: 10.3390/ijms232314590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 11/21/2022] [Accepted: 11/21/2022] [Indexed: 11/24/2022] Open
Abstract
Radiolabeled somatostatin subtype 2 receptor (SST2R)-antagonists have shown advantageous profiles for cancer theranostics compared with agonists. On the other hand, the newly introduced hybrid chelator (6-pentanoic acid)-6-(amino)methyl-1,4-diazepinetriacetate (DATA5m) rapidly binds Ga-68 (t1/2: 67.7 min) at much lower temperature, thus allowing for quick access to "ready-for-injection" [68Ga]Ga-tracers in hospitals. We herein introduce [68Ga]Ga-DATA5m-LM4 for PET/CT imaging of SST2R-positive human tumors. LM4 was obtained by 4Pal3/Tyr3-substitution in the known SST2R antagonist LM3 (H-DPhe-c[DCys-Tyr-DAph(Cbm)-Lys-Thr-Cys]-DTyr-NH2) and DATA5m was coupled at the N-terminus for labeling with radiogallium (Ga-67/68). [67Ga]Ga-DATA5m-LM4 was evaluated in HEK293-SST2R cells and mice models in a head-to-head comparison with [67Ga]Ga-DOTA-LM3. Clinical grade [68Ga]Ga-DATA5m-LM4 was prepared and injected in a neuroendocrine tumor (NET) patient for PET/CT imaging. DATA5m-LM4 displayed high SST2R binding affinity. [67Ga]Ga-DATA5m-LM4 showed markedly higher uptake in HEK293-SST2R cells versus [67Ga]Ga-DOTA-LM3 and was stable in vivo. In HEK293-SST2R xenograft-bearing mice, it achieved longer tumor retention and less kidney uptake than [67Ga]Ga-DOTA-LM3. [68Ga]Ga-DATA5m-LM4 accurately visualized tumor lesions with high contrast on PET/CT. In short, [68Ga]Ga-DATA5m-LM4 has shown excellent prospects for the PET/CT diagnosis of SST2R-positive tumors, further highlighting the benefits of Ga-68 labeling in a hospital environment via the DATA5m-chelator route.
Collapse
Affiliation(s)
| | - Berthold A. Nock
- Molecular Radiopharmacy, INRaSTES, NCSR “Demokritos”, GR-15310 Athens, Greece
| | - Lukas Greifenstein
- CURANOSTICUM Wiesbaden-Frankfurt, DKD Helios Klinik, D-65191 Wiesbaden, Germany
| | - Richard P. Baum
- CURANOSTICUM Wiesbaden-Frankfurt, DKD Helios Klinik, D-65191 Wiesbaden, Germany
| | - Frank Roesch
- Department Chemie, Standort TRIGA, Johannes Gutenberg-Universität Mainz, D-55126 Mainz, Germany
| | - Theodosia Maina
- Molecular Radiopharmacy, INRaSTES, NCSR “Demokritos”, GR-15310 Athens, Greece
- Correspondence: ; Tel.: +30-210-650-3908 (ext. 3891)
| |
Collapse
|
16
|
Lambidis E, Lumén D, Koskipahta E, Imlimthan S, Lopez BB, Sánchez AIF, Sarparanta M, Cheng RH, Airaksinen AJ. Synthesis and ex vivo biodistribution of two 68Ga-labeled tetrazine tracers: Comparison of pharmacokinetics. Nucl Med Biol 2022; 114-115:151-161. [PMID: 35680503 DOI: 10.1016/j.nucmedbio.2022.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 12/27/2022]
Abstract
Pretargeted PET imaging allows the use of radiotracers labeled with short-living PET radionuclides for tracing drugs with slow pharmacokinetics. Recently, especially methods based on bioorthogonal chemistry have been under intensive investigation for pretargeted PET imaging. The pharmacokinetics of the radiotracer is one of the factors that determine the success of the pretargeted strategy. Here, we report synthesis and biological evaluation of two 68Ga-labeled tetrazine (Tz)-based radiotracers, [68Ga]Ga-HBED-CC-PEG4-Tz ([68Ga]4) and [68Ga]Ga-DOTA-PEG4-Tz ([68Ga]6), aiming for development of new tracer candidates for pretargeted PET imaging based on the inverse electron demand Diels-Alder (IEDDA) ligation between a tetrazine and a strained alkene, such as trans-cyclooctene (TCO). Excellent radiochemical yield (RCY) was obtained for [68Ga]4 (RCY > 96%) and slightly lower for [68Ga]6 (RCY > 88%). Radiolabeling of HBED-CC-Tz proved to be faster and more efficient under milder conditions compared to the DOTA analogue. The two tracers exhibited excellent radiolabel stability both in vitro and in vivo. Moreover, [68Ga]4 was successfully used for radiolabeling two different TCO-functionalized nanoparticles in vitro: Hepatitis E virus nanoparticles (HEVNPs) and porous silicon nanoparticles (PSiNPs).
Collapse
Affiliation(s)
- Elisavet Lambidis
- Department of Chemistry, Radiochemistry, University of Helsinki, Helsinki FI-00014, Finland
| | - Dave Lumén
- Department of Chemistry, Radiochemistry, University of Helsinki, Helsinki FI-00014, Finland
| | - Elina Koskipahta
- Department of Chemistry, Radiochemistry, University of Helsinki, Helsinki FI-00014, Finland
| | - Surachet Imlimthan
- Department of Chemistry, Radiochemistry, University of Helsinki, Helsinki FI-00014, Finland
| | - Brianda B Lopez
- Department of Chemistry, Radiochemistry, University of Helsinki, Helsinki FI-00014, Finland
| | | | - Mirkka Sarparanta
- Department of Chemistry, Radiochemistry, University of Helsinki, Helsinki FI-00014, Finland
| | - R Holland Cheng
- Department of Molecular and Cellular Biology, University of California, Davis, CA 95616, USA
| | - Anu J Airaksinen
- Department of Chemistry, Radiochemistry, University of Helsinki, Helsinki FI-00014, Finland; Turku PET Centre, Department of Chemistry, University of Turku, Turku FI-20520, Finland.
| |
Collapse
|
17
|
Barta P, Kamaraj R, Kucharova M, Novy Z, Petrik M, Bendova K, Hajduch M, Pavek P, Trejtnar F. Preparation, In Vitro Affinity, and In Vivo Biodistribution of Receptor-Specific 68Ga-Labeled Peptides Targeting Vascular Endothelial Growth Factor Receptors. Bioconjug Chem 2022; 33:1825-1836. [PMID: 36197842 DOI: 10.1021/acs.bioconjchem.2c00272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
As angiogenesis plays a key role in tumor growth and metastasis, the angiogenic process has attracted scientific interest as a target for diagnostic and therapeutic agents. Factors influencing angiogenesis include the vascular endothelial growth factor (VEGF) family and the two associated receptor types (VEGFR-1 and VEGFR-2). VEGFR-1/-2 detection and quantification in cancer lesions are essential for tumor process management. As a result of the advantageous pharmacokinetics and image contrast, peptides radiolabeled with PET emitters have become interesting tools for the visualization of VEGFR-1/-2-positive tumors. In this study, we prepared 68Ga-labeled peptides containing 15 (peptide 1) and 23 (peptide 2) amino acids as new PET tracers for tumor angiogenic process imaging. METHODS The peptides were conjugated with NODAGA-tris(t-Bu ester) and subsequently radiolabeled with [68Ga]Ga-chloride. The prepared [68Ga]Ga-NODAGA-peptide 1 and [68Ga]Ga-NODAGA-peptide 2 were tested for radiochemical purity and saline/plasma stability. Consequently, the binding affinity toward VEGFRs was assessed in vitro on human glioblastoma and kidney carcinoma cells. The found peptide receptor affinity was compared with the calculated values in the PROtein binDIng enerGY prediction (PRODIGY) server. Finally, the biodistribution study was performed on BALB/c female mice to reveal the basic pharmacokinetic behavior of radiopeptides. RESULTS The in vitro affinity testing of [68Ga]Ga-NODAGA-peptides 1 and 2 showed retained receptor binding as characterized by equilibrium dissociation constant (KD) values in the range of 0.5-1.2 μM and inhibitory concentration 50% (IC50) values in the range of 3.0-5.6 μM. Better binding properties of peptide 2 to VEGFR-1/-2 were found in the PRODIGY server. The biodistribution study on mice showed remarkable accumulation of both peptides in the kidneys and urinary bladder with a short half-life after intravenous application. The in vitro plasma stability of [68Ga]Ga-NODAGA-peptide 2 was superior to that of [68Ga]Ga-NODAGA-peptide 1. CONCLUSIONS The obtained results demonstrated a high radiolabeling yield with no need for purification and preserved binding potency of 68Ga-labeled peptides 1 and 2 toward VEGFRs in cancer cells. The peptide-receptor protein interaction assessed in protein-peptide docking determined the strongest interaction of peptide 2 with domain 2 of VEGFR-2 in addition to a more acceptable plasma stability (t1/2 = 120 min) than that for peptide 1. We found both radiolabeled peptides very potent in their receptor binding, which makes them suitable imaging agents. The rapid transition of the radiopeptides into the urinary tract indicates suitable pharmacokinetic characteristics.
Collapse
Affiliation(s)
- Pavel Barta
- Faculty of Pharmacy in Hradec Kralove, Department of Biophysics and Physical Chemistry, Charles University, Hradec Kralove 500 05, Czech Republic
| | - Rajamanikkam Kamaraj
- Faculty of Pharmacy in Hradec Kralove, Department of Pharmacology and Toxicology, Charles University, Hradec Kralove 500 05, Czech Republic
| | - Monika Kucharova
- Faculty of Pharmacy in Hradec Kralove, Department of Biophysics and Physical Chemistry, Charles University, Hradec Kralove 500 05, Czech Republic
| | - Zbynek Novy
- Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Palacky University Olomouc, Olomouc 779 00, Czech Republic
| | - Milos Petrik
- Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Palacky University Olomouc, Olomouc 779 00, Czech Republic
| | - Katerina Bendova
- Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Palacky University Olomouc, Olomouc 779 00, Czech Republic
| | - Marian Hajduch
- Faculty of Medicine and Dentistry, Institute of Molecular and Translational Medicine, Palacky University Olomouc, Olomouc 779 00, Czech Republic
| | - Petr Pavek
- Faculty of Pharmacy in Hradec Kralove, Department of Pharmacology and Toxicology, Charles University, Hradec Kralove 500 05, Czech Republic
| | - Frantisek Trejtnar
- Faculty of Pharmacy in Hradec Kralove, Department of Pharmacology and Toxicology, Charles University, Hradec Kralove 500 05, Czech Republic
| |
Collapse
|
18
|
Hull A, Li Y, Bartholomeusz D, Hsieh W, Tieu W, Pukala TL, Staudacher AH, Bezak E. Preliminary Development and Testing of C595 Radioimmunoconjugates for Targeting MUC1 Cancer Epitopes in Pancreatic Ductal Adenocarcinoma. Cells 2022; 11:cells11192983. [PMID: 36230945 PMCID: PMC9563759 DOI: 10.3390/cells11192983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
Mucin 1 is a transmembrane glycoprotein which overexpresses cancer-specific epitopes (MUC1-CE) on pancreatic ductal adenocarcinoma (PDAC) cells. As PDAC is a low survival and highly aggressive malignancy, developing radioimmunoconjugates capable of targeting MUC1-CE could lead to improvements in PDAC outcomes. The aim of this study was to develop and perform preliminary testing of diagnostic and therapeutic radioimmunoconjugates for PDAC using an anti-MUC1 antibody, C595. Firstly, p-SCN-Bn-DOTA was conjugated to the C595 antibody to form a DOTA-C595 immunoconjugate. The stability and binding affinity of the DOTA-C595 conjugate was evaluated using mass spectrometry and ELISA. DOTA-C595 was radiolabelled to Copper-64, Lutetium-177, Gallium-68 and Technetium-99m to form novel radioimmunoconjugates. Cell binding assays were performed in PANC-1 (strong MUC1-CE expression) and AsPC-1 (weak MUC1-CE expression) cell lines using 64Cu-DOTA-C595 and 177Lu-DOTA-C595. An optimal molar ratio of 4:1 DOTA groups per C595 molecule was obtained from the conjugation process. DOTA-C595 labelled to Copper-64, Lutetium-177, and Technetium-99m with high efficiency, although the Gallium-68 labelling was low. 177Lu-DOTA-C595 demonstrated high cellular binding to the PANC-1 cell lines which was significantly greater than AsPC-1 binding at concentrations exceeding 100 nM (p < 0.05). 64Cu-DOTA-C595 showed similar binding to the PANC-1 and AsPC-1 cells with no significant differences observed between cell lines (p > 0.05). The high cellular binding of 177Lu-DOTA-C595 to MUC1-CE positive cell lines suggests promise as a therapeutic radioimmunoconjugate against PDAC while further work is required to harness the potential of 64Cu-DOTA-C595 as a diagnostic radioimmunoconjugate.
Collapse
Affiliation(s)
- Ashleigh Hull
- Allied Health and Human Performance Academic Unit, Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
- Department of PET, Nuclear Medicine & Bone Densitometry, Royal Adelaide Hospital, SA Medical Imaging, Adelaide, SA 5000, Australia
- Correspondence:
| | - Yanrui Li
- Allied Health and Human Performance Academic Unit, Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
| | - Dylan Bartholomeusz
- Department of PET, Nuclear Medicine & Bone Densitometry, Royal Adelaide Hospital, SA Medical Imaging, Adelaide, SA 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
| | - William Hsieh
- Allied Health and Human Performance Academic Unit, Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
- Department of PET, Nuclear Medicine & Bone Densitometry, Royal Adelaide Hospital, SA Medical Imaging, Adelaide, SA 5000, Australia
| | - William Tieu
- Molecular Imaging and Therapy Research Unit, South Australian Health and Medical Research Institute, Adelaide, SA 5000, Australia
- School of Physical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Tara L. Pukala
- School of Physical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Alexander H. Staudacher
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA 5000, Australia
| | - Eva Bezak
- Allied Health and Human Performance Academic Unit, Cancer Research Institute, University of South Australia, Adelaide, SA 5001, Australia
- School of Physical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia
| |
Collapse
|
19
|
Liu Y, Yu S, Xu T, Bodenko V, Orlova A, Oroujeni M, Rinne SS, Tolmachev V, Vorobyeva A, Gräslund T. Preclinical Evaluation of a New Format of 68Ga- and 111In-Labeled Affibody Molecule Z IGF-1R:4551 for the Visualization of IGF-1R Expression in Malignant Tumors Using PET and SPECT. Pharmaceutics 2022; 14:pharmaceutics14071475. [PMID: 35890370 PMCID: PMC9320461 DOI: 10.3390/pharmaceutics14071475] [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: 06/22/2022] [Revised: 07/11/2022] [Accepted: 07/13/2022] [Indexed: 02/04/2023] Open
Abstract
The Insulin-like growth factor-1 receptor (IGF-1R) is a molecular target for several monoclonal antibodies undergoing clinical evaluation as anticancer therapeutics. The non-invasive detection of IGF-1R expression in tumors might enable stratification of patients for specific treatment and improve the outcome of both clinical trials and routine treatment. The affibody molecule ZIGF-1R:4551 binds specifically to IGF-1R with subnanomolar affinity. The goal of this study was to evaluate the 68Ga and 111In-labeled affibody construct NODAGA-(HE)3-ZIGF-1R:4551 for the imaging of IGF-1R expression, using PET and SPECT. The labeling was efficient and provided stable coupling of both radionuclides. The two imaging probes, [68Ga]Ga-NODAGA-(HE)3-ZIGF-1R:4551 and [111In]In-NODAGA-(HE)3-ZIGF-1R:4551, demonstrated specific binding to IGF-1R-expressing human cancer cell lines in vitro and to IGF-1R-expressing xenografts in mice. Preclinical PET and SPECT/CT imaging demonstrated visualization of IGF-1R-expressing xenografts already one hour after injection. The tumor-to-blood ratios at 3 h after injection were 7.8 ± 0.2 and 8.0 ± 0.6 for [68Ga]Ga-NODAGA-(HE)3-ZIGF-1R:4551 and [111In]In-NODAGA-(HE)3-ZIGF-1R:4551, respectively. In conclusion, a molecular design of the ZIGF-1R:4551 affibody molecule, including placement of a (HE)3-tag on the N-terminus and site-specific coupling of a NODAGA chelator on the C-terminus, provides a tracer with improved imaging properties for visualization of IGF-1R in malignant tumors, using PET and SPECT.
Collapse
Affiliation(s)
- Yongsheng Liu
- Department of Immunology, Genetics and Pathology, Uppsala University, 75237 Uppsala, Sweden; (Y.L.); (T.X.); (M.O.); (A.V.)
| | - Shengze Yu
- Department of Protein Science, KTH Royal Institute of Technology, 10044 Stockholm, Sweden;
| | - Tianqi Xu
- Department of Immunology, Genetics and Pathology, Uppsala University, 75237 Uppsala, Sweden; (Y.L.); (T.X.); (M.O.); (A.V.)
| | - Vitalina Bodenko
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (V.B.); (A.O.)
| | - Anna Orlova
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (V.B.); (A.O.)
- Department of Medicinal Chemistry, Uppsala University, 75123 Uppsala, Sweden;
| | - Maryam Oroujeni
- Department of Immunology, Genetics and Pathology, Uppsala University, 75237 Uppsala, Sweden; (Y.L.); (T.X.); (M.O.); (A.V.)
- Affibody AB, 17165 Solna, Sweden
| | - Sara S. Rinne
- Department of Medicinal Chemistry, Uppsala University, 75123 Uppsala, Sweden;
| | - Vladimir Tolmachev
- Department of Immunology, Genetics and Pathology, Uppsala University, 75237 Uppsala, Sweden; (Y.L.); (T.X.); (M.O.); (A.V.)
- Research Centrum for Oncotheranostics, Research School of Chemistry and Applied Biomedical Sciences, Tomsk Polytechnic University, 634050 Tomsk, Russia; (V.B.); (A.O.)
- Correspondence: (V.T.); (T.G.); Tel.: +46-704-250782 (V.T.); +46-8790-9627 (T.G.)
| | - Anzhelika Vorobyeva
- Department of Immunology, Genetics and Pathology, Uppsala University, 75237 Uppsala, Sweden; (Y.L.); (T.X.); (M.O.); (A.V.)
| | - Torbjörn Gräslund
- Department of Protein Science, KTH Royal Institute of Technology, 10044 Stockholm, Sweden;
- Correspondence: (V.T.); (T.G.); Tel.: +46-704-250782 (V.T.); +46-8790-9627 (T.G.)
| |
Collapse
|
20
|
Porphyrins as Chelating Agents for Molecular Imaging in Nuclear Medicine. Molecules 2022; 27:molecules27103311. [PMID: 35630788 PMCID: PMC9148099 DOI: 10.3390/molecules27103311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 12/04/2022] Open
Abstract
Porphyrin ligands, showing a significant affinity for cancer cells, also have the ability to chelate metallic radioisotopes to form potential diagnostic radiopharmaceuticals. They can be applied in single-photon emission computed tomography (SPECT) and positron emission tomography (PET) to evaluate metabolic changes in the human body for tumor diagnostics. The aim of this paper is to present a short overview of the main metallic radionuclides complexed by porphyrin ligands and used in these techniques. These chelation reactions are discussed in terms of the complexation conditions and kinetics and the complex stability.
Collapse
|
21
|
Greifenstein L, Engelbogen N, Máthé D, Grus T, Rösch F, Bergmann R. Squaric Acid Bisphposphonates for Theranostics of Bone Metastasis - the Easy DOTA-Zoledronate. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2022; 2:870910. [PMID: 39354958 PMCID: PMC11440830 DOI: 10.3389/fnume.2022.870910] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/01/2022] [Indexed: 10/03/2024]
Abstract
Bisphosponates are an interesting molecular class and in recent years their application has found its way into radiopharmaceutical research and thus into molecular imaging. In addition to great imaging of bone metastases, bisphospnate-based tracers for imaging also have some significant drawbacks. For example, their synthesis is often difficult. Additionally, this can lead to complex and almost impossible purification and quality control. This has limited the production and labeling of suitable molecular and their widespread use to a few facilities. Our squaric acid-based approach provides a way to overcome these problems and makes the synthesis as well as the purification of the compounds much easier. In addition, we were able to demonstrate that labeling with 68Ga is possible under the typical conditions.
Collapse
Affiliation(s)
| | - Nils Engelbogen
- Institute of Nuclear Chemistry, Johannes Gutenberg University, Mainz, Germany
| | - Domokos Máthé
- Institute of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Tilmann Grus
- Institute of Nuclear Chemistry, Johannes Gutenberg University, Mainz, Germany
| | - Frank Rösch
- Institute of Nuclear Chemistry, Johannes Gutenberg University, Mainz, Germany
| | - Ralf Bergmann
- Institute of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden Rossendorf, Dresden Rossendorf, Germany
| |
Collapse
|
22
|
Holik HA, Ibrahim FM, Elaine AA, Putra BD, Achmad A, Kartamihardja AHS. The Chemical Scaffold of Theranostic Radiopharmaceuticals: Radionuclide, Bifunctional Chelator, and Pharmacokinetics Modifying Linker. Molecules 2022; 27:3062. [PMID: 35630536 PMCID: PMC9143622 DOI: 10.3390/molecules27103062] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/27/2022] [Accepted: 05/05/2022] [Indexed: 11/16/2022] Open
Abstract
Therapeutic radiopharmaceuticals have been researched extensively in the last decade as a result of the growing research interest in personalized medicine to improve diagnostic accuracy and intensify intensive therapy while limiting side effects. Radiometal-based drugs are of substantial interest because of their greater versatility for clinical translation compared to non-metal radionuclides. This paper comprehensively discusses various components commonly used as chemical scaffolds to build radiopharmaceutical agents, i.e., radionuclides, pharmacokinetic-modifying linkers, and chelators, whose characteristics are explained and can be used as a guide for the researcher.
Collapse
Affiliation(s)
- Holis Abdul Holik
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia; (F.M.I.); (A.A.E.); (B.D.P.)
| | - Faisal Maulana Ibrahim
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia; (F.M.I.); (A.A.E.); (B.D.P.)
| | - Angela Alysia Elaine
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia; (F.M.I.); (A.A.E.); (B.D.P.)
| | - Bernap Dwi Putra
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia; (F.M.I.); (A.A.E.); (B.D.P.)
| | - Arifudin Achmad
- Department of Nuclear Medicine and Molecular Theranostics, Faculty of Medicine, Universitas Padjadjaran/Hasan Sadikin General Hospital, Bandung 40161, Indonesia; (A.A.); (A.H.S.K.)
- Oncology and Stem Cell Working Group, Faculty of Medicine, Universitas Padjadjaran, Bandung 40161, Indonesia
| | - Achmad Hussein Sundawa Kartamihardja
- Department of Nuclear Medicine and Molecular Theranostics, Faculty of Medicine, Universitas Padjadjaran/Hasan Sadikin General Hospital, Bandung 40161, Indonesia; (A.A.); (A.H.S.K.)
| |
Collapse
|
23
|
Wang S, Gai Y, Sun L, Lan X, Zeng D, Xiang G, Ma X. Synthesis and evaluation of novel 1,4,7-triazacyclononane derivatives as Cu2+ and Ga3+ chelators. J Inorg Biochem 2022; 229:111719. [DOI: 10.1016/j.jinorgbio.2022.111719] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/31/2021] [Accepted: 01/02/2022] [Indexed: 12/25/2022]
|
24
|
Fersing C, Masurier N, Rubira L, Deshayes E, Lisowski V. AAZTA-Derived Chelators for the Design of Innovative Radiopharmaceuticals with Theranostic Applications. Pharmaceuticals (Basel) 2022; 15:234. [PMID: 35215346 PMCID: PMC8879111 DOI: 10.3390/ph15020234] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 02/12/2022] [Accepted: 02/14/2022] [Indexed: 02/06/2023] Open
Abstract
With the development of 68Ga and 177Lu radiochemistry, theranostic approaches in modern nuclear medicine enabling patient-centered personalized medicine applications have been growing in the last decade. In conjunction with the search for new relevant molecular targets, the design of innovative chelating agents to easily form stable complexes with various radiometals for theranostic applications has gained evident momentum. Initially conceived for magnetic resonance imaging applications, the chelating agent AAZTA features a mesocyclic seven-membered diazepane ring, conferring some of the properties of both acyclic and macrocyclic chelating agents. Described in the early 2000s, AAZTA and its derivatives exhibited interesting properties once complexed with metals and radiometals, combining a fast kinetic of formation with a slow kinetic of dissociation. Importantly, the extremely short coordination reaction times allowed by AAZTA derivatives were particularly suitable for short half-life radioelements (i.e., 68Ga). In view of these particular characteristics, the scope of this review is to provide a survey on the design, synthesis, and applications in the nuclear medicine/radiopharmacy field of AAZTA-derived chelators.
Collapse
Affiliation(s)
- Cyril Fersing
- Nuclear Medicine Department, Institut Régional du Cancer de Montpellier (ICM), University Montpellier, 34298 Montpellier, France; (L.R.); (E.D.)
- IBMM, University Montpellier, CNRS, ENSCM, 34293 Montpellier, France; (N.M.); (V.L.)
| | - Nicolas Masurier
- IBMM, University Montpellier, CNRS, ENSCM, 34293 Montpellier, France; (N.M.); (V.L.)
| | - Léa Rubira
- Nuclear Medicine Department, Institut Régional du Cancer de Montpellier (ICM), University Montpellier, 34298 Montpellier, France; (L.R.); (E.D.)
| | - Emmanuel Deshayes
- Nuclear Medicine Department, Institut Régional du Cancer de Montpellier (ICM), University Montpellier, 34298 Montpellier, France; (L.R.); (E.D.)
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Institut Régional du Cancer de Montpellier (ICM), University of Montpellier, 34298 Montpellier, France
| | - Vincent Lisowski
- IBMM, University Montpellier, CNRS, ENSCM, 34293 Montpellier, France; (N.M.); (V.L.)
- Department of Pharmacy, Lapeyronie Hospital, CHU Montpellier, 191 Av. du Doyen Gaston Giraud, 34295 Montpellier, France
| |
Collapse
|
25
|
Lepareur N. Cold Kit Labeling: The Future of 68Ga Radiopharmaceuticals? Front Med (Lausanne) 2022; 9:812050. [PMID: 35223907 PMCID: PMC8869247 DOI: 10.3389/fmed.2022.812050] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/07/2022] [Indexed: 12/11/2022] Open
Abstract
Over the last couple of decades, gallium-68 (68Ga) has gained a formidable interest for PET molecular imaging of various conditions, from cancer to infection, through cardiac pathologies or neuropathies. It has gained routine use, with successful radiopharmaceuticals such as somatostatin analogs ([68Ga]Ga-DOTATOC and [68Ga]GaDOTATATE) for neuroendocrine tumors, and PSMA ligands for prostate cancer. It represents a major clinical impact, particularly in the context of theranostics, coupled with their 177Lu-labeled counterparts. Beside those, a bunch of new 68Ga-labeled molecules are in the preclinical and clinical pipelines, with some of them showing great promise for patient care. Increasing clinical demand and regulatory issues have led to the development of automated procedures for the production of 68Ga radiopharmaceuticals. However, the widespread use of these radiopharmaceuticals may rely on simple and efficient radiolabeling methods, undemanding in terms of equipment and infrastructure. To make them technically and economically accessible to the medical community and its patients, it appears mandatory to develop a procedure similar to the well-established kit-based 99mTc chemistry. Already available commercial kits for the production of 68Ga radiopharmaceuticals have demonstrated the feasibility of using such an approach, thus paving the way for more kit-based 68Ga radiopharmaceuticals to be developed. This article discusses the development of 68Ga cold kit radiopharmacy, including technical issues, and regulatory aspects.
Collapse
Affiliation(s)
- Nicolas Lepareur
- Comprehensive Cancer Center Eugène Marquis, Rennes, France
- Univ Rennes, Inrae, Inserm, Institut NUMECAN (Nutrition, Métabolismes et Cancer), UMR_A 1341, UMR_S 1241, Rennes, France
| |
Collapse
|
26
|
Radiometals—Chemistry and radiolabeling. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00044-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
27
|
Hydroxypyridinones as a Very Promising Platform for Targeted Diagnostic and Therapeutic Radiopharmaceuticals. Molecules 2021. [DOI: 10.3390/molecules26226997
expr 973886017 + 973118332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Hydroxypyridinones (HOPOs) have been used in the chelation therapy of iron and actinide metals. Their application in metal-based radiopharmaceuticals has also been increasing in recent years. This review article focuses on how multidentate HOPOs can be used in targeted radiometal-based diagnostic and therapeutic radiopharmaceuticals. The general structure of radiometal-based targeted radiopharmaceuticals, a brief description of siderophores, the basic structure and properties of bidentate HOPO, some representative HOPO multidentate chelating agents, radiopharmaceuticals based on HOPO multidentate bifunctional chelators for gallium-68, thorium-227 and zirconium-89, as well as the future prospects of HOPO multidentate bifunctional chelators in other metal-based radiopharmaceuticals are described and discussed in turn. The HOPO metal-based radiopharmaceuticals that have shown good prospects in clinical and preclinical studies are gallium-68, thorium-227 and zirconium-89 radiopharmaceuticals. We expect HOPO multidentate bifunctional chelators to be a very promising platform for building novel targeted radiometal-based diagnostic and therapeutic radiopharmaceuticals.
Collapse
|
28
|
Zhou X, Dong L, Shen L. Hydroxypyridinones as a Very Promising Platform for Targeted Diagnostic and Therapeutic Radiopharmaceuticals. Molecules 2021; 26:6997. [PMID: 34834087 PMCID: PMC8619595 DOI: 10.3390/molecules26226997&set/a 916769719+956065658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Hydroxypyridinones (HOPOs) have been used in the chelation therapy of iron and actinide metals. Their application in metal-based radiopharmaceuticals has also been increasing in recent years. This review article focuses on how multidentate HOPOs can be used in targeted radiometal-based diagnostic and therapeutic radiopharmaceuticals. The general structure of radiometal-based targeted radiopharmaceuticals, a brief description of siderophores, the basic structure and properties of bidentate HOPO, some representative HOPO multidentate chelating agents, radiopharmaceuticals based on HOPO multidentate bifunctional chelators for gallium-68, thorium-227 and zirconium-89, as well as the future prospects of HOPO multidentate bifunctional chelators in other metal-based radiopharmaceuticals are described and discussed in turn. The HOPO metal-based radiopharmaceuticals that have shown good prospects in clinical and preclinical studies are gallium-68, thorium-227 and zirconium-89 radiopharmaceuticals. We expect HOPO multidentate bifunctional chelators to be a very promising platform for building novel targeted radiometal-based diagnostic and therapeutic radiopharmaceuticals.
Collapse
Affiliation(s)
- Xu Zhou
- HTA Co., Ltd., Beijing 102413, China;
- China Isotope & Radiation Corporation, Beijing 100089, China;
| | - Linlin Dong
- China Isotope & Radiation Corporation, Beijing 100089, China;
| | - Langtao Shen
- HTA Co., Ltd., Beijing 102413, China;
- China Isotope & Radiation Corporation, Beijing 100089, China;
- National Isotope Center of Engineering and Technology, China Institute of Atomic Energy, Beijing 102413, China
- Correspondence:
| |
Collapse
|
29
|
Zhou X, Dong L, Shen L. Hydroxypyridinones as a Very Promising Platform for Targeted Diagnostic and Therapeutic Radiopharmaceuticals. Molecules 2021; 26:6997. [PMID: 34834087 PMCID: PMC8619595 DOI: 10.3390/molecules26226997] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 11/13/2021] [Accepted: 11/17/2021] [Indexed: 01/30/2023] Open
Abstract
Hydroxypyridinones (HOPOs) have been used in the chelation therapy of iron and actinide metals. Their application in metal-based radiopharmaceuticals has also been increasing in recent years. This review article focuses on how multidentate HOPOs can be used in targeted radiometal-based diagnostic and therapeutic radiopharmaceuticals. The general structure of radiometal-based targeted radiopharmaceuticals, a brief description of siderophores, the basic structure and properties of bidentate HOPO, some representative HOPO multidentate chelating agents, radiopharmaceuticals based on HOPO multidentate bifunctional chelators for gallium-68, thorium-227 and zirconium-89, as well as the future prospects of HOPO multidentate bifunctional chelators in other metal-based radiopharmaceuticals are described and discussed in turn. The HOPO metal-based radiopharmaceuticals that have shown good prospects in clinical and preclinical studies are gallium-68, thorium-227 and zirconium-89 radiopharmaceuticals. We expect HOPO multidentate bifunctional chelators to be a very promising platform for building novel targeted radiometal-based diagnostic and therapeutic radiopharmaceuticals.
Collapse
Affiliation(s)
- Xu Zhou
- HTA Co., Ltd., Beijing 102413, China;
- China Isotope & Radiation Corporation, Beijing 100089, China;
| | - Linlin Dong
- China Isotope & Radiation Corporation, Beijing 100089, China;
| | - Langtao Shen
- HTA Co., Ltd., Beijing 102413, China;
- China Isotope & Radiation Corporation, Beijing 100089, China;
- National Isotope Center of Engineering and Technology, China Institute of Atomic Energy, Beijing 102413, China
| |
Collapse
|
30
|
Li L, Zhao R, Hong H, Li G, Zhang Y, Luo Y, Zha Z, Zhu J, Qiao J, Zhu L, Kung HF. 68Ga-labelled-exendin-4: New GLP1R targeting agents for imaging pancreatic β-cell and insulinoma. Nucl Med Biol 2021; 102-103:87-96. [PMID: 34695640 DOI: 10.1016/j.nucmedbio.2021.10.001] [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] [Received: 07/31/2021] [Revised: 09/13/2021] [Accepted: 10/05/2021] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Glucagon-like peptide-1 receptor (GLP1R) specifically expressed on the surface of pancreatic β-cells and insulinoma, is a potential biomarker for imaging β-cell mass (BCM). In this study, two new 68Ga-labelled GLP1R targeting agents were prepared and their biological properties for imaging BCM and insulinoma were evaluated. METHODS [68Ga]Ga-HBED-CC-MAL-Cys39-exendin-4 ([68Ga]Ga-4) and its dimer ([68Ga]Ga-5) were synthesized from corresponding precursors. Cell uptake studies were evaluated in INS-1 cells. Biodistribution and microPET studies were performed in male normal Sprague-Dawley rats, diabetic rats and insulinoma xenograft NOD/SCID mice. RESULTS [68Ga]Ga-4 and [68Ga]Ga-5 were efficiently radiolabelled by a simple one-step reaction without purification leading to high radiochemical yields and radiochemical purities (both >95%, decay corrected, n = 6, molar activity 15 GBq/μmol). They both showed excellent stability (~95%) in phosphate-buffered saline, pH 7.4, and in rat serum (~90%) for 2 h. Biodistribution studies and small animal PET/CT imaging showed that [68Ga]Ga-4 displayed specific uptake in rat pancreas and mouse insulinoma, and a reduced uptake in the pancreas of diabetic rat was observed (~62% reduction). Notably, it exhibited a rapid time-to-peak pancreatic uptake (0.96 ± 0.19%ID/g in 15 min) and fast clearance from the kidney (42% clearance in 30 min). Results suggested a favorable in vivo kinetics for human imaging studies. CONCLUSIONS [68Ga]Ga-4 targeting GLP1R of pancreatic β-cells may be a potentially useful PET agent and a suitable candidate for further structural modification studies. This agent has demonstrated several advantages, rapid time-to-peak pancreatic uptake and faster clearance from the kidney, factors may enhance diagnosis of diabetes and insulinoma.
Collapse
Affiliation(s)
- Linlin Li
- College of Chemistry, Beijing Normal University, Beijing, China
| | - Ruiyue Zhao
- College of Chemistry, Beijing Normal University, Beijing, China
| | - Haiyan Hong
- College of Chemistry, Beijing Normal University, Beijing, China
| | - Guangwen Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yan Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yang Luo
- College of Chemistry, Beijing Normal University, Beijing, China
| | - Zhihao Zha
- Department of Radiology, University of Pennsylvania, USA
| | - Jinxia Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Jinping Qiao
- College of Chemistry, Beijing Normal University, Beijing, China
| | - Lin Zhu
- College of Chemistry, Beijing Normal University, Beijing, China.
| | - Hank F Kung
- Department of Radiology, University of Pennsylvania, USA.
| |
Collapse
|
31
|
Choudhary N, Scheiber H, Zhang J, Patrick BO, de Guadalupe Jaraquemada-Peláez M, Orvig C. H 4HBEDpa: Octadentate Chelate after A. E. Martell. Inorg Chem 2021; 60:12855-12869. [PMID: 34424678 DOI: 10.1021/acs.inorgchem.1c01175] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
H4HBEDpa, a new octadentate chelator inspired by the 1960s ligand HBED of Arthur E. Martell, has been investigated for a selection of trivalent metal ions useful in diagnostic and therapeutic applications (Sc3+, Fe3+, Ga3+, In3+, and Lu3+). Complex formation equilibria were thoroughly investigated using combined potentiometric and UV-vis spectrophotometric titrations which revealed effective chelation and high metal-sequestering capacity, in particular for Fe3+, log KFeL = 36.62, [Fe(HBEDpa)]-. X-ray diffraction study of single crystals revealed that the ligand is preorganized and forms hexa-coordinated complexes with Fe3+ and Ga3+ at acidic pH. Density functional theory (DFT) calculations were applied to probe the geometries and energies of all the possible conformers of [M(HBEDpa)]- (M = Sc3+, Fe3+, Ga3+, In3+, and Lu3+). DFT calculations confirmed the experimental findings, indicating that [Fe(HBEDpa)]- is bound tightly in an asymmetric pattern as compared to the symmetrically bound and more open [Ga(HBEDpa)]-, prone to hydrolysis at higher pH. DFT calculations also showed that a large metal ion such as Lu3+ fully coordinates with HBEDpa4-, forming a binary octadentate complex in its lowest-energy form. Smaller metal ions form six or seven coordinate complexes with HBEDpa4-.
Collapse
Affiliation(s)
- Neha Choudhary
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada.,Life Sciences Division, TRIUMF, 4004 Wesbrook Mall, Vancouver, British Columbia V6T 2A3, Canada
| | - Hayden Scheiber
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada
| | - Jiale Zhang
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada
| | - Brian O Patrick
- Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada
| | - María de Guadalupe Jaraquemada-Peláez
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada
| | - Chris Orvig
- Medicinal Inorganic Chemistry Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia V6T 1Z1, Canada
| |
Collapse
|
32
|
Sneddon D, Cornelissen B. Emerging chelators for nuclear imaging. Curr Opin Chem Biol 2021; 63:152-162. [PMID: 34051509 DOI: 10.1016/j.cbpa.2021.03.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/28/2021] [Accepted: 03/09/2021] [Indexed: 11/25/2022]
Abstract
Chelators are necessary in nuclear medicine imaging to direct an inorganic radionuclide, a radiometal, to a desired target; unfortunately, there is no 'one-size-fits-all' chelator. As the toolbox of radiometals is expanding, new chelators are required to prevent off-target side effects. 1,4,7,10-Tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) is the current gold standard chelator for several radiometals, but typically, chelation requires harsh conditions, making it unsuitable to label biological vectors. The ideal chelator would allow labelling under mild conditions (near-neutral pH and low temperatures [∼37 °C]) and be both thermodynamically and kinetically stable. Over the past 2-3 years, several exciting chelators have been developed that have superior properties to make them worth investigating for future clinical applications.
Collapse
Affiliation(s)
- Deborah Sneddon
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, OX3 7LE, United Kingdom.
| | - Bart Cornelissen
- MRC Oxford Institute for Radiation Oncology, Department of Oncology, University of Oxford, OX3 7LE, United Kingdom
| |
Collapse
|
33
|
Cardoso ME, Tejería E, Zirbesegger K, Savio E, Terán M, Rey Ríos AM. Development and characterization of two novel 68 Ga-labelled neuropeptide Y short analogues with potential application in breast cancer imaging. Chem Biol Drug Des 2021; 98:182-191. [PMID: 33982434 DOI: 10.1111/cbdd.13864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 04/05/2021] [Accepted: 05/01/2021] [Indexed: 12/20/2022]
Abstract
In vivo receptor targeting with radiolabelled peptide-based probes is an attractive approach for the development of novel radiotracers for molecular imaging. This work presents the development and characterization of two novel neuropeptide Y analogues labelled with a positron emitter 68 Ga, for potential use in breast cancer imaging. Both analogues share the same amino acid sequence and were derivatized with NOTA through either a lysine linker (L1) or an acetylated lysine (L2). In both cases, a single product with radiochemical purity higher than 95% was obtained. The two complexes were hydrophilic, showed remarkable in vitro stability, good cellular uptake, binding affinity in the nanomolar range and high cellular internalization rate. Biodistribution studies revealed low blood uptake and elimination through the urinary tract. The addition of an acetyl group in the spacer increased the lipophilicity of C2 and modified the reactivity of the ε-amino group of the lysine which resulted in lower protein binding and lower percentage of injected dose in bladder and urine. The tumour versus muscle ratio was (3.8 ± 0.4) for 68 Ga-L1 and (4.7 ± 0.4) for 68 Ga-L2. These results encourage performing further studies in order to complete the evaluation of both tracers as potential radiopharmaceutical for breast cancer imaging.
Collapse
Affiliation(s)
- María Elena Cardoso
- Área de Radioquímica, Departamento Estrella Campos, Facultad de Química, Universidad de la República (UdelaR), Montevideo, Uruguay
- Área de Investigación y Desarrollo Biomédico y Químico Farmacéutico, Centro Uruguayo de Imagenología Molecular (CUDIM), Montevideo, Uruguay
- Graduate Program in Chemistry, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Emilia Tejería
- Área de Radioquímica, Departamento Estrella Campos, Facultad de Química, Universidad de la República (UdelaR), Montevideo, Uruguay
| | - Kevin Zirbesegger
- Área de Investigación y Desarrollo Biomédico y Químico Farmacéutico, Centro Uruguayo de Imagenología Molecular (CUDIM), Montevideo, Uruguay
| | - Eduardo Savio
- Área de Investigación y Desarrollo Biomédico y Químico Farmacéutico, Centro Uruguayo de Imagenología Molecular (CUDIM), Montevideo, Uruguay
| | - Mariella Terán
- Área de Radioquímica, Departamento Estrella Campos, Facultad de Química, Universidad de la República (UdelaR), Montevideo, Uruguay
| | - Ana María Rey Ríos
- Área de Radioquímica, Departamento Estrella Campos, Facultad de Química, Universidad de la República (UdelaR), Montevideo, Uruguay
| |
Collapse
|
34
|
Renard E, Moreau M, Bellaye PS, Guillemin M, Collin B, Prignon A, Denat F, Goncalves V. Positron Emission Tomography Imaging of Neurotensin Receptor-Positive Tumors with 68Ga-Labeled Antagonists: The Chelate Makes the Difference Again. J Med Chem 2021; 64:8564-8578. [PMID: 34107209 DOI: 10.1021/acs.jmedchem.1c00523] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Neurotensin receptor 1 (NTS1) is involved in the development and progression of numerous cancers, which makes it an interesting target for the development of diagnostic and therapeutic agents. A small molecule NTS1 antagonist, named [177Lu]Lu-IPN01087, is currently evaluated in phase I/II clinical trials for the targeted therapy of neurotensin receptor-positive cancers. In this study, we synthesized seven compounds based on the structure of NTS1 antagonists, bearing different chelating agents, and radiolabeled them with gallium-68 for PET imaging. These compounds were evaluated in vitro and in vivo in mice bearing a HT-29 xenograft. The compound [68Ga]Ga-bisNODAGA-16 showed a promising biodistribution profile with mainly signal in tumor (4.917 ± 0.776%ID/g, 2 h post-injection). Its rapid clearance from healthy tissues led to high tumor-to-organ ratios, resulting in highly contrasted PET images. These results were confirmed on subcutaneous xenografts of AsPC-1 tumor cells, a model of NTS1-positive human pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Emma Renard
- Institut de Chimie Moléculaire de l'Université de Bourgogne, ICMUB UMR CNRS 6302, Université Bourgogne Franche-Comté, Dijon 21000, France
| | - Mathieu Moreau
- Institut de Chimie Moléculaire de l'Université de Bourgogne, ICMUB UMR CNRS 6302, Université Bourgogne Franche-Comté, Dijon 21000, France
| | | | - Mélanie Guillemin
- Georges-François LECLERC Cancer Center - UNICANCER, Dijon 21000, France
| | - Bertrand Collin
- Georges-François LECLERC Cancer Center - UNICANCER, Dijon 21000, France
| | - Aurélie Prignon
- UMS28 Laboratoire d'Imagerie Moléculaire Positonique (LIMP), Sorbonne Université, Paris 75020, France
| | - Franck Denat
- Institut de Chimie Moléculaire de l'Université de Bourgogne, ICMUB UMR CNRS 6302, Université Bourgogne Franche-Comté, Dijon 21000, France
| | - Victor Goncalves
- Institut de Chimie Moléculaire de l'Université de Bourgogne, ICMUB UMR CNRS 6302, Université Bourgogne Franche-Comté, Dijon 21000, France
| |
Collapse
|
35
|
Zha Z, Choi SR, Ploessl K, Alexoff D, Zhao R, Zhu L, Kung HF. Radiolabeling Optimization and Preclinical Evaluation of the New PSMA Imaging Agent [ 18F]AlF-P16-093. Bioconjug Chem 2021; 32:1017-1026. [PMID: 33872489 DOI: 10.1021/acs.bioconjchem.1c00177] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Prostate-specific membrane antigen (PSMA)-targeted radioligands have played an increasing role in the diagnosis of prostate cancer. [68Ga]Ga-P16-093 is a PSMA-targeting agent for positron emission tomography imaging, currently under a Phase 2 clinical trial. In the present study, P16-093 was labeled with 18F via [18F]AlF2+ complex formation, and the biological properties of [18F]AlF-P16-093 were evaluated. Optimization of radiolabeling efficiency was performed by testing a series of parameters, including the amount of free ligand; the amount of Al3+; and the influence of solvent, pH, temperature, reaction time, and reaction volume. Optimal labeling results were achieved at pH 5 by reacting at 60 °C for 15 min in a vial containing 74-370 MBq of [18F]fluoride, 46 nmol of P16-093, 40 nmol of AlCl3·6 H2O, and 50% EtOH. [18F]AlF-P16-093 was prepared with a non-decay-corrected radiochemical yield of 54.4 ± 4.4% (n = 9) within 30 min (final radiochemical purity ≥95%). In vitro, [18F]AlF-P16-093 showed PSMA-specific high uptakes in PIP-PC3 cells. The binding affinity of [18F]AlF-P16-093 to PSMA was determined as Kd of 12.4 ± 2.0 nM. The tumor uptake in mice with a xenografted PSMA-expressing PIP-PC3 tumor was high (18.8 ± 5.14% ID/g at 1 h postinjection) and retained without washout for 2 h. In addition, tumor uptake was almost completely blocked by coinjecting a PSMA inhibitor, 2-PMPA. The bone activity at 1 h post injection was higher with [18F]AlF-P16-093 (2.83 ± 0.49% ID/g) in comparison to that of [68Ga]Ga-P16-093 (0.26 ± 0.07% ID/g). In summary, an efficient and simple radiosynthesis of [18F]AlF-P16-093 was achieved. [18F]AlF-P16-093 showed desirable in vivo pharmacokinetics and excellent PSMA-targeting properties for imaging PSMA expression in prostate cancer.
Collapse
Affiliation(s)
- Zhihao Zha
- Five Eleven Pharma Inc., Philadelphia, Pennsylvania 19104, United States
| | - Seok Rye Choi
- Five Eleven Pharma Inc., Philadelphia, Pennsylvania 19104, United States
| | - Karl Ploessl
- Five Eleven Pharma Inc., Philadelphia, Pennsylvania 19104, United States
| | - David Alexoff
- Five Eleven Pharma Inc., Philadelphia, Pennsylvania 19104, United States
| | - Ruiyue Zhao
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Lin Zhu
- College of Chemistry, Beijing Normal University, Beijing 100875, PR China
| | - Hank F Kung
- Five Eleven Pharma Inc., Philadelphia, Pennsylvania 19104, United States
- Department of Radiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
36
|
Herrero Álvarez N, Bauer D, Hernández-Gil J, Lewis JS. Recent Advances in Radiometals for Combined Imaging and Therapy in Cancer. ChemMedChem 2021; 16:2909-2941. [PMID: 33792195 DOI: 10.1002/cmdc.202100135] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Indexed: 12/14/2022]
Abstract
Nuclear medicine is defined as the use of radionuclides for diagnostic and therapeutic applications. The imaging modalities positron emission tomography (PET) and single-photon emission computed tomography (SPECT) are based on γ-emissions of specific energies. The therapeutic technologies are based on β- -particle-, α-particle-, and Auger electron emitters. In oncology, PET and SPECT are used to detect cancer lesions, to determine dosimetry, and to monitor therapy effectiveness. In contrast, radiotherapy is designed to irreparably damage tumor cells in order to eradicate or control the disease's progression. Radiometals are being explored for the development of diagnostic and therapeutic radiopharmaceuticals. Strategies that combine both modalities (diagnostic and therapeutic), referred to as theranostics, are promising candidates for clinical applications. This review provides an overview of the basic concepts behind therapeutic and diagnostic radiopharmaceuticals and their significance in contemporary oncology. Select radiometals that significantly impact current and upcoming cancer treatment strategies are grouped as clinically suitable theranostics pairs. The most important physical and chemical properties are discussed. Standard production methods and current radionuclide availability are provided to indicate whether a cost-efficient use in a clinical routine is feasible. Recent preclinical and clinical developments and outline perspectives for the radiometals are highlighted in each section.
Collapse
Affiliation(s)
- Natalia Herrero Álvarez
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - David Bauer
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Javier Hernández-Gil
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.,Biomedical MRI/MoSAIC, Department of Imaging and Pathology, Katholieke Universiteit, Herestraat 49, 3000, Leuven, Belgium
| | - Jason S Lewis
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.,Department of Radiology, Weill Cornell Medical College, 1300 York Avenue, New York, NY, 10065, USA.,Department of Pharmacology, Weill-Cornell Medical College, New York, NY, 10065, USA
| |
Collapse
|
37
|
Iudicello A, Genovese F, Di Iorio V, Cicoria G, Boschi S. An HPLC and UHPLC-HRMS approach to study PSMA-11 instability in aqueous solution. EJNMMI Radiopharm Chem 2021; 6:14. [PMID: 33761040 PMCID: PMC7990985 DOI: 10.1186/s41181-021-00122-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 01/22/2021] [Indexed: 12/18/2022] Open
Abstract
Background The stability of precursors and reagents is of utmost importance for developing a robust radiolabelling method that provides high and constant radiochemical yield and radiochemical purity. While performing the QC of the [68Ga]Ga-PSMA-11 injectable solutions according to Ph. Eur. Monograph that has recently been published, a trend to the instability of the standard PSMA-11, the same used as a precursor for [68Ga]Ga-PSMA-11 radiosynthesis, has been observed. This instability led to the formation of a side product in a time-dependent manner. The formation of this compound, besides making the implementation of the Ph. Eur. analytical method more difficult, negatively influenced the radiochemical yield and the radiochemical purity by increasing gallium-68 in colloidal and ionic forms. Results The nature of the side product was investigated by adding chelators, such as EDTA, to PSMA-11 solutions and using the combination of UHPLC-HRMS. The results led to the definition of the side product structure, as natFe-PSMA-11, from the combination of the high-affinity chelator HBED-CC, present in the molecule of PSMA-11, and environmental Fe (III). Conclusions Strategies to reduce the risk of low radiolabeling yields and to increase the stability of the PSMA-11 in an aqueous solution were also discussed.
Collapse
Affiliation(s)
- Antonella Iudicello
- Pharmaceutical Department, Azienda USL of Modena, Largo del Pozzo, 71, 41121, Modena, Italy. .,Oncology and Hematology Department, Nuclear Medicine Unit, Azienda Ospedaliero-Universitaria of Modena, Largo del Pozzo, 71, 41121, Modena, Italy.
| | - Filippo Genovese
- Centro Interdipartimentale Grandi Strumenti, University of Modena and Reggio Emilia, Via Campi 213/A, 41125, Modena, Italy
| | - Valentina Di Iorio
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) - IRCCS IRST, Via Piero Maroncelli, 40, 47014, Meldola, FC, Italy
| | - Gianfranco Cicoria
- Medical Physics Department, Azienda Ospedaliero-Universitaria of Modena, Largo del Pozzo, 71, 41121, Modena, Italy
| | - Stefano Boschi
- Department for Life Quality Studies, University of Bologna, Corso D'Augusto, 237, Rimini, 47921, Italy
| |
Collapse
|
38
|
Urbanová K, Seifert D, Vinšová H, Vlk M, Lebeda O. Simple new method for labelling of PSMA-11 with 68Ga in NaHCO 3. Appl Radiat Isot 2021; 172:109692. [PMID: 33770721 DOI: 10.1016/j.apradiso.2021.109692] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 02/22/2021] [Accepted: 03/13/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Prostate specific membrane antigen (PSMA) is a type II membrane protein widely expressed on the surface of prostate cancer cells. One of its functions is to act as a receptor mediating the ligand internalization. This PSMA property is employed in the diagnostics and therapy of prostate cancer. Over the years, small molecules with high affinity for PSMA have been developed and labelled with positron emitters (e.g. 68Ga, 18F, 11C, 64Cu, or 86Y). One of these radiolabelled ligands, [68Ga] PSMA-11, is one of the most widespread tracers for PET imaging of the prostate cancer. Many techniques have been proposed and tested for the 68Ga labelling of PSMA-11. The aim of our work was to design a labelling method of PSMA-11 that minimizes number of the used chemicals and steps, providing quantitative labelling yield at laboratory temperature and may be easily automated. METHODOLOGY A68Ge/68Ga generator eluate in 0.1 M HCl was loaded on an activated Oasis MCX cartridge, and the cartridge was then thoroughly washed with water. The radionuclide 68Ga was eluted from the cartridge with 0.1 M NaHCO3 (pH = 8.5, n = 36) or with the same solution with pH adjusted to 7.2-9.0 (n = 38). Precursor PSMA-11 was mixed directly with the cartridge eluate of 68Ga in 0.1 M NaHCO3 of given pH. For the stability test, samples of 68GaPSMA-11 in 0.1 M NaHCO3 (pH 8.5) were mixed in ratio 1 : 1 with the following solutions: 0.1 M NaHCO3 (pH 8.5), human serum, PBS and 0.9% NaCl. In order to estimate an effect of the time elapsed between 68Ga elution from the cartridge in 0.1 M NaHCO3 (pH 8.5) and the labelling onset of PSMA-11, the latter was initiated 0, 5, 10 and 20 min post elution and radiochemical yield was monitored. All the PSMA-11 labelled samples were subjected to radiochemical purity test using HPLC. The whole process starting from generator elution up to HPLC analysis commencement took 10-15 min. RESULTS Recovery of 68Ga from cartridge Oasis MCX using 0.1 M NaHCO3 at pH 8.5 was 71.5 ± 1.4%. Thirty six PSMA-11 samples (10 μg in reaction mixture) were labelled at pH 8.5 with total average radiochemical yield of 98 ± 2%. Recovery of 68Ga from cartridge Oasis MCX using 0.1 M NaHCO3 at variable pH of 7.2-9.0 was 62.5 ± 1.8% showing certain decrease with decreasing pH. A total of 138 samples of PSMA-11 were labelled with 68 Ga at variable pH (7.2-9.0) and four different amounts of PSMA-11 (1, 2.5, 5 and 10 μg) resulting in the labelling yields of 54.0 ± 5.3%, 88.2 ± 3.2%, 99.4 ± 0.3% and 99.9 ± 0.1%, respectively. Irrespective of the pH, the radiolabelling yield was quantitative for the molar ratio PSMA-11: 68Ga > 5000 : 1 in the reaction mixture. Stability tests in 0.1 M NaHCO3 (pH 8.5), human serum, PBS and 0.9% NaCl revealed no observable release of 68Ga from the 68Ga-PSMA-11 complex within 3 h. Similarly, the delay between the 68Ga elution from the Oasis MCX cartridge in 0.1 M NaHCO3 (pH 8.5) and start of the labelling of PSMA-11 labelling has no effect on the radiochemical yield. CONCLUSION A new method of labelling PSMA-11 ligand with 68Ga in 0.1 M NaHCO3 using Oasis MCX cartridges was proposed, developed and tested. The results demonstrated that it is rapid, simple, reproducible and easy to automate.
Collapse
Affiliation(s)
- Kamila Urbanová
- Department of Radiopharmaceuticals, Nuclear Physic Institute, Czech Academy of Sciences, Řež, Czech Republic; Department of Nuclear Chemistry, Faculty of Nuclear Sciences and Physical Engineering, Czech Technical University, Prague, Czech Republic
| | - Daniel Seifert
- Department of Radiopharmaceuticals, Nuclear Physic Institute, Czech Academy of Sciences, Řež, Czech Republic
| | - Hana Vinšová
- Department of Radiopharmaceuticals, Nuclear Physic Institute, Czech Academy of Sciences, Řež, Czech Republic
| | - Martin Vlk
- Department of Nuclear Chemistry, Faculty of Nuclear Sciences and Physical Engineering, Czech Technical University, Prague, Czech Republic
| | - Ondřej Lebeda
- Department of Radiopharmaceuticals, Nuclear Physic Institute, Czech Academy of Sciences, Řež, Czech Republic.
| |
Collapse
|
39
|
Häseli S, Holy M, Joksch M, Bergner C, Wree A, Kurth J, Cankaya A, Piel M, Krause BJ, Sitte HH, Rösch F. 68 Ga-Labelled Tropane Analogues for the Visualization of the Dopaminergic System. ChemMedChem 2021; 16:804-808. [PMID: 33245194 PMCID: PMC7984292 DOI: 10.1002/cmdc.202000820] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Indexed: 02/04/2023]
Abstract
The development of radiometal-labelled pharmaceuticals for neuroimaging could offer great potential due to easier handling during labelling and availability through radionuclide generator systems. Nonetheless, to date, no such tracers are available for positron emission tomography, primarily owing to the challenge of crossing the blood-brain barrier (BBB) and loss of affinity through chelator attachment. We have prepared a variety of 68 Ga-labelled phenyltropanes showing that, through a simple hydrocarbon-linker, it is possible to introduce a chelator onto the lead structure while maintaining its high affinity for hDAT (human dopamine transporter) and simultaneously achieving adequate lipophilicity. One of the candidates, [68 Ga]Ga-HBED-hexadiyne-tropane, showed an IC50 value of 66 nM, together with a log D7.4 of 0.96. A μPET study in a hemi-parkinsonian rat model showed a fast wash-out of the tracer, and no specific uptake in the brain, thus implying an inability to penetrate the BBB.
Collapse
Affiliation(s)
- Sascha Häseli
- Institute of Nuclear ChemistryJohannes Gutenberg-University MainzFritz-Strassmann-Weg 255128MainzGermany
| | - Marion Holy
- Institute of Pharmacology (Center for Physiology and Pharmacology)Medical University of ViennaWähringer Straße 13a1090WienAustria
| | - Markus Joksch
- Department of Nuclear MedicineRostock University Medical CenterGertrudenplatz 118057RostockGermany
| | - Carina Bergner
- Department of Nuclear MedicineRostock University Medical CenterGertrudenplatz 118057RostockGermany
| | - Andreas Wree
- Institute of AnatomyRostock University Medical CenterGertrudenstraße 918057RostockGermany
| | - Jens Kurth
- Department of Nuclear MedicineRostock University Medical CenterGertrudenplatz 118057RostockGermany
| | - Aylin Cankaya
- Institute of Nuclear ChemistryJohannes Gutenberg-University MainzFritz-Strassmann-Weg 255128MainzGermany
| | - Markus Piel
- Institute of Nuclear ChemistryJohannes Gutenberg-University MainzFritz-Strassmann-Weg 255128MainzGermany
| | - Bernd J. Krause
- Department of Nuclear MedicineRostock University Medical CenterGertrudenplatz 118057RostockGermany
| | - Harald H. Sitte
- Institute of Pharmacology (Center for Physiology and Pharmacology)Medical University of ViennaWähringer Straße 13a1090WienAustria
| | - Frank Rösch
- Institute of Nuclear ChemistryJohannes Gutenberg-University MainzFritz-Strassmann-Weg 255128MainzGermany
| |
Collapse
|
40
|
|
41
|
Witney TH, Blower PJ. The chemical tool-kit for molecular imaging with radionuclides in the age of targeted and immune therapy. Cancer Imaging 2021; 21:18. [PMID: 33516256 PMCID: PMC7847158 DOI: 10.1186/s40644-021-00385-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/20/2021] [Indexed: 02/05/2023] Open
Abstract
Nuclear medicine has evolved over the last half-century from a functional imaging modality using a handful of radiopharmaceuticals, many of unknown structure and mechanism of action, into a modern speciality that can properly be described as molecular imaging, with a very large number of specific radioactive probes of known structure that image specific molecular processes. The advances of cancer treatment in recent decades towards targeted and immune therapies, combined with recognition of heterogeneity of cancer cell phenotype among patients, within patients and even within tumours, has created a growing need for personalised molecular imaging to support treatment decision. This article describes the evolution of the present vast range of radioactive probes – radiopharmaceuticals – leveraging a wide variety of chemical disciplines, over the last half century. These radiochemical innovations have been inspired by the need to support personalised medicine and also by the parallel development in development of new radionuclide imaging technologies – from gamma scintigraphy, through single photon emission tomography (SPECT), through the rise of clinical positron emission tomography (PET) and PET-CT, and perhaps in the future, by the advent of total body PET. Thus, in the interdisciplinary world of nuclear medicine and molecular imaging, as quickly as radiochemistry solutions are developed to meet new needs in cancer imaging, new challenges emerge as developments in one contributing technology drive innovations in the others.
Collapse
Affiliation(s)
- Timothy H Witney
- School of Biomedical Engineering and Imaging Sciences, King's College London, 4th Floor Lambeth Wing, St Thomas' Hospital, London, SE1 7EH, England
| | - Philip J Blower
- School of Biomedical Engineering and Imaging Sciences, King's College London, 4th Floor Lambeth Wing, St Thomas' Hospital, London, SE1 7EH, England.
| |
Collapse
|
42
|
Design and synthesis of a new conjugate of a tris(3-hydroxy-4-pyridinone) chelator (KC18) for potential use as gallium-68-labeled prostate-specific membrane antigen (PSMA) radiopharmaceutical. RESULTS IN CHEMISTRY 2021. [DOI: 10.1016/j.rechem.2021.100240] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
|
43
|
Bailey DL, Sabanathan D, Aslani A, Campbell DH, Walsh BJ, Lengkeek NA. RetroSPECT: Gallium-67 as a Long-Lived Imaging Agent for Theranostics. ASIA OCEANIA JOURNAL OF NUCLEAR MEDICINE & BIOLOGY 2021; 9:1-8. [PMID: 33392343 PMCID: PMC7701228 DOI: 10.22038/aojnmb.2020.51714.1355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/28/2020] [Accepted: 10/31/2020] [Indexed: 11/06/2022]
Abstract
A limitation to the wider introduction of personalised dosimetry in theranostics is the relative paucity of imaging radionuclides with suitable physical and chemical properties to be paired with a long-lived therapeutic partner. As most of the beta-emitting therapeutic radionuclides emit gamma radiation as well they could potentially be used as the imaging radionuclide as well as the therapeutic radionuclide. However, the downsides are that the beta radiation will deliver a significant radiation dose as part of the treatment planning procedure, and the gamma radiation branching ratio is often quite low. Gallium-67 has been in use in nuclear medicine for over 50 years. However, the tremendous interest in gallium imaging in theranostics in recent times has focused on the PET radionuclide gallium-68. In this article it is suggested that the longer-lived gallium-67, which has desirable characteristics for imaging with the gamma camera and a suitably long half-life to match biological timescales for drug uptake and turnover, has been overlooked, in particular, for treatment planning with radionuclide therapy. Gallium-67 could also allow non-PET facilities to participate in theranostic imaging prior to treatment or for monitoring response after therapy. Gallium-67 could play a niche role in the future development of personalised medicine with theranostics.
Collapse
Affiliation(s)
- Dale L. Bailey
- Department of Nuclear Medicine, Royal North Shore Hospital, Sydney, Australia
- Faculty of Medicine & Health, University of Sydney, Sydney, Australia
| | | | - Alireza Aslani
- Department of Nuclear Medicine, Royal North Shore Hospital, Sydney, Australia
- Faculty of Medicine & Health, University of Sydney, Sydney, Australia
| | | | | | - Nigel A. Lengkeek
- Biosciences, Australian Nuclear Science & Technology Organisation (ANSTO), Sydney, Australia
| |
Collapse
|
44
|
|
45
|
Affiliation(s)
- Yulong Wang
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, Nanjing, People’s Republic of China
| | - Cunzheng Zhang
- Jiangsu Key Laboratory for Food Quality and Safety-State Key Laboratory Cultivation Base of Ministry of Science and Technology, Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, Nanjing, People’s Republic of China
| | - Fengquan Liu
- Institute of Plant Protection, Jiangsu Academy of Agricultural Science, Nanjing, People’s Republic of China
| |
Collapse
|
46
|
Demine S, Schulte ML, Territo PR, Eizirik DL. Beta Cell Imaging-From Pre-Clinical Validation to First in Man Testing. Int J Mol Sci 2020; 21:E7274. [PMID: 33019671 PMCID: PMC7582644 DOI: 10.3390/ijms21197274] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/21/2020] [Accepted: 09/28/2020] [Indexed: 12/14/2022] Open
Abstract
There are presently no reliable ways to quantify human pancreatic beta cell mass (BCM) in vivo, which prevents an accurate understanding of the progressive beta cell loss in diabetes or following islet transplantation. Furthermore, the lack of beta cell imaging hampers the evaluation of the impact of new drugs aiming to prevent beta cell loss or to restore BCM in diabetes. We presently discuss the potential value of BCM determination as a cornerstone for individualized therapies in diabetes, describe the presently available probes for human BCM evaluation, and discuss our approach for the discovery of novel beta cell biomarkers, based on the determination of specific splice variants present in human beta cells. This has already led to the identification of DPP6 and FXYD2ga as two promising targets for human BCM imaging, and is followed by a discussion of potential safety issues, the role for radiochemistry in the improvement of BCM imaging, and concludes with an overview of the different steps from pre-clinical validation to a first-in-man trial for novel tracers.
Collapse
Affiliation(s)
- Stephane Demine
- Indiana Biosciences Research Institute, Indianapolis, IN 46202, USA;
| | - Michael L. Schulte
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (M.L.S.); (P.R.T.)
| | - Paul R. Territo
- Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (M.L.S.); (P.R.T.)
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Decio L. Eizirik
- Indiana Biosciences Research Institute, Indianapolis, IN 46202, USA;
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| |
Collapse
|
47
|
Design and Applications of Bifunctional Small Molecules in Biology. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2020; 1869:140534. [PMID: 32871274 DOI: 10.1016/j.bbapap.2020.140534] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 08/17/2020] [Accepted: 08/27/2020] [Indexed: 12/12/2022]
|
48
|
Radiosynthesis and preclinical evaluation of [ 68Ga]Ga-NOTA-folate for PET imaging of folate receptor β-positive macrophages. Sci Rep 2020; 10:13593. [PMID: 32788595 PMCID: PMC7423886 DOI: 10.1038/s41598-020-70394-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 07/27/2020] [Indexed: 11/09/2022] Open
Abstract
Folate receptor β (FR-β), a marker expressed on macrophages, is a promising target for imaging of inflammation. Here, we report the radiosynthesis and preclinical evaluation of [68Ga]Ga-NOTA-folate (68Ga-FOL). After determining the affinity of 68Ga-FOL using cells expressing FR-β, we studied atherosclerotic mice with 68Ga-FOL and 18F-FDG PET/CT. In addition, we studied tracer distribution and co-localization with macrophages in aorta cryosections using autoradiography, histology, and immunostaining. The specificity of 68Ga-FOL was assessed in a blocking study with folate glucosamine. As a final step, human radiation doses were extrapolated from rat PET data. We were able to produce 68Ga-FOL with high radiochemical purity and moderate molar activity. Cell binding studies revealed that 68Ga-FOL had 5.1 nM affinity for FR-β. Myocardial uptake of 68Ga-FOL was 20-fold lower than that of 18F-FDG. Autoradiography and immunohistochemistry of the aorta revealed that 68Ga-FOL radioactivity co-localized with Mac-3–positive macrophage-rich atherosclerotic plaques. The plaque-to-healthy vessel wall ratio of 68Ga-FOL was significantly higher than that of 18F-FDG. Blocking studies verified that 68Ga-FOL was specific for FR. Based on estimations from rat data, the human effective dose was 0.0105 mSv/MBq. Together, these findings show that 68Ga-FOL represents a promising new FR-β–targeted tracer for imaging macrophage-associated inflammation.
Collapse
|
49
|
Xia Y, Zeng C, Zhao Y, Zhang X, Li Z, Chen Y. Comparative evaluation of 68Ga-labelled TATEs: the impact of chelators on imaging. EJNMMI Res 2020; 10:36. [PMID: 32297029 PMCID: PMC7158967 DOI: 10.1186/s13550-020-00620-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 03/19/2020] [Indexed: 01/01/2023] Open
Abstract
Background 68Ga-labelled peptides targeting somatostatin receptor 2 (SSTR2) have demonstrated encouraging results in managing patients with neuroendocrine tumours (NETs). In addition to metal chelation, bifunctional chelators have also been found to impact imaging outcomes due to their differences in stability, charge, hydrophilicity, etc. In the present work, a comparative pharmacokinetic evaluation and imaging characteristics were performed between 68Ga-labelled somatostatin analogues (TATE) using NOTA (1,4,7-triazacyclononane-1,4,7-triacetic acid) and DOTA (1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid) as bifunctional chelating agents (BFCAs). Results Both 68Ga-NOTA-TATE and 68Ga-DOTA-TATE were obtained with high radiochemical purity. 68Ga-NOTA-TATE demonstrated higher in vitro stability (≥ 99%) than 68Ga-DOTA-TATE (≥ 95%) after 3 h of incubation. The water solubilities (partition coefficients, − 1.76 ± 0.06 vs. − 2.72 ± 0.16) and plasma protein binding rates (12.12% vs. 30.6%) were lower for 68Ga-NOTA-TATE than for 68Ga-DOTA-TATE. Differential pharmacokinetics and comparable tumour affinities (within 1 h) were observed in AR42J tumour-bearing mice. Healthy volunteer imaging studies showed comparable distribution patterns of these two imaging agents. However, the maximum standardized uptake values (SUVmax) of the two tracers varied in each organ. The two PET agents demonstrated almost identical SUVmax values in the kidneys. 68Ga-NOTA-TATE did have a lower SUVmax in most other organs compared with 68Ga-DOTA-TATE, including the liver (4.2 vs. 10.1), potentially due to the lower protein binding rate. Conclusion 68Ga-NOTA-TATE and 68Ga-DOTA-TATE demonstrated comparable tumour uptake in an AR42J mouse model. An initial clinical study revealed that 68Ga-NOTA-TATE may have reduced background uptake in the major organs such as the liver. Although the subject numbers were limited, further investigation of 68Ga-NOTA-TATE is warranted for detecting SSTR2-positive neuroendocrine tumours.
Collapse
Affiliation(s)
- Yuxiao Xia
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, No. 25, Taiping St, Luzhou, 646000, Sichuan, People's Republic of China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, No. 25, Taiping St, Luzhou, 646000, Sichuan, People's Republic of China
| | - Chengrun Zeng
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, No. 25, Taiping St, Luzhou, 646000, Sichuan, People's Republic of China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, No. 25, Taiping St, Luzhou, 646000, Sichuan, People's Republic of China
| | - Yanhong Zhao
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, No. 25, Taiping St, Luzhou, 646000, Sichuan, People's Republic of China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, No. 25, Taiping St, Luzhou, 646000, Sichuan, People's Republic of China
| | - Xinyi Zhang
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, No. 25, Taiping St, Luzhou, 646000, Sichuan, People's Republic of China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, No. 25, Taiping St, Luzhou, 646000, Sichuan, People's Republic of China
| | - Zibo Li
- Biomedical Research Imaging Center, Department of Radiology, and UNC Lineberger Comprehensive Cancer Center, University of North Carolina-Chapel Hill, Chapel Hill, NC, 27514, USA.
| | - Yue Chen
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, No. 25, Taiping St, Luzhou, 646000, Sichuan, People's Republic of China. .,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, No. 25, Taiping St, Luzhou, 646000, Sichuan, People's Republic of China.
| |
Collapse
|
50
|
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
|