1
|
Marešová A, Jurášek M, Drašar PB, Dolenský B, Prokudina EA, Shalgunov V, Herth MM, Cumming P, Popkov A. A facile synthesis of precursor for the σ-1 receptor PET radioligand [ 18 F]FTC-146 and its radiofluorination. J Labelled Comp Radiopharm 2024; 67:59-66. [PMID: 38171540 DOI: 10.1002/jlcr.4081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 11/10/2023] [Accepted: 12/07/2023] [Indexed: 01/05/2024]
Abstract
The σ-1 receptor is a non-opioid transmembrane protein involved in various human pathologies including neurodegenerative diseases, inflammation, and cancer. The previously published ligand [18 F]FTC-146 is among the most promising tools for σ-1 molecular imaging by positron emission tomography (PET), with a potential for application in clinical diagnostics and research. However, the published six- or four-step synthesis of the tosyl ester precursor for its radiosynthesis is complicated and time-consuming. Herein, we present a simple one-step precursor synthesis followed by a one-step fluorine-18 labeling procedure that streamlines the preparation of [18 F]FTC-146. Instead of a tosyl-based precursor, we developed a one-step synthesis of the precursor analog AM-16 containing a chloride leaving group for the SN 2 reaction with 18 F-fluoride. 18 F-fluorination of AM-16 led to a moderate decay-corrected radiochemical yield (RCY = 7.5%) with molar activity (Am ) of 45.9 GBq/μmol. Further optimization of this procedure should enable routine radiopharmaceutical production of this promising PET tracer.
Collapse
Affiliation(s)
- Anna Marešová
- Department of Chemistry of Natural Compounds, University of Chemistry and Technology Prague, Praha 6, Czech Republic
| | - Michal Jurášek
- Department of Chemistry of Natural Compounds, University of Chemistry and Technology Prague, Praha 6, Czech Republic
| | - Pavel B Drašar
- Department of Chemistry of Natural Compounds, University of Chemistry and Technology Prague, Praha 6, Czech Republic
| | - Bohumil Dolenský
- Department of Analytical Chemistry, University of Chemistry and Technology Prague, Praha 6, Czech Republic
| | - Elena A Prokudina
- Department of Chemistry of Natural Compounds, University of Chemistry and Technology Prague, Praha 6, Czech Republic
| | - Vladimir Shalgunov
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
- PET and Cyclotron Unit, Copenhagen University Hospital, Copenhagen, Denmark
| | - Matthias M Herth
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Paul Cumming
- Department of Nuclear Medicine, University Hospital Bern, Bern, Switzerland
- School of Psychology and Counselling, Queensland University of Technology, Kevin Grove, Queensland, Australia
| | - Alexander Popkov
- Institute of Organic Chemistry, Johannes Kepler University, Linz, Austria
- Samo Biomedical Centre, Pardubice, Czech Republic
| |
Collapse
|
2
|
Bolcaen J, Nair S, Driver CHS, Boshomane TMG, Ebenhan T, Vandevoorde C. Novel Receptor Tyrosine Kinase Pathway Inhibitors for Targeted Radionuclide Therapy of Glioblastoma. Pharmaceuticals (Basel) 2021; 14:626. [PMID: 34209513 PMCID: PMC8308832 DOI: 10.3390/ph14070626] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma (GB) remains the most fatal brain tumor characterized by a high infiltration rate and treatment resistance. Overexpression and/or mutation of receptor tyrosine kinases is common in GB, which subsequently leads to the activation of many downstream pathways that have a critical impact on tumor progression and therapy resistance. Therefore, receptor tyrosine kinase inhibitors (RTKIs) have been investigated to improve the dismal prognosis of GB in an effort to evolve into a personalized targeted therapy strategy with a better treatment outcome. Numerous RTKIs have been approved in the clinic and several radiopharmaceuticals are part of (pre)clinical trials as a non-invasive method to identify patients who could benefit from RTKI. The latter opens up the scope for theranostic applications. In this review, the present status of RTKIs for the treatment, nuclear imaging and targeted radionuclide therapy of GB is presented. The focus will be on seven tyrosine kinase receptors, based on their central role in GB: EGFR, VEGFR, MET, PDGFR, FGFR, Eph receptor and IGF1R. Finally, by way of analyzing structural and physiological characteristics of the TKIs with promising clinical trial results, four small molecule RTKIs were selected based on their potential to become new therapeutic GB radiopharmaceuticals.
Collapse
Affiliation(s)
- Julie Bolcaen
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town 7131, South Africa;
| | - Shankari Nair
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town 7131, South Africa;
| | - Cathryn H. S. Driver
- Radiochemistry, South African Nuclear Energy Corporation, Pelindaba, Brits 0240, South Africa;
- Pre-Clinical Imaging Facility, Nuclear Medicine Research Infrastructure, Pelindaba, Brits 0242, South Africa;
| | - Tebatso M. G. Boshomane
- Department of Nuclear Medicine, University of Pretoria Steve Biko Academic Hospital, Pretoria 0001, South Africa;
| | - Thomas Ebenhan
- Pre-Clinical Imaging Facility, Nuclear Medicine Research Infrastructure, Pelindaba, Brits 0242, South Africa;
- Department of Nuclear Medicine, University of Pretoria Steve Biko Academic Hospital, Pretoria 0001, South Africa;
- Preclinical Drug Development Platform, Department of Science and Technology, North West University, Potchefstroom 2520, South Africa
| | - Charlot Vandevoorde
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town 7131, South Africa;
| |
Collapse
|
3
|
Neuber C, Tröster A, Löser R, Belter B, Schwalbe H, Pietzsch J. The Pyrazolo[3,4- d]pyrimidine-Based Kinase Inhibitor NVP-BHG712: Effects of Regioisomers on Tumor Growth, Perfusion, and Hypoxia in EphB4-Positive A375 Melanoma Xenografts. Molecules 2020; 25:molecules25215115. [PMID: 33153234 PMCID: PMC7662635 DOI: 10.3390/molecules25215115] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/29/2020] [Accepted: 11/01/2020] [Indexed: 12/02/2022] Open
Abstract
In a previous study, EphB4 was demonstrated to be a positive regulator of A375-melanoma growth but a negative regulator of tumor vascularization and perfusion. To distinguish between EphB4 forward and ephrinB2 reverse signaling, we used the commercially available EphB4 kinase inhibitor NVP-BHG712 (NVP), which was later identified as its regioisomer NVPiso. Since there have been reported significant differences between the inhibition profiles of NVP and NVPiso, we compared the influence of NVP and NVPiso on tumor characteristics under the same experimental conditions. Despite the different inhibitory profiles of NVP and NVPiso, the comparative study conducted here showed the same EphB4-induced effects in vivo as in the previous investigation. This confirmed the conclusion that EphB4-ephrinB2 reverse signaling is responsible for increased tumor growth as well as decreased tumor vascularization and perfusion. These results are further substantiated by microarrays showing differences between mock-transfected and EphB4-transfected (A375-EphB4) cells with respect to at least 9 angiogenesis-related proteins. Decreased expression of vascular endothelial growth factor (VEGF), angiotensin 1 (Ang-1), and protein kinase B (Akt/PKB), together with the increased expression of tissue inhibitor of metalloproteinase-1 (TIMP-1) and transforming growth factor beta-2 (TGF-β2), is consistent with the impaired vascularization of A375-EphB4 xenografts. Functional overexpression of EphB4 in A375-EphB4 cells was confirmed by activation of a variety of signaling pathways, including the Janus kinase/signal transducers and activators of transcription (JAK/STAT), rat sarcoma virus/rapidly accelerated fibrosarcoma/mitogen activated protein kinase kinase (Ras/Raf/MEK), and nuclear factor kappa-B (NFkB) pathways.
Collapse
Affiliation(s)
- Christin Neuber
- Institute of Radiopharmaceutical Cancer Research, Department Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany; (C.N.); (R.L.); (B.B.)
| | - Alix Tröster
- Centre for Biomolecular Magnetic Resonance (BMRZ), Institute of Organic Chemistry and Chemical Biology, Johann Wolfgang Goethe-University Frankfurt a. M., Max-von-Laue-Strasse 7, 60438 Frankfurt, Germany; (A.T.); (H.S.)
| | - Reik Löser
- Institute of Radiopharmaceutical Cancer Research, Department Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany; (C.N.); (R.L.); (B.B.)
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, 01069 Dresden, Germany
| | - Birgit Belter
- Institute of Radiopharmaceutical Cancer Research, Department Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany; (C.N.); (R.L.); (B.B.)
| | - Harald Schwalbe
- Centre for Biomolecular Magnetic Resonance (BMRZ), Institute of Organic Chemistry and Chemical Biology, Johann Wolfgang Goethe-University Frankfurt a. M., Max-von-Laue-Strasse 7, 60438 Frankfurt, Germany; (A.T.); (H.S.)
| | - Jens Pietzsch
- Institute of Radiopharmaceutical Cancer Research, Department Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstrasse 400, 01328 Dresden, Germany; (C.N.); (R.L.); (B.B.)
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, 01069 Dresden, Germany
- Correspondence: ; Tel.: +49-351-260-2622
| |
Collapse
|
4
|
Pretze M, Neuber C, Kinski E, Belter B, Köckerling M, Caflisch A, Steinbach J, Pietzsch J, Mamat C. Synthesis, radiolabelling and initial biological characterisation of 18F-labelled xanthine derivatives for PET imaging of Eph receptors. Org Biomol Chem 2020; 18:3104-3116. [PMID: 32253415 DOI: 10.1039/d0ob00391c] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Eph receptor tyrosine kinases, particularly EphA2 and EphB4, represent promising candidates for molecular imaging due to their essential role in cancer progression and therapy resistance. Xanthine derivatives were identified to be potent Eph receptor inhibitors with IC50 values in the low nanomolar range (1-40 nm). These compounds occupy the hydrophobic pocket of the ATP-binding site in the kinase domain. Based on lead compound 1, we designed two fluorine-18-labelled receptor tyrosine kinase inhibitors ([18F]2/3) as potential tracers for positron emission tomography (PET). Docking into the ATP-binding site allowed us to find the best position for radiolabelling. The replacement of the methyl group at the uracil residue ([18F]3) rather than the methyl group of the phenoxy moiety ([18F]2) by a fluoropropyl group was predicted to preserve the affinity of the lead compound 1. Herein, we point out a synthesis route to [18F]2 and [18F]3 and the respective tosylate precursors as well as a labelling procedure to insert fluorine-18. After radiolabelling, both radiotracers were obtained in approximately 5% radiochemical yield with high radiochemical purity (>98%) and a molar activity of >10 GBq μmol-1. In line with the docking studies, first cell experiments revealed specific, time-dependent binding and uptake of [18F]3 to EphA2 and EphB4-overexpressing A375 human melanoma cells, whereas [18F]2 did not accumulate at these cells. Since both tracers [18F]3 and [18F]2 are stable in rat blood, the novel radiotracers might be suitable for in vivo molecular imaging of Eph receptors with PET.
Collapse
Affiliation(s)
- Marc Pretze
- Helmholtz-Zentrum Dresden-Rossendorf, Institut für Radiopharmazeutische Krebsforschung, Bautzner Landstraße 400, D-01328 Dresden, Germany. and Technische Universität Dresden, Fakultät Chemie und Lebensmittelchemie, D-01062 Dresden, Germany
| | - Christin Neuber
- Helmholtz-Zentrum Dresden-Rossendorf, Institut für Radiopharmazeutische Krebsforschung, Bautzner Landstraße 400, D-01328 Dresden, Germany.
| | - Elisa Kinski
- Helmholtz-Zentrum Dresden-Rossendorf, Institut für Radiopharmazeutische Krebsforschung, Bautzner Landstraße 400, D-01328 Dresden, Germany.
| | - Birgit Belter
- Helmholtz-Zentrum Dresden-Rossendorf, Institut für Radiopharmazeutische Krebsforschung, Bautzner Landstraße 400, D-01328 Dresden, Germany.
| | - Martin Köckerling
- Universität Rostock, Institut für Chemie - Anorganische Festkörperchemie, Albert-Einstein-Straße 3a, D-18059 Rostock, Germany
| | - Amedeo Caflisch
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Jörg Steinbach
- Helmholtz-Zentrum Dresden-Rossendorf, Institut für Radiopharmazeutische Krebsforschung, Bautzner Landstraße 400, D-01328 Dresden, Germany. and Technische Universität Dresden, Fakultät Chemie und Lebensmittelchemie, D-01062 Dresden, Germany
| | - Jens Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf, Institut für Radiopharmazeutische Krebsforschung, Bautzner Landstraße 400, D-01328 Dresden, Germany. and Technische Universität Dresden, Fakultät Chemie und Lebensmittelchemie, D-01062 Dresden, Germany
| | - Constantin Mamat
- Helmholtz-Zentrum Dresden-Rossendorf, Institut für Radiopharmazeutische Krebsforschung, Bautzner Landstraße 400, D-01328 Dresden, Germany. and Technische Universität Dresden, Fakultät Chemie und Lebensmittelchemie, D-01062 Dresden, Germany
| |
Collapse
|
5
|
Neuber C, Belter B, Mamat C, Pietzsch J. Radiopharmacologist's and Radiochemist's View on Targeting the Eph/Ephrin Receptor Tyrosine Kinase System. ACS OMEGA 2020; 5:16318-16331. [PMID: 32685795 PMCID: PMC7364440 DOI: 10.1021/acsomega.0c01058] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 05/25/2020] [Indexed: 05/06/2023]
Abstract
In the past decade, there have been extensive efforts to open up the Eph/ephrin subfamily of the receptor tyrosine kinase family for diagnostic and therapeutic applications. Besides classical pharmaceutical developments, which focus either on drugs targeting the extracellular ligand binding domains or on the intracellular tyrosine kinase domains of these receptors, there also have been first radiopharmaceutical approaches. Here the focus is on the development of specific and selective probes for molecular imaging, particularly by means of positron emission tomography, and the functional characterization of the Eph/ephrin subfamily in certain target tissues. The aim of this mini-review is to summarize the different approaches toward Eph-targeting radiotracers by using antibodies, peptides, and small molecules and to discuss their radiopharmacological characterization. With regard to the small molecules, further considerations will focus on the design and synthesis of nonradioactive reference compounds and precursors as well as on radiolabeling strategies.
Collapse
Affiliation(s)
- Christin Neuber
- Helmholtz-Zentrum
Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer
Research, Department of Radiopharmaceutical
and Chemical Biology, 01328 Dresden, Germany
| | - Birgit Belter
- Helmholtz-Zentrum
Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer
Research, Department of Radiopharmaceutical
and Chemical Biology, 01328 Dresden, Germany
| | - Constantin Mamat
- Helmholtz-Zentrum
Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer
Research, Department of Radionuclide Theragnostics, 01328 Dresden, Germany
- Technische
Universität Dresden, School of Science,
Faculty of Chemistry and Food Chemistry, 01062 Dresden, Germany
| | - Jens Pietzsch
- Helmholtz-Zentrum
Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer
Research, Department of Radiopharmaceutical
and Chemical Biology, 01328 Dresden, Germany
- Technische
Universität Dresden, School of Science,
Faculty of Chemistry and Food Chemistry, 01062 Dresden, Germany
- E-mail:
| |
Collapse
|
6
|
Löser R, Bader M, Kuchar M, Wodtke R, Lenk J, Wodtke J, Kuhne K, Bergmann R, Haase-Kohn C, Urbanová M, Steinbach J, Pietzsch J. Synthesis, 18F-labelling and radiopharmacological characterisation of the C-terminal 30mer of Clostridium perfringens enterotoxin as a potential claudin-targeting peptide. Amino Acids 2018; 51:219-244. [PMID: 30264172 DOI: 10.1007/s00726-018-2657-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 09/17/2018] [Indexed: 12/26/2022]
Abstract
The cell surface receptor claudin-4 (Cld-4) is upregulated in various tumours and represents an important emerging target for both diagnosis and treatment of solid tumours of epithelial origin. The C-terminal fragment of the Clostridium perfringens enterotoxin cCPE290-319 appears as a suitable ligand for targeting Cld-4. The synthesis of this 30mer peptide was attempted via several approaches, which has revealed sequential SPPS using three pseudoproline dipeptide building blocks to be the most efficient one. Labelling with fluorine-18 was achieved on solid phase using N-succinimidyl 4-[18F]fluorobenzoate ([18F]SFB) and 4-[18F]fluorobenzoyl chloride as 18F-acylating agents, which was the most advantageous when [18F]SFB was reacted with the resin-bound 30mer containing an N-terminal 6-aminohexanoic spacer. Binding to Cld-4 was demonstrated via surface plasmon resonance using a protein construct containing both extracellular loops of Cld-4. In addition, cell binding experiments were performed for 18F-labelled cCPE290-319 with the Cld-4 expressing tumour cell lines HT-29 and A431 that were complemented by fluorescence microscopy studies using the corresponding fluorescein isothiocyanate-conjugated peptide. The 30mer peptide proved to be sufficiently stable in blood plasma. Studying the in vivo behaviour of 18F-labelled cCPE290-319 in healthy mice and rats by dynamic PET imaging and radiometabolite analyses has revealed that the peptide is subject to substantial liver uptake and rapid metabolic degradation in vivo, which limits its suitability as imaging probe for tumour-associated Cld-4.
Collapse
Affiliation(s)
- Reik Löser
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany.
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Mommsenstraße 4, 01062, Dresden, Germany.
| | - Miriam Bader
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Mommsenstraße 4, 01062, Dresden, Germany
| | - Manuela Kuchar
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Mommsenstraße 4, 01062, Dresden, Germany
| | - Robert Wodtke
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Mommsenstraße 4, 01062, Dresden, Germany
| | - Jens Lenk
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Mommsenstraße 4, 01062, Dresden, Germany
| | - Johanna Wodtke
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Konstantin Kuhne
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Mommsenstraße 4, 01062, Dresden, Germany
| | - Ralf Bergmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Cathleen Haase-Kohn
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Marie Urbanová
- Department of Physics and Measurements, University of Chemistry and Technology, 166 28, Prague, Czech Republic
| | - Jörg Steinbach
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Mommsenstraße 4, 01062, Dresden, Germany
| | - Jens Pietzsch
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden Rossendorf, Bautzner Landstraße 400, 01328, Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, Mommsenstraße 4, 01062, Dresden, Germany
| |
Collapse
|
7
|
Kuchar M, Neuber C, Belter B, Bergmann R, Lenk J, Wodtke R, Kniess T, Steinbach J, Pietzsch J, Löser R. Evaluation of Fluorine-18-Labeled α1(I)-N-Telopeptide Analogs as Substrate-Based Radiotracers for PET Imaging of Melanoma-Associated Lysyl Oxidase. Front Chem 2018; 6:121. [PMID: 29755969 PMCID: PMC5932954 DOI: 10.3389/fchem.2018.00121] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 03/30/2018] [Indexed: 12/28/2022] Open
Abstract
Accumulating evidence suggests an unequivocal role of lysyl oxidases as key players of tumor progression and metastasis, which renders this enzyme family highly attractive for targeted non-invasive functional imaging of tumors. Considering their function in matrix remodeling, malignant melanoma appears as particularly interesting neoplasia in this respect. For the development of radiotracers that enable PET imaging of the melanoma-associated lysyl oxidase activity, substrates derived from the type I collagen α1 N-telopeptide were labeled with fluorine-18 using N-succinimidyl 4-[18F]fluorobenzoate ([18F]SFB) as prosthetic reagent. With regards to potential crosslinking to tumor-associated collagen in vivo, their interaction with triple-helical type I collagen was studied by SPR. A mouse model of human melanoma was established on the basis of the A375 cell line, for which the expression of the oncologically relevant lysyl oxidase isoforms LOX and LOXL2 was demonstrated in Western blot and immunohistochemical experiments. The radiopharmacological profiles of the peptidic radiotracers were evaluated in normal rats and A375 melanoma-bearing mice by ex vivo metabolite analysis, whole-body biodistribution studies and dynamic PET imaging. Out of three 18F-labeled telopeptide analogs, the one with the most favorable substrate properties has shown favorable tumor uptake and tumor-to-muscle ratio. Lysyl oxidase-mediated tumor uptake was proven by pharmacological inhibition using β-aminopropionitrile and by employing negative-control analogs of impeded or abolished targeting capability. The latter were obtained by substituting the lysine residue by ornithine and norleucine, respectively. Comparing the tumor uptake of the lysine-containing peptide with that of the non-functional analogs indicate the feasibility of lysyl oxidase imaging in melanoma using substrate-based radiotracers.
Collapse
Affiliation(s)
- Manuela Kuchar
- Helmholtz-Zentrum Dresden Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Faculty of Chemistry and Food Chemistry, School of Science, Technische Unversität Dresden, Dresden, Germany
| | - Christin Neuber
- Helmholtz-Zentrum Dresden Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Birgit Belter
- Helmholtz-Zentrum Dresden Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Ralf Bergmann
- Helmholtz-Zentrum Dresden Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Jens Lenk
- Helmholtz-Zentrum Dresden Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Faculty of Chemistry and Food Chemistry, School of Science, Technische Unversität Dresden, Dresden, Germany
| | - Robert Wodtke
- Helmholtz-Zentrum Dresden Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Faculty of Chemistry and Food Chemistry, School of Science, Technische Unversität Dresden, Dresden, Germany
| | - Torsten Kniess
- Helmholtz-Zentrum Dresden Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany
| | - Jörg Steinbach
- Helmholtz-Zentrum Dresden Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Faculty of Chemistry and Food Chemistry, School of Science, Technische Unversität Dresden, Dresden, Germany
| | - Jens Pietzsch
- Helmholtz-Zentrum Dresden Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Faculty of Chemistry and Food Chemistry, School of Science, Technische Unversität Dresden, Dresden, Germany
| | - Reik Löser
- Helmholtz-Zentrum Dresden Rossendorf, Institute of Radiopharmaceutical Cancer Research, Dresden, Germany.,Faculty of Chemistry and Food Chemistry, School of Science, Technische Unversität Dresden, Dresden, Germany
| |
Collapse
|
8
|
Kranz M, Bergmann R, Kniess T, Belter B, Neuber C, Cai Z, Deng G, Fischer S, Zhou J, Huang Y, Brust P, Deuther-Conrad W, Pietzsch J. Bridging from Brain to Tumor Imaging: (S)-(-)- and (R)-(+)-[ 18F]Fluspidine for Investigation of Sigma-1 Receptors in Tumor-Bearing Mice. Molecules 2018; 23:E702. [PMID: 29558382 PMCID: PMC6017399 DOI: 10.3390/molecules23030702] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 03/12/2018] [Accepted: 03/18/2018] [Indexed: 12/12/2022] Open
Abstract
Sigma-1 receptors (Sig1R) are highly expressed in various human cancer cells and hence imaging of this target with positron emission tomography (PET) can contribute to a better understanding of tumor pathophysiology and support the development of antineoplastic drugs. Two Sig1R-specific radiolabeled enantiomers (S)-(-)- and (R)-(+)-[18F]fluspidine were investigated in several tumor cell lines including melanoma, squamous cell/epidermoid carcinoma, prostate carcinoma, and glioblastoma. Dynamic PET scans were performed in mice to investigate the suitability of both radiotracers for tumor imaging. The Sig1R expression in the respective tumors was confirmed by Western blot. Rather low radiotracer uptake was found in heterotopically (subcutaneously) implanted tumors. Therefore, a brain tumor model (U87-MG) with orthotopic implantation was chosen to investigate the suitability of the two Sig1R radiotracers for brain tumor imaging. High tumor uptake as well as a favorable tumor-to-background ratio was found. These results suggest that Sig1R PET imaging of brain tumors with [18F]fluspidine could be possible. Further studies with this tumor model will be performed to confirm specific binding and the integrity of the blood-brain barrier (BBB).
Collapse
Affiliation(s)
- Mathias Kranz
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 04318 Leipzig, Germany.
- Department of Diagnostic Radiology, PET Center, Yale University School of Medicine, New Haven, CT 06519, USA.
| | - Ralf Bergmann
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany.
| | - Torsten Kniess
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany.
| | - Birgit Belter
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany.
| | - Christin Neuber
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany.
| | - Zhengxin Cai
- Department of Diagnostic Radiology, PET Center, Yale University School of Medicine, New Haven, CT 06519, USA.
| | - Gang Deng
- Department of Neurosurgery and Biomedical Engineering, Yale University School of Medicine, New Haven, CT 06519, USA.
| | - Steffen Fischer
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 04318 Leipzig, Germany.
| | - Jiangbing Zhou
- Department of Neurosurgery and Biomedical Engineering, Yale University School of Medicine, New Haven, CT 06519, USA.
| | - Yiyun Huang
- Department of Diagnostic Radiology, PET Center, Yale University School of Medicine, New Haven, CT 06519, USA.
| | - Peter Brust
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 04318 Leipzig, Germany.
| | - Winnie Deuther-Conrad
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 04318 Leipzig, Germany.
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany.
- Technische Universität Dresden, School of Science, Faculty of Chemistry and Food Chemistry, 01062 Dresden, Germany.
| |
Collapse
|
9
|
Neuber C, Belter B, Meister S, Hofheinz F, Bergmann R, Pietzsch HJ, Pietzsch J. Overexpression of Receptor Tyrosine Kinase EphB4 Triggers Tumor Growth and Hypoxia in A375 Melanoma Xenografts: Insights from Multitracer Small Animal Imaging Experiments. Molecules 2018; 23:E444. [PMID: 29462967 PMCID: PMC6017846 DOI: 10.3390/molecules23020444] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 02/13/2018] [Accepted: 02/16/2018] [Indexed: 12/15/2022] Open
Abstract
Experimental evidence has associated receptor tyrosine kinase EphB4 with tumor angiogenesis also in malignant melanoma. Considering the limited in vivo data available, we have conducted a systematic multitracer and multimodal imaging investigation in EphB4-overexpressing and mock-transfected A375 melanoma xenografts. Tumor growth, perfusion, and hypoxia were investigated by positron emission tomography. Vascularization was investigated by fluorescence imaging in vivo and ex vivo. The approach was completed by magnetic resonance imaging, radioluminography ex vivo, and immunohistochemical staining for blood and lymph vessel markers. Results revealed EphB4 to be a positive regulator of A375 melanoma growth, but a negative regulator of tumor vascularization. Resulting in increased hypoxia, this physiological characteristic is considered as highly unfavorable for melanoma prognosis and therapy outcome. Lymphangiogenesis, by contrast, was not influenced by EphB4 overexpression. In order to distinguish between EphB4 forward and EphrinB2, the natural EphB4 ligand, reverse signaling a specific EphB4 kinase inhibitor was applied. Blocking experiments show EphrinB2 reverse signaling rather than EphB4 forward signaling to be responsible for the observed effects. In conclusion, functional expression of EphB4 is considered a promising differentiating characteristic, preferentially determined by non-invasive in vivo imaging, which may improve personalized theranostics of malignant melanoma.
Collapse
Affiliation(s)
- Christin Neuber
- Department Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstrasse 400, 01314 Dresden, Germany.
| | - Birgit Belter
- Department Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstrasse 400, 01314 Dresden, Germany.
| | - Sebastian Meister
- Department Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstrasse 400, 01314 Dresden, Germany.
| | - Frank Hofheinz
- Department Positron Emission Tomography, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstrasse 400, 01314 Dresden, Germany.
| | - Ralf Bergmann
- Department Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstrasse 400, 01314 Dresden, Germany.
| | - Hans-Jürgen Pietzsch
- Department Radionuclide Theragnostics, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstrasse 400, 01314 Dresden, Germany.
| | - Jens Pietzsch
- Department Radiopharmaceutical and Chemical Biology, Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstrasse 400, 01314 Dresden, Germany.
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, 01062 Dresden, Germany.
| |
Collapse
|
10
|
Chen Y, Zhang H, Zhang Y. Targeting receptor tyrosine kinase EphB4 in cancer therapy. Semin Cancer Biol 2017; 56:37-46. [PMID: 28993206 DOI: 10.1016/j.semcancer.2017.10.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 09/13/2017] [Accepted: 10/04/2017] [Indexed: 12/12/2022]
Abstract
Eph receptors and their Eph receptor-interacting (ephrin) ligands together form an important cell communication system with diverse roles. Experimental evidence demonstrated Eph receptor bidirectional signaling with both tumor-suppressing and tumor-promoting activities in cancer cells. The tyrosine kinase EphB4, a member of the Eph receptor family, has been associated with tumor angiogenesis, growth and metastasis, thus making it a valuable and attractive target for drug design for therapeutic applications. In the past decade, many studies have focused on elucidating the structure and function of EphB4 in complex with its ligand ephrinB2 for their role in carcinogenesis. Meanwhile, an array of compounds targeting EphB4 have been studied and several selective inhibitors have been tested in clinical studies. This review discusses the structure and function of the EphB4 receptor, analyzes its potential as a target for anticancer therapy, and summarizes the information about inhibitors of EphB4 kinase activity. Conclusively, EphB4 is a challenging but promising therapeutic target in cancer.
Collapse
Affiliation(s)
- Yinnan Chen
- School of Molecular Sciences, Arizona State University, Tempe, AZ 85287, USA
| | - Hongmei Zhang
- Department of Endocrinology, Xinhua Hospital affiliated to Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai 200092, PR China.
| | - Yanmin Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, No. 76, Yanta West Street, #54, Xi'an, Shaanxi Province 710061, PR China.
| |
Collapse
|
11
|
Wiemer J, Steinbach J, Pietzsch J, Mamat C. Preparation of a novel radiotracer targeting the EphB4 receptor via radiofluorination using spiro azetidinium salts as precursor. J Labelled Comp Radiopharm 2017; 60:489-498. [DOI: 10.1002/jlcr.3526] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 05/10/2017] [Accepted: 05/29/2017] [Indexed: 12/26/2022]
Affiliation(s)
- Jens Wiemer
- Institut für Radiopharmazeutische Krebsforschung; Helmholtz-Zentrum Dresden-Rossendorf; Dresden Germany
| | - Jörg Steinbach
- Institut für Radiopharmazeutische Krebsforschung; Helmholtz-Zentrum Dresden-Rossendorf; Dresden Germany
- Fachbereich Chemie und Lebensmittelchemie; Technische Universität Dresden; Dresden Germany
| | - Jens Pietzsch
- Institut für Radiopharmazeutische Krebsforschung; Helmholtz-Zentrum Dresden-Rossendorf; Dresden Germany
- Fachbereich Chemie und Lebensmittelchemie; Technische Universität Dresden; Dresden Germany
| | - Constantin Mamat
- Institut für Radiopharmazeutische Krebsforschung; Helmholtz-Zentrum Dresden-Rossendorf; Dresden Germany
- Fachbereich Chemie und Lebensmittelchemie; Technische Universität Dresden; Dresden Germany
| |
Collapse
|
12
|
Mamat C, Pretze M, Gott M, Köckerling M. Synthesis, dynamic NMR characterization and XRD studies of novel N, N'-substituted piperazines for bioorthogonal labeling. Beilstein J Org Chem 2016; 12:2478-2489. [PMID: 28144316 PMCID: PMC5238536 DOI: 10.3762/bjoc.12.242] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Accepted: 10/31/2016] [Indexed: 12/12/2022] Open
Abstract
Novel, functionalized piperazine derivatives were successfully synthesized and fully characterized by 1H/13C/19F NMR, MS, elemental analysis and lipophilicity. All piperazine compounds occur as conformers resulting from the partial amide double bond. Furthermore, a second conformational shape was observed for all nitro derivatives due to the limited change of the piperazine chair conformation. Therefore, two coalescence points were determined and their resulting activation energy barriers were calculated using 1H NMR. To support this result, single crystals of 1-(4-nitrobenzoyl)piperazine (3a, monoclinic, space group C2/c, a = 24.587(2), b = 7.0726(6), c = 14.171(1) Å, β = 119.257(8)°, V = 2149.9(4) Å3, Z = 4, Dobs = 1.454 g/cm3) and the alkyne derivative 4-(but-3-yn-1-yl)-1-(4-fluorobenzoyl)piperazine (4b, monoclinic, space group P21/n, a = 10.5982(2), b = 8.4705(1), c = 14.8929(3) Å, β = 97.430(1)°, V = 1325.74(4) Å3, Z = 4, Dobs = 1.304 g/cm3) were obtained from a saturated ethyl acetate solution. The rotational conformation of these compounds was also verified by XRD. As proof of concept for future labeling purposes, both nitropiperazines were reacted with [18F]F–. To test the applicability of these compounds as possible 18F-building blocks, two biomolecules were modified and chosen for conjugation either using the Huisgen-click reaction or the traceless Staudinger ligation.
Collapse
Affiliation(s)
- Constantin Mamat
- Helmholtz-Zentrum Dresden-Rossendorf, Institut für Radiopharmazeutische Krebsforschung, Bautzner Landstraße 400, D-01328 Dresden, Germany; Technische Universität Dresden, Fachrichtung Chemie und Lebensmittelchemie, D-01062 Dresden, Germany
| | - Marc Pretze
- Medizinische Fakultät Mannheim der Universität Heidelberg, Institut für Klinische Radiologie und Nuklearmedizin, Theodor-Kutzner-Ufer 1-3, D-68167 Mannheim, Germany
| | - Matthew Gott
- Helmholtz-Zentrum Dresden-Rossendorf, Institut für Radiopharmazeutische Krebsforschung, Bautzner Landstraße 400, D-01328 Dresden, Germany
| | - Martin Köckerling
- Universität Rostock, Institut für Chemie - Festkörperchemie, Albert-Einstein-Straße 3a, D-18059 Rostock, Germany
| |
Collapse
|
13
|
Herwig N, Belter B, Wolf S, Haase-Kohn C, Pietzsch J. Interaction of extracellular S100A4 with RAGE prompts prometastatic activation of A375 melanoma cells. J Cell Mol Med 2016; 20:825-35. [PMID: 26928771 PMCID: PMC4831350 DOI: 10.1111/jcmm.12808] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 01/08/2016] [Indexed: 12/19/2022] Open
Abstract
S100A4, a member of the S100 protein family of EF-hand calcium-binding proteins, is overexpressed in various tumour entities, including melanoma, and plays an important role in tumour progression. Several studies in epithelial and mesenchymal tumours revealed a correlation between extracellular S100A4 and metastasis. However, exact mechanisms how S100A4 stimulates metastasis in melanoma are still unknown. From a pilot experiment on baseline synthesis and secretion of S100A4 in human melanoma cell lines, which are in broad laboratory use, A375 wild-type cells and, additionally, newly generated A375 cell lines stably transfected with human S100A4 (A375-hS100A4) or human receptor for advanced glycation endproducts (A375-hRAGE), were selected to investigate the influence of extracellular S100A4 on cell motility, adhesion, migration and invasion in more detail. We demonstrated that A375 cells actively secrete S100A4 in the extracellular space via an endoplasmic reticulum-Golgi-dependent pathway. S100A4 overexpression and secretion resulted in prometastatic activation of A375 cells. Moreover, we determined the influence of S100A4-RAGE interaction and its blockade on A375, A375-hS100A4, A375-hRAGE cells, and showed that interaction of RAGE with extracellular S100A4 contributes to the observed activation of A375 cells. This investigation reveals additional molecular targets for therapeutic approaches aiming at blockade of ligand binding to RAGE or RAGE signalling to inhibit melanoma metastasis.
Collapse
Affiliation(s)
- Nadine Herwig
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.,Department of Chemistry and Food Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Birgit Belter
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Susann Wolf
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Cathleen Haase-Kohn
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany.,Department of Chemistry and Food Chemistry, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
14
|
Recent Advances in the Development and Application of Radiolabeled Kinase Inhibitors for PET Imaging. Molecules 2015; 20:22000-27. [PMID: 26690113 PMCID: PMC6332294 DOI: 10.3390/molecules201219816] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 11/18/2015] [Accepted: 12/01/2015] [Indexed: 12/20/2022] Open
Abstract
Over the last 20 years, intensive investigation and multiple clinical successes targeting protein kinases, mostly for cancer treatment, have identified small molecule kinase inhibitors as a prominent therapeutic class. In the course of those investigations, radiolabeled kinase inhibitors for positron emission tomography (PET) imaging have been synthesized and evaluated as diagnostic imaging probes for cancer characterization. Given that inhibitor coverage of the kinome is continuously expanding, in vivo PET imaging will likely find increasing applications for therapy monitoring and receptor density studies both in- and outside of oncological conditions. Early investigated radiolabeled inhibitors, which are mostly based on clinically approved tyrosine kinase inhibitor (TKI) isotopologues, have now entered clinical trials. Novel radioligands for cancer and PET neuroimaging originating from novel but relevant target kinases are currently being explored in preclinical studies. This article reviews the literature involving radiotracer design, radiochemistry approaches, biological tracer evaluation and nuclear imaging results of radiolabeled kinase inhibitors for PET reported between 2010 and mid-2015. Aspects regarding the usefulness of pursuing selective vs. promiscuous inhibitor scaffolds and the inherent challenges associated with intracellular enzyme imaging will be discussed.
Collapse
|
15
|
Ebert K, Wiemer J, Caballero J, Köckerling M, Steinbach J, Pietzsch J, Mamat C. Development of indazolylpyrimidine derivatives as high-affine EphB4 receptor ligands and potential PET radiotracers. Bioorg Med Chem 2015; 23:6025-35. [PMID: 26189032 DOI: 10.1016/j.bmc.2015.06.040] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 06/05/2015] [Accepted: 06/11/2015] [Indexed: 01/06/2023]
Abstract
Due to their essential role in the pathogenesis of cancer, members of the Eph (erythropoietin-producing hepatoma cell line-A2) receptor tyrosine kinase family represent promising candidates for molecular imaging. Thus, the development and preparation of novel radiotracers for the noninvasive imaging of the EphB4 receptor via positron emission tomography (PET) is described. First in silico investigations with the indazolylpyrimidine lead compound which is known to be highly affine to EphB4 were executed to identify favorable labeling positions for an introduction of fluorine-18 to retain the affinity. Based on this, reference compounds as well as precursors were developed and labeled with carbon-11 and fluorine-18, respectively. For this purpose, a protecting group strategy essentially had to be generated to prevent unwanted methylation and to enable the introduction of fluorine-18. Further, a convenient radiolabeling strategy using [(11)C]methyl iodide was established which afforded the isotopically labeled radiotracer in 30-35% RCY (d.c.) which is identical with the original inhibitor molecule. A spiro ammonium precursor was prepared for radiolabeling with fluorine-18. Unfortunately, the labeling did not lead to the desired (18)F-radiotracer under the chosen conditions.
Collapse
Affiliation(s)
- Kristin Ebert
- Helmholtz-Zentrum Dresden-Rossendorf, Institut für Radiopharmazeutische Krebsforschung, Bautzner Landstraße 400, D-01328 Dresden, Germany
| | - Jens Wiemer
- Helmholtz-Zentrum Dresden-Rossendorf, Institut für Radiopharmazeutische Krebsforschung, Bautzner Landstraße 400, D-01328 Dresden, Germany
| | - Julio Caballero
- Universidad de Talca, Centro de Bioinformática y Simulación Molecular, 2 Norte 685, Casilla 721, Talca, Chile
| | - Martin Köckerling
- Universität Rostock, Institut für Chemie, Anorganische Festkörperchemie, Albert-Einstein-Straße 3a, D-18057 Rostock, Germany
| | - Jörg Steinbach
- Helmholtz-Zentrum Dresden-Rossendorf, Institut für Radiopharmazeutische Krebsforschung, Bautzner Landstraße 400, D-01328 Dresden, Germany; TU Dresden, Fachbereich Chemie und Lebensmittelchemie, D-01062 Dresden, Germany
| | - Jens Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf, Institut für Radiopharmazeutische Krebsforschung, Bautzner Landstraße 400, D-01328 Dresden, Germany; TU Dresden, Fachbereich Chemie und Lebensmittelchemie, D-01062 Dresden, Germany
| | - Constantin Mamat
- Helmholtz-Zentrum Dresden-Rossendorf, Institut für Radiopharmazeutische Krebsforschung, Bautzner Landstraße 400, D-01328 Dresden, Germany; TU Dresden, Fachbereich Chemie und Lebensmittelchemie, D-01062 Dresden, Germany.
| |
Collapse
|
16
|
Puttick S, Stringer BW, Day BW, Bruce ZC, Ensbey KS, Mardon K, Cowin GJ, Thurecht KJ, Whittaker AK, Fay M, Boyd AW, Rose S. EphA2 as a Diagnostic Imaging Target in Glioblastoma: A Positron Emission Tomography/Magnetic Resonance Imaging Study. Mol Imaging 2015. [DOI: 10.2310/7290.2015.00008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Simon Puttick
- From the Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, and Centre for Advanced Imaging, The University of Queensland, St Lucia; QIMR Berghofer Medical Research Institute, Herston; Australian National Imaging Facility, Queensland Node, Brisbane; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Queensland Node, Brisbane; Queensland Health – Royal Brisbane and Women's Hospital, Herston; School of Medicine, The University of Queensland,
| | - Brett W. Stringer
- From the Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, and Centre for Advanced Imaging, The University of Queensland, St Lucia; QIMR Berghofer Medical Research Institute, Herston; Australian National Imaging Facility, Queensland Node, Brisbane; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Queensland Node, Brisbane; Queensland Health – Royal Brisbane and Women's Hospital, Herston; School of Medicine, The University of Queensland,
| | - Bryan W. Day
- From the Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, and Centre for Advanced Imaging, The University of Queensland, St Lucia; QIMR Berghofer Medical Research Institute, Herston; Australian National Imaging Facility, Queensland Node, Brisbane; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Queensland Node, Brisbane; Queensland Health – Royal Brisbane and Women's Hospital, Herston; School of Medicine, The University of Queensland,
| | - Zara C. Bruce
- From the Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, and Centre for Advanced Imaging, The University of Queensland, St Lucia; QIMR Berghofer Medical Research Institute, Herston; Australian National Imaging Facility, Queensland Node, Brisbane; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Queensland Node, Brisbane; Queensland Health – Royal Brisbane and Women's Hospital, Herston; School of Medicine, The University of Queensland,
| | - Kathleen S. Ensbey
- From the Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, and Centre for Advanced Imaging, The University of Queensland, St Lucia; QIMR Berghofer Medical Research Institute, Herston; Australian National Imaging Facility, Queensland Node, Brisbane; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Queensland Node, Brisbane; Queensland Health – Royal Brisbane and Women's Hospital, Herston; School of Medicine, The University of Queensland,
| | - Karine Mardon
- From the Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, and Centre for Advanced Imaging, The University of Queensland, St Lucia; QIMR Berghofer Medical Research Institute, Herston; Australian National Imaging Facility, Queensland Node, Brisbane; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Queensland Node, Brisbane; Queensland Health – Royal Brisbane and Women's Hospital, Herston; School of Medicine, The University of Queensland,
| | - Gary J. Cowin
- From the Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, and Centre for Advanced Imaging, The University of Queensland, St Lucia; QIMR Berghofer Medical Research Institute, Herston; Australian National Imaging Facility, Queensland Node, Brisbane; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Queensland Node, Brisbane; Queensland Health – Royal Brisbane and Women's Hospital, Herston; School of Medicine, The University of Queensland,
| | - Kristofer J. Thurecht
- From the Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, and Centre for Advanced Imaging, The University of Queensland, St Lucia; QIMR Berghofer Medical Research Institute, Herston; Australian National Imaging Facility, Queensland Node, Brisbane; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Queensland Node, Brisbane; Queensland Health – Royal Brisbane and Women's Hospital, Herston; School of Medicine, The University of Queensland,
| | - Andrew K. Whittaker
- From the Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, and Centre for Advanced Imaging, The University of Queensland, St Lucia; QIMR Berghofer Medical Research Institute, Herston; Australian National Imaging Facility, Queensland Node, Brisbane; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Queensland Node, Brisbane; Queensland Health – Royal Brisbane and Women's Hospital, Herston; School of Medicine, The University of Queensland,
| | - Michael Fay
- From the Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, and Centre for Advanced Imaging, The University of Queensland, St Lucia; QIMR Berghofer Medical Research Institute, Herston; Australian National Imaging Facility, Queensland Node, Brisbane; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Queensland Node, Brisbane; Queensland Health – Royal Brisbane and Women's Hospital, Herston; School of Medicine, The University of Queensland,
| | - Andrew W. Boyd
- From the Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, and Centre for Advanced Imaging, The University of Queensland, St Lucia; QIMR Berghofer Medical Research Institute, Herston; Australian National Imaging Facility, Queensland Node, Brisbane; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Queensland Node, Brisbane; Queensland Health – Royal Brisbane and Women's Hospital, Herston; School of Medicine, The University of Queensland,
| | - Stephen Rose
- From the Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, and Centre for Advanced Imaging, The University of Queensland, St Lucia; QIMR Berghofer Medical Research Institute, Herston; Australian National Imaging Facility, Queensland Node, Brisbane; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Queensland Node, Brisbane; Queensland Health – Royal Brisbane and Women's Hospital, Herston; School of Medicine, The University of Queensland,
| |
Collapse
|
17
|
Optical imaging of COX-2: studies on an autofluorescent 2,3-diaryl-substituted indole-based cyclooxygenase-2 inhibitor. Biochem Biophys Res Commun 2015; 458:40-5. [PMID: 25637530 DOI: 10.1016/j.bbrc.2015.01.057] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2015] [Accepted: 01/14/2015] [Indexed: 12/14/2022]
Abstract
This study aimed at in vivo visualization of cyclooxygenase-2 (COX-2) by optical imaging using a representative compound of a class of autofluorescent 2,3-diaryl-substituted indole-based selective COX-2 inhibitors (2,3-diaryl-indole coxibs). COX-2 was successfully visualized in mice models with phorbol myristate ester (TPA)-induced inflammation or bearing xenografted human melanoma cells by 2-[4-(aminosulfonyl)phenyl]-3-(4-methoxyphenyl)-1H-indole (C1). COX-2 protein expression in both TPA-induced inflammatory sites and human melanoma xenografts was confirmed by immunoblotting. Control experiments using surrogate markers, sham injections, and non-COX-2 expressing melanoma cells further confirmed specificity of tissue association of C1. The merging of therapeutic and diagnostic properties of 2,3-diaryl-indole coxibs may widen the range of applications of COX-2-targeted treatment, e.g., for in situ-guided surgery and ex vivo diagnostics.
Collapse
|
18
|
Xie F, Kniess T, Neuber C, Deuther-Conrad W, Mamat C, Lieberman BP, Liu B, Mach RH, Brust P, Steinbach J, Pietzsch J, Jia H. Novel indole-based sigma-2 receptor ligands: synthesis, structure–affinity relationship and antiproliferative activity. MEDCHEMCOMM 2015. [DOI: 10.1039/c5md00079c] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Novel sigma-2 ligand 1b induced G1 phase cell cycle arrest in DU145 cells and displayed antiproliferative activity.
Collapse
|
19
|
Pretze M, Mosch B, Bergmann R, Steinbach J, Pietzsch J, Mamat C. Radiofluorination and first radiopharmacological characterization of a SWLAY peptide-based ligand targeting EphA2. J Labelled Comp Radiopharm 2014; 57:660-5. [DOI: 10.1002/jlcr.3237] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 07/24/2014] [Accepted: 08/24/2014] [Indexed: 12/11/2022]
Affiliation(s)
- Marc Pretze
- Institut für Radiopharmazeutische Krebsforschung; Helmholtz-Zentrum Dresden-Rossendorf; Bautzner Landstraße 400 D-01328 Dresden Germany
- Fachbereich Chemie und Lebensmittelchemie; Technische Universität Dresden; D-01062 Dresden Germany
| | - Birgit Mosch
- Institut für Radiopharmazeutische Krebsforschung; Helmholtz-Zentrum Dresden-Rossendorf; Bautzner Landstraße 400 D-01328 Dresden Germany
| | - Ralf Bergmann
- Institut für Radiopharmazeutische Krebsforschung; Helmholtz-Zentrum Dresden-Rossendorf; Bautzner Landstraße 400 D-01328 Dresden Germany
| | - Jörg Steinbach
- Institut für Radiopharmazeutische Krebsforschung; Helmholtz-Zentrum Dresden-Rossendorf; Bautzner Landstraße 400 D-01328 Dresden Germany
- Fachbereich Chemie und Lebensmittelchemie; Technische Universität Dresden; D-01062 Dresden Germany
| | - Jens Pietzsch
- Institut für Radiopharmazeutische Krebsforschung; Helmholtz-Zentrum Dresden-Rossendorf; Bautzner Landstraße 400 D-01328 Dresden Germany
- Fachbereich Chemie und Lebensmittelchemie; Technische Universität Dresden; D-01062 Dresden Germany
| | - Constantin Mamat
- Institut für Radiopharmazeutische Krebsforschung; Helmholtz-Zentrum Dresden-Rossendorf; Bautzner Landstraße 400 D-01328 Dresden Germany
- Fachbereich Chemie und Lebensmittelchemie; Technische Universität Dresden; D-01062 Dresden Germany
| |
Collapse
|
20
|
Experimental hypoxia does not influence gene expression and protein synthesis of Eph receptors and ephrin ligands in human melanoma cells in vitro. Melanoma Res 2014; 23:85-95. [PMID: 23358429 DOI: 10.1097/cmr.0b013e32835e58f3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Eph receptor tyrosine kinases and their ephrin ligands are considered to play important roles in melanoma progression and metastasis. Moreover, hypoxia is known to contribute to melanoma metastasis. In this study, the influence of experimental hypoxia on the expression and synthesis of EphA2 and EphB4, and their corresponding ligands ephrinA1, ephrinA5, and ephrinB2 was studied systematically in four human melanoma cell lines in vitro. Melanoma cell monolayer and spheroid cultures were used as both extrinsic and intrinsic hypoxia models. Hypoxic conditions were confirmed by analyzing hypoxia-inducible factors 1α or 2α expression, vascular endothelial growth factor expression, and cellular uptake of [F]fluoromisonidazole. In normoxia, EphA2, EphB4, ephrinA1, ephrinA5, and ephrinB2 expression was detectable in all cell lines to varying extents. Considerable protein synthesis of EphA2 was detected in all cell lines. However, no effect of experimental hypoxia on both Eph/ephrin expression and protein synthesis was observed. This contributes critically to the debate on the hypothesis that hypoxia regulates the Eph/ephrin system in melanoma.
Collapse
|
21
|
Pretze M, Kuchar M, Bergmann R, Steinbach J, Pietzsch J, Mamat C. An efficient bioorthogonal strategy using CuAAC click chemistry for radiofluorinations of SNEW peptides and the role of copper depletion. ChemMedChem 2013; 8:935-45. [PMID: 23559494 DOI: 10.1002/cmdc.201300053] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Indexed: 12/20/2022]
Abstract
The EphB2 receptor is known to be overexpressed in various types of cancer and is therefore a promising target for tumor cell imaging by positron emission tomography (PET). In this regard, imaging could facilitate the early detection of EphB2-overexpressing tumors, monitoring responses to therapy directed toward EphB2, and thus improvement in patient outcomes. We report the synthesis and evaluation of several fluorine-18-labeled peptides containing the SNEW amino acid motif, with high affinity for the EphB2 receptor, for their potential as radiotracers in the non-invasive imaging of cancer using PET. For the purposes of radiofluorination, EphB2-antagonistic SNEW peptides were varied at the C terminus by the introduction of L-cysteine, and further by alkyne- or azide-modified amino acids. In addition, two novel bifunctional and bioorthogonal labeling building blocks [(18)F]AFP and [(18)F]BFP were applied, and their capacity to introduce fluorine-18 was compared with that of the established building block [(18)F]FBAM. Copper-assisted Huisgen 1,3-dipolar cycloaddition, which belongs to the set of bioorthogonal click chemistry reactions, was used to introduce both novel building blocks into azide- or alkyne-modified SNEW peptides under mild conditions. Finally, the depletion of copper immediately after radiolabeling is a highly important step of this novel methodology.
Collapse
Affiliation(s)
- Marc Pretze
- Institut für Radiopharmazeutische Krebsforschung, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | | | | | | | | | | |
Collapse
|
22
|
Incerti M, Tognolini M, Russo S, Pala D, Giorgio C, Hassan-Mohamed I, Noberini R, Pasquale EB, Vicini P, Piersanti S, Rivara S, Barocelli E, Mor M, Lodola A. Amino acid conjugates of lithocholic acid as antagonists of the EphA2 receptor. J Med Chem 2013; 56:2936-47. [PMID: 23489211 DOI: 10.1021/jm301890k] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The Eph receptor-ephrin system is an emerging target for the development of novel antiangiogenetic agents. We recently identified lithocholic acid (LCA) as a small molecule able to block EphA2-dependent signals in cancer cells, suggesting that its (5β)-cholan-24-oic acid scaffold can be used as a template to design a new generation of improved EphA2 antagonists. Here, we report the design and synthesis of an extended set of LCA derivatives obtained by conjugation of its carboxyl group with different α-amino acids. Structure-activity relationships indicate that the presence of a lipophilic amino acid side chain is fundamental to achieve good potencies. The l-Trp derivative (20, PCM126) was the most potent antagonist of the series disrupting EphA2-ephrinA1 interaction and blocking EphA2 phosphorylation in prostate cancer cells at low μM concentrations, thus being significantly more potent than LCA. Compound 20 is among the most potent small-molecule antagonists of the EphA2 receptor.
Collapse
Affiliation(s)
- Matteo Incerti
- Dipartimento di Farmacia, Università degli Studi di Parma, Viale delle Scienze 27/A, I-43124 Parma, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Eph receptors and their ligands: promising molecular biomarkers and therapeutic targets in prostate cancer. Biochim Biophys Acta Rev Cancer 2013; 1835:243-57. [PMID: 23396052 DOI: 10.1016/j.bbcan.2013.01.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 01/21/2013] [Accepted: 01/25/2013] [Indexed: 01/01/2023]
Abstract
Although at present, there is a high incidence of prostate cancer, particularly in the Western world, mortality from this disease is declining and occurs primarily only from clinically significant late stage tumors with a poor prognosis. A major current focus of this field is the identification of new biomarkers which can detect earlier, and more effectively, clinically significant tumors from those deemed "low risk", as well as predict the prognostic course of a particular cancer. This strategy can in turn offer novel avenues for targeted therapies. The large family of Receptor Tyrosine Kinases, the Ephs, and their binding partners, the ephrins, has been implicated in many cancers of epithelial origin through stimulation of oncogenic transformation, tumor angiogenesis, and promotion of increased cell survival, invasion and migration. They also show promise as both biomarkers of diagnostic and prognostic value and as targeted therapies in cancer. This review will briefly discuss the complex roles and biological mechanisms of action of these receptors and ligands and, with regard to prostate cancer, highlight their potential as biomarkers for both diagnosis and prognosis, their application as imaging agents, and current approaches to assessing them as therapeutic targets. This review demonstrates the need for future studies into those particular family members that will prove helpful in understanding the biology and potential as targets for treatment of prostate cancer.
Collapse
|