1
|
Weidner P, Saar D, Söhn M, Schroeder T, Yu Y, Zöllner FG, Ponelies N, Zhou X, Zwicky A, Rohrbacher FN, Pattabiraman VR, Tanriver M, Bauer A, Ahmed H, Ametamey SM, Riffel P, Seger R, Bode JW, Wade RC, Ebert MPA, Kragelund BB, Burgermeister E. Myotubularin-related-protein-7 inhibits mutant (G12V) K-RAS by direct interaction. Cancer Lett 2024; 588:216783. [PMID: 38462034 DOI: 10.1016/j.canlet.2024.216783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/19/2024] [Accepted: 03/03/2024] [Indexed: 03/12/2024]
Abstract
Inhibition of K-RAS effectors like B-RAF or MEK1/2 is accompanied by treatment resistance in cancer patients via re-activation of PI3K and Wnt signaling. We hypothesized that myotubularin-related-protein-7 (MTMR7), which inhibits PI3K and ERK1/2 signaling downstream of RAS, directly targets RAS and thereby prevents resistance. Using cell and structural biology combined with animal studies, we show that MTMR7 binds and inhibits RAS at cellular membranes. Overexpression of MTMR7 reduced RAS GTPase activities and protein levels, ERK1/2 phosphorylation, c-FOS transcription and cancer cell proliferation in vitro. We located the RAS-inhibitory activity of MTMR7 to its charged coiled coil (CC) region and demonstrate direct interaction with the gastrointestinal cancer-relevant K-RASG12V mutant, favouring its GDP-bound state. In mouse models of gastric and intestinal cancer, a cell-permeable MTMR7-CC mimicry peptide decreased tumour growth, Ki67 proliferation index and ERK1/2 nuclear positivity. Thus, MTMR7 mimicry peptide(s) could provide a novel strategy for targeting mutant K-RAS in cancers.
Collapse
Affiliation(s)
- Philip Weidner
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Daniel Saar
- Structural Biology and NMR Laboratory (SBiNLab) and the Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Michaela Söhn
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Torsten Schroeder
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Yanxiong Yu
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Frank G Zöllner
- Computer Assisted Clinical Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Mannheim Institute for Intelligent Systems in Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; Cooperative Core Facility Animal Scanner ZI, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Norbert Ponelies
- Orthopaedics & Trauma Surgery, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Xiaobo Zhou
- Department of Medicine I, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - André Zwicky
- Laboratory of Organic Chemistry, Department of Chemistry and Applied Bioscience of ETH, Zurich, Switzerland
| | - Florian N Rohrbacher
- Laboratory of Organic Chemistry, Department of Chemistry and Applied Bioscience of ETH, Zurich, Switzerland
| | - Vijaya R Pattabiraman
- Laboratory of Organic Chemistry, Department of Chemistry and Applied Bioscience of ETH, Zurich, Switzerland
| | - Matthias Tanriver
- Laboratory of Organic Chemistry, Department of Chemistry and Applied Bioscience of ETH, Zurich, Switzerland
| | - Alexander Bauer
- Structural Biology and NMR Laboratory (SBiNLab) and the Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Hazem Ahmed
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences of ETH, Zurich, Switzerland
| | - Simon M Ametamey
- Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences of ETH, Zurich, Switzerland
| | - Philipp Riffel
- Clinic of Radiology and Nuclear Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Rony Seger
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Jeffrey W Bode
- Laboratory of Organic Chemistry, Department of Chemistry and Applied Bioscience of ETH, Zurich, Switzerland
| | - Rebecca C Wade
- Heidelberg Institute for Theoretical Studies (HITS), Heidelberg, Germany; Heidelberg University, Zentrum für Molekulare Biologie (ZMBH), DKFZ-ZMBH Alliance, and Interdisciplinary Center for Scientific Computing (IWR), Heidelberg, Germany
| | - Matthias P A Ebert
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany; DKFZ-Hector Institute at the University Medical Center, Mannheim, Germany
| | - Birthe B Kragelund
- Structural Biology and NMR Laboratory (SBiNLab) and the Linderstrøm-Lang Centre for Protein Science, Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| | - Elke Burgermeister
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
2
|
Ahmed H, Wallimann R, Gisler L, Elghazawy NH, Gruber S, Keller C, Liang SH, Sippl W, Haider A, Ametamey SM. Characterization of ( R)- and ( S)-[ 18F]OF-NB1 in Rodents as Positron Emission Tomography Probes for Imaging GluN2B Subunit-Containing N-Methyl-d-Aspartate Receptors. ACS Chem Neurosci 2023; 14:4323-4334. [PMID: 38060344 DOI: 10.1021/acschemneuro.3c00519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023] Open
Abstract
The N-methyl-d-aspartate receptor (NMDAR) subtype 2B (GluN1/2B) is implicated in various neuropathologies. Given the lack of a validated radiofluorinated positron emission tomography (PET) probe for the imaging of GluN1/2B receptors, we comprehensively investigated the enantiomers of [18F]OF-NB1 in rodents. Particularly, the (R)- and (S)- enantiomers were evaluated using in silico docking, in vitro autoradiography, in vivo PET imaging, and ex vivo biodistribution studies. A select panel of GluN1/2B antagonists (CP-101,606, CERC-301, and eliprodil) and the off-target sigma-1 receptor ligands (fluspidine and SA4503) were used to determine the specificity and selectivity of the tested enantiomers. Additionally, a nonmetal-mediated radiofluorination strategy was devised that harnesses the potential of diaryliodoniums in the nucleophilic radiofluorination of nonactivated aromatic compounds. Both enantiomers exhibited known GluN1/2B binding patterns; however, the R-enantiomer showed higher GluN1/2B-specific accumulation in rodent autoradiography and higher brain uptake in PET imaging experiments compared to the S-enantiomer. Molecular simulation studies provided further insights with respect to the difference in binding, whereby a reduced ligand-receptor interaction was observed for the S-enantiomer. Nonetheless, both enantiomers showed dose dependency when two different doses (1 and 5 mg/kg) of the GluN1/2B antagonist, CP-101,606, were used in the PET imaging study. Taken together, (R)-[18F]OF-NB1 appears to exhibit the characteristics of a suitable PET probe for imaging of GluN2B-containing NMDARs in clinical studies.
Collapse
Affiliation(s)
- Hazem Ahmed
- Institute of Pharmaceutical Sciences, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland
| | - Rahel Wallimann
- Institute of Pharmaceutical Sciences, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland
| | - Livio Gisler
- Institute of Pharmaceutical Sciences, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland
| | - Nehal H Elghazawy
- Institute of Pharmacy, Department of Medicinal Chemistry, Martin-Luther-University Halle-Wittenberg, W.-Langenbeck-Str. 4, 06120 Halle, Germany
| | - Stefan Gruber
- Institute of Pharmaceutical Sciences, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland
| | - Claudia Keller
- Institute of Pharmaceutical Sciences, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland
| | - Steven H Liang
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Wolfgang Sippl
- Institute of Pharmacy, Department of Medicinal Chemistry, Martin-Luther-University Halle-Wittenberg, W.-Langenbeck-Str. 4, 06120 Halle, Germany
| | - Achi Haider
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, Massachusetts 02114, United States
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Simon M Ametamey
- Institute of Pharmaceutical Sciences, ETH Zurich, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland
| |
Collapse
|
3
|
Korff M, Chaudhary A, Li Y, Zhou X, Zhao C, Rong J, Chen J, Xiao Z, Elghazawy NH, Sippl W, Davenport AT, Daunais JB, Wang L, Abate C, Ahmed H, Crowe R, Schmidt TJ, Liang SH, Ametamey SM, Wünsch B, Haider A. Synthesis and Biological Evaluation of Enantiomerically Pure ( R) - and ( S) -[18F]OF-NB1 for Imaging the GluN2B Subunit-Containing NMDA Receptors. J Med Chem 2023; 66:16018-16031. [PMID: 37979148 DOI: 10.1021/acs.jmedchem.3c01441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2023]
Abstract
GluN2B subunit-containing N-methyl-d-aspartate (NMDA) receptors have been implicated in various neurological disorders. Nonetheless, a validated fluorine-18 labeled positron emission tomography (PET) ligand for GluN2B imaging in the living human brain is currently lacking. The aim of this study was to develop a novel synthetic approach that allows an enantiomerically pure radiosynthesis of the previously reported PET radioligands (R)-[18F]OF-NB1 and (S)-[18F]OF-NB1 as well as to assess their in vitro and in vivo performance characteristics for imaging the GluN2B subunit-containing NMDA receptor in rodents. A novel synthetic approach was successfully developed, which allows for the enantiomerically pure radiosynthesis of (R)-[18F]OF-NB1 and (S)-[18F]OF-NB1 and the translation of the probe to the clinic. While both enantiomers were selective over sigma2 receptors in vitro and in vivo, (R)-[18F]OF-NB1 showed superior GluN2B subunit specificity by in vitro autoradiography and higher volumes of distribution in the rodent brain by small animal PET studies.
Collapse
Affiliation(s)
- Marvin Korff
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Road, Atlanta, Georgia 30322, United States
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, Münster D-48149, Germany
| | - Ahmad Chaudhary
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Road, Atlanta, Georgia 30322, United States
| | - Yinlong Li
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Road, Atlanta, Georgia 30322, United States
| | - Xin Zhou
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Road, Atlanta, Georgia 30322, United States
| | - Chunyu Zhao
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Road, Atlanta, Georgia 30322, United States
| | - Jian Rong
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Road, Atlanta, Georgia 30322, United States
| | - Jiahui Chen
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Road, Atlanta, Georgia 30322, United States
| | - Zhiwei Xiao
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Road, Atlanta, Georgia 30322, United States
| | - Nehal H Elghazawy
- Institute of Pharmacy, Department of Medicinal Chemistry, Martin-Luther-University Halle-Wittenberg, W.-Langenbeck-Str. 4, Halle 06120, Germany
| | - Wolfgang Sippl
- Institute of Pharmacy, Department of Medicinal Chemistry, Martin-Luther-University Halle-Wittenberg, W.-Langenbeck-Str. 4, Halle 06120, Germany
| | - April T Davenport
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston Salem, North Carolina 27157, United States
| | - James B Daunais
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston Salem, North Carolina 27157, United States
| | - Lu Wang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Carmen Abate
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| | - Hazem Ahmed
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| | - Ron Crowe
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Road, Atlanta, Georgia 30322, United States
| | - Thomas J Schmidt
- Institut für Pharmazeutische Biologie und Phytochemie, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, Münster D-48149, Germany
| | - Steven H Liang
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Road, Atlanta, Georgia 30322, United States
| | - Simon M Ametamey
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Vladimir-Prelog-Weg 4, Zurich 8093, Switzerland
| | - Bernhard Wünsch
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, Münster D-48149, Germany
| | - Achi Haider
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Road, Atlanta, Georgia 30322, United States
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, Zurich 8091, Switzerland
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States
| |
Collapse
|
4
|
Ritter N, Disse P, Aymanns I, Mücher L, Schreiber JA, Brenker C, Strünker T, Schepmann D, Budde T, Strutz-Seebohm N, Ametamey SM, Wünsch B, Seebohm G. Downstream Allosteric Modulation of NMDA Receptors by 3-Benzazepine Derivatives. Mol Neurobiol 2023; 60:7238-7252. [PMID: 37542648 PMCID: PMC10657792 DOI: 10.1007/s12035-023-03526-1] [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: 10/19/2022] [Accepted: 07/20/2023] [Indexed: 08/07/2023]
Abstract
N-Methyl-D-aspartate receptors (NMDARs) composed of different splice variants display distinct pH sensitivities and are crucial for learning and memory, as well as for inflammatory or injury processes. Dysregulation of the NMDAR has been linked to diseases like Parkinson's, Alzheimer's, schizophrenia, and drug addiction. The development of selective receptor modulators, therefore, constitutes a promising approach for numerous therapeutical applications. Here, we identified (R)-OF-NB1 as a promising splice variant selective NMDAR antagonist. We investigated the interaction of (R)-OF-NB1 and NMDAR from a biochemical, bioinformatical, and electrophysiological perspective to characterize the downstream allosteric modulation of NMDAR by 3-benzazepine derivatives. The allosteric modulatory pathway starts at the ifenprodil binding pocket in the amino terminal domain and immobilizes the connecting α5-helix to the ligand binding domain, resulting in inhibition. In contrast, the exon 5 splice variant GluN1-1b elevates the NMDARs flexibility and promotes the open state of its ligand binding domain.
Collapse
Affiliation(s)
- Nadine Ritter
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, 48149, Münster, Germany.
- Chembion, University of Münster, 48149, Münster, Germany.
| | - Paul Disse
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, 48149, Münster, Germany
- Chembion, University of Münster, 48149, Münster, Germany
| | - Isabel Aymanns
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, 48149, Münster, Germany
| | - Lena Mücher
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, 48149, Münster, Germany
| | - Julian A Schreiber
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, 48149, Münster, Germany
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstr. 48, 48149, Münster, Germany
| | - Christoph Brenker
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Domagkstr. 11, 48149, Münster, Germany
| | - Timo Strünker
- Chembion, University of Münster, 48149, Münster, Germany
- Centre of Reproductive Medicine and Andrology, University Hospital Münster, University of Münster, Domagkstr. 11, 48149, Münster, Germany
| | - Dirk Schepmann
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstr. 48, 48149, Münster, Germany
| | - Thomas Budde
- Institute of Physiology I, University of Münster, 48149, Münster, Germany
| | - Nathalie Strutz-Seebohm
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, 48149, Münster, Germany
| | - Simon M Ametamey
- Institute of Pharmaceutical Sciences, ETH Zürich, Zürich, Switzerland
| | - Bernhard Wünsch
- Chembion, University of Münster, 48149, Münster, Germany
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstr. 48, 48149, Münster, Germany
| | - Guiscard Seebohm
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, 48149, Münster, Germany
- Chembion, University of Münster, 48149, Münster, Germany
| |
Collapse
|
5
|
Wang X, Wang T, Fan X, Zhang Z, Wang Y, Li Z. A Molecular Toolbox of Positron Emission Tomography Tracers for General Anesthesia Mechanism Research. J Med Chem 2023; 66:6463-6497. [PMID: 37145921 DOI: 10.1021/acs.jmedchem.2c01965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
With appropriate radiotracers, positron emission tomography (PET) allows direct or indirect monitoring of the spatial and temporal distribution of anesthetics, neurotransmitters, and biomarkers, making it an indispensable tool for studying the general anesthesia mechanism. In this Perspective, PET tracers that have been recruited in general anesthesia research are introduced in the following order: 1) 11C/18F-labeled anesthetics, i.e., PET tracers made from inhaled and intravenous anesthetics; 2) PET tracers targeting anesthesia-related receptors, e.g., neurotransmitters and voltage-gated ion channels; and 3) PET tracers for studying anesthesia-related neurophysiological effects and neurotoxicity. The radiosynthesis, pharmacodynamics, and pharmacokinetics of the above PET tracers are mainly discussed to provide a practical molecular toolbox for radiochemists, anesthesiologists, and those who are interested in general anesthesia.
Collapse
Affiliation(s)
- Xiaoxiao Wang
- Center for Molecular Imaging and Translational Medicine, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian 361102, China
| | - Tao Wang
- Center for Molecular Imaging and Translational Medicine, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian 361102, China
| | - Xiaowei Fan
- Center for Molecular Imaging and Translational Medicine, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian 361102, China
| | - Zhao Zhang
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yingwei Wang
- Department of Anesthesiology, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Zijing Li
- Center for Molecular Imaging and Translational Medicine, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, Department of Laboratory Medicine, School of Public Health, Xiamen University, Xiamen, Fujian 361102, China
| |
Collapse
|
6
|
Ritter N, Disse P, Wünsch B, Seebohm G, Strutz-Seebohm N. Pharmacological Potential of 3-Benzazepines in NMDAR-Linked Pathophysiological Processes. Biomedicines 2023; 11:1367. [PMID: 37239037 PMCID: PMC10216354 DOI: 10.3390/biomedicines11051367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/22/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
The number of N-Methyl-D-aspartate receptor (NMDAR) linked neurodegenerative diseases such as Alzheimer's disease and dementia is constantly increasing. This is partly due to demographic change and presents new challenges to societies. To date, there are no effective treatment options. Current medications are nonselective and can lead to unwanted side effects in patients. A promising therapeutic approach is the targeted inhibition of NMDARs in the brain. NMDARs containing different subunits and splice variants display different physiological properties and play a crucial role in learning and memory, as well as in inflammatory or injury processes. They become overactivated during the course of the disease, leading to nerve cell death. Until now, there has been a lack of understanding of the general functions of the receptor and the mechanism of inhibition, which need to be understood in order to develop inhibitors. Ideal compounds should be highly targeted and even splice-variant-selective. However, a potent and splice-variant-selective NMDAR-targeting drug has yet to be developed. Recently developed 3-benzazepines are promising inhibitors for further drug development. The NMDAR splice variants GluN1-1b-4b carry a 21-amino-acid-long, flexible exon 5. Exon 5 lowers the NMDAR's sensitivity to allosteric modulators by probably acting as an NMDAR modulator itself. The role of exon 5 in NMDAR modulation is still poorly understood. In this review, we summarize the structure and pharmacological relevance of tetrahydro-3-benzazepines.
Collapse
Affiliation(s)
- Nadine Ritter
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, D-48149 Münster, Germany; (P.D.); (G.S.); (N.S.-S.)
- Chembion, University of Münster, D-48149 Münster, Germany;
| | - Paul Disse
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, D-48149 Münster, Germany; (P.D.); (G.S.); (N.S.-S.)
- Chembion, University of Münster, D-48149 Münster, Germany;
| | - Bernhard Wünsch
- Chembion, University of Münster, D-48149 Münster, Germany;
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstr. 48, D-48149 Münster, Germany
| | - Guiscard Seebohm
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, D-48149 Münster, Germany; (P.D.); (G.S.); (N.S.-S.)
- Chembion, University of Münster, D-48149 Münster, Germany;
| | - Nathalie Strutz-Seebohm
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Münster, D-48149 Münster, Germany; (P.D.); (G.S.); (N.S.-S.)
| |
Collapse
|
7
|
Haider A, Elghazawy NH, Dawoud A, Gebhard C, Wichmann T, Sippl W, Hoener M, Arenas E, Liang SH. Translational molecular imaging and drug development in Parkinson's disease. Mol Neurodegener 2023; 18:11. [PMID: 36759912 PMCID: PMC9912681 DOI: 10.1186/s13024-023-00600-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 01/23/2023] [Indexed: 02/11/2023] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder that primarily affects elderly people and constitutes a major source of disability worldwide. Notably, the neuropathological hallmarks of PD include nigrostriatal loss and the formation of intracellular inclusion bodies containing misfolded α-synuclein protein aggregates. Cardinal motor symptoms, which include tremor, rigidity and bradykinesia, can effectively be managed with dopaminergic therapy for years following symptom onset. Nonetheless, patients ultimately develop symptoms that no longer fully respond to dopaminergic treatment. Attempts to discover disease-modifying agents have increasingly been supported by translational molecular imaging concepts, targeting the most prominent pathological hallmark of PD, α-synuclein accumulation, as well as other molecular pathways that contribute to the pathophysiology of PD. Indeed, molecular imaging modalities such as positron emission tomography (PET) and single-photon emission computed tomography (SPECT) can be leveraged to study parkinsonism not only in animal models but also in living patients. For instance, mitochondrial dysfunction can be assessed with probes that target the mitochondrial complex I (MC-I), while nigrostriatal degeneration is typically evaluated with probes designed to non-invasively quantify dopaminergic nerve loss. In addition to dopaminergic imaging, serotonin transporter and N-methyl-D-aspartate (NMDA) receptor probes are increasingly used as research tools to better understand the complexity of neurotransmitter dysregulation in PD. Non-invasive quantification of neuroinflammatory processes is mainly conducted by targeting the translocator protein 18 kDa (TSPO) on activated microglia using established imaging agents. Despite the overwhelming involvement of the brain and brainstem, the pathophysiology of PD is not restricted to the central nervous system (CNS). In fact, PD also affects various peripheral organs such as the heart and gastrointestinal tract - primarily via autonomic dysfunction. As such, research into peripheral biomarkers has taken advantage of cardiac autonomic denervation in PD, allowing the differential diagnosis between PD and multiple system atrophy with probes that visualize sympathetic nerve terminals in the myocardium. Further, α-synuclein has recently gained attention as a potential peripheral biomarker in PD. This review discusses breakthrough discoveries that have led to the contemporary molecular concepts of PD pathophysiology and how they can be harnessed to develop effective imaging probes and therapeutic agents. Further, we will shed light on potential future trends, thereby focusing on potential novel diagnostic tracers and disease-modifying therapeutic interventions.
Collapse
Affiliation(s)
- Achi Haider
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA.
- Department of Radiology and Imaging Sciences, Emory University, 101 Woodruff Circle, Atlanta, GA, 30322, USA.
| | - Nehal H Elghazawy
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Main Entrance of Al-Tagamoa Al-Khames, Cairo, 11835, Egypt
- Molecular Genetics Research Team (MGRT), Pharmaceutical Biology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Main Entrance of Al-Tagamoa Al-Khames, Cairo, 11835, Egypt
| | - Alyaa Dawoud
- Biochemistry Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Main Entrance of Al-Tagamoa Al-Khames, Cairo, 11835, Egypt
- Molecular Genetics Research Team (MGRT), Pharmaceutical Biology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo, Main Entrance of Al-Tagamoa Al-Khames, Cairo, 11835, Egypt
| | - Catherine Gebhard
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
| | - Thomas Wichmann
- Department of Neurology/School of Medicine, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
| | - Wolfgang Sippl
- Institute of Pharmacy, Department of Medicinal Chemistry, Martin-Luther-University Halle-Wittenberg, W.-Langenbeck-Str. 4, 06120, Halle, Germany
| | - Marius Hoener
- Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Ernest Arenas
- Karolinska Institutet, MBB, Molecular Neurobiology, Stockholm, Sweden
| | - Steven H Liang
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA.
- Department of Radiology and Imaging Sciences, Emory University, 101 Woodruff Circle, Atlanta, GA, 30322, USA.
| |
Collapse
|
8
|
Bengs S, Warnock GI, Portmann A, Mikail N, Rossi A, Ahmed H, Etter D, Treyer V, Gisler L, Pfister SK, Jie CVML, Meisel A, Keller C, Liang SH, Schibli R, Mu L, Buechel RR, Kaufmann PA, Ametamey SM, Gebhard C, Haider A. Rest/stress myocardial perfusion imaging by positron emission tomography with 18F-Flurpiridaz: A feasibility study in mice. J Nucl Cardiol 2023; 30:62-73. [PMID: 35484467 PMCID: PMC9984310 DOI: 10.1007/s12350-022-02968-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 03/15/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Myocardial perfusion imaging by positron emission tomography (PET-MPI) is the current gold standard for quantification of myocardial blood flow. 18F-flurpiridaz was recently introduced as a valid alternative to currently used PET-MPI probes. Nonetheless, optimum scan duration and time interval for image analysis are currently unknown. Further, it is unclear whether rest/stress PET-MPI with 18F-flurpiridaz is feasible in mice. METHODS Rest/stress PET-MPI was performed with 18F-flurpiridaz (0.6-3.0 MBq) in 27 mice aged 7-8 months. Regadenoson (0.1 µg/g) was used for induction of vasodilator stress. Kinetic modeling was performed using a metabolite-corrected arterial input function. Image-derived myocardial 18F-flurpiridaz uptake was assessed for different time intervals by placing a volume of interest in the left ventricular myocardium. RESULTS Tracer kinetics were best described by a two-tissue compartment model. K1 ranged from 6.7 to 20.0 mL·cm-3·min-1, while myocardial volumes of distribution (VT) were between 34.6 and 83.6 mL·cm-3. Of note, myocardial 18F-flurpiridaz uptake (%ID/g) was significantly correlated with K1 at rest and following pharmacological vasodilation for all time intervals assessed. However, while Spearman's coefficients (rs) ranged between 0.478 and 0.681, R2 values were generally low. In contrast, an excellent correlation of myocardial 18F-flurpiridaz uptake with VT was obtained, particularly when employing the averaged myocardial uptake from 20 to 40 min post tracer injection (R2 ≥ 0.98). Notably, K1 and VT were similarly sensitive to pharmacological vasodilation induction. Further, mean stress-to-rest ratios of K1, VT, and %ID/g 18F-flurpiridaz were virtually identical, suggesting that %ID/g 18F-flurpiridaz can be used to estimate coronary flow reserve (CFR) in mice. CONCLUSION Our findings suggest that a simplified assessment of relative myocardial perfusion and CFR, based on image-derived tracer uptake, is feasible with 18F-flurpiridaz in mice, enabling high-throughput mechanistic CFR studies in rodents.
Collapse
Affiliation(s)
- Susan Bengs
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, 8952, Schlieren, Switzerland
| | - Geoffrey I Warnock
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, 8952, Schlieren, Switzerland
| | - Angela Portmann
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, 8952, Schlieren, Switzerland
| | - Nidaa Mikail
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, 8952, Schlieren, Switzerland
| | - Alexia Rossi
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, 8952, Schlieren, Switzerland
| | - Hazem Ahmed
- Institute of Pharmaceutical Sciences, ETH Zurich, 8093, Zurich, Switzerland
| | - Dominik Etter
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, 8952, Schlieren, Switzerland
| | - Valerie Treyer
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Livio Gisler
- Institute of Pharmaceutical Sciences, ETH Zurich, 8093, Zurich, Switzerland
| | - Stefanie K Pfister
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
| | - Caitlin V M L Jie
- Institute of Pharmaceutical Sciences, ETH Zurich, 8093, Zurich, Switzerland
| | - Alexander Meisel
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, 8952, Schlieren, Switzerland
| | - Claudia Keller
- Institute of Pharmaceutical Sciences, ETH Zurich, 8093, Zurich, Switzerland
| | - Steven H Liang
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA
| | - Roger Schibli
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
- Institute of Pharmaceutical Sciences, ETH Zurich, 8093, Zurich, Switzerland
| | - Linjing Mu
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
- Institute of Pharmaceutical Sciences, ETH Zurich, 8093, Zurich, Switzerland
| | - Ronny R Buechel
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Philipp A Kaufmann
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Simon M Ametamey
- Institute of Pharmaceutical Sciences, ETH Zurich, 8093, Zurich, Switzerland
| | - Catherine Gebhard
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, 8952, Schlieren, Switzerland
| | - Achi Haider
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland.
- Center for Molecular Cardiology, University of Zurich, 8952, Schlieren, Switzerland.
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
9
|
Korff M, Chaudhary A, Li Y, Zhou X, Zhao C, Rong J, Chen J, Xiao Z, Elghazawy NH, Sippl W, Davenport AT, Daunais JB, Wang L, Abate C, Ahmed H, Crowe R, Liang SH, Ametamey SM, Wünsch B, Haider A. Synthesis and Biological Evaluation of Enantiomerically Pure ( R)- and ( S)-[ 18F]OF-NB1 for Imaging the GluN2B Subunit-Containing NMDA receptors. RESEARCH SQUARE 2023:rs.3.rs-2516002. [PMID: 36747738 PMCID: PMC9901044 DOI: 10.21203/rs.3.rs-2516002/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
GluN2B subunit-containing N-methyl-d-aspartate (NMDA) receptors have been implicated in various neurological disorders. Nonetheless, a validated fluorine-18 labeled positron emission tomography (PET) ligand for GluN2B imaging in the living human brain is currently lacking. As part of our PET ligand development program, we have recently reported on the preclinical evaluation of [18F]OF-NB1 - a GluN2B PET ligand with promising attributes for potential clinical translation. However, the further development of [18F]OF-NB1 is currently precluded by major limitations in the radiolabeling procedure. These limitations include the use of highly corrosive reactants and racemization during the radiosynthesis. As such, the aim of this study was to develop a synthetic approach that allows an enantiomerically pure radiosynthesis of (R)-[18F]OF-NB1 and (S)-[18F]OF-NB1, as well as to assess their in vitro and in vivo performance characteristics for imaging the GluN2B subunit-containing NMDA receptor in rodents. A two-step radiosynthesis involving radiofluorination of the boronic acid pinacol ester, followed by coupling to the 3-benzazepine core structure via reductive amination was employed. The new synthetic approach yielded enantiomerically pure (R)-[18F]OF-NB1 and (S)-[18F]OF-NB1, while concurrently circumventing the use of corrosive reactants. In vitro autoradiograms with mouse and rat brain sections revealed a higher selectivity of (R)-[18F]OF-NB1 over (S)-[18F]OFNB1 for GluN2B-rich brain regions. In concert with these observations, blockade studies with commercially available GluN2B antagonist, CP101606, showed a significant signal reduction, which was more pronounced for (R)-[18F]OF-NB1 than for (S)-[18F]OF-NB1. Conversely, blockade experiments with sigma2 ligand, FA10, did not result in a significant reduction of tracer binding for both enantiomers. PET imaging experiments with CD1 mice revealed a higher brain uptake and retention for (R)-[18F]OF-NB1, as assessed by visual inspection and volumes of distribution from Logan graphical analyses. In vivo blocking experiments with sigma2 ligand, FA10, did not result in a significant reduction of the brain signal for both enantiomers, thus corroborating the selectivity over sigma2 receptors. In conclusion, we have developed a novel synthetic approach that is suitable for upscale to human use and allows the enantiomerically pure radiosynthesis of (R)-[18F]OF-NB1 and (S)-[18F]OF-NB1. While both enantiomers were selective over sigma2 receptors in vitro and in vivo, (R)-[18F]OF-NB1 showed superior GluN2B subunit specificity by in vitro autoradiography and higher volumes of distribution in small animal PET studies.
Collapse
Affiliation(s)
- Marvin Korff
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Road, Atlanta, GA 30322, USA
| | - Ahmad Chaudhary
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Road, Atlanta, GA 30322, USA
| | - Yinlong Li
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Road, Atlanta, GA 30322, USA
| | - Xin Zhou
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Road, Atlanta, GA 30322, USA
| | - Chunyu Zhao
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Road, Atlanta, GA 30322, USA
| | - Jian Rong
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Road, Atlanta, GA 30322, USA
| | - Jiahui Chen
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Road, Atlanta, GA 30322, USA
| | - Zhiwei Xiao
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Road, Atlanta, GA 30322, USA
| | - Nehal H Elghazawy
- Institute of Pharmacy, Department of Medicinal Chemistry, Martin-Luther-University Halle-Wittenberg, W.-Langenbeck-Str. 4, 06120 Halle, Germany
| | - Wolfgang Sippl
- Institute of Pharmacy, Department of Medicinal Chemistry, Martin-Luther-University Halle-Wittenberg, W.-Langenbeck-Str. 4, 06120 Halle, Germany
| | - April T Davenport
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - James B Daunais
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston Salem, NC 27157, USA
| | - Lu Wang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Carmen Abate
- Dipartimento di Farmacia-Scienze Del Farmaco, Università Degli Studi di Bari ALDO MORO, Via Orabona 4, Bari 70125, Italy
| | - Hazem Ahmed
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland
| | - Ron Crowe
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Road, Atlanta, GA 30322, USA
| | - Steven H Liang
- Department of Radiology and Imaging Sciences, Emory University, 1364 Clifton Road, Atlanta, GA 30322, USA
| | - Simon M Ametamey
- Center for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Vladimir-Prelog-Weg 4, 8093 Zurich, Switzerland
| | - Bernhard Wünsch
- Institut für Pharmazeutische und Medizinische Chemie, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
| | - Ahmed Haider
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA 02114, USA
| |
Collapse
|
10
|
Ahmed H, Zheng MQ, Smart K, Fang H, Zhang L, Emery PR, Gao H, Ropchan J, Haider A, Tamagnan G, Carson RE, Ametamey SM, Huang Y. Evaluation of ( rac)-, ( R)-, and ( S)- 18F-OF-NB1 for Imaging GluN2B Subunit-Containing N-Methyl-d-Aspartate Receptors in Nonhuman Primates. J Nucl Med 2022; 63:1912-1918. [PMID: 35710735 PMCID: PMC9730915 DOI: 10.2967/jnumed.122.263977] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/07/2022] [Indexed: 01/11/2023] Open
Abstract
Despite 2 decades of research, no N-methyl-d-aspartate (NMDA) glutamate receptor (GluN) subtype 2B (GluN1/2B) radioligand is yet clinically validated. Previously, we reported on (rac)-18F-OF-NB1 as a promising GluN1/2B PET probe in rodents and its successful application for the visualization of GluN2B-containing NMDA receptors in postmortem brain tissues of patients with amyotrophic lateral sclerosis. In the current work, we report on the in vivo characterization of (rac)-, (R)-, and (S)-18F-OF-NB1 in nonhuman primates. Methods: PET scans were performed on rhesus monkeys. Plasma profiling was used to obtain the arterial input function. Regional brain time-activity curves were generated and fitted with the 1- and 2-tissue-compartment models and the multilinear analysis 1 method, and the corresponding regional volumes of distribution were calculated. Blocking studies with the GluN1/2B ligand Co 101244 (0.25 mg/kg) were performed for the enantiopure radiotracers. Receptor occupancy, nonspecific volume of distribution, and regional binding potential (BP ND) were obtained. Potential off-target binding toward σ1 receptors was assessed for (S)-18F-OF-NB1 using the σ1 receptor ligand FTC-146. Results: Free plasma fraction was moderate, ranging from 12% to 16%. All radiotracers showed high and heterogeneous brain uptake, with the highest levels in the cortex. (R)-18F-OF-NB1 showed the highest uptake and slowest washout kinetics of all tracers. The 1-tissue-compartment model and multilinear analysis 1 method fitted the regional time-activity curves well for all tracers and produced reliable regional volumes of distribution, which were higher for (R)- than (S)-18F-OF-NB1. Receptor occupancy by Co 101244 was 85% and 96% for (S)-18F-OF-NB1 and (R)-18F-OF-NB1, respectively. Pretreatment with FTC-146 at both a low (0.027 mg/kg) and high (0.125 mg/kg) dose led to a similar reduction (48% and 49%, respectively) in specific binding of (S)-18F-OF-NB1. Further, pretreatment with both Co 101244 and FTC-146 did not result in a further reduction in specific binding compared with Co 101244 alone in the same monkey (82% vs. 81%, respectively). Regional BP ND values ranged from 1.3 in the semiovale to 3.4 in the cingulate cortex for (S)-18F-OF-NB1. Conclusion: Both (R)- and (S)-18F-OF-NB1 exhibited high binding specificity to GluN2B subunit-containing NMDA receptors. The fast washout kinetics, good regional BP ND values, and high plasma free fraction render (S)-18F-OF-NB1 an attractive radiotracer for clinical translation.
Collapse
Affiliation(s)
- Hazem Ahmed
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
- PET Center, Yale University, New Haven, Connecticut; and
| | | | - Kelly Smart
- PET Center, Yale University, New Haven, Connecticut; and
| | - Hanyi Fang
- PET Center, Yale University, New Haven, Connecticut; and
- Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Li Zhang
- PET Center, Yale University, New Haven, Connecticut; and
| | - Paul R Emery
- PET Center, Yale University, New Haven, Connecticut; and
| | - Hong Gao
- PET Center, Yale University, New Haven, Connecticut; and
| | - Jim Ropchan
- PET Center, Yale University, New Haven, Connecticut; and
| | - Achi Haider
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | | | | | - Simon M Ametamey
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland;
| | - Yiyun Huang
- PET Center, Yale University, New Haven, Connecticut; and
| |
Collapse
|
11
|
Ni R, Müller Herde A, Haider A, Keller C, Louloudis G, Vaas M, Schibli R, Ametamey SM, Klohs J, Mu L. In vivo Imaging of Cannabinoid Type 2 Receptors: Functional and Structural Alterations in Mouse Model of Cerebral Ischemia by PET and MRI. Mol Imaging Biol 2022; 24:700-709. [PMID: 34642898 PMCID: PMC9581861 DOI: 10.1007/s11307-021-01655-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 09/09/2021] [Accepted: 09/14/2021] [Indexed: 02/07/2023]
Abstract
PURPOSE Stroke is one of the most prevalent vascular diseases. Non-invasive molecular imaging methods have the potential to provide critical insights into the temporal dynamics and follow alterations of receptor expression and metabolism in ischemic stroke. The aim of this study was to assess the cannabinoid type 2 receptor (CB2R) levels in transient middle cerebral artery occlusion (tMCAO) mouse models at subacute stage using positron emission tomography (PET) with our novel tracer [18F]RoSMA-18-d6 and structural imaging by magnetic resonance imaging (MRI). PROCEDURES Our recently developed CB2R PET tracer [18F]RoSMA-18-d6 was used for imaging neuroinflammation at 24 h after reperfusion in tMCAO mice. The RNA expression levels of CB2R and other inflammatory markers were analyzed by quantitative real-time polymerase chain reaction using brain tissues from tMCAO (1 h occlusion) and sham-operated mice. [18F]fluorodeoxyglucose (FDG) was included for evaluation of the cerebral metabolic rate of glucose (CMRglc). In addition, diffusion-weighted imaging and T2-weighted imaging were performed for anatomical reference and delineating the lesion in tMCAO mice. RESULTS mRNA expressions of inflammatory markers TNF-α, Iba1, MMP9 and GFAP, CNR2 were increased to 1.3-2.5 fold at 24 h after reperfusion in the ipsilateral compared to contralateral hemisphere of tMCAO mice, while mRNA expression of the neuronal marker MAP-2 was markedly reduced to ca. 50 %. Reduced [18F]FDG uptake was observed in the ischemic striatum of tMCAO mouse brain at 24 h after reperfusion. Although higher activity of [18F]RoSMA-18-d6 in ex vivo biodistribution studies and higher standard uptake value ratio (SUVR) were detected in the ischemic ipsilateral compared to contralateral striatum in tMCAO mice, the in vivo specificity of [18F]RoSMA-18-d6 was confirmed only in the CB2R-rich spleen. CONCLUSIONS This study revealed an increased [18F]RoSMA-18-d6 measure of CB2R and a reduced [18F]FDG measure of CMRglc in the ischemic striatum of tMCAO mice at subacute stage. [18F]RoSMA-18-d6 might be a promising PET tracer for detecting CB2R alterations in animal models of neuroinflammation without neuronal loss.
Collapse
Affiliation(s)
- Ruiqing Ni
- Institute for Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
- Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Adrienne Müller Herde
- Department of Chemistry and Applied Biosciences, ETH Zurich, HCI H427 Vladimir-Prelog-Weg 1-5/10, 8093, Zurich, Switzerland
| | - Achi Haider
- Department of Chemistry and Applied Biosciences, ETH Zurich, HCI H427 Vladimir-Prelog-Weg 1-5/10, 8093, Zurich, Switzerland
| | - Claudia Keller
- Department of Chemistry and Applied Biosciences, ETH Zurich, HCI H427 Vladimir-Prelog-Weg 1-5/10, 8093, Zurich, Switzerland
| | - Georgios Louloudis
- Institute for Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
| | - Markus Vaas
- Institute for Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
| | - Roger Schibli
- Department of Chemistry and Applied Biosciences, ETH Zurich, HCI H427 Vladimir-Prelog-Weg 1-5/10, 8093, Zurich, Switzerland
| | - Simon M Ametamey
- Department of Chemistry and Applied Biosciences, ETH Zurich, HCI H427 Vladimir-Prelog-Weg 1-5/10, 8093, Zurich, Switzerland
| | - Jan Klohs
- Institute for Biomedical Engineering, University of Zurich & ETH Zurich, Zurich, Switzerland
| | - Linjing Mu
- Department of Chemistry and Applied Biosciences, ETH Zurich, HCI H427 Vladimir-Prelog-Weg 1-5/10, 8093, Zurich, Switzerland.
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland.
| |
Collapse
|
12
|
Smart K, Zheng MQ, Ahmed H, Fang H, Xu Y, Cai L, Holden D, Kapinos M, Haider A, Felchner Z, Ropchan JR, Tamagnan G, Innis RB, Pike VW, Ametamey SM, Huang Y, Carson RE. Comparison of three novel radiotracers for GluN2B-containing NMDA receptors in non-human primates: (R)-[ 11C]NR2B-Me, (R)-[ 18F]of-Me-NB1, and (S)-[ 18F]of-NB1. J Cereb Blood Flow Metab 2022; 42:1398-1409. [PMID: 35209743 PMCID: PMC9274863 DOI: 10.1177/0271678x221084416] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/04/2022] [Accepted: 02/08/2022] [Indexed: 11/16/2022]
Abstract
The NMDA receptor GluN2B subunit is a target of interest in neuropsychiatric disorders but to date there is no selective radiotracer available to quantify its availability in vivo. Here we report direct comparisons in non-human primates of three GluN2B-targeting radioligands: (R)-[11C]NR2B-Me, (R)-[18F]OF-Me-NB1, and (S)-[18F]OF-NB1. Plasma free fraction, metabolism, tissue distribution and kinetics, and quantitative kinetic modeling methods and parameters were evaluated in two adult rhesus macaques. Free fraction in plasma was <2% for (R)-[11C]NR2B-Me and (R)-[18F]OF-Me-NB1 and higher for (S)-[18F]OF-NB1 (15%). All radiotracers showed good brain uptake and distribution throughout grey matter, with substantial (>68%) blockade across the brain by the GluN2B-targeting drug Co-101,244 (0.25 mg/kg), including in the cerebellum. Time-activity curves were well-fitted by the one-tissue compartment model, with volume of distribution values of 20-40 mL/cm3 for (R)-[11C]NR2B-Me, 8-16 mL/cm3 for (R)-[18F]OF-Me-NB1, and 15-35 mL/cm3 for (S)-[18F]OF-NB1. Estimates of regional non-displaceable binding potential were in the range of 2-3 for (R)-[11C]NR2B-Me and (S)-[18F]-OF-NB1, and 0.5-1 for (R)-[18F]OF-Me-NB1. Altogether, each radiotracer showed an acceptable profile for quantitative imaging of GluN2B. (S)-[18F]OF-NB1 has particularly promising imaging characteristics for potential translation into humans. However, the source of unexpected displaceable binding in the cerebellum for each of these compounds requires further investigation.
Collapse
Affiliation(s)
- Kelly Smart
- Yale School of Medicine, Yale PET Center, New Haven, Connecticut, USA
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, USA
| | - Ming-Qiang Zheng
- Yale School of Medicine, Yale PET Center, New Haven, Connecticut, USA
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, USA
| | - Hazem Ahmed
- Yale School of Medicine, Yale PET Center, New Haven, Connecticut, USA
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, USA
- Institute of Pharmaceutical Sciences, Zurich, Switzerland
| | - Hanyi Fang
- Yale School of Medicine, Yale PET Center, New Haven, Connecticut, USA
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, USA
- Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yuping Xu
- Yale School of Medicine, Yale PET Center, New Haven, Connecticut, USA
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, USA
- Jiangsu Institute of Nuclear Medicine, Jiangsu, China
| | - Lisheng Cai
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland, USA
| | - Daniel Holden
- Yale School of Medicine, Yale PET Center, New Haven, Connecticut, USA
| | - Michael Kapinos
- Yale School of Medicine, Yale PET Center, New Haven, Connecticut, USA
| | - Ahmed Haider
- Institute of Pharmaceutical Sciences, Zurich, Switzerland
| | - Zachary Felchner
- Yale School of Medicine, Yale PET Center, New Haven, Connecticut, USA
| | - Jim R Ropchan
- Yale School of Medicine, Yale PET Center, New Haven, Connecticut, USA
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, USA
| | - Gilles Tamagnan
- Yale School of Medicine, Yale PET Center, New Haven, Connecticut, USA
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland, USA
| | - Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, Maryland, USA
| | - Simon M Ametamey
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, USA
| | - Yiyun Huang
- Yale School of Medicine, Yale PET Center, New Haven, Connecticut, USA
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, USA
| | - Richard E Carson
- Yale School of Medicine, Yale PET Center, New Haven, Connecticut, USA
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, USA
| |
Collapse
|
13
|
Rischka L, Vraka C, Pichler V, Rasul S, Nics L, Gryglewski G, Handschuh P, Murgaš M, Godbersen GM, Silberbauer LR, Unterholzner J, Wotawa C, Haider A, Ahmed H, Schibli R, Mindt T, Mitterhauser M, Wadsak W, Hahn A, Lanzenberger R, Hacker M, Ametamey SM. First-in-Humans Brain PET Imaging of the GluN2B-Containing N-methyl-d-aspartate Receptor with ( R)- 11C-Me-NB1. J Nucl Med 2022; 63:936-941. [PMID: 34620732 PMCID: PMC9157734 DOI: 10.2967/jnumed.121.262427] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/26/2021] [Indexed: 11/16/2022] Open
Abstract
The N-methyl-d-aspartate receptor (NMDAR) plays a crucial role in neurodegenerative diseases such as Alzheimer disease and in the treatment of major depression by fast-acting antidepressants such as ketamine. Given their broad implications, GluN2B-containing NMDARs have been of interest as diagnostic and therapeutic targets. Recently, (R)-11C-Me-NB1 was investigated preclinically and shown to be a promising radioligand for imaging GluN2B subunits. Here, we report on the performance characteristics of this radioligand in a first-in-humans PET study. Methods: Six healthy male subjects were scanned twice on a fully integrated PET/MR scanner with (R)-11C-Me-NB1 for 120 min. Brain uptake and tracer distribution over time were investigated by SUVs. Test-retest reliability was assessed with the absolute percentage difference and the coefficient of variation. Exploratory total volumes of distribution (VT) were computed using an arterial input function and the Logan plot as well as a constrained 2-tissue-compartment model with the ratio of rate constants between plasma and tissue compartments (K1/k2) coupled (2TCM). SUV was correlated with VT to investigate its potential as a surrogate marker of GluN2B expression. Results: High and heterogeneous radioligand uptake was observed across the entire gray matter with reversible kinetics within the scan time. SUV absolute percentage difference ranged from 6.9% to 8.5% and coefficient of variation from 4.9% to 6.0%, indicating a high test-retest reliability. A moderate correlation was found between SUV averaged from 70 to 90 min and VT using Logan plot (Spearman ρ = 0.44). Correlation between VT Logan and 2TCM was r = 0.76. Conclusion: The radioligand (R)-11C-Me-NB1 was highly effective in mapping GluN2B-enriched NMDARs in the human brain. With a heterogeneous uptake and a high test-retest reliability, this radioligand offers promise to deepen our understanding of the GluN2B-containing NMDAR in the pathophysiology and treatment of neuropsychiatric disease such as Alzheimer disease and major depression. Additionally, it could help in the selection of appropriate doses of GluN2B-targeting drugs.
Collapse
Affiliation(s)
- Lucas Rischka
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Chrysoula Vraka
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Verena Pichler
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
- Department of Pharmaceutical Chemistry, University of Vienna, Vienna, Austria
| | - Sazan Rasul
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Lukas Nics
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Gregor Gryglewski
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Patricia Handschuh
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Matej Murgaš
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Godber M Godbersen
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Leo R Silberbauer
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Jakob Unterholzner
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Christoph Wotawa
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Achi Haider
- Centre for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Zurich, Switzerland
| | - Hazem Ahmed
- Centre for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Zurich, Switzerland
| | - Roger Schibli
- Centre for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Zurich, Switzerland
| | - Thomas Mindt
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria
- Institute of Inorganic Chemistry, University of Vienna, Vienna, Austria; and
| | - Markus Mitterhauser
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria
| | - Wolfgang Wadsak
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
- Center for Biomarker Research in Medicine (CBmed), Graz, Austria
| | - Andreas Hahn
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria
| | - Rupert Lanzenberger
- Department of Psychiatry and Psychotherapy, Medical University of Vienna, Vienna, Austria;
| | - Marcus Hacker
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria;
| | - Simon M Ametamey
- Centre for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Zurich, Switzerland;
| |
Collapse
|
14
|
Zheng M, Ahmed H, Smart K, Xu Y, Holden D, Kapinos M, Felchner Z, Haider A, Tamagnan G, Carson RE, Huang Y, Ametamey SM. Characterization in nonhuman primates of (R)-[ 18F]OF-Me-NB1 and (S)-[ 18F]OF-Me-NB1 for imaging the GluN2B subunits of the NMDA receptor. Eur J Nucl Med Mol Imaging 2022; 49:2153-2162. [PMID: 35107627 PMCID: PMC9165293 DOI: 10.1007/s00259-022-05698-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 01/19/2022] [Indexed: 11/04/2022]
Abstract
PURPOSE GluN2B containing N-methyl-D-aspartate receptors (NMDARs) play an essential role in neurotransmission and are a potential treatment target for multiple neurological and neurodegenerative diseases, including stroke, Alzheimer's disease, and Parkinson's disease. (R)-[18F]OF-Me-NB1 was reported to be more specific and selective than (S)-[18F]OF-Me-NB1 for the GluN2B subunits of the NMDAR based on their binding affinity to GluN2B and sigma-1 receptors. Here we report a comprehensive evaluation of (R)-[18F]OF-Me-NB1 and (S)-[18F]OF-Me-NB1 in nonhuman primates. METHODS The radiosynthesis of (R)-[18F]OF-Me-NB1 and (S)-[18F]OF-Me-NB1 started from 18F-fluorination of the boronic ester precursor, followed by removal of the acetyl protecting group. PET scans in two rhesus monkeys were conducted on the Focus 220 scanner. Blocking studies were performed after treatment of the animals with the GluN2B antagonist Co101,244 or the sigma-1 receptor antagonist FTC-146. One-tissue compartment (1TC) model and multilinear analysis-1 (MA1) method with arterial input function were used to obtain the regional volume of distribution (VT, mL/cm3). Occupancy values by the two blockers were obtained by the Lassen plot. Regional non-displaceable binding potential (BPND) was calculated from the corresponding baseline VT and the VND derived from the occupancy plot of the Co101,244 blocking scans. RESULTS (R)- and (S)-[18F]OF-Me-NB1 were produced in > 99% radiochemical and enantiomeric purity, with molar activity of 224.22 ± 161.69 MBq/nmol at the end of synthesis (n = 10). Metabolism was moderate, with ~ 30% parent compound remaining for (R)-[18F]OF-Me-NB1 and 20% for (S)-[18F]OF-Me-NB1 at 30 min postinjection. Plasma free fraction was 1-2%. In brain regions, both (R)- and (S)-[18F]OF-Me-NB1 displayed fast uptake with slower clearance for the (R)- than (S)-enantiomer. For (R)-[18F]OF-Me-NB1, both the 1TC model and MA1 method gave reliable estimates of regional VT values, with MA1 VT (mL/cm3) values ranging from 8.9 in the cerebellum to 12.8 in the cingulate cortex. Blocking with 0.25 mg/kg of Co101,244 greatly reduced the uptake of (R)-[18F]OF-Me-NB1 across all brain regions, resulting in occupancy of 77% and VND of 6.36, while 0.027 mg/kg of FTC-146 reduced specific binding by 30%. Regional BPND, as a measure of specific binding signals, ranged from 0.40 in the cerebellum to 1.01 in the cingulate cortex. CONCLUSIONS In rhesus monkeys, (R)-[18F]OF-Me-NB1 exhibited fast kinetics and heterogeneous uptake across brain regions, while the (S)-enantiomer displayed a narrower dynamic range of uptake across regions. A Blocking study with a GluN2B antagonist indicated binding specificity. The value of BPND was > 0.5 in most brain regions, suggesting good in vivo specific binding signals. Taken together, results from the current study demonstrated the potential of (R)-[18F]OF-Me-NB1 as a useful radiotracer for imaging the GluN2B receptors.
Collapse
Affiliation(s)
| | - Hazem Ahmed
- PET Center, Yale University, New Haven, CT, USA
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Kelly Smart
- PET Center, Yale University, New Haven, CT, USA
| | - Yuping Xu
- PET Center, Yale University, New Haven, CT, USA
- Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China
| | | | | | | | - Achi Haider
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | | | | | - Yiyun Huang
- PET Center, Yale University, New Haven, CT, USA.
| | - Simon M Ametamey
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
15
|
He Y, Schild M, Grether U, Benz J, Leibrock L, Heer D, Topp A, Collin L, Kuhn B, Wittwer M, Keller C, Gobbi LC, Schibli R, Mu L. Development of High Brain-Penetrant and Reversible Monoacylglycerol Lipase PET Tracers for Neuroimaging. J Med Chem 2022; 65:2191-2207. [PMID: 35089028 DOI: 10.1021/acs.jmedchem.1c01706] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Monoacylglycerol lipase (MAGL) is one of the key enzymes in the endocannabinoid system. Inhibition of MAGL has been proposed as an attractive approach for the treatment of various diseases. In this study, we designed and successfully synthesized two series of piperazinyl pyrrolidin-2-one derivatives as novel reversible MAGL inhibitors. (R)-[18F]13 was identified through the preliminary evaluation of two carbon-11-labeled racemic structures [11C]11 and [11C]16. In dynamic positron-emission tomography (PET) scans, (R)-[18F]13 showed a heterogeneous distribution and matched the MAGL expression pattern in the mouse brain. High brain uptake and brain-to-blood ratio were achieved by (R)-[18F]13 in comparison with previously reported reversible MAGL PET radiotracers. Target occupancy studies with a therapeutic MAGL inhibitor revealed a dose-dependent reduction of (R)-[18F]13 accumulation in the mouse brain. These findings indicate that (R)-[18F]13 ([18F]YH149) is a highly promising PET probe for visualizing MAGL non-invasively in vivo and holds great potential to support drug development.
Collapse
Affiliation(s)
- Yingfang He
- Center for Radiopharmaceutical Sciences, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, CH-8093 Zurich, Switzerland
| | - Matthias Schild
- Center for Radiopharmaceutical Sciences, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, CH-8093 Zurich, Switzerland
| | - Uwe Grether
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Jörg Benz
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Lea Leibrock
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Dominik Heer
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Andreas Topp
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Ludovic Collin
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Bernd Kuhn
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Matthias Wittwer
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Claudia Keller
- Center for Radiopharmaceutical Sciences, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, CH-8093 Zurich, Switzerland
| | - Luca C Gobbi
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, CH-4070 Basel, Switzerland
| | - Roger Schibli
- Center for Radiopharmaceutical Sciences, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, CH-8093 Zurich, Switzerland
| | - Linjing Mu
- Center for Radiopharmaceutical Sciences, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, CH-8093 Zurich, Switzerland.,Department of Nuclear Medicine, University Hospital Zurich, CH-8091 Zurich, Switzerland
| |
Collapse
|
16
|
Ni R. Positron Emission Tomography in Animal Models of Alzheimer's Disease Amyloidosis: Translational Implications. Pharmaceuticals (Basel) 2021; 14:1179. [PMID: 34832961 PMCID: PMC8623863 DOI: 10.3390/ph14111179] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/13/2021] [Accepted: 11/15/2021] [Indexed: 12/30/2022] Open
Abstract
Animal models of Alzheimer's disease amyloidosis that recapitulate cerebral amyloid-beta pathology have been widely used in preclinical research and have greatly enabled the mechanistic understanding of Alzheimer's disease and the development of therapeutics. Comprehensive deep phenotyping of the pathophysiological and biochemical features in these animal models is essential. Recent advances in positron emission tomography have allowed the non-invasive visualization of the alterations in the brain of animal models and in patients with Alzheimer's disease. These tools have facilitated our understanding of disease mechanisms and provided longitudinal monitoring of treatment effects in animal models of Alzheimer's disease amyloidosis. In this review, we focus on recent positron emission tomography studies of cerebral amyloid-beta accumulation, hypoglucose metabolism, synaptic and neurotransmitter receptor deficits (cholinergic and glutamatergic system), blood-brain barrier impairment, and neuroinflammation (microgliosis and astrocytosis) in animal models of Alzheimer's disease amyloidosis. We further propose the emerging targets and tracers for reflecting the pathophysiological changes and discuss outstanding challenges in disease animal models and future outlook in the on-chip characterization of imaging biomarkers towards clinical translation.
Collapse
Affiliation(s)
- Ruiqing Ni
- Institute for Biomedical Engineering, ETH & University of Zurich, 8093 Zurich, Switzerland;
- Institute for Regenerative Medicine, University of Zurich, 8952 Zurich, Switzerland
| |
Collapse
|
17
|
Takamura Y, Kakuta H. In Vivo Receptor Visualization and Evaluation of Receptor Occupancy with Positron Emission Tomography. J Med Chem 2021; 64:5226-5251. [PMID: 33905258 DOI: 10.1021/acs.jmedchem.0c01714] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Positron emission tomography (PET) is useful for noninvasive in vivo visualization of disease-related receptors, for evaluation of receptor occupancy to determine an appropriate drug dosage, and for proof-of-concept of drug candidates in translational research. For these purposes, the specificity of the PET tracer for the target receptor is critical. Here, we review work in this area, focusing on the chemical structures of reported PET tracers, their Ki/Kd values, and the physical properties relevant to target receptor selectivity. Among these physical properties, such as cLogP, cLogD, molecular weight, topological polar surface area, number of hydrogen bond donors, and pKa, we focus especially on LogD and LogP as important physical properties that can be easily compared across a range of studies. We discuss the success of PET tracers in evaluating receptor occupancy and consider likely future developments in the field.
Collapse
Affiliation(s)
- Yuta Takamura
- Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 1-1-1, Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| | - Hiroki Kakuta
- Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 1-1-1, Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| |
Collapse
|
18
|
Sun JY, Kumata K, Chen Z, Zhang YD, Chen JH, Hatori A, Fu HL, Rong J, Deng XY, Yamasaki T, Xie L, Hu K, Fujinaga M, Yu QZ, Shao T, Collier TL, Josephson L, Shao YH, Du YF, Wang L, Xu H, Zhang MR, Liang SH. Synthesis and preliminary evaluation of novel 11C-labeled GluN2B-selective NMDA receptor negative allosteric modulators. Acta Pharmacol Sin 2021; 42:491-498. [PMID: 32661351 PMCID: PMC8027431 DOI: 10.1038/s41401-020-0456-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/03/2020] [Indexed: 12/30/2022]
Abstract
N-methyl-D-aspartate receptors (NMDARs) play critical roles in the physiological function of the mammalian central nervous system (CNS), including learning, memory, and synaptic plasticity, through modulating excitatory neurotransmission. Attributed to etiopathology of various CNS disorders and neurodegenerative diseases, GluN2B is one of the most well-studied subtypes in preclinical and clinical studies on NMDARs. Herein, we report the synthesis and preclinical evaluation of two 11C-labeled GluN2B-selective negative allosteric modulators (NAMs) containing N,N-dimethyl-2-(1H-pyrrolo[3,2-b]pyridin-1-yl)acetamides for positron emission tomography (PET) imaging. Two PET ligands, namely [11C]31 and [11C]37 (also called N2B-1810 and N2B-1903, respectively) were labeled with [11C]CH3I in good radiochemical yields (decay-corrected 28% and 32% relative to starting [11C]CO2, respectively), high radiochemical purity (>99%) and high molar activity (>74 GBq/μmol). In particular, PET ligand [11C]31 demonstrated moderate specific binding to GluN2B subtype by in vitro autoradiography studies. However, because in vivo PET imaging studies showed limited brain uptake of [11C]31 (up to 0.5 SUV), further medicinal chemistry and ADME optimization are necessary for this chemotype attributed to low binding specificity and rapid metabolism in vivo.
Collapse
Affiliation(s)
- Ji-Yun Sun
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| | - Katsushi Kumata
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Zhen Chen
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| | - Yi-Ding Zhang
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Jia-Hui Chen
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| | - Akiko Hatori
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Hua-Long Fu
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| | - Jian Rong
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| | - Xiao-Yun Deng
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| | - Tomoteru Yamasaki
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Lin Xie
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Kuan Hu
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Masayuki Fujinaga
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Qing-Zhen Yu
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| | - Tuo Shao
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| | - Thomas Lee Collier
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| | - Lee Josephson
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| | - Yi-Han Shao
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, 73019, USA
| | - Yun-Fei Du
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, 300072, China
| | - Lu Wang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou, 510630, China
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA
| | - Hao Xu
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou, 510630, China.
| | - Ming-Rong Zhang
- Department of Advanced Nuclear Medicine Sciences, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan.
| | - Steven H Liang
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, MA, 02114, USA.
| |
Collapse
|
19
|
Ahmed H, Wallimann R, Haider A, Hosseini V, Gruber S, Robledo M, Nguyen TAN, Herde AM, Iten I, Keller C, Vogel V, Schibli R, Wünsch B, Mu L, Ametamey SM. Preclinical Development of 18F-OF-NB1 for Imaging GluN2B-Containing N-Methyl-d-Aspartate Receptors and Its Utility as a Biomarker for Amyotrophic Lateral Sclerosis. J Nucl Med 2021; 62:259-265. [PMID: 32737247 DOI: 10.2967/jnumed.120.246785] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 06/26/2020] [Indexed: 01/05/2023] Open
Abstract
As part of our continuous efforts to develop a suitable 18F-labeled PET radioligand with improved characteristics for imaging the N-methyl-d-aspartate receptors (NMDARs) subtype 2B (GluN1/2B), we investigated in the current work ortho-fluorinated (OF) and meta-fluorinated (MF) analogs of 18F-para-fluorinated (PF)-NB1, a 3-benzazepine-based radiofluorinated probe. Methods: OF-NB1 and MF-NB1 were prepared using a multistep synthesis, and their binding affinities toward GluN2B subunits and selectivity over σ1 receptors (σ1Rs) were determined via competitive binding assays. 18F-OF-NB1 was synthesized via copper-mediated radiofluorination and was evaluated in Wistar rats by in vitro autoradiography, PET imaging, ex vivo biodistribution, metabolite experiments, and receptor occupancy studies using CP-101,606, an established GluN2B antagonist. To determine in vivo selectivity, 18F-OF-NB1 was validated in wild-type and σ1R knock-out mice. Translational relevance was assessed in autoradiographic studies using postmortem human brain tissues from healthy individuals and ALS patients, the results of which were corroborated by immunohistochemistry. Results: The binding affinity values for OF-NB1 and MF-NB1 toward the GluN2B subunits were 10.4 ± 4.7 and 590 ± 36 nM, respectively. For σ1R binding, OF-NB1 and MF-NB1 exhibited inhibition constants of 410 and 2,700 nM, respectively. OF-NB1, which outperformed MF-NB1, was radiolabeled with 18F to afford 18F-OF-NB1 in more than 95% radiochemical purity and molar activities of 192 ± 33 GBq/μmol. In autoradiography experiments, 18F-OF-NB1 displayed a heterogeneous and specific binding in GluN2B subunit-rich brain regions such as the cortex, striatum, hypothalamus, and hippocampus. PET imaging studies in Wistar rats showed a similar heterogeneous uptake, and no brain radiometabolites were detected. A dose-dependent blocking effect was observed with CP-101,606 (0.5-15 mg/kg) and resulted in a 50% receptor occupancy of 8.1 μmol/kg. Postmortem autoradiography results revealed lower expression of the GluN2B subunits in ALS brain tissue sections than in healthy controls, in line with immunohistochemistry results. Conclusion:18F-OF-NB1 is a highly promising PET probe for imaging the GluN2B subunits of the N-methyl-d-aspartate receptor. It possesses utility for receptor occupancy studies and has potential for PET imaging studies in ALS patients and possibly other brain disorders.
Collapse
Affiliation(s)
- Hazem Ahmed
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Rahel Wallimann
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Achi Haider
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Vahid Hosseini
- Laboratory of Applied Mechanobiology, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
| | - Stefan Gruber
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Marvin Robledo
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Thi A N Nguyen
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | | | - Irina Iten
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Claudia Keller
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Viola Vogel
- Laboratory of Applied Mechanobiology, Institute of Translational Medicine, Department of Health Sciences and Technology, ETH Zürich, Zurich, Switzerland
| | - Roger Schibli
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland; and
| | - Bernhard Wünsch
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Münster, Germany
| | - Linjing Mu
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland; and
| | - Simon M Ametamey
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
20
|
Goud NS, Bhattacharya A, Joshi RK, Nagaraj C, Bharath RD, Kumar P. Carbon-11: Radiochemistry and Target-Based PET Molecular Imaging Applications in Oncology, Cardiology, and Neurology. J Med Chem 2021; 64:1223-1259. [PMID: 33499603 DOI: 10.1021/acs.jmedchem.0c01053] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The positron emission tomography (PET) molecular imaging technique has gained its universal value as a remarkable tool for medical diagnosis and biomedical research. Carbon-11 is one of the promising radiotracers that can report target-specific information related to its pharmacology and physiology to understand the disease status. Currently, many of the available carbon-11 (t1/2 = 20.4 min) PET radiotracers are heterocyclic derivatives that have been synthesized using carbon-11 inserted different functional groups obtained from primary and secondary carbon-11 precursors. A spectrum of carbon-11 PET radiotracers has been developed against many of the upregulated and emerging targets for the diagnosis, prognosis, prediction, and therapy in the fields of oncology, cardiology, and neurology. This review focuses on the carbon-11 radiochemistry and various target-specific PET molecular imaging agents used in tumor, heart, brain, and neuroinflammatory disease imaging along with its associated pathology.
Collapse
Affiliation(s)
- Nerella Sridhar Goud
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Ahana Bhattacharya
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Raman Kumar Joshi
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Chandana Nagaraj
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Rose Dawn Bharath
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| | - Pardeep Kumar
- Department of Neuroimaging and Interventional Radiology (NIIR), National Institute of Mental Health and Neuro Sciences (NIMHANS), Bengaluru 560 029, India
| |
Collapse
|
21
|
He Y, Whitehead DM, Briard E, Numao S, Mu L, Schibli R, Ametamey SM, Auberson YP. Evaluation of 5H-Thiazolo[3,2-α]pyrimidin-5-ones as Potential GluN2A PET Tracers. ChemMedChem 2020; 15:2448-2461. [PMID: 32544308 DOI: 10.1002/cmdc.202000340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Indexed: 11/10/2022]
Abstract
We describe here our efforts to develop a PET tracer for imaging GluN2A-containing NMDA receptors, based on a 5H-thiazolo[3,2-α]pyrimidin-5-one scaffold. The metabolic stability and overall properties could be optimized satisfactorily, although binding affinities remained a limiting factor for in vivo imaging. We nevertheless identified 7-(((2-fluoroethyl)(3-fluorophenyl)amino)-methyl)-3-(2-(hydroxymethyl)cyclopropyl)-2-methyl-5H-thiazolo-[3,2-α]pyrimidin-5-one ([18 F]7b) as a radioligand providing good-quality images in autoradiographic studies, as well as a tritiated derivative, 2-(7-(((2-fluoroethyl)(4-fluorophenyl)amino)methyl)-2-methyl-5-oxo-5H-thiazolo[3,2-α]pyrimidin-3-yl)cyclopropane-1-carbonitrile ([3 H2 ]15b), which was used for the successful development of a radioligand binding assay. These are valuable new tools for the study of GluN2A-containing NMDA receptors, and for the optimization of allosteric modulators binding to the pharmacophore located at the dimer interface of the GluN1-GluN2A ligand-binding domain.
Collapse
Affiliation(s)
- Yingfang He
- Department of Chemistry and Applied Biosciences ETH Zürich, Vladimir-Prelog-Weg 4, 8093, Zürich, Switzerland
| | - David M Whitehead
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Fabrikstrasse 2, 4056, Basel, Switzerland
| | - Emmanuelle Briard
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Fabrikstrasse 2, 4056, Basel, Switzerland
| | - Shin Numao
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, Fabrikstrasse 2, 4056, Basel, Switzerland
| | - Linjing Mu
- Department of Chemistry and Applied Biosciences ETH Zürich, Vladimir-Prelog-Weg 4, 8093, Zürich, Switzerland
| | - Roger Schibli
- Department of Chemistry and Applied Biosciences ETH Zürich, Vladimir-Prelog-Weg 4, 8093, Zürich, Switzerland
| | - Simon M Ametamey
- Department of Chemistry and Applied Biosciences ETH Zürich, Vladimir-Prelog-Weg 4, 8093, Zürich, Switzerland
| | - Yves P Auberson
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Fabrikstrasse 2, 4056, Basel, Switzerland
| |
Collapse
|
22
|
Kim JH, Marton J, Ametamey SM, Cumming P. A Review of Molecular Imaging of Glutamate Receptors. Molecules 2020; 25:molecules25204749. [PMID: 33081223 PMCID: PMC7587586 DOI: 10.3390/molecules25204749] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 12/22/2022] Open
Abstract
Molecular imaging with positron emission tomography (PET) and single photon emission computed tomography (SPECT) is a well-established and important in vivo technique to evaluate fundamental biological processes and unravel the role of neurotransmitter receptors in various neuropsychiatric disorders. Specific ligands are available for PET/SPECT studies of dopamine, serotonin, and opiate receptors, but corresponding development of radiotracers for receptors of glutamate, the main excitatory neurotransmitter in mammalian brain, has lagged behind. This state of affairs has persisted despite the central importance of glutamate neurotransmission in brain physiology and in disorders such as stroke, epilepsy, schizophrenia, and neurodegenerative diseases. Recent years have seen extensive efforts to develop useful ligands for molecular imaging of subtypes of the ionotropic (N-methyl-D-aspartate (NMDA), kainate, and AMPA/quisqualate receptors) and metabotropic glutamate receptors (types I, II, and III mGluRs). We now review the state of development of radioligands for glutamate receptor imaging, placing main emphasis on the suitability of available ligands for reliable in vivo applications. We give a brief account of the radiosynthetic approach for selected molecules. In general, with the exception of ligands for the GluN2B subunit of NMDA receptors, there has been little success in developing radiotracers for imaging ionotropic glutamate receptors; failure of ligands for the PCP/MK801 binding site in vivo doubtless relates their dependence on the open, unblocked state of the ion channel. Many AMPA and kainite receptor ligands with good binding properties in vitro have failed to give measurable specific binding in the living brain. This may reflect the challenge of developing brain-penetrating ligands for amino acid receptors, compounded by conformational differences in vivo. The situation is better with respect to mGluR imaging, particularly for the mGluR5 subtype. Several successful PET ligands serve for investigations of mGluRs in conditions such as schizophrenia, depression, substance abuse and aging. Considering the centrality and diversity of glutamatergic signaling in brain function, we have relatively few selective and sensitive tools for molecular imaging of ionotropic and metabotropic glutamate receptors. Further radiopharmaceutical research targeting specific subtypes and subunits of the glutamate receptors may yet open up new investigational vistas with broad applications in basic and clinical research.
Collapse
Affiliation(s)
- Jong-Hoon Kim
- Neuroscience Research Institute, Gachon University, Incheon 21565, Korea
- Gachon Advanced Institute for Health Science and Technology, Graduate School, Incheon 21565, Korea
- Department of Psychiatry, Gil Medical Center, Gachon University College of Medicine, Gachon University, Incheon 21565, Korea
- Correspondence: (J.-H.K.); (P.C.); Tel.: +41-31-664-0498 (P.C.); Fax: +41-31-632-7663 (P.C.)
| | - János Marton
- ABX Advanced Biochemical Compounds, Biomedizinische Forschungsreagenzien GmbH, Heinrich-Glaeser-Strasse 10-14, D-1454 Radeberg, Germany;
| | - Simon Mensah Ametamey
- Centre for Radiopharmaceutical Sciences ETH-PSI-USZ, Institute of Pharmaceutical Sciences ETH, Vladimir-Prelog-Weg 4, CH-8093 Zürich, Switzerland;
| | - Paul Cumming
- Department of Nuclear Medicine, University of Bern, Inselspital, Freiburgstrasse 18, CH-3010 Bern, Switzerland
- School of Psychology and Counselling, Queensland University of Technology, Brisbane QLD 4059, Australia
- Correspondence: (J.-H.K.); (P.C.); Tel.: +41-31-664-0498 (P.C.); Fax: +41-31-632-7663 (P.C.)
| |
Collapse
|
23
|
Ahmed H, Haider A, Ametamey SM. N-Methyl-D-Aspartate (NMDA) receptor modulators: a patent review (2015-present). Expert Opin Ther Pat 2020; 30:743-767. [PMID: 32926646 DOI: 10.1080/13543776.2020.1811234] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION - The NMDA receptor is implicated in various diseases including neurodegenerative, neurodevelopmental and mood disorders. However, only a limited number of clinically approved NMDA receptor modulators are available. Today, apparent NMDA receptor drug development strategies entail 1) exploring the unknown chemical space to identify novel scaffolds; 2) using the clinically available NMDA receptor modulators to expand the therapeutic indication space; 3) and to trace physiological functions of the NMDA receptor. AREAS COVERED - The current review reflects on the functional and pharmacological facets of NMDA receptors and the current clinical status quo of NMDA receptor modulators. Patent literature covering 2015 till April 2020 is discussed with emphasis on new indications. EXPERT OPINION - Supporting evidence shows that subtype-selective NMDA receptor antagonists show an improved safety profile compared to broad-spectrum channel blockers. Although GluN2B-selective antagonists are by far the most extensively investigated subtype-selective modulators, they have shown only modest clinical efficacy so far. To overcome the limitations that have hampered the clinical development of previous subtype-selective NMDA receptor antagonists, future studies with improved animal models that better reflect human NMDA receptor pathophysiology are warranted. The increased availability of subtype-selective probes will allow target engagement studies and proper dose finding in future clinical trials.
Collapse
Affiliation(s)
- Hazem Ahmed
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich , Zurich, Switzerland
| | - Ahmed Haider
- Department of Nuclear Medicine, University Hospital Zurich , Zurich, Switzerland.,Center for Molecular Cardiology, University of Zurich , Schlieren, Switzerland
| | - Simon M Ametamey
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich , Zurich, Switzerland
| |
Collapse
|
24
|
van der Aart J, Yaqub M, Kooijman EJM, Bakker J, Langermans JAM, Schuit RC, Hofman MBM, Christiaans JAM, Lammertsma AA, Windhorst AD, van Berckel BNM. Evaluation of the Novel PET Tracer [ 11C]HACH242 for Imaging the GluN2B NMDA Receptor in Non-Human Primates. Mol Imaging Biol 2020; 21:676-685. [PMID: 30306318 DOI: 10.1007/s11307-018-1284-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
PURPOSE There are currently no positron emission tomography (PET) radiotracers for the GluN2B (NR2B) binding sites of brain N-methyl-D-aspartate (NMDA) receptors. In rats, the GluN2B antagonist Ro25-6981 reduced the binding of N-((5-(4-fluoro-2-[11C]methoxyphenyl)pyridin-3-yl)methyl)cyclopentanamin ([11C]HACH242). This paper reports the evaluation of [11C]HACH242 PET in non-human primates at baseline and following administration of the GluN2B negative allosteric modulator radiprodil. PROCEDURES Eight 90-min dynamic [11C]HACH242 PET scans were acquired in three male anaesthetised rhesus monkeys, including a retest session of subject 1, at baseline and 10 min after intravenous 10 mg/kg radiprodil. Standardised uptake values (SUV) were calculated for 9 brain regions. Arterial blood samples were taken at six timepoints to characterise pharmacokinetics in blood and plasma. Reliable input functions for kinetic modelling could not be generated due to variability in the whole-blood radioactivity measurements. RESULTS [11C]HACH242 entered the brain and displayed fairly uniform uptake. The mean (± standard deviation, SD) Tmax was 17 ± 7 min in baseline scans and 24 ± 15 min in radiprodil scans. The rate of radioligand metabolism in plasma (primarily to polar metabolites) was high, with mean parent fractions of 26 ± 10 % at 20 min and 8 ± 5 % at 85 min. Radiprodil increased [11C]HACH242 whole-brain SUV in the last PET frame by 25 %, 1 %, 3 and 17 % for subjects 1, 2, 3 and retest of subject 1, respectively. The mean brain to plasma ratio was 5.4 ± 2.6, and increased by 39 to 110 % in the radiprodil condition, partly due to lower parent plasma radioactivity of -11 to -56 %. CONCLUSIONS The present results show that [11C]HACH242 has a suitable kinetic profile in the brain and low accumulation of lipophilic radiometabolites. Radiprodil did not consistently change [11C]HACH242 brain uptake. These findings may be explained by variations in cerebral blood flow, a low fraction of specifically bound tracer, or interactions with endogenous NMDA receptor ligands at the binding site. Further experiments of ligand interactions are necessary to facilitate the development of radiotracers for in vivo imaging of the ionotropic NMDA receptor.
Collapse
Affiliation(s)
- Jasper van der Aart
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands. .,Centre for Human Drug Research, Leiden, The Netherlands.
| | - Maqsood Yaqub
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands
| | - Esther J M Kooijman
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands
| | - Jaco Bakker
- Animal Science Department, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Jan A M Langermans
- Animal Science Department, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Robert C Schuit
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands
| | - Mark B M Hofman
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands
| | - Johannes A M Christiaans
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands
| | - Adriaan A Lammertsma
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands
| | - Albert D Windhorst
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands
| | - Bart N M van Berckel
- Department of Radiology & Nuclear Medicine, Amsterdam UMC, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Thum S, Schepmann D, Reinoso RF, Alvarez I, Ametamey SM, Wünsch B. Synthesis and pharmacological evaluation of fluorinated benzo[7]annulen-7-amines as GluN2B-selective NMDA receptor antagonists. J Labelled Comp Radiopharm 2020; 62:354-379. [PMID: 30850991 DOI: 10.1002/jlcr.3718] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/24/2019] [Accepted: 02/28/2019] [Indexed: 01/30/2023]
Abstract
Because of their neuroprotective potential, GluN2B-selective ligands are of great interest for the treatment of various neurological and neurodegenerative disorders. Fluorinated benzo[7]annulen-7-amines, capable for PET, were synthesized by combining fluorinated phenylalkylamines with differently substituted ketones. Relationships between substitution pattern and GluN2B affinity as well as selectivity towards σ1 and σ2 receptors were investigated. Two promising ligands (18a and 20c) were selected for further pharmacological evaluation. Besides a slight serotonin transporter (SERT), norepinephrine transporter (NET), and hERG affinity, they did not show interaction with other targets. Furthermore, the pKa value of a set fluorinated ligands, bearing the fluorine atom in different positions, was determined.
Collapse
Affiliation(s)
- Simone Thum
- Institute of Pharmaceutical and Medicinal Chemistry, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Dirk Schepmann
- Institute of Pharmaceutical and Medicinal Chemistry, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | | | | | - Simon M Ametamey
- Center for Radiopharmaceutical Science of ETH, PSI, and USZ, ETH-Hönggerberg, Zürich, Switzerland
| | - Bernhard Wünsch
- Institute of Pharmaceutical and Medicinal Chemistry, Westfälische Wilhelms-Universität Münster, Münster, Germany.,Cells-in-Motion Cluster of Excellence (EXC 1003-CiM), Westfälische Wilhelms-Universität Münster, Germany
| |
Collapse
|
26
|
Cai L, Liow JS, Morse CL, Telu S, Davies R, Frankland MP, Zoghbi SS, Cheng K, Hall MD, Innis RB, Pike VW. Evaluation of 11C-NR2B-SMe and Its Enantiomers as PET Radioligands for Imaging the NR2B Subunit Within the NMDA Receptor Complex in Rats. J Nucl Med 2020; 61:1212-1220. [PMID: 31924728 DOI: 10.2967/jnumed.119.235143] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 12/19/2019] [Indexed: 01/01/2023] Open
Abstract
[S-methyl-11C](±)-7-methoxy-3-(4-(4-(methylthio)phenyl)butyl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-ol (11C-NR2B-SMe) and its enantiomers were synthesized as candidates for imaging the NR2B subunit within the N-methyl-d-aspartate receptor with PET. Methods: Brains were scanned with PET for 90 min after intravenous injection of one of the candidate radioligands into rats. To detect any NR2B-specific binding of radioligand in brain, various preblocking or displacing agents were evaluated for their impact on the PET brain imaging data. Radiometabolites from brain and other tissues were measured ex vivo and in vitro. Results: Each radioligand gave high early whole-brain uptake of radioactivity, followed by a brief fast decline and then a slow final decline. 11C-(S)-NR2B-SMe was studied extensively. Ex vivo measurements showed that radioactivity in rat brain at 30 min after radioligand injection was virtually unchanged radioligand. Only less lipophilic radiometabolites appeared in plasma. High-affinity NR2B ligands, Ro-25-6981, ifenprodil, and CO101244, showed increasing preblocking of whole-brain radioactivity retention with increasing dose (0.01-3.00 mg/kg, intravenously). Five σ1 antagonists (FTC146, BD1407, F3, F4, and NE100) and 4 σ1 agonists ((+)-pentazocine, (±)-PPCC, PRE-084, and (+)-SKF10047) were ineffective preblocking agents, except FTC146 and F4 at a high dose. Two potent σ1 receptor agonists, TC1 and SA4503, showed dose-dependent preblocking effects in the presence or absence of pharmacologic σ1 receptor blockade with FTC146. Conclusion: 11C-(S)-NR2B-SMe has adequate NR2B-specific PET signal in rat brain to warrant further evaluation in higher species. TC1 and SA4503 likely have off-target binding to NR2B in vivo.
Collapse
Affiliation(s)
- Lisheng Cai
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland; and
| | - Jeih-San Liow
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland; and
| | - Cheryl L Morse
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland; and
| | - Sanjay Telu
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland; and
| | - Riley Davies
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland; and
| | - Michael P Frankland
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland; and
| | - Sami S Zoghbi
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland; and
| | - Ken Cheng
- NCATS Chemical Genomics Center, National Institutes of Health, Rockville, Maryland
| | - Matthew D Hall
- NCATS Chemical Genomics Center, National Institutes of Health, Rockville, Maryland
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland; and
| | - Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland; and
| |
Collapse
|
27
|
Ahmed H, Haider A, Varisco J, Stanković M, Wallimann R, Gruber S, Iten I, Häne S, Müller Herde A, Keller C, Schibli R, Schepmann D, Mu L, Wünsch B, Ametamey SM. Structure-Affinity Relationships of 2,3,4,5-Tetrahydro-1 H-3-benzazepine and 6,7,8,9-Tetrahydro-5 H-benzo[7]annulen-7-amine Analogues and the Discovery of a Radiofluorinated 2,3,4,5-Tetrahydro-1 H-3-benzazepine Congener for Imaging GluN2B Subunit-Containing N-Methyl-d-aspartate Receptors. J Med Chem 2019; 62:9450-9470. [PMID: 31657559 DOI: 10.1021/acs.jmedchem.9b00812] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Aspiring to develop a positron emission tomography (PET) imaging agent for the GluN2B subunits of the N-methyl-d-aspartate receptor (NMDAR), a key therapeutic target for drug development toward several neurological disorders, we synthesized a series of 2,3,4,5-tetrahydro-1H-3-benzazepine and 6,7,8,9-tetrahydro-5H-benzo[7]annulen-7-amine analogues. After in vitro testing via competition binding assay and autoradiography, [18F]PF-NB1 emerged as the best performing tracer with respect to specificity and selectivity over σ1 and σ2 receptors and was thus selected for further in vivo evaluation. Copper-mediated radiofluorination was accomplished in good radiochemical yields and high molar activities. Extensive in vivo characterization was performed in Wistar rats comprising PET imaging, biodistribution, receptor occupancy, and metabolites studies. [18F]PF-NB1 binding was selective to GluN2B-rich forebrain regions and was specifically blocked by the GluN2B antagonist, CP-101,606, in a dose-dependent manner with no brain radiometabolites. [18F]PF-NB1 is a promising fluorine-18 PET tracer for imaging the GluN2B subunits of the NMDAR and has utility for receptor occupancy studies.
Collapse
Affiliation(s)
- Hazem Ahmed
- Institute of Pharmaceutical Sciences , ETH Zurich , Vladimir-Prelog-Weg 4 , 8093 Zurich , Switzerland
| | - Achi Haider
- Institute of Pharmaceutical Sciences , ETH Zurich , Vladimir-Prelog-Weg 4 , 8093 Zurich , Switzerland
| | - Jasmine Varisco
- Institute of Pharmaceutical Sciences , ETH Zurich , Vladimir-Prelog-Weg 4 , 8093 Zurich , Switzerland
| | - Maja Stanković
- Institute of Pharmaceutical Sciences , ETH Zurich , Vladimir-Prelog-Weg 4 , 8093 Zurich , Switzerland
| | - Rahel Wallimann
- Institute of Pharmaceutical Sciences , ETH Zurich , Vladimir-Prelog-Weg 4 , 8093 Zurich , Switzerland
| | - Stefan Gruber
- Institute of Pharmaceutical Sciences , ETH Zurich , Vladimir-Prelog-Weg 4 , 8093 Zurich , Switzerland
| | - Irina Iten
- Institute of Pharmaceutical Sciences , ETH Zurich , Vladimir-Prelog-Weg 4 , 8093 Zurich , Switzerland
| | - Surya Häne
- Institute of Pharmaceutical Sciences , ETH Zurich , Vladimir-Prelog-Weg 4 , 8093 Zurich , Switzerland
| | - Adrienne Müller Herde
- Institute of Pharmaceutical Sciences , ETH Zurich , Vladimir-Prelog-Weg 4 , 8093 Zurich , Switzerland
| | - Claudia Keller
- Institute of Pharmaceutical Sciences , ETH Zurich , Vladimir-Prelog-Weg 4 , 8093 Zurich , Switzerland
| | - Roger Schibli
- Institute of Pharmaceutical Sciences , ETH Zurich , Vladimir-Prelog-Weg 4 , 8093 Zurich , Switzerland
- Department of Nuclear Medicine , University Hospital Zurich , 8091 Zurich , Switzerland
| | - Dirk Schepmann
- Institute of Pharmaceutical and Medicinal Chemistry , University of Münster , Corrensstr. 48 , 48149 Münster , Germany
| | - Linjing Mu
- Institute of Pharmaceutical Sciences , ETH Zurich , Vladimir-Prelog-Weg 4 , 8093 Zurich , Switzerland
- Department of Nuclear Medicine , University Hospital Zurich , 8091 Zurich , Switzerland
| | - Bernhard Wünsch
- Institute of Pharmaceutical and Medicinal Chemistry , University of Münster , Corrensstr. 48 , 48149 Münster , Germany
| | - Simon M Ametamey
- Institute of Pharmaceutical Sciences , ETH Zurich , Vladimir-Prelog-Weg 4 , 8093 Zurich , Switzerland
| |
Collapse
|
28
|
Classification of Visual Cortex Plasticity Phenotypes following Treatment for Amblyopia. Neural Plast 2019; 2019:2564018. [PMID: 31565045 PMCID: PMC6746165 DOI: 10.1155/2019/2564018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 04/04/2019] [Accepted: 05/13/2019] [Indexed: 12/25/2022] Open
Abstract
Monocular deprivation (MD) during the critical period (CP) has enduring effects on visual acuity and the functioning of the visual cortex (V1). This experience-dependent plasticity has become a model for studying the mechanisms, especially glutamatergic and GABAergic receptors, that regulate amblyopia. Less is known, however, about treatment-induced changes to those receptors and if those changes differentiate treatments that support the recovery of acuity versus persistent acuity deficits. Here, we use an animal model to explore the effects of 3 visual treatments started during the CP (n = 24, 10 male and 14 female): binocular vision (BV) that promotes good acuity versus reverse occlusion (RO) and binocular deprivation (BD) that causes persistent acuity deficits. We measured the recovery of a collection of glutamatergic and GABAergic receptor subunits in the V1 and modeled recovery of kinetics for NMDAR and GABAAR. There was a complex pattern of protein changes that prompted us to develop an unbiased data-driven approach for these high-dimensional data analyses to identify plasticity features and construct plasticity phenotypes. Cluster analysis of the plasticity phenotypes suggests that BV supports adaptive plasticity while RO and BD promote a maladaptive pattern. The RO plasticity phenotype appeared more similar to adults with a high expression of GluA2, and the BD phenotypes were dominated by GABAA α1, highlighting that multiple plasticity phenotypes can underlie persistent poor acuity. After 2-4 days of BV, the plasticity phenotypes resembled normals, but only one feature, the GluN2A:GluA2 balance, returned to normal levels. Perhaps, balancing Hebbian (GluN2A) and homeostatic (GluA2) mechanisms is necessary for the recovery of vision.
Collapse
|
29
|
Thum S, Schepmann D, Ayet E, Pujol M, Nieto FR, Ametamey SM, Wünsch B. Tetrahydro-3-benzazepines with fluorinated side chains as NMDA and σ 1 receptor antagonists: Synthesis, receptor affinity, selectivity and antiallodynic activity. Eur J Med Chem 2019; 177:47-62. [PMID: 31129453 DOI: 10.1016/j.ejmech.2019.05.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/23/2019] [Accepted: 05/12/2019] [Indexed: 11/24/2022]
Abstract
The class of tetrahydro-1H-3-benzazepines was systematically modified in 1-, 3- and 7-position. In particular, a F-atom was introduced in β- or γ-position of the 4-phenylbutyl side chain in 3-position. Ligands with the F-atom in γ-position possess higher GluN2B affinity than analogs bearing the F-atom in β-position. This effect was attributed to the reduced basicity of β-fluoro amines. 3-Benzazepines with a benzylic OH moiety show moderate GluN2B affinity, but considerable selectivity over the σ2 receptor. However, removal of the benzylic OH moiety led to increased GluN2B affinity, but reduced GluN2B/σ2 selectivity. With respect to GluN2B affinity the phenol 17b with a γ-fluorophenylbutyl moiety in 3-position represents the most interesting fluorinated ligand (Ki(GluN2B) = 16 nM). Most of the synthesized ligands reveal either similar GluN2B and σ1 affinity or higher σ1 affinity than GluN2B affinity. The methyl ether 16b shows high σ1 affinity (Ki(σ1) = 6.6 nM) and high selectivity over a broad panel of receptors and transporters. The high antiallodynic activity in the mouse capsaicin assay proved the σ1 antagonistic activity of 16b.
Collapse
Affiliation(s)
- Simone Thum
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
| | - Dirk Schepmann
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster, Corrensstraße 48, D-48149 Münster, Germany
| | - Eva Ayet
- Esteve Pharmaceuticals S.A, Baldiri Reixach 4-8, 08028 Barcelona, Spain
| | - Marta Pujol
- Esteve Pharmaceuticals S.A, Baldiri Reixach 4-8, 08028 Barcelona, Spain
| | - Francisco R Nieto
- Department of Pharmacology and Institute of Neuroscience, School of Medicine and Biomedical Research Center, University of Granada, Avenida de la Investigación 11, 18016 Granada, Spain
| | - Simon M Ametamey
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Bernhard Wünsch
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster, Corrensstraße 48, D-48149 Münster, Germany; Cells-in-Motion Cluster of Excellence (EXC 1003-CiM), Westfälische Wilhelms-Universität Münster, Germany.
| |
Collapse
|
30
|
Haider A, Herde AM, Krämer SD, Varisco J, Keller C, Frauenknecht K, Auberson YP, Temme L, Robaa D, Sippl W, Schibli R, Wünsch B, Mu L, Ametamey SM. Preclinical Evaluation of Benzazepine-Based PET Radioligands ( R)- and ( S)- 11C-Me-NB1 Reveals Distinct Enantiomeric Binding Patterns and a Tightrope Walk Between GluN2B- and σ 1-Receptor-Targeted PET Imaging. J Nucl Med 2019; 60:1167-1173. [PMID: 30683765 PMCID: PMC6681693 DOI: 10.2967/jnumed.118.221051] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/14/2018] [Indexed: 01/15/2023] Open
Abstract
The study aims to investigate the performance characteristics of the enantiomers of 11C-Me-NB1, a recently reported PET imaging probe that targets the GluN2B subunit of N-methyl-d-aspartate (NMDA) receptors. Methods: Reference compound Me-NB1 (inhibition constant for hGluN1/GluN2B, 5.4 nM) and the phenolic precursor were prepared via multistep synthesis. Following chiral resolution by high-performance liquid chromatography, enantiopure precursor compounds, (R)-NB1 and (S)-NB1, were labeled with 11C and validated in rodents using in vitro/ex vivo autoradiography, PET experiments, and dose-response studies. To illustrate the translational relevance, (R)-11C-Me-NB1 was validated in autoradiographic studies using postmortem human GluN2B-rich cortical and GluN2B-deficient cerebellar brain slices. To determine target engagement, receptor occupancy was assessed at different plasma concentrations of CP101,606, a GluN2B receptor antagonist. Results: The radiosynthesis of (R)- and (S)-11C-Me-NB1 was accomplished in 42% ± 9% (decay-corrected) radiochemical yields. Molar activity ranged from 40 to 336 GBq/μmol, and an excellent radiochemical purity of greater than 99% was achieved. Although (R)-11C-Me-NB1 displayed heterogeneous accumulation with high selectivity for the GluN2B-rich forebrain, (S)-11C-Me-NB1 revealed a homogeneous distribution across all brain regions in rodent brain autoradiograms and predominantly exhibited σ1-receptor binding. Similar to rodent brain, (R)-11C-Me-NB1 showed in postmortem human brain tissues higher binding in the cortex than in the cerebellum. Coincubation of the GluN2B-antagonist CERC-301 (1 μM) reduced cortical but not cerebellar binding, demonstrating the specificity of (R)-11C-Me-NB1 binding to the human GluN2B-containing NMDA receptor. In vivo specificity of (R)-11C-Me-NB1 in the GluN2B-expressing cortex, striatum, thalamus, and hippocampus was demonstrated by PET imaging in rodents. Applying GluN2B-antagonist eliprodil, an evident dose-response behavior was observed with (R)-11C-Me-NB1 but not with (S)-11C-Me-NB1. Our findings further underline the tightrope walk between GluN2B- and σ1-receptor-targeted imaging, illustrated by the entirely different receptor binding behavior of the 2 radioligand enantiomers. Conclusion: (R)-11C-Me-NB1 is a highly selective and specific PET radioligand for imaging the GluN2B subunit of the NMDA receptor. The entirely different receptor binding behavior of (R)-11C-Me-NB1 and (S)-11C-Me-NB1 raises awareness of a delicate balance that is underlying the selective targeting of either GluN2B-carrying NMDA or σ1-receptors.
Collapse
Affiliation(s)
- Achi Haider
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | | | - Stefanie D Krämer
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Jasmine Varisco
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Claudia Keller
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Katrin Frauenknecht
- Institute of Neuropathology, University of Zurich/University Hospital Zurich, Zurich, Switzerland
| | - Yves P Auberson
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Louisa Temme
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Münster, Germany
| | - Dina Robaa
- Institute of Pharmacy, Martin-Luther University of Halle-Wittenberg, Halle/Saale, Germany; and
| | - Wolfgang Sippl
- Institute of Pharmacy, Martin-Luther University of Halle-Wittenberg, Halle/Saale, Germany; and
| | - Roger Schibli
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Bernhard Wünsch
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Münster, Germany
| | - Linjing Mu
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland
| | - Simon M Ametamey
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
31
|
Jakobsson J, Gourni E, Khanapur S, Brito B, Riss PJ. Synthesis and Characterization in Rodent Brain of the Subtype-Selective NR2B NMDA Receptor Ligand [ 11C]Ro04-5595 as a Potential Radiotracer for Positron Emission Tomography. ACS OMEGA 2019; 4:9925-9931. [PMID: 31460083 PMCID: PMC6648642 DOI: 10.1021/acsomega.9b00357] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 04/22/2019] [Indexed: 06/10/2023]
Abstract
The NR2B subunit of the N-methyl-d-aspartate (NMDA) receptor has been implicated in controlling synaptic plasticity, memory, and learning. Herein, we describe an 11C-labeled PET radiotracer based on 1-(4-chlorophenethyl)-6-methoxy-2-methyl-1,2,3,4-tetrahydroisoquinolin-7-ol, Ro04-5595. The radiotracer was evaluated in rats using PET. The PET study showed a good pharmacokinetic profile with rapid uptake and washout over 90 min. Complementary high-resolution autoradiographic images using [3H]Ro04-5595 demonstrated strong binding in NR2B receptor-rich regions and low binding in cerebellum where NR2B concentration is low. We conclude to have developed a selective NR2B receptor radioligand suitable for quantitative and qualitative imaging of a NR2B receptor distribution in vitro and in vivo.
Collapse
Affiliation(s)
- Jimmy
E. Jakobsson
- Realomics
SRI, Kjemisk Institutt, Universitetet i
Oslo, Sem Sælands vei 26, Kjemibygningen, 0371 Oslo, Norway
| | - Eleni Gourni
- Realomics
SRI, Kjemisk Institutt, Universitetet i
Oslo, Sem Sælands vei 26, Kjemibygningen, 0371 Oslo, Norway
| | - Shivashankar Khanapur
- Realomics
SRI, Kjemisk Institutt, Universitetet i
Oslo, Sem Sælands vei 26, Kjemibygningen, 0371 Oslo, Norway
| | - Beatriz Brito
- Realomics
SRI, Kjemisk Institutt, Universitetet i
Oslo, Sem Sælands vei 26, Kjemibygningen, 0371 Oslo, Norway
| | - Patrick J. Riss
- Realomics
SRI, Kjemisk Institutt, Universitetet i
Oslo, Sem Sælands vei 26, Kjemibygningen, 0371 Oslo, Norway
- Klinik
for Kirurgi og Nevrofag, Oslo Universitets
Sykehus HF-Rikshospitalet, Postboks 4950
Nydalen, 0424 Oslo, Norway
- Norsk
Medisinsk Syklotronsenter AS, Gaustad, Postboks 4950 Nydalen, 0424 Oslo, Norway
| |
Collapse
|
32
|
Fu H, Tang W, Chen Z, Belov VV, Zhang G, Shao T, Zhang X, Yu Q, Rong J, Deng X, Han W, Myers SJ, Giffenig P, Wang L, Josephson L, Shao Y, Davenport AT, Daunais JB, Papisov M, Yuan H, Li Z, Traynelis SF, Liang SH. Synthesis and Preliminary Evaluations of a Triazole-Cored Antagonist as a PET Imaging Probe ([ 18F]N2B-0518) for GluN2B Subunit in the Brain. ACS Chem Neurosci 2019; 10:2263-2275. [PMID: 30698943 PMCID: PMC6727982 DOI: 10.1021/acschemneuro.8b00591] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
GluN2B is the most studied subunit of N-methyl-d-aspartate receptors (NMDARs) and implicated in the pathologies of various central nervous system disorders and neurodegenerative diseases. As pan NMDAR antagonists often produce debilitating side effects, new approaches in drug discovery have shifted to subtype-selective NMDAR modulators, especially GluN2B-selective antagonists. While positron emission tomography (PET) studies of GluN2B-selective NMDARs in the living brain would enable target engagement in drug development and improve our understanding in the NMDAR signaling pathways between normal and disease conditions, a suitable PET ligand is yet to be identified. Herein we developed an 18F-labeled potent antagonist, 2-((1-(4-[18F]fluoro-3-methylphenyl)-1 H-1,2,3-triazol-4-yl)methoxy)-5-methoxypyrimidine ([18F]13; also called [18F]N2B-0518) as a PET tracer for imaging the GluN2B subunit. The radiofluorination of [18F]13 was efficiently achieved by our spirocyclic iodonium ylide (SCIDY) method. In in vitro autoradiography studies, [18F]13 displayed highly region-specific binding in brain sections of rat and nonhuman primate, which was in accordance with the expression of GluN2B subunit. Ex vivo biodistribution in mice revealed that [18F]13 could penetrate the blood-brain barrier with moderate brain uptake (3.60% ID/g at 2 min) and rapid washout. Altogether, this work provides a GluN2B-selective PET tracer bearing a new chemical scaffold and shows high specificity to GluN2B subunit in vitro, which may pave the way for the development of a new generation of GluN2B PET ligands.
Collapse
Affiliation(s)
- Hualong Fu
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States
| | - Weiting Tang
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, P. R. China
| | - Zhen Chen
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States
| | - Vasily V. Belov
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, and the Shriners Burns Hospital, Boston, Massachusetts 02114, United States
| | - Genwei Zhang
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Tuo Shao
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States
| | - Xiaofei Zhang
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States
| | - Qingzhen Yu
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States
| | - Jian Rong
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States
| | - Xiaoyun Deng
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States
| | - Wei Han
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
- Department of Neurology, Children’s Hospital of Chongqing Medical University, Chongqing, 400014, P. R. China
| | - Scott J. Myers
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Pilar Giffenig
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, and the Shriners Burns Hospital, Boston, Massachusetts 02114, United States
| | - Lu Wang
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States
- Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University & Institute of Molecular and Functional Imaging, Jinan University, Guangzhou 510630, P. R. China
| | - Lee Josephson
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States
| | - Yihan Shao
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - April T. Davenport
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston Salem, North Carolina 27157, United States
| | - James B. Daunais
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston Salem, North Carolina 27157, United States
| | - Mikhail Papisov
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, and the Shriners Burns Hospital, Boston, Massachusetts 02114, United States
| | - Hongjie Yuan
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Zijing Li
- State Key Laboratory of Molecular Vaccinology, Molecular Diagnosis & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, P. R. China
| | - Stephen F. Traynelis
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322, United States
| | - Steven H. Liang
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States
| |
Collapse
|
33
|
Börgel F, Galla F, Lehmkuhl K, Schepmann D, Ametamey SM, Wünsch B. Pharmacokinetic properties of enantiomerically pure GluN2B selective NMDA receptor antagonists with 3-benzazepine scaffold. J Pharm Biomed Anal 2019; 172:214-222. [PMID: 31060034 DOI: 10.1016/j.jpba.2019.04.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 04/11/2019] [Accepted: 04/15/2019] [Indexed: 01/12/2023]
Abstract
Recently, the eutomers of highly potent GluN2B selective NMDA receptor antagonists with 3-benzazepine scaffold were identified. Herein, pharmacokinetic properties regarding lipophilicity, plasma protein binding (PPB) and metabolism are analyzed. The logD7.4 values of 1.68 for phenol 1 and 2.46 for methyl ether 2 are in a very good range for CNS agents. A very similar logD7.4 value was recorded for the prototypical GluN2B antagonist ifenprodil (logD7.4 = 1.49). The herein developed high performance affinity chromatography (HPAC) method using human serum albumin as stationary phase led to PPB of 3-benzazepines (R)-1-3 and (S)-1-3 of 76-98%. Upon incubation with mouse liver microsomes, (R)-1-3 and (S)-1-3 showed moderate to high metabolic stability. The (R)-configured eutomers turned out to be metabolically more stable than their (S)-configured distomers. During phase I metabolism of 3-benzazepines 1-3 hydroxylations at both aromatic rings, the aliphatic side chain and the seven-membered ring were observed. O-demethylation of methyl ether (S)-2 was faster than O-demethylation of its enantiomer (R)-2. In phase I biotransformation the phenol eutomer (R)-1 showed comparable stability as ifenprodil. In phase II biotransformation, glucuronidation of the phenolic (only 1) and benzylic hydroxy groups was observed. Both enantiomers formed the same type of metabolites, respectively, but in different amounts. Whereas, the benzylic hydroxy group of (R)-2 was glucuronidated preferably, predominant benzylic glucuronidation of (S)-3 was detected. Mouse liver microsomes produced the glucuronide of phenol 1 (main metabolite) in larger amounts than rat liver microsomes.
Collapse
Affiliation(s)
- Frederik Börgel
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster, Corrensstraße 48, D-48149, Münster, Germany
| | - Fabian Galla
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster, Corrensstraße 48, D-48149, Münster, Germany
| | - Kirstin Lehmkuhl
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster, Corrensstraße 48, D-48149, Münster, Germany
| | - Dirk Schepmann
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster, Corrensstraße 48, D-48149, Münster, Germany
| | - Simon M Ametamey
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Bernhard Wünsch
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster, Corrensstraße 48, D-48149, Münster, Germany; Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), Westfälische Wilhelms-Universität, Münster, Germany.
| |
Collapse
|
34
|
Fu H, Chen Z, Josephson L, Li Z, Liang SH. Positron Emission Tomography (PET) Ligand Development for Ionotropic Glutamate Receptors: Challenges and Opportunities for Radiotracer Targeting N-Methyl-d-aspartate (NMDA), α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid (AMPA), and Kainate Receptors. J Med Chem 2019; 62:403-419. [PMID: 30110164 PMCID: PMC6393217 DOI: 10.1021/acs.jmedchem.8b00714] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Ionotropic glutamate receptors (iGluRs) mediate excitatory neurotransmission within the mammalian central nervous system. iGluRs exist as three main groups: N-methyl-d-aspartate receptors (NMDARs), α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs), and kainate receptors. The past decades have witnessed a remarkable development of PET tracers targeting different iGluRs including NMDARs and AMPARs, and several of the tracers have advanced to clinical imaging studies. Here, we assess the recent development of iGluR PET probes, focusing on tracer design, brain kinetics, and performance in PET imaging studies. Furthermore, this review will not only present challenges in the tracer development but also provide novel approaches in conjunction with most recent drug discovery efforts on these iGluRs, including subtype-selective NMDAR and transmembrane AMPAR regulatory protein modulators and positive allosteric modulators (PAMs) of AMPARs. These approaches, if successful as PET tracers, may provide fundamental knowledge to understand the roles of iGluR receptors under physiological and pathological conditions.
Collapse
Affiliation(s)
- Hualong Fu
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St., Boston, MA 02114 USA
| | - Zhen Chen
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St., Boston, MA 02114 USA
| | - Lee Josephson
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St., Boston, MA 02114 USA
| | - Zijing Li
- State Key Laboratory of Molecular Vaccinology, Molecular Diagnosis & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, P. R. China
| | - Steven H. Liang
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital and Harvard Medical School, 55 Fruit St., Boston, MA 02114 USA
| |
Collapse
|
35
|
Haider A, Iten I, Ahmed H, Herde AM, Gruber S, Krämer SD, Keller C, Schibli R, Wünsch B, Mu L, Ametamey SM. Identification and Preclinical Evaluation of a Radiofluorinated Benzazepine Derivative for Imaging the GluN2B Subunit of the Ionotropic NMDA Receptor. J Nucl Med 2018; 60:jnumed.118.212134. [PMID: 30030340 PMCID: PMC8833860 DOI: 10.2967/jnumed.118.212134] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 07/04/2018] [Indexed: 01/06/2023] Open
Abstract
The previously reported carbon-11 labeled GluN2B PET radioligand 11C-Me-NB1 served as a starting point for derivatization and led to the successful development of a radiofluorinated analogue designated (R)-18F-OF-Me-NB1. Given the short physical half-life of 20.3 min for carbon-11, (R)-18F-OF-Me-NB1 with a physical half-life of 109.8 min would allow satellite distribution to nuclear medicine facilities without an on-site cyclotron. Methods: Two fluorinated Me-NB1 derivatives, OF-Me-NB1 and PF-Me-NB1, were synthesized. Upon chiral resolution, the respective enantiomers were radiolabeled with carbon-11 and assessed in a proof-of-concept study by applying in vitro autoradiography on rodent brain sections. Based on the autoradiograms, (R)-OF-Me-NB1 was selected for radiofluorination and preclinical evaluation by ex vivo autoradiography, PET imaging, biodistribution and metabolite studies in Wistar rats. To rule out off-target binding to the σ1 receptor, the brain uptake of (R)-18F-OF-Me-NB1 in wild-type mice was compared with σ1 receptor knock-out mice. Results: Autoradiographic assessment revealed that both enantiomers of 11C-PF-Me-NB1 distributed homogenously across all brain regions on rodent brain sections. In contrast, the two enantiomers of 11C-OF-Me-NB1 exhibited an entirely different behaviour. While (S)-11C-OF-Me-NB1 bound virtually to all brain regions with considerable σ1 receptor binding, (R)-11C-OF-Me-NB1 exhibited high selectivity and specificity for the GluN2B-rich rat forebrain. These findings were confirmed for the radiofluorinated analogue (R)-11C-OF-Me-NB1, which was obtained via copper-mediated radiofluorination in radiochemical yields of 13-25% and molar activities ranging from 61-168 GBq/µmol. PET imaging and biodistribution studies in Wistar rats indicated appropriate pharmacokinetic profile and high in vivo specific binding of (R)-18F-OF-Me-NB1 as revealed by blocking studies with GluN2B-antagonist CP101,606. Off-target binding to the σ1 receptor was excluded by PET imaging with σ1 receptor knock-out mice. Receptor occupancy experiments with CP101,606 revealed a D50-value of 8.3 µmol/kg (intravenous). Conclusion: (R)-18F-OF-Me-NB1 is a promising radiofluorinated probe that exhibits specificity and selectivity for the GluN2B-containing N-methyl-D-aspartate (NMDA) complex and enables in vivo target occupancy studies in rodents.
Collapse
Affiliation(s)
- Ahmed Haider
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Irina Iten
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Hazem Ahmed
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | | | - Stefan Gruber
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | | | - Claudia Keller
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Roger Schibli
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland; and
| | - Bernhard Wünsch
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Münster, Germany
| | - Linjing Mu
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
- Department of Nuclear Medicine, University Hospital Zurich, Zurich, Switzerland; and
| | - Simon M. Ametamey
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
36
|
Börgel F, Szermerski M, Schreiber JA, Temme L, Strutz‐Seebohm N, Lehmkuhl K, Schepmann D, Ametamey SM, Seebohm G, Schmidt TJ, Wünsch B. Synthesis and Pharmacological Evaluation of Enantiomerically Pure GluN2B Selective NMDA Receptor Antagonists. ChemMedChem 2018; 13:1580-1587. [DOI: 10.1002/cmdc.201800214] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 05/16/2018] [Indexed: 11/10/2022]
Affiliation(s)
- Frederik Börgel
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster Corrensstraße 48 48149 Münster Germany
| | - Marina Szermerski
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster Corrensstraße 48 48149 Münster Germany
| | - Julian A. Schreiber
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster Corrensstraße 48 48149 Münster Germany
| | - Louisa Temme
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster Corrensstraße 48 48149 Münster Germany
| | - Nathalie Strutz‐Seebohm
- Myocellular Electrophysiology and Molecular Biology Institute for Genetics of Heart Diseases (IfGH) Department of Cardiovascular Medicine University Hospital Münster 48149 Münster Germany
| | - Kirstin Lehmkuhl
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster Corrensstraße 48 48149 Münster Germany
| | - Dirk Schepmann
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster Corrensstraße 48 48149 Münster Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003—CiM) Westfälische Wilhelms-Universität Münster Münster Germany
| | - Simon M. Ametamey
- Department of Chemistry and Applied Biosciences Institute of Pharmaceutical Sciences ETH Zürich Zürich Switzerland
| | - Guiscard Seebohm
- Myocellular Electrophysiology and Molecular Biology Institute for Genetics of Heart Diseases (IfGH) Department of Cardiovascular Medicine University Hospital Münster 48149 Münster Germany
| | - Thomas J. Schmidt
- Institut für Pharmazeutische Biologie und Phytochemie der Universität Münster Corrensstraße 48 48149 Münster Germany
| | - Bernhard Wünsch
- Institut für Pharmazeutische und Medizinische Chemie der Universität Münster Corrensstraße 48 48149 Münster Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003—CiM) Westfälische Wilhelms-Universität Münster Münster Germany
| |
Collapse
|
37
|
Auberson YP, Briard E, Rudolph B, Kaupmann K, Smith P, Oberhauser B. PET Imaging of T Cells: Target Identification and Feasibility Assessment. ChemMedChem 2018; 13:1566-1579. [DOI: 10.1002/cmdc.201800241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 05/23/2018] [Indexed: 01/21/2023]
Affiliation(s)
- Yves P. Auberson
- Global Discovery Chemistry Novartis Institutes for BioMedical Research 141 Klybeckstrasse 4057 Basel Switzerland
| | - Emmanuelle Briard
- Global Discovery Chemistry Novartis Institutes for BioMedical Research 141 Klybeckstrasse 4057 Basel Switzerland
| | - Bettina Rudolph
- Translational Medicine, Pharmacokinetics Sciences Novartis Institutes for BioMedical Research Novartis Campus 4056 Basel Switzerland
| | - Klemens Kaupmann
- Autoimmunity, Transplantation & Inflammation Novartis Institutes for BioMedical Research Novartis Campus 4056 Basel Switzerland
| | - Paul Smith
- Autoimmunity, Transplantation & Inflammation Novartis Institutes for BioMedical Research Novartis Campus 4056 Basel Switzerland
| | - Berndt Oberhauser
- Global Discovery Chemistry Novartis Institutes for BioMedical Research 141 Klybeckstrasse 4057 Basel Switzerland
| |
Collapse
|
38
|
Taddio MF, Mu L, Keller C, Schibli R, Krämer SD. Physiologically Based Pharmacokinetic Modelling with Dynamic PET Data to Study the In Vivo Effects of Transporter Inhibition on Hepatobiliary Clearance in Mice. CONTRAST MEDIA & MOLECULAR IMAGING 2018; 2018:5849047. [PMID: 29967572 PMCID: PMC6008768 DOI: 10.1155/2018/5849047] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 02/20/2018] [Indexed: 01/16/2023]
Abstract
Physiologically based pharmacokinetic modelling (PBPK) is a powerful tool to predict in vivo pharmacokinetics based on physiological parameters and data from in vivo studies and in vitro assays. In vivo PBPK modelling in laboratory animals by noninvasive imaging could help to improve the in vivo-in vivo translation towards human pharmacokinetics modelling. We evaluated the feasibility of PBPK modelling with PET data from mice. We used data from two of our PET tracers under development, [11C]AM7 and [11C]MT107. PET images suggested hepatobiliary excretion which was reduced after cyclosporine administration. We fitted the time-activity curves of blood, liver, gallbladder/intestine, kidney, and peripheral tissue to a compartment model and compared the resulting pharmacokinetic parameters under control conditions ([11C]AM7 n = 2; [11C]MT107, n = 4) and after administration of cyclosporine ([11C]MT107, n = 4). The modelling revealed a significant reduction in [11C]MT107 hepatobiliary clearance from 35.2 ± 10.9 to 17.1 ± 5.6 μl/min after cyclosporine administration. The excretion profile of [11C]MT107 was shifted from predominantly hepatobiliary (CLH/CLR = 3.8 ± 3.0) to equal hepatobiliary and renal clearance (CLH/CLR = 0.9 ± 0.2). Our results show the potential of PBPK modelling for characterizing the in vivo effects of transporter inhibition on whole-body and organ-specific pharmacokinetics.
Collapse
Affiliation(s)
- Marco F. Taddio
- Radiopharmaceutical Science and Biopharmacy, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Linjing Mu
- Department of Nuclear Medicine, University Hospital Zurich, Switzerland
| | - Claudia Keller
- Radiopharmaceutical Science and Biopharmacy, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Roger Schibli
- Radiopharmaceutical Science and Biopharmacy, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Stefanie D. Krämer
- Radiopharmaceutical Science and Biopharmacy, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
39
|
Szermerski M, Börgel F, Schepmann D, Haider A, Betzel T, Ametamey SM, Wünsch B. Fluorinated GluN2B Receptor Antagonists with a 3-Benzazepine Scaffold Designed for PET Studies. ChemMedChem 2018; 13:1058-1068. [PMID: 29522653 DOI: 10.1002/cmdc.201700819] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 02/05/2018] [Indexed: 11/08/2022]
Abstract
To analyze the N-methyl-d-aspartate (NMDA) receptor distribution in the central nervous system, fluorinated ligands that selectively address the ifenprodil binding site of GluN2B-subunit-containing NMDA receptors were developed. Various strategies to introduce a fluorine atom into the potent GluN2B ligand 2 (3-(4-phenylbutyl)-2,3,4,5-tetrahydro-1H-3-benzazepin-1,7-diol) were pursued, including replacement of the benzylic OH moiety with a fluorine atom (13) and introduction of fluoroethoxy moieties at various positions (14 (7-position), 17 (9-position), 18a-c (1-position)). With respect to GluN2B affinity and selectivity over related receptors, the fluoroethoxy derivatives 14 and 18a are the most promising ligands. Radiosynthesis of fluoroethoxy derivative [18 F]14 was performed by nucleophilic substitution of the phenol 2 with 2-[18 F]fluoroethyl tosylate. On rat brain slices the fluorinated PET tracer [18 F]14 accumulated in regions with high density of NMDA receptors containing GluN2B subunits. The bound radioactivity could not be replaced by (S)-glutamate. However, the GluN2B ligands eliprodil, Ro 25-6981, and the non-labeled 3-benzazepine 14 were able to abolish the specific binding of [18 F]14.
Collapse
Affiliation(s)
- Marina Szermerski
- Institut für Pharmazeutische und Medizinische Chemie der, Universität Münster, Corrensstraße 48, 48149, Münster, Germany
| | - Frederik Börgel
- Institut für Pharmazeutische und Medizinische Chemie der, Universität Münster, Corrensstraße 48, 48149, Münster, Germany
| | - Dirk Schepmann
- Institut für Pharmazeutische und Medizinische Chemie der, Universität Münster, Corrensstraße 48, 48149, Münster, Germany
| | - Achi Haider
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Thomas Betzel
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Simon M Ametamey
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Bernhard Wünsch
- Institut für Pharmazeutische und Medizinische Chemie der, Universität Münster, Corrensstraße 48, 48149, Münster, Germany
- Cells-in-Motion Cluster of Excellence (EXC 1003-CiM), Westfälische Wilhelms-Universität, Münster, Germany
| |
Collapse
|
40
|
Warnock G, Sommerauer M, Mu L, Pla Gonzalez G, Geistlich S, Treyer V, Schibli R, Buck A, Krämer SD, Ametamey SM. A first-in-man PET study of [ 18F]PSS232, a fluorinated ABP688 derivative for imaging metabotropic glutamate receptor subtype 5. Eur J Nucl Med Mol Imaging 2017; 45:1041-1051. [PMID: 29177707 DOI: 10.1007/s00259-017-3879-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 11/06/2017] [Indexed: 12/21/2022]
Abstract
PURPOSE Non-invasive imaging of metabotropic glutamate receptor 5 (mGlu5) in the brain using PET is of interest in e.g., anxiety, depression, and Parkinson's disease. Widespread application of the most widely used mGlu5 tracer, [11C]ABP688, is limited by the short physical half-life of carbon-11. [18F]PSS232 is a fluorinated analog with promising preclinical properties and high selectivity and specificity for mGlu5. In this first-in-man study, we evaluated the brain uptake pattern and kinetics of [18F]PSS232 in healthy volunteers. METHODS [18F]PSS232 PET was performed with ten healthy male volunteers aged 20-40 years. Seven of the subjects received a bolus injection and the remainder a bolus/infusion protocol. Cerebral blood flow was determined in seven subjects using [15O]water PET. Arterial blood activity was measured using an online blood counter. Tracer kinetics were evaluated by compartment modeling and parametric maps were generated for both tracers. RESULTS At 90 min post-injection, 59.2 ± 11.1% of total radioactivity in plasma corresponded to intact tracer. The regional first pass extraction fraction of [18F]PSS232 ranged from 0.41 ± 0.06 to 0.55 ± 0.03 and brain distribution pattern matched that of [11C]ABP688. Uptake kinetics followed a simple two-tissue compartment model. The volume of distribution of total tracer (V T, ml/cm3) ranged from 1.18 ± 0.20 for white matter to 2.91 ± 0.51 for putamen. The respective mean distribution volume ratios (DVR) with cerebellum as the reference tissue were 0.88 ± 0.06 and 2.12 ± 0.10, respectively. The tissue/cerebellum ratios of a bolus/infusion protocol (30/70 dose ratio) were close to the DVR values. CONCLUSIONS Brain uptake of [18F]PSS232 matched the distribution of mGlu5 and followed a two-tissue compartment model. The well-defined kinetics and the possibility to use reference tissue models, obviating the need for arterial blood sampling, make [18F]PSS232 a promising fluorine-18 labeled radioligand for measuring mGlu5 density in humans.
Collapse
Affiliation(s)
- Geoffrey Warnock
- Department of Nuclear Medicine, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Michael Sommerauer
- Department of Nuclear Medicine, University Hospital Zurich, 8091, Zurich, Switzerland.,Department of Neurology, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Linjing Mu
- Department of Nuclear Medicine, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Gloria Pla Gonzalez
- Radiopharmaceutical Science, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog Weg 4, 8093, Zurich, Switzerland
| | - Susanne Geistlich
- Radiopharmaceutical Science, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog Weg 4, 8093, Zurich, Switzerland
| | - Valerie Treyer
- Department of Nuclear Medicine, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Roger Schibli
- Radiopharmaceutical Science, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog Weg 4, 8093, Zurich, Switzerland
| | - Alfred Buck
- Department of Nuclear Medicine, University Hospital Zurich, 8091, Zurich, Switzerland
| | - Stefanie D Krämer
- Radiopharmaceutical Science, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog Weg 4, 8093, Zurich, Switzerland
| | - Simon M Ametamey
- Radiopharmaceutical Science, Institute of Pharmaceutical Sciences, Department of Chemistry and Applied Biosciences, ETH Zurich, Vladimir-Prelog Weg 4, 8093, Zurich, Switzerland.
| |
Collapse
|