1
|
Recent advances in radiotracers targeting norepinephrine transporter: structural development and radiolabeling improvements. J Neural Transm (Vienna) 2020; 127:851-873. [PMID: 32274584 PMCID: PMC7223405 DOI: 10.1007/s00702-020-02180-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 03/21/2020] [Indexed: 12/13/2022]
Abstract
The norepinephrine transporter (NET) is a major target for the evaluation of the cardiac sympathetic nerve system in patients with heart failure and Parkinson's disease. It is also used in the therapeutic applications against certain types of neuroendocrine tumors, as exemplified by the clinically used 123/131I-MIBG as theranostic single-photon emission computed tomography (SPECT) agent. With the development of more advanced positron emission tomography (PET) technology, more radiotracers targeting NET have been reported, with superior temporal and spatial resolutions, along with the possibility of functional and kinetic analysis. More recently, fluorine-18-labelled NET tracers have drawn increasing attentions from researchers, due to their longer radiological half-life relative to carbon-11 (110 min vs. 20 min), reduced dependence on on-site cyclotrons, and flexibility in the design of novel tracer structures. In the heart, certain NET tracers provide integral diagnostic information on sympathetic innervation and the nerve status. In the central nervous system, such radiotracers can reveal NET distribution and density in pathological conditions. Most radiotracers targeting cardiac NET-function for the cardiac application consistent of derivatives of either norepinephrine or MIBG with its benzylguanidine core structure, e.g. 11C-HED and 18F-LMI1195. In contrast, all NET tracers used in central nervous system applications are derived from clinically used antidepressants. Lastly, possible applications of NET as selective tracers over organic cation transporters (OCTs) in the kidneys and other organs controlled by sympathetic nervous system will also be discussed.
Collapse
|
2
|
Ermert J, Benešová M, Hugenberg V, Gupta V, Spahn I, Pietzsch HJ, Liolios C, Kopka K. Radiopharmaceutical Sciences. Clin Nucl Med 2020. [DOI: 10.1007/978-3-030-39457-8_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
3
|
Central noradrenaline transporter availability in highly obese, non-depressed individuals. Eur J Nucl Med Mol Imaging 2017; 44:1056-1064. [PMID: 28066877 DOI: 10.1007/s00259-016-3590-3] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 11/28/2016] [Indexed: 10/20/2022]
Abstract
PURPOSE The brain noradrenaline (NA) system plays an important role in the central nervous control of energy balance and is thus implicated in the pathogenesis of obesity. The specific processes modulated by this neurotransmitter which lead to obesity and overeating are still a matter of debate. METHODS We tested the hypothesis that in vivo NA transporter (NAT) availability is changed in obesity by using positron emission tomography (PET) and S,S-[11C]O-methylreboxetine (MRB) in twenty subjects comprising ten highly obese (body mass index BMI > 35 kg/m2), metabolically healthy, non-depressed individuals and ten non-obese (BMI < 30 kg/m2) healthy controls. RESULTS Overall, we found no significant differences in binding potential (BPND) values between obese and non-obese individuals in the investigated brain regions, including the NAT-rich thalamus (0.40 ± 0.14 vs. 0.41 ± 0.18; p = 0.84) though additional discriminant analysis correctly identified individual group affiliation based on regional BPND in all but one (control) case. Furthermore, inter-regional correlation analyses indicated different BPND patterns between both groups but this did not survive testing for multiple comparions. CONCLUSIONS Our data do not find an overall involvement of NAT changes in human obesity. However, preliminary secondary findings of distinct regional and associative patterns warrant further investigation.
Collapse
|
4
|
Moriguchi S, Kimura Y, Ichise M, Arakawa R, Takano H, Seki C, Ikoma Y, Takahata K, Nagashima T, Yamada M, Mimura M, Suhara T. PET Quantification of the Norepinephrine Transporter in Human Brain with (S,S)-18F-FMeNER-D2. J Nucl Med 2016; 58:1140-1145. [DOI: 10.2967/jnumed.116.178913] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 12/05/2016] [Indexed: 11/16/2022] Open
|
5
|
[(18)F]FMeNER-D2: A systematic in vitro analysis of radio-metabolism. Nucl Med Biol 2016; 43:490-5. [PMID: 27236284 DOI: 10.1016/j.nucmedbio.2016.05.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 05/06/2016] [Accepted: 05/10/2016] [Indexed: 11/23/2022]
Abstract
INTRODUCTION The norepinephrine transporter (NET) presents an important target for therapy and diagnosis of ADHD and other neurodegenerative and psychiatric diseases. Thus, PET is the diagnostic method of choice, using radiolabeled NET-ligands derived from reboxetine. So far, [(18)F]FMeNER-D2 showed best pharmacokinetic and -dynamic properties. However, the disadvantage of reboxetine derived PET tracers is their high metabolic cleavage-resulting in impeding signals in the PET scans, which hamper a proper quantification of the NET in cortical areas. METHODS Metabolic stability testing was performed in vitro using a plethora of human and murine enzymes. RESULTS No metabolism was observed using monoamine oxidase A and B or catechol-O-methyl transferase. Incubation of [(18)F]FMeNER-D2 with CYP450-enzymes, predominantly located in the liver, led to a significant and fast metabolism of the tracer. Moreover, the arising three radiometabolites were found to be more polar than [(18)F]FMeNER-D2. Surprisingly, definitely no formation of free [(18)F]fluoride was observed. CONCLUSION According to our in vitro data, the interfering uptake in cortical regions might be attributed to these emerging radiometabolites but does not reflect bonding in bone due to defluorination. Further research on these radiometabolites is necessary to elucidate the in vivo situation. This might include an analysis of human blood samples after injection of [(18)F]FMeNER-D2, to enable a better correction of the PET-input function.
Collapse
|
6
|
Rami-Mark C, Berroterán-Infante N, Philippe C, Foltin S, Vraka C, Hoepping A, Lanzenberger R, Hacker M, Mitterhauser M, Wadsak W. Radiosynthesis and first preclinical evaluation of the novel norepinephrine transporter pet-ligand [(11)C]ME@HAPTHI. EJNMMI Res 2015; 5:113. [PMID: 26061602 PMCID: PMC4467816 DOI: 10.1186/s13550-015-0113-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 05/22/2015] [Indexed: 11/16/2022] Open
Abstract
Background The norepinephrine transporter (NET) has been demonstrated to be relevant to a multitude of neurological, psychiatric and cardiovascular pathologies. Due to the wide range of possible applications for PET imaging of the NET together with the limitations of currently available radioligands, novel PET tracers for imaging of the cerebral NET with improved pharmacological and pharmacodynamic properties are needed. Methods The present study addresses the radiosynthesis and first preclinical evaluation of the novel NET PET tracer [11C]Me@HAPTHI by describing its affinity, selectivity, metabolic stability, plasma free fraction, blood–brain barrier (BBB) penetration and binding behaviour in in vitro autoradiography. Results [11C]Me@HAPTHI was prepared and displayed outstanding affinity and selectivity as well as excellent in vitro metabolic stability, and it is likely to penetrate the BBB. Moreover, selective NET binding in in vitro autoradiography was observed in human brain and rat heart tissue samples. Conclusions All preclinical results and radiosynthetic key-parameters indicate that the novel benzothiadiazole dioxide-based PET tracer [11C]Me@HAPTHI is a feasible and improved NET radioligand and might prospectively facilitate clinical NET imaging. Electronic supplementary material The online version of this article (doi:10.1186/s13550-015-0113-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christina Rami-Mark
- Department of Biomedical Imaging and Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria,
| | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Joshi EM, Need A, Schaus J, Chen Z, Benesh D, Mitch C, Morton S, Raub TJ, Phebus L, Barth V. Efficiency gains in tracer identification for nuclear imaging: can in vivo LC-MS/MS evaluation of small molecules screen for successful PET tracers? ACS Chem Neurosci 2014; 5:1154-63. [PMID: 25247893 DOI: 10.1021/cn500073j] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Positron emission tomography (PET) imaging has become a useful noninvasive technique to explore molecular biology within living systems; however, the utility of this method is limited by the availability of suitable radiotracers to probe specific targets and disease biology. Methods to identify potential areas of improvement in the ability to predict small molecule performance as tracers prior to radiolabeling would speed the discovery of novel tracers. In this retrospective analysis, we characterized the brain penetration or peak SUV (standardized uptake value), binding potential (BP), and brain exposure kinetics across a series of known, nonradiolabeled PET ligands using in vivo LC-MS/MS (liquid chromatography coupled to mass spectrometry) and correlated these parameters with the reported PET ligand performance in nonhuman primates and humans available in the literature. The PET tracers studied included those reported to label G protein-coupled receptors (GPCRs), intracellular enzymes, and transporters. Additionally, data for each tracer was obtained from a mouse brain uptake assay (MBUA), previously published, where blood-brain barrier (BBB) penetration and clearance parameters were assessed and compared against similar data collected on a broad compound set of central nervous system (CNS) therapeutic compounds. The BP and SUV identified via nonradiolabeled LC-MS/MS, while different from the published values observed in the literature PET tracer data, allowed for an identification of initial criteria values we sought to facilitate increased potential for success from our early discovery screening paradigm. Our analysis showed that successful, as well as novel, clinical PET tracers exhibited BP of greater than 1.5 and peak SUVs greater than approximately 150% at 5 min post dose in rodents. The brain kinetics appeared similar between both techniques despite differences in tracer dose, suggesting linearity across these dose ranges. The assessment of tracers in a CNS exposure model, the mouse brain uptake assessment (MBUA), showed that those compound with initial brain-to-plasma ratios >2 and unbound fraction in brain homogenate >0.01 were more likely to be clinically successful PET ligands. Taken together, early incorporation of a LC/MS/MS cold tracer discovery assay and a parallel MBUA can be an useful screening paradigm to prioritize and rank order potential novel PET radioligands during early tracer discovery efforts. Compounds considered for continued in vivo PET assessments can be identified quickly by leveraging in vitro affinity and selectivity measures, coupled with data from a MBUA, primarily the 5 min brain-to-plasma ratio and unbound fraction data. Coupled utilization of these data creates a strategy to efficiently screen for the identification of appropriate chemical space to invest in for radiotracer discovery.
Collapse
Affiliation(s)
- Elizabeth M. Joshi
- Eli Lilly and Co., Lilly Research Laboratories, Indianapolis, Indiana 46285, United States
| | - Anne Need
- Eli Lilly and Co., Lilly Research Laboratories, Indianapolis, Indiana 46285, United States
| | - John Schaus
- Eli Lilly and Co., Lilly Research Laboratories, Indianapolis, Indiana 46285, United States
| | - Zhaogen Chen
- Eli Lilly and Co., Lilly Research Laboratories, Indianapolis, Indiana 46285, United States
| | - Dana Benesh
- Eli Lilly and Co., Lilly Research Laboratories, Indianapolis, Indiana 46285, United States
| | - Charles Mitch
- Eli Lilly and Co., Lilly Research Laboratories, Indianapolis, Indiana 46285, United States
| | - Stuart Morton
- Eli Lilly and Co., Lilly Research Laboratories, Indianapolis, Indiana 46285, United States
| | - Thomas J. Raub
- Eli Lilly and Co., Lilly Research Laboratories, Indianapolis, Indiana 46285, United States
| | - Lee Phebus
- Eli Lilly and Co., Lilly Research Laboratories, Indianapolis, Indiana 46285, United States
| | - Vanessa Barth
- Eli Lilly and Co., Lilly Research Laboratories, Indianapolis, Indiana 46285, United States
| |
Collapse
|
8
|
Mark C, Bornatowicz B, Mitterhauser M, Hendl M, Nics L, Haeusler D, Lanzenberger R, Berger ML, Spreitzer H, Wadsak W. Development and automation of a novel NET-PET tracer: [11C]Me@APPI. Nucl Med Biol 2013; 40:295-303. [DOI: 10.1016/j.nucmedbio.2012.11.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 11/07/2012] [Accepted: 11/15/2012] [Indexed: 10/27/2022]
|
9
|
Kanegawa N, Kiyono Y, Sugitaa T, Kuge Y, Fujibayasi Y, Saji H. Norepinephrine Transporter Imaging in the Brain of a Rat Model of Depression Using Radioiodinated (2S, αS)-2-(α-(2-iodophenoxy)benzyl)morpholine. Mol Imaging 2012. [DOI: 10.2310/7290.2011.00049] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
To visualize the norepinephrine transporters (NETs) in various brain diseases, we developed radioiodinated (2S,αS)-2-(α-(2-iodophenoxy)benzyl)morpholine ((S,S)-IPBM). This radioligand achieved the basic requirements for NET imaging. In this study, we assessed the potential of radioiodinated (S,S)-IPBM as an imaging biomarker of NET to obtain diagnostic information about depression in relation to NET expression in the brain using a rat depression model. The ex vivo autoradiographic experiments using the (S,S)-[125I]IPBM showed significantly lower accumulation of radioactivity in the locus coeruleus (LC) and the anteroventricular thalamic nucleus (AVTN) of the depression group than in those of the control group. Consequently, in vitro autoradiographic experiments showed that NET maximum binding (Bmax) values in the LC and AVTN, known as NET-rich regions, were significantly decreased in the rat model of depression when compared to those of the control rats. In addition, there was an extremely good correlation between NET Bmax and (S,S)-IPBM accumulation ( r = .98), an indication of radioiodinated IPBM as a quantitative NET imaging biomarker. The reduction in(S,S)-[125I]IPBM accumulation in the rat model of depression correlated with that of NET density. These results suggest that (S,S)-[123I]IPBM has potential as an imaging biomarker of NET to obtain diagnostic information about major depression.
Collapse
Affiliation(s)
- Naoki Kanegawa
- From the Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Japan; Biomedical Imaging Research Center, University of Fukui, Fukui, Japan; Radioisotopes Research Laboratory, Kyoto University Hospital, Faculty of Medicine, Kyoto University, Japan; Central Institute of Isotope Science, Hokkaido University, Hokkaido, Japan; and Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | - Yasushi Kiyono
- From the Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Japan; Biomedical Imaging Research Center, University of Fukui, Fukui, Japan; Radioisotopes Research Laboratory, Kyoto University Hospital, Faculty of Medicine, Kyoto University, Japan; Central Institute of Isotope Science, Hokkaido University, Hokkaido, Japan; and Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | - Taku Sugitaa
- From the Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Japan; Biomedical Imaging Research Center, University of Fukui, Fukui, Japan; Radioisotopes Research Laboratory, Kyoto University Hospital, Faculty of Medicine, Kyoto University, Japan; Central Institute of Isotope Science, Hokkaido University, Hokkaido, Japan; and Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | - Yuji Kuge
- From the Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Japan; Biomedical Imaging Research Center, University of Fukui, Fukui, Japan; Radioisotopes Research Laboratory, Kyoto University Hospital, Faculty of Medicine, Kyoto University, Japan; Central Institute of Isotope Science, Hokkaido University, Hokkaido, Japan; and Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | - Yasushisa Fujibayasi
- From the Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Japan; Biomedical Imaging Research Center, University of Fukui, Fukui, Japan; Radioisotopes Research Laboratory, Kyoto University Hospital, Faculty of Medicine, Kyoto University, Japan; Central Institute of Isotope Science, Hokkaido University, Hokkaido, Japan; and Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| | - Hideo Saji
- From the Department of Patho-Functional Bioanalysis, Graduate School of Pharmaceutical Sciences, Kyoto University, Japan; Biomedical Imaging Research Center, University of Fukui, Fukui, Japan; Radioisotopes Research Laboratory, Kyoto University Hospital, Faculty of Medicine, Kyoto University, Japan; Central Institute of Isotope Science, Hokkaido University, Hokkaido, Japan; and Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan
| |
Collapse
|
10
|
Gulyás B, Vas Á, Tóth M, Takano A, Varrone A, Cselényi Z, Schain M, Mattsson P, Halldin C. Age and disease related changes in the translocator protein (TSPO) system in the human brain: Positron emission tomography measurements with [11C]vinpocetine. Neuroimage 2011; 56:1111-21. [DOI: 10.1016/j.neuroimage.2011.02.020] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Revised: 02/02/2011] [Accepted: 02/05/2011] [Indexed: 01/06/2023] Open
|
11
|
Gallezot JD, Weinzimmer D, Nabulsi N, Lin SF, Fowles K, Sandiego C, McCarthy TJ, Maguire RP, Carson RE, Ding YS. Evaluation of [(11)C]MRB for assessment of occupancy of norepinephrine transporters: Studies with atomoxetine in non-human primates. Neuroimage 2010; 56:268-79. [PMID: 20869448 DOI: 10.1016/j.neuroimage.2010.09.040] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2010] [Accepted: 09/16/2010] [Indexed: 12/15/2022] Open
Abstract
[(11)C]MRB is one of the most promising radioligands used to measure brain norepinephrine transporters (NET) with positron emission tomography (PET). The objective of this study was to evaluate the suitability of [(11)C]MRB for drug occupancy studies of NET using atomoxetine (ATX), a NET uptake inhibitor used in the treatment of depression and attention-deficit hyperactivity disorder (ADHD). A second goal of the study was identification of a suitable reference region. Ten PET studies were performed in three anesthetized rhesus monkeys following an infusion of ATX or placebo. [(11)C]MRB arterial input functions and ATX plasma levels were also measured. A dose-dependent reduction of [(11)C]MRB volume of distribution was observed after correction for [(11)C]MRB plasma free fraction. ATX IC(50) was estimated to be 31 ± 10ng/mL plasma. This corresponds to an effective dose (ED(50)) of 0.13mg/kg, which is much lower than the therapeutic dose of ATX in ADHD (1.0-1.5mg/kg). [(11)C]MRB binding potential BP(ND) in the thalamus was estimated to be 1.8 ± 0.3. Defining a reference region for a NET radiotracer is challenging due to the widespread and relatively uniform distribution of NET in the brain. Three regions were evaluated for use as reference region: caudate, putamen and occipital cortex. Caudate was found to be the most suitable for preclinical drug occupancy studies in rhesus monkeys. The IC(50) estimate obtained using MRTM2 BP(ND) without arterial blood sampling was 21 ± 3ng/mL (using caudate as the reference region). This study demonstrated that [(11)C]MRB is suitable for drug occupancy studies of NET.
Collapse
|
12
|
The norepinephrine transporter (NET) radioligand (S,S)-[18F]FMeNER-D2 shows significant decreases in NET density in the human brain in Alzheimer's disease: A post-mortem autoradiographic study. Neurochem Int 2010; 56:789-98. [DOI: 10.1016/j.neuint.2010.03.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2010] [Accepted: 03/01/2010] [Indexed: 11/19/2022]
|