100 results on '"Wakana Mori"'
Search Results
2. Automated radiosynthesis and in vivo evaluation of 18F-labeled analog of the photosensitizer ADPM06 for planning photodynamic therapy
- Author
-
Kazunori Kawamura, Tomoteru Yamasaki, Masayuki Fujinaga, Tomomi Kokufuta, Yiding Zhang, Wakana Mori, Yusuke Kurihara, Masanao Ogawa, Kaito Tsukagoe, Nobuki Nengaki, and Ming-Rong Zhang
- Subjects
18F ,BF2-chelated tetraaryl-azadipyrromethenes ,Photosensitizer ,Photodynamic therapy ,Medical physics. Medical radiology. Nuclear medicine ,R895-920 ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Abstract Background A family of BF2-chelated tetraaryl-azadipyrromethenes was developed as non-porphyrin photosensitizers for photodynamic therapy. Among the developed photosensitizers, ADPM06 exhibited excellent photochemical and photophysical properties. Molecular imaging is a useful tool for photodynamic therapy planning and monitoring. Radiolabeled photosensitizers can efficiently address photosensitizer biodistribution, providing helpful information for photodynamic therapy planning. To evaluate the biodistribution of ADPM06 and predict its pharmacokinetics on photodynamic therapy with light irradiation immediately after administration, we synthesized [18F]ADPM06 and evaluated its in vivo properties. Results [18F]ADPM06 was automatically synthesized by Lewis acid-assisted isotopic 18F-19F exchange using ADPM06 and tin (IV) chloride at room temperature for 10 min. Radiolabeling was carried out using 0.4 μmol of ADPM06 and 200 μmol of tin (IV) chloride. The radiosynthesis time was approximately 60 min, and the radiochemical purity was > 95% at the end of the synthesis. The decay-corrected radiochemical yield from [18F]F− at the start of synthesis was 13 ± 2.7% (n = 5). In the biodistribution study of male ddY mice, radioactivity levels in the heart, lungs, liver, pancreas, spleen, kidney, small intestine, muscle, and brain gradually decreased over 120 min after the initial uptake. The mean radioactivity level in the thighbone was the highest among all organs investigated and increased for 120 min after injection. Upon co-injection with ADPM06, the radioactivity levels in the blood and brain significantly increased, whereas those in the heart, lung, liver, pancreas, kidney, small intestine, muscle, and thighbone of male ddY mice were not affected. In the metabolite analysis of the plasma at 30 min post-injection in female BALB/c-nu/nu mice, the percentage of radioactivity corresponding to [18F]ADPM06 was 76.3 ± 1.6% (n = 3). In a positron emission tomography study using MDA-MB-231-HTB-26 tumor-bearing mice (female BALB/c-nu/nu), radioactivity accumulated in the bone at a relatively high level and in the tumor at a moderate level for 60 min after injection. Conclusions We synthesized [18F]ADPM06 using an automated 18F-labeling synthesizer and evaluated the initial uptake and pharmacokinetics of ADPM06 using biodistribution of [18F]ADPM06 in mice to guide photodynamic therapy with light irradiation.
- Published
- 2023
- Full Text
- View/download PDF
3. Automated radiosynthesis of two 18F-labeled tracers containing 3-fluoro-2-hydroxypropyl moiety, [18F]FMISO and [18F]PM-PBB3, via [18F]epifluorohydrin
- Author
-
Takayuki Ohkubo, Yusuke Kurihara, Masanao Ogawa, Nobuki Nengaki, Masayuki Fujinaga, Wakana Mori, Katsushi Kumata, Masayuki Hanyu, Kenji Furutsuka, Hiroki Hashimoto, Kazunori Kawamura, and Ming-Rong Zhang
- Subjects
18F ,[18F]Epifluorohydrin ,[18F]FMISO ,[18F]PM-PBB3 ,Positron emission tomography (PET) ,Medical physics. Medical radiology. Nuclear medicine ,R895-920 ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Abstract Background [18F]Fluoromisonidazole ([18F]FMISO) and 1-[18F]fluoro-3-((2-((1E,3E)-4-(6-(methylamino)pyridine-3-yl)buta-1,3-dien-1-yl)benzo[d]thiazol-6-yl)oxy)propan-2-ol ([18F]PM-PBB3 or [18F]APN-1607) are clinically used radiotracers for imaging hypoxia and tau pathology, respectively. Both radiotracers were produced by direct 18F-fluorination using the corresponding tosylate precursors 1 or 2 and [18F]F−, followed by the removal of protecting groups. In this study, we synthesized [18F]FMISO and [18F]PM-PBB3 by 18F-fluoroalkylation using [18F]epifluorohydrin ([18F]5) for clinical applications. Results First, [18F]5 was synthesized by the reaction of 1,2-epoxypropyl tosylate (8) with [18F]F− and was purified by distillation. Subsequently, [18F]5 was reacted with 2-nitroimidazole (6) or PBB3 (7) as a precursor for 18F-labeling, and each reaction mixture was purified by preparative high-performance liquid chromatography and formulated to obtain the [18F]FMISO or [18F]PM-PBB3 injection. All synthetic sequences were performed using an automated 18F-labeling synthesizer. The obtained [18F]FMISO showed sufficient radioactivity (0.83 ± 0.20 GBq at the end of synthesis (EOS); n = 8) with appropriate radiochemical yield based on [18F]F− (26 ± 7.5 % at EOS, decay-corrected; n = 8). The obtained [18F]PM-PBB3 also showed sufficient radioactivity (0.79 ± 0.10 GBq at EOS; n = 11) with appropriate radiochemical yield based on [18F]F− (16 ± 3.2 % at EOS, decay-corrected; n = 11). Conclusions Both [18F]FMISO and [18F]PM-PBB3 injections were successfully synthesized with sufficient radioactivity by 18F-fluoroalkylation using [18F]5.
- Published
- 2021
- Full Text
- View/download PDF
4. Development of a highly-specific 18F-labeled irreversible positron emission tomography tracer for monoacylglycerol lipase mapping
- Author
-
Zhen Chen, Wakana Mori, Jian Rong, Michael A. Schafroth, Tuo Shao, Richard S. Van, Daisuke Ogasawara, Tomoteru Yamasaki, Atsuto Hiraishi, Akiko Hatori, Jiahui Chen, Yiding Zhang, Kuan Hu, Masayuki Fujinaga, Jiyun Sun, Qingzhen Yu, Thomas L. Collier, Yihan Shao, Benjamin F. Cravatt, Lee Josephson, Ming-Rong Zhang, and Steven H. Liang
- Subjects
Monoacylglycerol lipase (MAGL) ,Central nervous system (CNS) ,2-Arachidonylglycerol (2-AG) ,Arachidonic acid (AA) ,Positron emission tomography (PET) ,Fluorine-18 ,Therapeutics. Pharmacology ,RM1-950 - Abstract
As a serine hydrolase, monoacylglycerol lipase (MAGL) is principally responsible for the metabolism of 2-arachidonoylglycerol (2-AG) in the central nervous system (CNS), leading to the formation of arachidonic acid (AA). Dysfunction of MAGL has been associated with multiple CNS disorders and symptoms, including neuroinflammation, cognitive impairment, epileptogenesis, nociception and neurodegenerative diseases. Inhibition of MAGL provides a promising therapeutic direction for the treatment of these conditions, and a MAGL positron emission tomography (PET) probe would greatly facilitate preclinical and clinical development of MAGL inhibitors. Herein, we design and synthesize a small library of fluoropyridyl-containing MAGL inhibitor candidates. Pharmacological evaluation of these candidates by activity-based protein profiling identified 14 as a lead compound, which was then radiolabeled with fluorine-18 via a facile SNAr reaction to form 2-[18F]fluoropyridine scaffold. Good blood–brain barrier permeability and high in vivo specific binding was demonstrated for radioligand [18F]14 (also named as [18F]MAGL-1902). This work may serve as a roadmap for clinical translation and further design of potent 18F-labeled MAGL PET tracers.
- Published
- 2021
- Full Text
- View/download PDF
5. Radiosynthesis and in vivo evaluation of 11C-labeled BMS-193885 and its desmethyl analog as PET tracers for neuropeptide Y1 receptors
- Author
-
Kazunori Kawamura, Wakana Mori, Masayuki Fujinaga, Tomoteru Yamasaki, Yiding Zhang, Hidekatsu Wakizaka, Akiko Hatori, Lin Xie, Katsushi Kumata, Takayuki Ohkubo, Yusuke Kurihara, Masanao Ogawa, Nobuki Nengaki, and Ming-Rong Zhang
- Subjects
Carbon-11 ,Positron emission tomography ,Neuropeptide Y1 receptor ,BMS-193885 ,Medical physics. Medical radiology. Nuclear medicine ,R895-920 ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Abstract Background Neuropeptide Y (NPY) has been implicated in a wide variety of physiological processes, including feeding, learning, memory, emotion, cardiovascular homeostasis, hormone secretion, and circadian rhythms. NPY Yl receptor (NPY Y1-R) is the most widely studied NPY receptor, and is involved in many of these processes. BMS-193885 (1) was previously developed as a potent and selective NPY Y1-R antagonist, which has good systemic bioavailability and brain penetration. To evaluate the characteristics of 1 in vivo, we developed 11C-labeled BMS-193885 ([11C]1) and its desmethyl analog ([11C]2) for potential use as two new positron emission tomography (PET) tracers. Results [11C]1 was synthesized from [11C]methyl iodide using 2. [11C]2 was synthesized from [11C]phosgene using its aniline and amine derivatives. The mean ± S.D. decay-corrected radiochemical yields of [11C]1 and [11C]2 from 11CO2 at the end of radionuclide production were 23 ± 3.2% (n = 6) and 24 ± 1.5% (n = 4), respectively. In biodistribution on mice, radioactivity levels for both tracers were relatively high in the kidney, small intestine, and liver at 60 min post-injection. The radioactivity levels in the kidney, lung, and spleen of mice at 30 min post-injection with [11C]1 were significantly reduced by pretreatment with 1 (10 mg/kg), and levels of [11C]1 in the brain of mice were significantly increased by pretreatment with the P-glycoprotein and breast cancer resistance protein inhibitor elacridar (10 mg/kg). In metabolite analysis using mouse plasma, [11C]1 and [11C]2 were rapidly metabolized within 30 min post-injection, and [11C]1 was mainly metabolized into unlabeled 2 and radiolabeled components. Conclusion [11C]1 and [11C]2 were successfully synthesized with sufficient amount of radioactivity and high quality for use in vivo. Our study of [11C]1 and its desmethyl analog [11C]2 was useful in that it helped to elucidate the in vivo characteristics of 1.
- Published
- 2019
- Full Text
- View/download PDF
6. Pharmacokinetic Evaluation of [C]CEP-32496 in Nude Mice Bearing BRAF Mutation-Induced Melanomas
- Author
-
Cuiping Jiang MMSc, Lin Xie MD, PhD, Yiding Zhang BSc, Masayuki Fujinaga PhD, Wakana Mori MSc, Yusuke Kurihara PhD, Tomoteru Yamasaki PhD, Feng Wang MD, PhD, and Ming-Rong Zhang MD, PhD
- Subjects
Biology (General) ,QH301-705.5 ,Medical technology ,R855-855.5 - Abstract
CEP-32496, also known as RXDX-105 or Agerafenib, is a new orally active inhibitor for the mutated v-raf murine sarcoma viral oncogene homolog B1 (BRAF V600E ), which has attracted considerable attention in clinical trials for the treatment of human cancers. Here, we used carbon-11-labeled CEP-32496 ([ 11 C]CEP-32496) as a positron emission tomography (PET) radiotracer to evaluate its pharmacokinetic properties and explore its potential for in vivo imaging. Following radiotracer synthesis, we performed in vitro binding assays and autoradiography of [ 11 C]CEP-32496 in the A375 melanoma cell line and on tumor tissue sections from mice harboring the BRAF V600E mutation. These were followed by PET scans and biodistribution studies on nude mice bearing subcutaneous A375 cell-induced melanoma. [ 11 C]CEP-32496 showed high binding affinity for BRAF V600E -positive A375 melanoma cells and densely accumulated in the respective tissue sections; this could be blocked by the BRAF V600E selective antagonist sorafenib and by unlabeled CEP-32496. The PET and biodistribution results revealed that [ 11 C]CEP-32496 accumulated continuously but slowly into the tumor within a period of 0 to 60 minutes postinjection in A375-melanoma-bearing nude mice. Metabolite analysis showed high in vivo stability of [ 11 C]CEP-32496 in plasma. Our results indicate that [ 11 C]CEP-32496 has excellent specificity and affinity for the BRAF V600E mutation in vitro, while its noninvasive personalized diagnostic role needs to be studied further.
- Published
- 2018
- Full Text
- View/download PDF
7. First demonstration of in vivo mapping for regional brain monoacylglycerol lipase using PET with [11C]SAR127303.
- Author
-
Tomoteru Yamasaki, Wakana Mori, Yiding Zhang, Akiko Hatori, Masayuki Fujinaga, Hidekatsu Wakizaka, Yusuke Kurihara, Lu Wang, Nobuki Nengaki, Tomoyuki Ohya, Steven H. Liang, and Ming-Rong Zhang
- Published
- 2018
- Full Text
- View/download PDF
8. Automated radiosynthesis and in vivo evaluation of [18F]ADPM06 as a photosensitizer for photodynamic therapy
- Author
-
Kazunori Kawamura, Tomoteru Yamasaki, Masayuki Fujinaga, Kokufuta Tomomi, Yiding Zhang, Wakana Mori, Yusuke Kurihara, Masanao Ogawa, Kaito Tsukagoe, Nobuki Nengaki, and Ming-Rong Zhang
- Abstract
Background A family of BF2-chelated tetraaryl-azadipyrromethenes was developed as non-porphyrin photosensitizers for photodynamic therapy. Among the developed photosensitizers, ADPM06 exhibited excellent photochemical and photophysical properties. Molecular imaging is a useful tool for photodynamic therapy planning and monitoring. Radiolabeled photosensitizers can efficiently address photosensitizer biodistribution, providing helpful information for photodynamic therapy planning. To evaluate the biodistribution of ADPM06 and predict its pharmacokinetics on photodynamic therapy, we synthesized [18F]ADPM06 and evaluated its in vivo properties. Results [18F]ADPM06 was automatically synthesized by Lewis acid-assisted isotopic 18F-19F exchange using ADPM06 and tin (IV) chloride at room temperature for 10 min. Radiolabeling was carried out using 0.4 µmol of ADPM06 and 200 µmol of tin (IV) chloride. The radiosynthesis time was approximately 60 min, and the radiochemical purity was > 95% at the end of the synthesis. The decay-corrected radiochemical yield from [18F]F- at the end of irradiation was 13 ± 2.7% (n = 5). In the biodistribution study, radioactivity levels in the heart, lungs, liver, pancreas, spleen, kidney, small intestine, muscle, and brain gradually decreased over 120 min after the initial uptake. The mean radioactivity level in the bone was the highest among all organs investigated and increased for 120 min after injection. Upon co-injection with ADPM06, the radioactivity levels in the blood, heart, and brain significantly increased, whereas those in the lung, liver, pancreas, kidney, small intestine, muscle, and bone were not affected. In the metabolite study of the plasma in mice, the percentage of radioactivity corresponding to [18F]ADPM06 was 76.3 ± 1.6% (n = 3). In a positron emission tomography study using MDA-MB-231-HTB-26 tumor-bearing mice, radioactivity accumulated in the bone at a relatively high level and in the tumor at a moderate level for 60 min after injection. Conclusions We synthesized [18F]ADPM06 using an automated 18F-labeling synthesizer and evaluated the biodistribution of [18F]ADPM06 in mice, which may be useful for predicting the pharmacokinetics of ADPM06 in photodynamic therapy.
- Published
- 2023
- Full Text
- View/download PDF
9. Imaging of transmembrane AMPA receptor regulatory protein by positron emission tomography
- Author
-
Qingzhen, Yu, Katsushi, Kumata, Jian, Rong, Zhen, Chen, Tomoteru, Yamasaki, Jiahui, Chen, Zhiwei, Xiao, Hideki, Ishii, Atsuto, Hiraishi, Tuo, Shao, Zhang, Yiding, Kuan, Hu, Lin, Xie, Masayuki, Fujinaga, Chunyu, Zhao, Wakana, Mori, Thomas, Collier, Ahmed, Haider, Susumu, Tomita, Zhang, Ming-Rong, and Steven, Liang
- Abstract
The transmembrane α-amino-3-hydroxyl-5-methyl-4-isoxazolepropionic acid (AMPA) receptor regulatory protein γ-8 (TARP γ-8) constitutes an auxiliary subunit of AMPA receptors, which mediates various brain functions including learning and memory. TARP γ-8 has emerged as a promising therapeutic target for central nervous system disorders. Despite considerable efforts, previously reported TARP γ-8 PET radioligands, such as [ 11C]TARP-1903 and [11C]TARP-1811 series, were plagued by limited brain uptake and/or high nonspecific binding in vivo. Herein, we developed two novel 11C-labeled probes, [11C]8 and [11C]15 (also named as [11C]TARP2105), of which the latter exhibited a reasonable brain uptake as well as specific binding toward TARP γ-8 both in vitro and in vivo, as confirmed by blocking experiments with the commercially available TARP γ-8 inhibitor, JNJ-55511118 in the TARP γ-8-rich hippocampus. Overall, [11C]15 exhibited promising tracer characteristics and proved to be a lead positron-emission tomography ligand for the non-invasive quantification of TARP γ-8 in the mammalian brain.
- Published
- 2022
10. A novel monoacylglycerol lipase-targeted 18F-labeled probe for positron emission tomography imaging of brown adipose tissue in the energy network
- Author
-
Ran Cheng, Masayuki Fujinaga, Jing Yang, Jian Rong, Ahmed Haider, Daisuke Ogasawara, Richard S. Van, Tuo Shao, Zhen Chen, Xiaofei Zhang, Erick R. Calderon Leon, Yiding Zhang, Wakana Mori, Katsushi Kumata, Tomoteru Yamasaki, Lin Xie, Shaofa Sun, Lu Wang, Chongzhao Ran, Yihan Shao, Benjamin Cravatt, Lee Josephson, Ming-Rong Zhang, and Steven H. Liang
- Subjects
Pharmacology ,Pharmacology (medical) ,General Medicine - Published
- 2022
- Full Text
- View/download PDF
11. Upregulation of Striatal Metabotropic Glutamate Receptor Subtype 1 (mGluR1) in Rats with Excessive Glutamate Release Induced by N-Acetylcysteine
- Author
-
Tomoteru Yamasaki, Maki Okada, Atsuto Hiraishi, Wakana Mori, Yiding Zhang, Masayuki Fujinaga, Hidekatsu Wakizaka, Yusuke Kurihara, Nobuki Nengaki, and Ming-Rong Zhang
- Subjects
General Neuroscience ,Animals ,Glutamic Acid ,Receptors, Metabotropic Glutamate ,Toxicology ,Acetylcysteine ,Rats ,Up-Regulation - Abstract
The aim of this study is to investigate the changes in expression of metabotropic glutamate (Glu) receptor subtype 1 (mGluR1), a key molecule involved in neuroexcitetoxicity, during excessive Glu release in the brain by PET imaging. An animal model of excessive Glu release in the brain was produced by intraperitoneally implanting an Alzet osmotic pump containing N-acetylcysteine (NAC), an activator of the cysteine/Glu antiporter, into the abdomen of rats. Basal Glu concentration in the brain was measured by microdialysis, which showed that basal Glu concentration in NAC-treated rats (0.31 µM) was higher than that in saline-treated rats (0.17 µM) at day 7 after the implantation of the osmotic pump. Similarly, PET studies with [
- Published
- 2022
- Full Text
- View/download PDF
12. Novel Reversible-Binding PET Ligands for Imaging Monoacylglycerol Lipase Based on the Piperazinyl Azetidine Scaffold
- Author
-
Rong, Jian, Wakana, Mori, Xia, Xiaotian, A. Schafroth, Michael, Zhao, Chunyu, S. Van, Richard, Tomoteru, Yamasaki, Chen, Jiahui, Xiao, Zhiwei, Haider, Ahmed, Ogasawara, Daisuke, Atsuto, Hiraishi, Shao, Tuo, Zhang, Yiding, Chen, Zhen, Pang, Fuwen, Kuan, Hu, Lin, Xie, Masayuki, Fujinaga, Katsushi, Kumata, Gou, Yuancheng, Fang, Yang, Gu, Shuyin, Wei, Huiyi, Bao, Liang, Xu, Hao, L. Collier, Thomas, Shao, Yihan, E. Carson, Richard, F. Cravatt, Benjamin, Lu, Wang, ming-rong, zhang, Liang, Huan, and Zhang, Ming-Rong
- Abstract
Monoacylglycerol lipase (MAGL) is a 33 kDa serine protease primarily responsible for hydrolyzing 2-arachidonoylglycerol into the proinflammatory eicosanoid precursor arachidonic acid in the central nervous system. Inhibition of MAGL constitutes an attractive therapeutic concept for treating psychiatric disorders and neurodegenerative diseases. Herein, we present the design and synthesis of multiple reversible MAGL inhibitor candidates based on a piperazinyl azetidine scaffold. Compounds 10 and 15 were identified as the best-performing reversible MAGL inhibitors by pharmacological evaluations, thus channeling their radiolabeling with fluorine-18 in high radiochemical yields and favorable molar activity. Furthermore, evaluation of [18F]10 and [18F]15 ([18F]MAGL-2102) by autoradiography and positron emission tomography (PET) imaging in rodents and nonhuman primates demonstrated favorable brain uptakes, heterogeneous radioactivity distribution, good specific binding, and adequate brain kinetics, and [18F]15 demonstrated a better performance. In conclusion, [18F]15 was found to be a suitable PET radioligand for the visualization of MAGL, harboring potential for the successful translation into humans.
- Published
- 2021
13. Novel Reversible-Binding PET Ligands for Imaging Monoacylglycerol Lipase Based on the Piperazinyl Azetidine Scaffold
- Author
-
Ahmed Haider, Jian Rong, Steven H. Liang, Huiyi Wei, Daisuke Ogasawara, Masayuki Fujinaga, Tuo Shao, Wakana Mori, Shuyin Gu, Hao Xu, Michael A. Schafroth, Lin Xie, Yuancheng Gou, Ming-Rong Zhang, Thomas Lee Collier, Jiahui Chen, Katsushi Kumata, Zhen Chen, Richard Van, Yihan Shao, Lu Wang, Zhiwei Xiao, Xiaotian Xia, Kuan Hu, Bao Liang, Tomoteru Yamasaki, Richard E. Carson, Yiding Zhang, Fuwen Pang, Benjamin F. Cravatt, Yang Fang, Chunyu Zhao, and Atsuto Hiraishi
- Subjects
Models, Molecular ,Azetidine ,Ligands ,01 natural sciences ,Article ,Proinflammatory cytokine ,Structure-Activity Relationship ,03 medical and health sciences ,chemistry.chemical_compound ,Drug Discovery ,Radioligand ,Animals ,Distribution (pharmacology) ,030304 developmental biology ,Serine protease ,0303 health sciences ,Binding Sites ,Dose-Response Relationship, Drug ,Molecular Structure ,biology ,010405 organic chemistry ,Haplorhini ,Monoacylglycerol Lipases ,Rats ,0104 chemical sciences ,3. Good health ,Monoacylglycerol lipase ,Eicosanoid ,chemistry ,Biochemistry ,Positron-Emission Tomography ,biology.protein ,Azetidines ,Molecular Medicine ,Arachidonic acid ,Radiopharmaceuticals - Abstract
Monoacylglycerol lipase (MAGL) is a 33 kDa serine protease primarily responsible for hydrolyzing 2-arachidonoylglycerol into the proinflammatory eicosanoid precursor arachidonic acid in the central nervous system. Inhibition of MAGL constitutes an attractive therapeutic concept for treating psychiatric disorders and neurodegenerative diseases. Herein, we present the design and synthesis of multiple reversible MAGL inhibitor candidates based on a piperazinyl azetidine scaffold. Compounds 10 and 15 were identified as the best-performing reversible MAGL inhibitors by pharmacological evaluations, thus channeling their radiolabeling with fluorine-18 in high radiochemical yields and favorable molar activity. Furthermore, evaluation of [18F]10 and [18F]15 ([18F]MAGL-2102) by autoradiography and positron emission tomography (PET) imaging in rodents and nonhuman primates demonstrated favorable brain uptakes, heterogeneous radioactivity distribution, good specific binding, and adequate brain kinetics, and [18F]15 demonstrated a better performance. In conclusion, [18F]15 was found to be a suitable PET radioligand for the visualization of MAGL, harboring potential for the successful translation into humans.
- Published
- 2021
- Full Text
- View/download PDF
14. Automated radiosynthesis of 18F-fluoromethylated tracers using the simplified one-pot 18F-fluoromethylation via [18F]fluoromethyl tosylate
- Author
-
Kazunori, Kawamura, Wakana, Mori, Masayuki, Fujinaga, Kurihara, Yusuke, Masanao, Ogawa, Kaito, Tsukagoe, Ohkubo, Takayuki, Hiroki, Hashimoto, Nobuki, Nengaki, and Zhang, Ming-Rong
- Abstract
Background/Aims: [18F]Fluoroalkyl groups are essential labeling units because they are considered as surrogates for [11C]methyl moieties, and are coupled to the same functional units as the [11C]methyl group. Many [18F]fluoroalkyated PET tracers have been developed. In general, 18F-fluoroalkylation using [18F]fluoroalkyl reagents requires multi-step radiosynthesis procedures and a multi-pot 18F-labeling synthesizer. To overcome these limitations, a straightforward one-pot method for 18F-fluoroethylation without azeotropic drying of [18F]F- was developed [1]. We have used an improved one-pot 18F-fluoroethylaion method to synthesize 18F-fluoroethylated tracers [2]. In this study, we further modified this one-pot method suitable for 18F-fluoromethylation, and simplified the automated radiosynthesis of two [18F]fluoromethylated tracers using this method. Methods: We synthesized [18F]fluoromethyl tosylate in a mixture of 18F- in K222/K2CO3 acetonitrile solution including 2% water, bis(tosyloxy)methane and cesium carbonate. Without purification of [18F]fluoromethyl tosylate, we directly added a labeling precursor to this mixture for the simplified one-pot 18F-fluoromethylation. Using this procedure equipped to a 18F-labeling synthesizer, [18F]FCho (a PET tracer for imaging tumor) and [18F]FMeNER-D2 (a PET tracer for imaging norepinephrine transporter) were automatically synthesized by the reactions of their corresponding labeling precursors with [18F]fluoromethyl tosylate, respectively. Results: Using the simplified one-pot 18F-fluoromethylation procedure in the automated radiosynthesis, we achieved [18F]FCho and [18F]FMeNER-D2 in approximately 10% of radiochemical yield from 18F- at the end of irradiation (EOI). Radiosynthesis times and radiochemical purities of two 18F-labeled tracers were approximately 60 min after EOI and over 95%, respectively. Conclusions: We have successively synthesized [18F]FCho and [18F]FMeNER-D2 using the simplified one-pot 18F-fluoromethylation method, and achieved automation for all radiosynthesis processes using an 18F-labeling synthesizer. The present method provides a shorter synthesis time and automated procedures with one-pot for the 18F-fluoromethylation strategy., 第13回世界核医学会(13th Congress of the World Federation of Nuclear Medicine and Biology)
- Published
- 2022
15. Development of metabotropic glutamate receptor 1-targeted radiopharmaceuticals for theranostics of melanoma
- Author
-
Lin, Xie, Masayuki, Hanyu, Masayuki, Fujinaga, Zhang, Lulu, Zhang, Yiding, Wakana, Mori, Kuan, Hu, Katsuyuki, Minegishi, Kotaro, Nagatsu, and Zhang, Ming-Rong
- Abstract
Background/Aims: Metabotropic glutamate receptor 1 (mGluR1), a key mediator of glutamatergic signaling, is frequently expressed as an oncoprotein and has been an attractive target to overcome most solid tumors, such as melanoma (1). Here, a novel small-molecular radiopharmaceutical pair, 3-iodo-N-[4-[6-(methylamino)pyrimidin-4-yl]-1,3-thiazol-2-yl]-N-[11C]methylbenzamide ([11C]1) and 3-211At-astato-N-[4-[6-(methylamino)pyrimidin-4-yl]-1,3-thiazol-2-yl]-N-methylbenzamide ([211At]1), was designed and developed to target the mGluR1 for theranostics of melanomas. Methods: [ 11C]1 was synthesized by reacting a N-desmethyl precursor with [11C]CH3OTf in the presence of NaOH at room temperature for 5 min. Radiolabeling with [211At]1 was performed by reaction of aryl tin precursor with NCS-containing 211At/MeOH solution according to the method reported by our laboratory (2). The theranostic potentials of the radiopharmaceutical pair were explored for PET imaging and radiotherapy in mGluR1-positive B16F10 melanoma-bearing mice. Results: [ 11C]1 and [211At]1 were obtained with a radiochemical purity of greater than 99% and radiochemical yields of 19 ± 8 % and 46 ± 3%, respectively, based on the total radioactivity of used radionuclides. In vivo PET imaging of [ 11C]1 clearly visualized the targeted melanomas with a good tumor-to-background contrast. Ex vivo biodistribution study verified the persistent increase of [ 11C]1, which reached 12.29 ± 2.44 %ID/g tissue at 90 min in the targeted melanomas, and rapidly cleared from nontarget organs after intravenous injection. In the therapeutic studies, [211At]1 exhibited unequivocal and durable antitumor efficacy with only a single treatment (2.96 MBq) in the melanoma model, compared to the controls (0.31 ± 0.08 cm3 vs. 10.44 ± 1.61 cm3 at 16 days post-therapy). No decrease in body weight and no liver and kidney damage were observed through the examination period in melanoma mice injected with 2.96 MBq of [211At]1. Conclusions: The novel small-molecular radiopharmaceutical pair successfully visualized the mGluR1-positive melanomas by [ 11C]1 with high contrast PET images, and further treated the melanoma by [211At]1 without significant toxicity. The results highlight the good potential of using [ 11C]1 and [211At]1 as theranostic agents for the management of mGluR1-positive tumors and should be further investigated in the theranostic field of oncology in the clinic., 第13回世界核医学会(13th orld Federation of Nuclear Medicine and Biology)
- Published
- 2022
16. Development of a highly-specific 18F-labeled irreversible positron emission tomography tracer for monoacylglycerol lipase mapping
- Author
-
Yiding Zhang, Jiahui Chen, Atsuto Hiraishi, Steven H. Liang, Tomoteru Yamasaki, Ming-Rong Zhang, Benjamin F. Cravatt, Wakana Mori, Lee Josephson, Richard Van, Jian Rong, Tuo Shao, Masayuki Fujinaga, Michael A. Schafroth, Akiko Hatori, Thomas Lee Collier, Jiyun Sun, Qingzhen Yu, Kuan Hu, Zhen Chen, Yihan Shao, and Daisuke Ogasawara
- Subjects
Central nervous system ,RM1-950 ,Pharmacology ,Epileptogenesis ,Central nervous system (CNS) ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,medicine ,Radioligand ,General Pharmacology, Toxicology and Pharmaceutics ,Neuroinflammation ,030304 developmental biology ,0303 health sciences ,medicine.diagnostic_test ,Serine hydrolase ,Fluorine-18 ,Monoacylglycerol lipase ,medicine.anatomical_structure ,Monoacylglycerol lipase (MAGL) ,chemistry ,Positron emission tomography ,030220 oncology & carcinogenesis ,Arachidonic acid (AA) ,Positron emission tomography (PET) ,Arachidonic acid ,Therapeutics. Pharmacology ,2-Arachidonylglycerol (2-AG) - Abstract
As a serine hydrolase, monoacylglycerol lipase (MAGL) is principally responsible for the metabolism of 2-arachidonoylglycerol (2-AG) in the central nervous system (CNS), leading to the formation of arachidonic acid (AA). Dysfunction of MAGL has been associated with multiple CNS disorders and symptoms, including neuroinflammation, cognitive impairment, epileptogenesis, nociception and neurodegenerative diseases. Inhibition of MAGL provides a promising therapeutic direction for the treatment of these conditions, and a MAGL positron emission tomography (PET) probe would greatly facilitate preclinical and clinical development of MAGL inhibitors. Herein, we design and synthesize a small library of fluoropyridyl-containing MAGL inhibitor candidates. Pharmacological evaluation of these candidates by activity-based protein profiling identified 14 as a lead compound, which was then radiolabeled with fluorine-18 via a facile SNAr reaction to form 2-[18F]fluoropyridine scaffold. Good blood–brain barrier permeability and high in vivo specific binding was demonstrated for radioligand [18F]14 (also named as [18F]MAGL-1902). This work may serve as a roadmap for clinical translation and further design of potent 18F-labeled MAGL PET tracers.
- Published
- 2021
17. [11C]phosgene: Synthesis and application for development of PET radiotracers
- Author
-
Fukumura, Toshimitsu, Mori, Wakana, Ogawa, Masanao, Fujinaga, Masayuki, Ming-Rong, Zhang, Toshimitsu, Fukumura, Wakana, Mori, Masanao, Ogawa, Masayuki, Fujinaga, and Zhang, Ming-Rong
- Abstract
Carbon-11-labeled phosgene ([11C]phosgene, [11C]COCl2) is a useful labeling agent that connects two heteroatoms by inserting [11C]carbonyl (11C=O) function in carbamates, ureas, and carbonates, which are components of biologically important heterocyclic compounds and functional groups in drugs as a linker of fragments with in vivo stability. Development of 11C-labeled PET tracers has been performed using [11C]phosgene as a labeling agent. However, [11C]phosgene has not been frequently used for 11C-labeling because preparation of [11C]phosgene required dedicated synthesis apparatus (not commercially available) and had problems in reproducibility and reliability. In our laboratory, an improved method for synthesizing [11C]phosgene using a carbon tetrachloride detection tube kit in environmental air analysis and the automated synthesis system for preparing [11C]phosgene have been developed in 2009. This apparatus has been used for routine synthesis of 11C-labeled tracers 1–4 times/week. Using [11C]phosgene we have developed and produced many PET radiotracers containing [11C]urea and [11C]carbamate moieties. In this review, we report the performance of our method for preparing [11C]phosgene, including automated synthesis apparatus developed in house, and the application of [11C]phosgene for development and production of 11C-labeled PET tracers.
- Published
- 2021
18. [11C]phosgene: Synthesis and application for development of PET radiotracers
- Author
-
Masayuki Fujinaga, Wakana Mori, Masanao Ogawa, Ming-Rong Zhang, and Toshimitsu Fukumura
- Subjects
Cancer Research ,Chemistry ,Improved method ,Combinatorial chemistry ,030218 nuclear medicine & medical imaging ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,030220 oncology & carcinogenesis ,Urea ,Carbon tetrachloride ,Molecular Medicine ,Radiology, Nuclear Medicine and imaging ,Pet tracer ,Phosgene ,Synthesis system - Abstract
Carbon-11-labeled phosgene ([11C]phosgene, [11C]COCl2) is a useful labeling agent that connects two heteroatoms by inserting [11C]carbonyl (11C=O) function in carbamates, ureas, and carbonates, which are components of biologically important heterocyclic compounds and functional groups in drugs as a linker of fragments with in vivo stability. Development of 11C-labeled PET tracers has been performed using [11C]phosgene as a labeling agent. However, [11C]phosgene has not been frequently used for 11C-labeling because preparation of [11C]phosgene required dedicated synthesis apparatus (not commercially available) and had problems in reproducibility and reliability. In our laboratory, an improved method for synthesizing [11C]phosgene using a carbon tetrachloride detection tube kit in environmental air analysis and the automated synthesis system for preparing [11C]phosgene have been developed in 2009. This apparatus has been used for routine synthesis of 11C-labeled tracers 1-4 times/week. Using [11C]phosgene we have developed and produced many PET radiotracers containing [11C]urea and [11C]carbamate moieties. In this review, we report the performance of our method for preparing [11C]phosgene, including automated synthesis apparatus developed in house, and the application of [11C]phosgene for development and production of 11C-labeled PET tracers.
- Published
- 2021
- Full Text
- View/download PDF
19. Theranostics of melanoma targeting metabotropic glutamate receptor 1 with a novel small-molecular radiopharmaceutical pair
- Author
-
Lin, Xie, Masayuki, Hanyu, Masayuki, Fujinaga, Zhang, Lulu, Zhang, Yiding, Wakana, Mori, Kuan, Hu, Katsuyuki, Minegishi, Kotaro, Nagatsu, Kazunori, Kawamura, and Zhang, Ming-Rong
- Abstract
Objectives: Metabotropic glutamate receptor 1 (GRM1) is aberrantly overexpressed in a wide variety of human solid tumors, such as melanoma,but not in normal peripheral organs (1). Therefore, GRM1 can be a widely–applicable target for the theranostics in oncology. Here, a novel small-molecular radiopharmaceutical pair, 3-iodo- N-[4-[6-(methylamino)pyrimidin-4-yl]-1,3-thiazol-2-yl]-N-[11C]methylbenzamide ([11C]1) and 3-211At-astato-N-[4-[6-(methylamino)pyrimidin-4-yl]-1,3-thiazol-2-yl]-N-methylbenzamide ([211At]1), was designed and developed to target the GRM1 for the theranostics in melanomas (Fig.1). Method: [ 11C]1 was synthesized by reacting a N-desmethyl precursor with [11C]CH3OTf in the presence of NaOH at room temperature for 5 min. Radiolabeling of [211At]1 was performed by reaction of aryl tin precursor with NCS-containing 211At/MeOH solution. The theranostic potentials of the radiopharmaceutical pair were explored for PET imaging and radiotherapy in GRM1-positive B16F10 melanoma-bearing mice. Results: [ 11C]1 and [211At]1 were obtained with a radiochemical purity of greater than 99% and radiochemical yields of 19 ± 8 % and 46 ± 3%, respectively, based on the total radioactivity of used radionuclides. In vivo PET imaging of [ 11C]1 clearly visualized the targeted melanomas with a good tumor-to-background contrast (Fig. 1A). Ex vivo biodistribution study verified the persistent increase of [ 11C]1, which reached 12.29 ± 2.44 %ID/g tissue at 90 min in the targeted melanomas, and rapidly cleared from nontarget organs after intravenous injection. In the therapeutic studies, [211At]1 exhibited unequivocal and durable antitumor efficacy with only a single treatment (2.96 MBq) in the melanoma model, compared to the controls (0.38 ± 0.02 cm3 vs. 7.88 ± 1.28 cm3 at 13 days post-therapy) (Fig. 1B). No decrease in body weight, and no liver and kidney damage were observed through the examination period in melanoma mice injected with 2.96 MBq of [211At]1. Conclusion: The novel small-molecular radiopharmaceutical pair successfully visualized the GRM1-positive melanomas by [ 11C]1 with high contrast PET images, and further treated by [211At]1 without significant toxicity. The results highlight the good potential of using [ 11C]1 and [211At]1 as theranostic agents for the management of GRM1-positive tumors and should be further investigated in the theranostic field of oncology in the clinic., SNMMI2022
- Published
- 2022
20. Synthesis and evaluation of PET ligands for monoacylglycerol lipase in brain
- Author
-
Wakana, Mori, Kurihara, Yusuke, Tomoteru, Yamasaki, Akiko, Hatori, Zhang, Yiding, Masayuki, Fujinaga, and Zhang, Ming-Rong
- Abstract
Objectives: Monoacylglycerol lipase (MAGL) is an attractive target in development of pharmaceuticals for central nervous system disorders and neuroinflammation. So far, to visualize MAGL, we have developed several irreversible-type PET ligands [1,2]. Of these, PET ligands containing azetidine moieties have showed unexpected brain kinetics different from that of the typical irreversible-type PET ligand. Here, to understand relationship between the chemical structure and brain kinetics, we synthesized three new ligands including the azetidine ([11C]1), pyrrolidine ([11C]2) and piperidine ([11C]3) moieties and compared their kinetics in the brain. Methods: Compounds 1–3 and their precursors 5–7 for radiolabeling were synthesized from various pyrazole heterocyclic compounds in 6–7 steps. Synthesis of [11C]1–3 was performed using a home-made automated synthesizer equipped with [11C]COCl2 that was prepared from cyclotron-produced [11C]CO2[3]. As a general procedure exemplified for [11C]1 (Scheme 1), [11C]COCl2 was trapped into a solution of 1,1,1,3,3,3-hexafluoro-2-propanol (4; 3.1 μL) and 1,2,2,6,6-pentamethylpiperidine (PMP; 5.4 μL) in THF (200 μL) at 0 °C. The reaction mixture was heated at 30 °C for 3 min. A solution of corresponding pyrazole 5–7 (1.0 mg) and PMP (2.2 μL) in THF (200 μL) was added into the mixture and this mixture was heated at 30 °C for 3 min and then the solvent was removed at 80 °C for 5 min. The residue dissolved in the preparative HPLC solvent was applied to the semi-preparative HPLC column. Brain kinetics of these radioligands were evaluated by small-animal PET imaging using rats. Results: Starting from 22–24 GBq of [11C]CO2, [11C]1–3 were synthesized with 16–24% radiochemical yields (decay-corrected), 49–60 GBq/μmol molar activity, and ≥98% radiochemical purity. The average total synthesis time from the end of bombardment was 44 min. PET imaging with [11C]1 showed high radioactive uptake with a moderate clearance in MAGL-rich brain regions. In contrast, although radioactive uptake of [11C]3 in the brain indicated similar distribution to that of [11C]1, radioactive clearance from brain was not detected during PET scan. Conclusions: We successfully synthesized several 11C-labeled pyrazole heterocyclic compounds and compared their brain kinetics by PET imaging using rats. The result of present work suggested that the big heterocycle PET ligand for MAGL was more stable in vivo.
- Published
- 2022
21. Automated radiosynthesis of the 18F-labeled BF2-chelated tetraaryl-azadipyrromethenes photosensitizer using isotopic exchange
- Author
-
Kazunori, Kawamura, Tomoteru, Yamasaki, Atsuto, Hiraishi, Zhang, Yiding, Lin, Xie, Masayuki, Fujinaga, Wakana, Mori, Kurihara, Yusuke, Masanao, Ogawa, Kaito, Tsukagoe, Nobuki, Nengaki, and Zhang, Ming-Rong
- Abstract
Objectives: A family of the BF2-chelated tetraaryl-azadipyrromethenes (ADPMs) was developed as a nonporphyrin photosensitizer (PS) for photodynamic therapy (PDT) [1]. Among ADPMs, ADPM06 displayed excellent photochemical and photophysical properties [1]. In addition, PDT using ADPM06 elicited impressive complete response rates in various tumor models when a short drug-light interval was applied [2]. Molecular imaging is a promising PDT planning and monitoring tool, and the PS biodistribution is a relevant issue for PDT planning that radiolabeled PSs may address efficiently. To evaluate efficiency for PDT using ADPM06 and also side effects of ADPM, we synthesized [18F]ADPM06 using an automated 18F-labeling synthesizer, and evaluated its in vivo properties. Methods: [18F]ADPM06 was synthesized using an automated 18F-labeling synthesizer by Lewis acid-assisted isotopic 18F-19F exchange [3]. The [18F]F– was extracted from a Sep-Pak Accell Plus QMA Carbonate Plus Light cartridge with a mixture of tetrabutylanmmonium bicarbonate aqueous solution and acetonitrile. The solution was concentrated by evaporation at 100 °C for 10 min under nitrogen gas flow. After the reaction vessel was cooled, the mixture of ADPM06 in acetonitrile and tin(IV) chloride (SnCl4) solution was added to the reaction vessel, and then was agitated using magnetic stirrer at room temperature for 10 min. After the reaction, the mixture was diluted with water for injection, and transferred to the injector for semi-preparative radio-HPLC. The HPLC fractions were collected in a flask, to which Tween 80 in ethanol was added prior to radiosynthesis. The solution was subsequently evaporated to dryness and the residue was dissolved in physiological saline. The product was analyzed by HPLC with radioactivity and UV-VIS detection. The in vivo biodistribution study was performed using mice. Results: To radio-synthesize efficiently [18F]ADPM06, we performed semi-automated radiosynthesis. By increasing the concentration of SnCl4 from 100 to 400 μmol, the radiochemical conversion (RCC) of [18F]ADPM06 from [18F]TBAF was increased until 60%. Also, the RCC using 0.8 μmol of ADPM06 was 1.2-fold higher than that using 0.4 μmol of ADPM06. In the radiosynthesis using an automated 18F-labeling synthesizer, we successfully synthesized [18F]ADPM06 for in vivo applications. The radiochemical yield (RCY) from [18F]F- was 13 ± 2.7 % (n = 5; 0.4 μmol of ADPM06, 200 μmol of SnCl4) at the end of irradiation. The radiosynthesis time was within 60 min, and radiochemical purity remained >95% after maintaining it for 1 hour after the end of synthesis. In the biodistribution study within 120 min after the injection, radioactivity levels in heart, lung, liver, pancreas, spleen, kidney, small intestine, muscle, and brain gradually decreased after initial uptake. Conclusions: We enabled to synthesize [18F]ADPM06 using an automated 18F-labeling synthesizer, and to evaluate biodistribution of [18F]ADPM06 in mice.
- Published
- 2022
22. Demonstration of intracellular pH-weighting PET imaging using a new-type PET probe responsible for monoacylglycerol lipase activity in the brain
- Author
-
Tomoteru, Yamasaki, Wakana, Mori, Ohkubo, Takayuki, Atsuto, Hiraishi, Kurihara, Yusuke, Nobuki, Nengaki, and Zhang, Ming-Rong
- Abstract
Objectives: The brain acidosis is caused by intracellular hyper-accumulation of acidic sources (H+, lactate, and carbonic acid) by switching the cellular energy metabolism from aerobic to anaerobic by the hypoxia. Such intra cellular acidosis give curucial injury to central nurves system of the brain. Therefore, monitoring intracellular pH would be very important to diagnosis neuronal condition. Recently, covalent inhibitors for monoacylglycerol lipase (MAGL), an enzyme intracellur lacated on neuron and astrocyte in the brain and regulates endocannabinoid system, were identified by Bulter et al [1]. Among these inhibitors, 1,1,1,3,3,3-hexafluoropropan-2-yl 3-(1-phenyl-1H-pyrazol-3-yl)azetidine-1-carboxylate (1) showed reversible inhibitory effect to MAGL. The purpose of this study is to establish quantification method of hydrolysis rate of compound 1 mediated by MAGL and to demonstrate pH-weighted PET imaging in the brain of ischemic rat. Methods: To estimate interaction between compound 1 and MAGL, docking simulations were conducted comparing to similar chemical structural irreversible-type inhibitor (2). In addition, to evaluate influents of pH shifts, molecular dynamics (MD) simulations of compound 1 were also performed under the neutral (pH 7) or acidic (pH 6) conditions. Radiosynthesis of [11C]1 and [11C]2 was described in another presentation in this meeting (Mori W, et a1.). To confirm MAGL-hydrolysis of [11C]1, in vitro assessments using rat brain homogenates were conducted. PET imaging with [11C]1 was carried out using middle cerebral artery occlusion (MCAO) rat as an acute hypoxia model and hydrolysis rate (KH) of [11C]1 with MAGL was estimated by monoexponential fitting on time-activity curves of ipsilateral region. Results: MD simulations predicted that azetidine carbamate moiety of 1 was easily hydrolyzed by MAGL due to close distance from water molecule, compared to 2 containing piperidine carbamate. Moreover, the acylated azetidine in 1 has been shown to react differently than piperidine ring size amide in 2 due to decreased planarity in the amide moiety itself, conveyed by the ring strain associated with the azetidine itself. Additionally, it was simulated that the hydrolysis rate of 1 would be slower under the acidic condition because of changing interaction of 1 against water molecule. In vitro assessments showed that generation rate of 11CO2, the final product derived from hydrolysis of [11C]1, would become slower depending on pH shifts. In PET study with [11C]1 using MCAO rat, KH value in ipsilateral region was significantly slower than that in contralateral region. Conclusions: We successfully established the method for quantifying hydrolysis rate of MAGL using new-type PET probe and demonstrated pH-weighted imaging in vivo.
- Published
- 2022
23. Upregulation of striatal metabotropic glutamate receptor subtype 1 (mGluR1) in rats with excessive glutamate release induced by N-acetylcysteine
- Author
-
Tomoteru, Yamasaki, Maki, Okada, Atsuto, Hiraishi, Wakana, Mori, Zhang, Yiding, Masayuki, Fujinaga, Hidekatsu, Wakizaka, Kurihara, Yusuke, Nobuki, Nengaki, Zhang, Ming-Rong, Tomoteru, Yamasaki, Maki, Okada, Atsuto, Hiraishi, Wakana, Mori, Zhang, Yiding, Masayuki, Fujinaga, Hidekatsu, Wakizaka, Kurihara, Yusuke, Nobuki, Nengaki, and Zhang, Ming-Rong
- Abstract
Aim of this study is to investigate the changes in expression of metabotropic glutamate (Glu) receptor subtype 1 (mGluR1), a key molecule involved in neuroexcitetoxicity, during excessive Glu release in the brain by PET imaging. An animal model of excessive Glu release in the brain was produced by intraperitoneally implanting an Alzet osmotic pump containing N-acetylcysteine (NAC), an activator of the cysteine/Glu antiporter, into the abdomen of rats. Basal Glu concentration in the brain was measured by microdialysis, which showed that basal Glu concentration in NAC-treated rats (0.31 µM) was higher than that in saline-treated rats (0.17 µM) at day 7 after the implantation of the osmotic pump. Similarly, PET studies with [11C]ITDM, a useful radioligand for mGluR1 imaging exhibited that the striatal binding potential (BPND) of [11C]ITDM for mGluR1 in PET assessments was increased in NAC-treated animals at day 7 after implantation (2.30) compared with before implantation (1.92). The dynamic changes in striatal BPND during the experimental period were highly correlated with basal Glu concentration. In conclusion, density of mGluR1 is rapidly upregulated by increases in basal Glu concentration, suggesting that mGluR1 might to be a potential biomarker of abnormal conditions in the brain.
- Published
- 2022
24. Simple and rapid automated synthesis of [11C]L-glutamine
- Author
-
Masayuki, Fujinaga, Kurihara, Yusuke, Masanao, Ogawa, Katsushi, Kumata, Wakana, Mori, Zhang, Ming-Rong, Masayuki, Fujinaga, Kurihara, Yusuke, Masanao, Ogawa, Katsushi, Kumata, Wakana, Mori, and Zhang, Ming-Rong
- Abstract
Objectives: L-Glutamine is involved in various metabolic processes. Recently, glutaminolysis has been recognized to play important roles in activation of many cancer cells. Thus, [11C]L-glutamine is a useful PET tracer for elucidating the mechanisms of glutamine utilization in cancer cells. To date, synthesis of [11C]L-glutamine has been reported by several groups [1]. However, their synthesis methods required long synthesis time (60 min) and complicated synthesis device for separating and purifying the reaction mixture by solid phase extractions and HPLC system. Herein, we developed a simple and rapid method for synthesizing [11C]L-glutamine using a fully-automated synthesis system without HPLC purification. Methods: [11C]L-glutamine was synthesized by using a fully-automated synthesis system (Scheme 1). The precursor 1 was purchased from commercial sources. Before irradiation of [11C]CO2, the mixture of 18-crown-6 (8 mg in 900 L of CH3CN) and Cs2CO3 (3 mg in 150 L of water) was azeotropically dried in reaction vial and then added CH3CN (300 L). After the automatically produced [11C]HCN was trapped into the above solution, precursor 1 (3.5 mg) in CH3CN (300 L) was added and the reaction mixture was heated at 90 oC for 8 min, followed by complete removal of the reaction solvent. TFA-H2SO4 (4 : 1, 500 L) was added to the residue and this reaction mixture was heated at 80 oC for 5 min. After the reaction, the mixture was diluted with diethyl ether (1.5 mL) and passed through a silica plus Sep-Pak cartridge to trap [11C]L-glutamine. The cartridge was washed with diethyl ether (10 mL) and then eluted with phosphate buffer (5 mL). The [11C]L-glutamine solution was obtained by removal of diethyl ether from the eluant and was adjusted to suitable pH value for use by addition of phosphate buffer (4 mL). The final radioactive product was analyzed by HPLC with radioactivity and UV-VIS detectors. Scheme 1. Syntheses of [11C]L-glutamine Results: The [11C]cyanation of precu
- Published
- 2022
25. The fully-automated synthesis of [11C]CF3-aryl derivatives with [11C]fluoroform
- Author
-
Masayuki, Fujinaga, Masanao, Ogawa, Nobuki, Nengaki, Katsushi, Kumata, Wakana, Mori, Zhang, Ming-Rong, Masayuki, Fujinaga, Masanao, Ogawa, Nobuki, Nengaki, Katsushi, Kumata, Wakana, Mori, and Zhang, Ming-Rong
- Abstract
Objectives: The trifluoromethyl group can improve physicochemical properties, such as metabolic stability, lipophilicity and pharmacokinetics of targeted probes. Radiolabeling by trifluoromethyl group is an attractive method to synthesize a useful PET probe. Recently, Haskali et al. have developed the synthetic method of [11C]fluoroform from [11C]CH4 with a CoF3 column and the application route of [11C]fluoroform for various trifluoromethylation reactions [1]. However, those application reactions were not automated and the synthesis of [11C]CF3-aryl derivatives is difficult because preparation of [11C]CuCF3 as a trifluoromethylation reagent was conducted in a glove box to avoid air and moisture. Herein, we developed a synthetic method of [11C]CF3-aryl derivatives using a fully-automated system equipped with [11C]CuCF3 without glove box. Methods: [11C]Trifluoromethylation of aryl precursor was performed by using a fully-automated synthesis system (Scheme 1). [11C]Fluoroform was produced by passing [11C]CH4 through a column that was precoated with cobalt(III) fluoride (24 g) and heated at 350 oC. CuOtBu was generated by mixing t-BuOK (15 mol) with CuBr (5 mol) in DMF (0.3 mL) in a sealed vial under N2. Preparation of [11C]CuCF3 was performed by bubbling [11C]fluoroform into the CuOtBu solution at -45 oC. After the radioactivity of [11C]fluoroform reached a plateau, the reaction mixture was warmed to room temperature for 2 min and then added Et3N-3HF (0.82 L) in DMF (0.2 mL). The trifluoromethylation was performed by the reaction of [11C]CuCF3 with aryl precursor (10 mg) at room temperature or 130 oC for 5 min. After the preparative HPLC mobile phase (1.0 mL) was added, the reaction mixture was applied to the HPLC system for separation. The HPLC fraction of [11C]1 was directly collected in a vial. The radioactive product was analyzed by HPLC with radioactivity and UV-VIS detectors. Scheme 1. Preparation of [11C]CuCF3 and synthesis of [11C]CF3-aryl derivatives Result
- Published
- 2022
26. Development of a highly-specific 18F-labeled irreversible positron emission tomography tracer for monoacylglycerol lipase mapping
- Author
-
Chen, Zhen, Mori, Wakana, Rong, Jian, A. Schafroth, Michael, Shao, Tuo, S. Van, Richard, Ogasawara, Daisuke, Yamasaki, Tomoteru, Hiraishi, Atsuto, Hatori, Akiko, Chen, Jiahui, Zhang, Yiding, Kuan, Hu, Fujinaga, Masayuki, Sun, Jiyun, Yu, Qingzhen, L. Collier, Thomas, Shao, Yihan, F. Cravatt, Benjamin, Josephson, Lee, Ming-Rong, Zhang, Liang, Huan, Wakana, Mori, Tomoteru, Yamasaki, Atsuto, Hiraishi, Akiko, Hatori, Masayuki, Fujinaga, and Zhang, Ming-Rong
- Abstract
Monoacylglycerol lipase (MAGL) is a serine hydrolase, which metabolizes 2-arachidonoylglycerol (2-AG) to arachidonic acid (AA) in the central nervous system (CNS). Dysfunction of MAGL has been associated with multiple CNS disorders and symptoms, including neuroinflammation, cognitive impairment, epileptogenesis, nociception and neurodegenerative diseases. Inhibition of MAGL provides a promising therapeutic direction for the treatment of these conditions, and a MAGL positron emission tomography (PET) probe would greatly facilitate preclinical and clinical development of MAGL inhibitors. Herein, we design and synthesize a small library of fluoropyridyl-containing MAGL inhibitor candidates. Pharmacological evaluation of these candidates by activity-based protein profiling identified 14 as a lead compound, which was then radiolabeled with fluorine-18 via a facile SNAr reaction to form 2-[18F]fluoropyridinyl scaffold. Excellent blood−brain barrier penetration and high in vivo binding specificity was validated for radioligand [18F]14 (also named as [18F]MAGL-1902). This work may serve as a roadmap for clinical translation and further design of potent 18F-labeled MAGL PET tracers. KEY WORDS Monoacylglycerol lipase (MAGL); Central nervous system (CNS); 2-Arachidonylglycerol (2-AG); Arachidonic acid (AA); Positron emission tomography (PET); Fluorine-18
- Published
- 2020
27. Identification and Development of a New Positron Emission Tomography Ligand 4-(2-Fluoro-4-[11C]methoxyphenyl)-5-((1-methyl-1H-pyrazol-3-yl)methoxy)picolinamide for Imaging Metabotropic Glutamate Receptor Subtype 2 (mGlu2)
- Author
-
Xiaoyun Deng, Kuan Hu, Douglas J. Sheffler, Masayuki Fujinaga, Zhen Chen, Ming-Rong Zhang, Yihan Shao, Richard Van, Steven H. Liang, Wakana Mori, Nicholas D. P. Cosford, Xiaofei Zhang, Katsushi Kumata, Nobuki Nengaki, Tomoteru Yamasaki, Akiko Hatori, Masanao Ogawa, Lin Xie, Yiding Zhang, and Hidekatsu Wakizaka
- Subjects
medicine.diagnostic_test ,Ligand ,Chemistry ,Stereochemistry ,Allosteric regulation ,In vivo ,Metabotropic glutamate receptor ,Positron emission tomography ,Drug Discovery ,Lipophilicity ,medicine ,Molecular Medicine ,Metabotropic glutamate receptor 2 ,Receptor - Abstract
Metabotropic glutamate receptor 2 (mGlu2) is a known target for treating several central nervous system (CNS) disorders. To develop a viable positron emission tomography (PET) ligand for mGlu2, we identified new candidates 5a-i that are potent negative allosteric modulators (NAMs) of mGlu2. Among these candidates, 4-(2-fluoro-4-methoxyphenyl)-5-((1-methyl-1H-pyrazol-3-yl)methoxy)picolinamide (5i, also named as [11C]MG2-1812) exhibited high potency, high subtype selectivity, and favorable lipophilicity. Compound 5i was labeled with positron-emitting carbon-11 (11C) to obtain [11C]5i in high radiochemical yield and high molar activity by O-[11C]methylation of the phenol precursor 12 with [11C]CH3I. In vitro autoradiography with [11C]5i showed heterogeneous radioactive accumulation in the brain tissue sections, ranked in the order: cortex > striatum > hippocampus > cerebellum ≫ thalamus > pons. PET study of [11C]5i indicated in vivo specific binding of mGlu2 in the rat brain. Based on the [11C]5i scaffold, further optimization for new candidates is underway to identify a more suitable ligand for imaging mGlu2.
- Published
- 2020
- Full Text
- View/download PDF
28. Identification and Development of a New Positron Emission Tomography Ligand 4-(2-Fluoro-4-[11C]methoxyphenyl)-5-((1-methyl-1H-pyrazol-3-yl)methoxy)picolinamide for Imaging Metabotropic Glutamate Receptor Subtype 2 (mGlu2)
- Author
-
Yamasaki, Tomoteru, Zhang, Xiaofei, Kumata, Katsushi, Zhang, Yiding, Deng, Xiaoyun, Fujinaga, Masayuki, Chen, Zhen, Mori, Wakana, Kuan, Hu, Wakizaka, Hidekatsu, Hatori, Akiko, Xie, Lin, Ogawa, Masanao, Nengaki, Nobuki, Van, Richard, Shao, Yihan, J. Sheffler, Douglas, D. P. Cosford, Nicholas, Liang, Huan, Ming-Rong, Zhang, Tomoteru, Yamasaki, Katsushi, Kumata, Masayuki, Fujinaga, Wakana, Mori, Hidekatsu, Wakizaka, Akiko, Hatori, Lin, Xie, Masanao, Ogawa, Nobuki, Nengaki, and Zhang, Ming-Rong
- Abstract
Metabotropic glutamate receptor 2 (mGlu2) is a known target for treating several central nervous system (CNS) disorders. To develop a viable positron emission tomography (PET) ligand for mGlu2, we identified new candidates 5a–i that are potent negative allosteric modulators (NAMs) of mGlu2. Among these candidates, 4-(2-fluoro-4-methoxyphenyl)-5-((1-methyl-1H-pyrazol-3-yl)methoxy)picolinamide (5i, also named as [11C]MG2-1812) exhibited high potency, high subtype selectivity, and favorable lipophilicity. Compound 5i was labeled with positron-emitting carbon-11 (11C) to obtain [11C]5i in high radiochemical yield and high molar activity by O-[11C]methylation of the phenol precursor 12 with [11C]CH3I. In vitro autoradiography with [11C]5i showed heterogeneous radioactive accumulation in the brain tissue sections, ranked in the order: cortex > striatum > hippocampus > cerebellum ≫ thalamus > pons. PET study of [11C]5i indicated in vivo specific binding of mGlu2 in the rat brain. Based on the [11C]5i scaffold, further optimization for new candidates is underway to identify a more suitable ligand for imaging mGlu2.
- Published
- 2020
29. Development of an In Vivo Method to Estimate Effective Drug Doses and Quantify Fatty Acid Amide Hydrolase in Rodent Brain using Positron Emission Tomography Tracer [11C]DFMC
- Author
-
Masayuki Fujinaga, Tomoyuki Ohya, Nobuki Nengaki, Ming-Rong Zhang, Yiding Zhang, Tomoteru Yamasaki, Hidekatsu Wakizaka, Wakana Mori, and Tatsuya Kikuchi
- Subjects
0301 basic medicine ,Pharmacology ,Drug doses ,Chromatography ,medicine.diagnostic_test ,Chemistry ,Rat brain ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,In vivo ,Fatty acid amide hydrolase ,Positron emission tomography ,TRACER ,medicine ,Molecular Medicine ,Graphical analysis ,030217 neurology & neurosurgery ,ED50 - Abstract
Fatty acid amide hydrolase (FAAH) is a key enzyme in the endocannabinoid system. N-(3,4-Dimethylisoxazol-5-yl)piperazine-4-[4-(2-fluoro-4-[11C]methylphenyl)thiazol-2-yl]-1-carboxamide ([11C]DFMC) was developed as an irreversible-type positron emission tomography (PET) tracer for FAAH. Here, we attempted to noninvasively estimate rate constant k3 (rate of transfer to the specifically-bound compartment) as a direct index for FAAH in the rat brain. First, the two-tissue compartment model analysis including three parameters [K1−k3, two-tissue compartment model for the irreversible-type radiotracer (2TCMi)] in PET study with [11C]DFMC was conducted, which provided 0.21 ± 0.04 ml·cm−3·min−1 of the net uptake value (Ki), an indirect index for FAAH, in the FAAH-richest region (the cingulate cortex). Subsequently, to noninvasively estimate Ki value, the reference model analysis (Patlak graphical analysis reference model) was tried using a time-activity curve of the spinal cord. In that result, the noninvasive Ki value (KREF) was concisely estimated with high correlation (r > 0.95) to Ki values based on 2TCMi. Using estimated KREF value, we tried to obtain calculated-k3 based on previously defined equations. The calculated k3 was successfully estimated with high correlation (r = 0.95) to direct k3 in 2TCMi. Finally, the dose relationship study using calculated k3 demonstrated that in vivo ED50 value of [3-(3-carbamoylphenyl)phenyl] N-cyclohexylcarbamate, a major inhibitor of FAAH, was 66.4 µg/kg in rat brain. In conclusion, we proposed the calculated k3 as an alternative index corresponding to regional FAAH concentrations and suggested that PET with [11C]DFMC enables occupancy study for new pharmaceuticals targeting FAAH. SIGNIFICANCE STATEMENT In the present study, we proposed calculated k3 as an alternative index corresponding with fatty acid amide hydrolase concentration. By using calculated k3, in vivo ED50 of [3-(3-carbamoylphenyl)phenyl] N-cyclohexylcarbamate was successfully estimated to be 66.4 µg/kg for rats. Thus, we demonstrated the pharmacological utility of positron emission tomography with N-(3,4-dimethylisoxazol-5-yl)piperazine-4-[4-(2-fluoro-4-[11C]methylphenyl)thiazol-2-yl]-1-carboxamide.
- Published
- 2020
- Full Text
- View/download PDF
30. Development of an In Vivo Method to Estimate Effective Drug Doses and Quantify Fatty Acid Amide Hydrolase in Rodent Brain using Positron Emission Tomography Tracer N-(3,4-dimethylisoxazol-5-yl)piperazine-4-[4-(2-fluoro-4-[11C]methylphenyl)thiazol-2-yl]-1-carboxamide ([11C]DFMC)
- Author
-
Yamasaki, Tomoteru, Ohya, Tomoyuki, Mori, Wakana, Zhang, Yiding, Wakizaka, Hidekatsu, Nengaki, Nobuki, Fujinaga, Masayuki, Kikuchi, Tatsuya, Ming-Rong, Zhang, Tomoteru, Yamasaki, Tomoyuki, Ohya, Wakana, Mori, Hidekatsu, Wakizaka, Nobuki, Nengaki, Masayuki, Fujinaga, Tatsuya, Kikuchi, and Zhang, Ming-Rong
- Subjects
lipids (amino acids, peptides, and proteins) - Abstract
Fatty acid amide hydrolase (FAAH) is a key enzyme in the endocannabinoid system. N-(3,4-Dimethylisoxazol-5-yl)piperazine-4-[4-(2-fluoro-4-[11C]methylphenyl)thiazol-2-yl]-1-carboxamide ([11C]DFMC) was developed as an irreversible-type positron emission tomography (PET) ligand for FAAH. Here, we attempted to noninvasively estimate rate constant k3 as a direct index for FAAH in the rat brain. First, the two-tissue compartment model analysis including three parameters (K1–k3, 2TCMi) in PET study with [11C]DFMC was conducted, which provided 0.21 ± 0.04 mL·cm−3·min−1 of the net uptake value (Ki), an indirect index for FAAH, in the FAAH-richest region (the cingulate cortex). Subsequently, to noninvasively estimate Ki value, the reference model analysis (Patlak Reference, PGAREF) was tried using a time-activity curve of the spinal cord. In that result, the noninvasive Ki value (KREF) was concisely estimated with high correlation (r > 0.95) to Ki values based on 2TCMi. Using estimated KREF value, we tried to obtain calculated-k3 based on previously defined equations. The caluculated-k3 was successfully estimated with high correlation (r = 0.95) to direct k3 in 2TCMi. Finally, the dose relationship study using calculated-k3 demonstrated that in vivo ED50 value of URB597, a major inhibitor of FAAH, was 66.4 µg/kg in rat brain. In conclusion, we proposed the calculated-k3 as an alternative index corresponding to regional FAAH concentrations and suggested that PET with [11C]DFMC enables occupancy study for new pharmaceuticals targeting FAAH.
- Published
- 2020
31. Radiosynthesis and evaluation of 4-(6-[18F]Fluoro-4-(5-isopropoxy-1H-indazol-3-yl)pyridin-2-yl)morpholine as a novel radiotracer candidate targeting leucine-rich repeat kinase 2
- Author
-
Masayuki Hanyu, Yiding Zhang, Nobuki Nengaki, Wakana Mori, Hong Zhang, Yasushi Hattori, Katsushi Kumata, Tomoteru Yamasaki, Masayuki Fujinaga, and Ming-Rong Zhang
- Subjects
Pharmacology ,Biodistribution ,010405 organic chemistry ,Stereochemistry ,Organic Chemistry ,Radiosynthesis ,Pharmaceutical Science ,01 natural sciences ,Biochemistry ,LRRK2 ,0104 chemical sciences ,Chemistry ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,chemistry ,In vivo ,Morpholine ,Drug Discovery ,Molecular Medicine ,Protecting group ,IC50 ,Lead compound ,030217 neurology & neurosurgery - Abstract
Mutations that increase leucine-rich repeat kinase 2 (LRRK2) activity in the brain are associated with Parkinson's disease. Here, we synthesized a novel compound 4-(6-fluoro-4-(5-isopropoxy-1H-indazol-3-yl)pyridin-2-yl)morpholine (FIPM) and labeled it with fluorine-18 ((18)F), to develop a positron emission tomography (PET) tracer for in vivo visualization of LRRK2 in the brain. FIPM showed high in vitro binding affinity for LRRK2 (IC(50) = 8.0 nM). [(18)F]FIPM was prepared in 5% radiochemical yield (n = 5), by inserting (18)F into a pyridine ring, followed by removal of the protecting group. After HPLC separation and formulation, [(18)F]FIPM was acquired with >97% radiochemical purity and 103–300 GBq μmol(–1) of molar activity at the end of radiosynthesis. Biodistribution and small-animal PET studies in mice indicated a low in vivo specific binding of [(18)F]FIPM. While [(18)F]FIPM presented limited potential as an in vivo PET tracer for LRRK2, we suggested that it can be used as a lead compound for developing new radiotracers with improved in vivo brain properties.
- Published
- 2020
- Full Text
- View/download PDF
32. Radiosynthesis and evaluation of acetamidobenzoxazolone based radioligand [11C]N′-MPB for visualization of 18 kDa TSPO in brain
- Author
-
Anjani K. Tiwari, Anil K. Mishra, Yiding Zhang, Wakana Mori, Neelam Kumari, Nobuki Nengaki, Masayuki Fujinaga, Ming-Rong Zhang, Tomoteru Yamasaki, Akiko Hatori, and Hong Zhang
- Subjects
Biodistribution ,medicine.diagnostic_test ,biology ,Microglia ,010405 organic chemistry ,Chemistry ,Radiosynthesis ,General Chemistry ,Pharmacology ,01 natural sciences ,Catalysis ,In vitro ,0104 chemical sciences ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Positron emission tomography ,Materials Chemistry ,medicine ,Translocator protein ,biology.protein ,Radioligand ,030217 neurology & neurosurgery ,Neuroinflammation - Abstract
The 18 kDa translocator protein (TSPO) is a viable target for imaging of inflammation in brain. In the recent past we have explored a pharmacophore skeleton acetamidobenzoxazolone for positron emission tomography (PET) imaging of TSPO expression in brain. Here we evaluated a new radioligand for visualization of TSPO, namely [11C]N-(2-methoxyoxyphenyl)-N-methyl-2-(5-nitro-2-oxobenzo[d]oxazol-3(2H)-yl)acetamide ([11C]N′-MPB). This PET ligand exhibited high binding affinity towards TSPO (Ki = 4.9 nM) and a suitable lipophilicity (log D) of 2.08 for brain imaging. A biodistribution study on mice showed high accumulation of radioactivity in TSPO-rich organs, such as the lungs, heart, kidneys, and adrenal glands. Metabolite analysis of rat brain homogenate showed 98% intact [11C]N′-MPB at 30 min after injection. To determine the specific binding of the radioligand with TSPO on neuroinflammation of the brain, in vitro autoradiography and PET studies were performed in an ischemic rat model. In vitro autoradiography indicated significantly increased binding on the ipsilateral side compared with that on the contralateral side of ischemic rat brains. This result was supported firmly by the contrast of radioactivity in PET images. Displacement experiments with PK11195 minimized the difference in radioactivity uptake between the two sides. In summary, [11C]N′-MPB is a potential PET imaging radioligand for TSPO and, consequently, for gaining more insight for up-regulation of microglia during neuroinflammation.
- Published
- 2020
- Full Text
- View/download PDF
33. Simple and rapid automated synthesis of L-[11C]glutamine
- Author
-
Masayuki Fujinaga, Yusuke Kurihara, Masanao Ogawa, Katsushi Kumata, Wakana Mori, and Ming-Rong Zhang
- Subjects
Cancer Research ,Molecular Medicine ,Radiology, Nuclear Medicine and imaging - Published
- 2022
- Full Text
- View/download PDF
34. Design, Synthesis, and Evaluation of 18F-Labeled Monoacylglycerol Lipase Inhibitors as Novel Positron Emission Tomography Probes
- Author
-
Jian Rong, Tuo Shao, Xiaofei Zhang, Jun-An Ma, Mikhail Papisov, Yiding Zhang, Genwei Zhang, Zhen Chen, Masayuki Fujinaga, Yihan Shao, Xiaoyun Deng, Daisuke Ogasawara, Lu Wang, Michael A. Schafroth, Ming-Rong Zhang, Akiko Hatori, Qingzhen Yu, Richard Van, Thomas Lee Collier, Hualong Fu, Kuan Hu, Vasily Belov, Benjamin F. Cravatt, Wakana Mori, Pilar Giffenig, Steven H. Liang, and Lee Josephson
- Subjects
Fluorine Radioisotopes ,Contrast Media ,01 natural sciences ,Article ,03 medical and health sciences ,Drug Discovery ,medicine ,Animals ,Spiro Compounds ,Tissue Distribution ,Tissue distribution ,Enzyme Inhibitors ,030304 developmental biology ,0303 health sciences ,Binding Sites ,medicine.diagnostic_test ,Extramural ,Chemistry ,Brain ,Ligand (biochemistry) ,Monoacylglycerol Lipases ,Rats ,0104 chemical sciences ,3. Good health ,Molecular Docking Simulation ,Monoacylglycerol lipase ,010404 medicinal & biomolecular chemistry ,Biochemistry ,Design synthesis ,Blood-Brain Barrier ,Positron emission tomography ,Drug Design ,Isotope Labeling ,Positron-Emission Tomography ,Molecular Medicine - Abstract
Dysfunction of monoacylglycerol lipase (MAGL) is associated with several psychopathological disorders, including drug addiction and neurodegenerative diseases. Herein we design, synthesize and evaluate several irreversible fluorine-containing MAGL inhibitors for positron emission tomography (PET) ligand development. Compound 6 (identified from a therapeutic agent) was advanced for (18)F-labeling via a novel spirocyclic iodonium ylide (SCIDY) strategy, which demonstrated high brain permeability and excellent specific binding. This work supports further development of novel (18)F-labeled MAGL PET probes.
- Published
- 2019
- Full Text
- View/download PDF
35. Design, Synthesis, and Evaluation of 18F‑Labeled Monoacylglycerol Lipase Inhibitors as Novel Positron Emission Tomography Probes
- Author
-
Chen, Zhen, Mori, Wakana, Fu, Hualong, A. Schafroth, Michael, Hatori, Akiko, Shao, Tuo, Zhang, Genwei, S. Van, Richard, Zhang, Yiding, Hu, Kuan, Fujinaga, Masayuki, Ming-Rong, Zhang, Wakana, Mori, Akiko, Hatori, Kuan, Hu, Masayuki, Fujinaga, and Zhang, Ming-Rong
- Abstract
Dysfunction of monoacylglycerol lipase (MAGL) is associated with several psychopathological disorders, including drug addiction and neurodegenerative diseases. Herein we design, synthesize, and evaluate several irreversible fluorine-containing MAGL inhibitors for positron emission tomography (PET) ligand development. Compound 6 (identified from a therapeutic agent) was advanced for 18F-labeling via a novel spirocyclic iodonium ylide (SCIDY) strategy, which demonstrated high brain permeability and excellent specific binding. This work supports further development of novel 18F-labeled MAGL PET probes.
- Published
- 2019
36. Radiosynthesis and evaluation of a novel monoacylglycerol lipase radiotracer: 1,1,1,3,3,3-hexafluoropropan-2-yl-3-(1-benzyl-1H-pyrazol-3-yl)azetidine-1-[11C]carboxylate
- Author
-
Akiko Hatori, Ming-Rong Zhang, Tomoteru Yamasaki, Lin Xie, Masayuki Fujinaga, Wakana Mori, Katsushi Kumata, Yiding Zhang, Kuan Hu, and Yusuke Kurihara
- Subjects
Biodistribution ,010405 organic chemistry ,Chemistry ,Stereochemistry ,Hydrochloride ,Organic Chemistry ,Clinical Biochemistry ,Azetidine ,Radiosynthesis ,Pharmaceutical Science ,01 natural sciences ,Biochemistry ,0104 chemical sciences ,Monoacylglycerol lipase ,010404 medicinal & biomolecular chemistry ,chemistry.chemical_compound ,In vivo ,Drug Discovery ,Lipophilicity ,Molecular Medicine ,Carboxylate ,Molecular Biology - Abstract
Monoacylglycerol lipase (MAGL) is a major serine hydrolase that hydrolyses 2-arachidonoylglycerol (2-AG) into arachidonic acid (AA) and glycerol in the brain. Because 2-AG and AA are endogenous biologically active ligands in the brain, the inhibition of MAGL is an attractive therapeutic target for neurodegenerative diseases. In this study, to visualize MAGL via positron emission tomography (PET), we report a new carbon-11-labeled radiotracer, namely 1,1,1,3,3,3-hexafluoropropan-2-yl-3-(1-benzyl-1H-pyrazol-3-yl)azetidine-1-[11C]carboxylate ([11C]6). Compound 6 exhibited high in vitro binding affinity (IC50 = 0.41 nM) to MAGL in the brain with a suitable lipophilicity (cLogD = 3.29). [11C]6 was synthesized by reacting 1,1,1,3,3,3-hexafluoropropanol (7) with [11C]phosgene ([11C]COCl2), followed by a reaction with 3-(1-benzyl-1H-pyrazol-3-yl)azetidine hydrochloride (8), which resulted in a 15.0 ± 6.8% radiochemical yield (decay-corrected, n = 7) based on [11C]CO2 and a 45 min synthesis time from the end of bombardment. A biodistribution study in mice showed high uptake of radioactivity in MAGL-rich organs, including the lungs, heart, and kidneys. More than 90% of the total radioactivity was irreversibly bound in the brain homogenate of rats 5 min and 30 min after the radiotracer injection. PET summation images of rat brains showed high radioactivity in all brain regions. Pretreatment with 6 or MAGL-selective inhibitor JW642 significantly reduced the uptake of radioactivity in the brain. [11C]6 is a promising PET tracer which offers in vivo specific binding and selectivity for MAGL in rodent brains.
- Published
- 2019
- Full Text
- View/download PDF
37. Design, Synthesis, and Evaluation of Reversible and Irreversible Monoacylglycerol Lipase Positron Emission Tomography (PET) Tracers Using a 'Tail Switching' Strategy on a Piperazinyl Azetidine Skeleton
- Author
-
Yiding Zhang, Zhen Chen, Ran Cheng, Tuo Shao, Yihan Shao, Qingzhen Yu, Jun-An Ma, Kuan Hu, Xiaoyun Deng, Akiko Hatori, Steven H. Liang, Lee Josephson, Hualong Fu, Kenneth Dahl, Neil Vasdev, Michael A. Schafroth, Masayuki Fujinaga, Lin Xie, Christopher J. Fowler, Shuyin Gu, Thomas Lee Collier, Lu Wang, Benjamin F. Cravatt, Jian Rong, Daisuke Ogasawara, Tomoteru Yamasaki, Bao Liang, Yuancheng Gou, Ming-Rong Zhang, Xiaofei Zhang, Katsushi Kumata, Genwei Zhang, Wakana Mori, and Jingjin Chen
- Subjects
Azetidine ,Proof of Concept Study ,01 natural sciences ,Piperazines ,Article ,Rats, Sprague-Dawley ,Mice ,Radioligand Assay ,03 medical and health sciences ,chemistry.chemical_compound ,In vivo ,Drug Discovery ,medicine ,Animals ,Tissue Distribution ,Enzyme Inhibitors ,Binding selectivity ,030304 developmental biology ,Mice, Knockout ,0303 health sciences ,Dose-Response Relationship, Drug ,medicine.diagnostic_test ,Neurodegeneration ,Serine hydrolase ,medicine.disease ,Endocannabinoid system ,Monoacylglycerol Lipases ,Rats ,0104 chemical sciences ,Molecular Docking Simulation ,Monoacylglycerol lipase ,010404 medicinal & biomolecular chemistry ,chemistry ,Positron emission tomography ,Drug Design ,Positron-Emission Tomography ,Biophysics ,Azetidines ,Molecular Medicine ,Radiopharmaceuticals - Abstract
Monoacylglycerol lipase (MAGL) is a serine hydrolase that degrades 2-arachidonoylglycerol (2-AG) in the endocannabinoid system (eCB). Selective inhibition of MAGL has emerged as a potential therapeutic approach for the treatment of diverse pathological conditions including chronic pain, inflammation, cancer and neurodegeneration. Herein we disclose a novel array of reversible and irreversible MAGL inhibitors by means of tail switching on a piperazinyl azetidine scaffold. We developed a lead irreversible-binding MAGL inhibitor 8, and reversible-binding compounds 17 and 37 which are amenable for radiolabeling with (11)C or (18)F. [(11)C]8 ([(11)C]MAGL-2–11) exhibited high brain uptake and excellent binding specificity in the brain towards MAGL. Reversible radioligands [(11)C]17 ([(11)C]PAD) and [(18)F]37 ([(18)F]MAGL-4–11) also demonstrated excellent in vivo binding specificity towards MAGL in peripheral organs. This work may pave the way for the development of MAGL-targeted positron emission tomography (PET) tracers with tunability in reversible and irreversible binding mechanism.
- Published
- 2019
- Full Text
- View/download PDF
38. Synthesis and evaluation of 4-(2-fluoro-4-[11C]methoxyphenyl)-5-((2-methylpyridin-4-yl)methoxy)picolinamide for PET imaging of the metabotropic glutamate receptor 2 in the rat brain
- Author
-
Ming-Rong Zhang, Tomoteru Yamasaki, Nobuki Nengaki, Wakana Mori, Masayuki Fujinaga, Katsushi Kumata, Yiding Zhang, Lin Xie, and Akiko Hatori
- Subjects
Allosteric modulator ,010405 organic chemistry ,Chemistry ,Organic Chemistry ,Clinical Biochemistry ,Glutamate receptor ,Pharmaceutical Science ,Human brain ,Striatum ,01 natural sciences ,Biochemistry ,Molecular biology ,In vitro ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,medicine.anatomical_structure ,In vivo ,Drug Discovery ,Forebrain ,medicine ,Molecular Medicine ,Metabotropic glutamate receptor 2 ,Molecular Biology - Abstract
Metabotropic glutamate receptor 2 (mGluR2) has been suggested as a therapeutic target for treating schizophrenia-like symptoms arising from increased glutamate transmission in the human forebrain. However, no reliable positron emission tomography (PET) radiotracer allowing for in vivo visualization of mGluR2 in the human brain is currently available. In this study, we synthesized 4-(2-fluoro-4-[11C]methoxyphenyl)-5-((2-methylpyridin-4-yl)methoxy)picolinamide ([11C]1) and evaluated its potential as a PET tracer for imaging mGluR2 in the rodent brain. Compound 1, a negative allosteric modulator (NAM) of mGluR2, showed high in vitro binding affinity (IC50: 26 nM) for mGluR2 overexpressed in human cells. [11C]1 was synthesized by O-[11C]methylation of the phenol precursor 2 with [11C]methyl iodide. After the reaction, HPLC purification and formulation, [11C]1 of 7.4 ± 2.8 GBq (n = 8) was obtained from [11C]carbon dioxide of 22.5 ± 4.8 GBq (n = 8) with >99% radiochemical purity and 70 ± 32 GBq/μmol (n = 8) molar activity at the end of synthesis. In vitro autoradiography for rat brains showed that [11C]1 binding was heterogeneously distributed in the cerebral cortex, striatum, hippocampus, and cerebellum. This pattern is consistent with the regional distribution pattern of mGluR2 in the rodent brain. The radioactivity was significantly reduced by self- or MNI-137 (a mGluR2 NAM) blocking. Small-animal PET studies indicated a low in vivo specific binding of [11C]1 in the rat brain. The brain uptake was increased in a P-glycoprotein and breast cancer resistant protein double knockout mouse, when compared to a wild-type mouse. While [11C]1 presented limited potential as an in vivo PET tracer for mGluR2, we suggested that it can be used as a lead compound for developing new radiotracers with improved in vivo brain properties.
- Published
- 2019
- Full Text
- View/download PDF
39. Neuroprotective effects of minocycline and KML29, a potent inhibitor of monoacylglycerol lipase, in an experimental stroke model: a small-animal positron emission tomography study
- Author
-
Yusuke Kurihara, Ming-Rong Zhang, Steven H. Liang, Akiko Hatori, Yiding Zhang, Wakana Mori, Jian Rong, Hidekatsu Wakizaka, Tomoteru Yamasaki, Lu Wang, and Masanao Ogawa
- Subjects
Male ,Central nervous system ,Ischemia ,Medicine (miscellaneous) ,Minocycline ,Striatum ,Arachidonic Acids ,Pharmacology ,Neuroprotection ,Brain Ischemia ,Glycerides ,Rats, Sprague-Dawley ,Piperidines ,Cortex (anatomy) ,medicine ,Animals ,Benzodioxoles ,Carbon Radioisotopes ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) ,Ischemic Stroke ,business.industry ,Glutamate receptor ,Brain ,Infarction, Middle Cerebral Artery ,medicine.disease ,Cell Hypoxia ,Monoacylglycerol Lipases ,Rats ,Monoacylglycerol lipase ,Stroke ,Disease Models, Animal ,medicine.anatomical_structure ,Neuroprotective Agents ,PET ,Positron-Emission Tomography ,MAGL ,KML29 ,business ,Tomography, X-Ray Computed ,TSPO ,medicine.drug ,Endocannabinoids ,Research Paper - Abstract
Hypoxia caused by ischemia induces acidosis and neuroexcitotoxicity, resulting in neuronal death in the central nervous system (CNS). Monoacylglycerol lipase (MAGL) is a modulator of 2-arachidonoylglycerol (2-AG), which is involved in retrograde inhibition of glutamate release in the endocannabinoid system. In the present study, we used positron emission tomography (PET) to monitor MAGL-positive neurons and neuroinflammation in the brains of ischemic rats. Additionally, we performed PET imaging to evaluate the neuroprotective effects of an MAGL inhibitor in an ischemic injury model. Methods: Ischemic-injury rat models were induced by intraluminal right middle cerebral artery occlusion (MCAO). PET studies of the brains of the ischemic rats were performed at several experimental time points (pre-occlusion, days 2, 4, and 7 after the MCAO surgery) using [11C]SAR127303 for MAGL and [18F]FEBMP for 18 kDa translocator protein (TSPO, a hall-mark of neuroinflammation). Medication using minocycline (a well-known neuroprotective agent) or KML29 (a potent MAGL inhibitor) was given immediately after the MCAO surgery and then daily over the subsequent three days. Results: PET imaging of the ischemic rats using [11C]SAR127303 showed an acute decline of radioactive accumulation in the ipsilateral side at two days after MCAO surgery (ratio of the area under the curve between the ipsilateral and contralateral sides: 0.49 ± 0.04 in the cortex and 0.73 ± 0.02 in the striatum). PET imaging with [18F]FEBMP, however, showed a moderate increase in accumulation of radioactivity in the ipsilateral hemisphere on day 2 (1.36 ± 0.11), and further increases on day 4 (1.72 ± 0.15) and day 7 (1.99 ± 0.06). Treatment with minocycline or KML29 eased the decline in radioactive accumulation of [11C]SAR127303 for MAGL (minocycline-treated group: 0.82 ± 0.06 in the cortex and 0.81 ± 0.05 in the striatum; KML29-treated group: 0.72 ± 0.07 in the cortex and 0.88 ± 0.04 in the striatum) and increased uptake of [18F]FEBMP for TSPO (minocycline-treated group: 1.52 ± 0.21 in the cortex and 1.56 ± 0.11 in the striatum; KML29-treated group: 1.63 ± 0.09 in the cortex and 1.50 ± 0.17 in the striatum). In MCAO rats, minocycline treatment showed a neuroprotective effect in the sensorimotor cortex suffering from severe hypoxic injury, whereas KML29 treatment saved neurons in the striatum, including bundles of myelinated axons. Conclusions: PET imaging allowed visualization of the different neuroprotective effects of minocycline and KML29, and indicated that combination pharmacotherapy using these drugs may be an effective therapy in acute ischemia.
- Published
- 2021
40. Radiosynthesis of 18F-fluoroethylated tracers via a simplified one-pot 18F-fluoroethylation method using [18F]fluoroethyl tosylate
- Author
-
Kawamura, Kazunori, Kumata, Katsushi, Mori, Wakana, Fujinaga, Masayuki, Kurihara, Yusuke, Ogawa, Masanao, Ohkubo, Takayuki, Furutsuka, Kenji, Hashimoto, Hiroki, Nengaki, Nobuki, Ming-Rong, Zhang, Kazunori, Kawamura, Katsushi, Kumata, Wakana, Mori, Masayuki, Fujinaga, Masanao, Ogawa, Kenji, Furutsuka, Hiroki, Hashimoto, Nobuki, Nengaki, Zhang, Ming-Rong, Kawamura, Kazunori, Kumata, Katsushi, Mori, Wakana, Fujinaga, Masayuki, Kurihara, Yusuke, Ogawa, Masanao, Ohkubo, Takayuki, Furutsuka, Kenji, Hashimoto, Hiroki, Nengaki, Nobuki, Ming-Rong, Zhang, Kazunori, Kawamura, Katsushi, Kumata, Wakana, Mori, Masayuki, Fujinaga, Masanao, Ogawa, Kenji, Furutsuka, Hiroki, Hashimoto, Nobuki, Nengaki, and Zhang, Ming-Rong
- Abstract
Recently, a straightforward one-pot method for 18F-fluoroethylation without azeotropic drying of cyclotron-produced [18F]F– was developed. In this study, we have attempted to simplify the automated radiosynthesis of two [18F]fluoroethylated tracers, [18F]FEDAC and [18F]FET, using a desmethyl labeling precursor and [18F]fluoroethyl tosylate, based on the above-mentioned method. The radiochemical yields of [18F]FEDAC and [18F]FET were 26 ± 3.7% (n = 5) and 14 ± 2.2% (n = 4), respectively, based on total [18F]F– at the end of irradiation.
- Published
- 2021
41. Automated radiosynthesis of two 18F-labeled tracers containing 3-fluoro-2-hydroxypropyl moiety, [18F]FMISO and [18F]PM-PBB3, via [18F]epifluorohydrin
- Author
-
Ohkubo, Takayuki, Kurihara, Yusuke, Masanao, Ogawa, Nobuki, Nengaki, Masayuki, Fujinaga, Wakana, Mori, Katsushi, Kumata, Masayuki, Hanyu, Kenji, Furutsuka, Hiroki, Hashimoto, Kazunori, Kawamura, Zhang, Ming-Rong, Ohkubo, Takayuki, Kurihara, Yusuke, Masanao, Ogawa, Nobuki, Nengaki, Masayuki, Fujinaga, Wakana, Mori, Katsushi, Kumata, Masayuki, Hanyu, Kenji, Furutsuka, Hiroki, Hashimoto, Kazunori, Kawamura, and Zhang, Ming-Rong
- Abstract
Background [ 18 F]Fluoromisonidazole ([ 18 F]FMISO) and 1-[ 18 F]fluoro-3-((2-((1 E ,3 E )-4-(6-(methylamino)pyridine-3-yl)buta-1,3-dien-1-yl)benzo[d]thiazol-6-yl)oxy)propan-2-ol ([18 F]PM-PBB3 or [ 18 F]APN-1607) are clinically used radiotracers for imaging hypoxia and tau pathology, respectively. Both radiotracers were produced by direct 18 F-fluorination using the corresponding tosylate precursors 1 or 2 and [ 18 F]F - , followed by the removal of protecting groups. In this study, we synthesized [ 18F]FMISO and [ 18 F]PM-PBB3 by 18 F-fluoroalkylation using [ 18 F]epifluorohydrin ([ 18 F] 5 ) for clinical applications. Results First, [ 18 F] 5 was synthesized by the reaction of 1,2-epoxypropyl tosylate ( 8 ) with [ 18 F]F - and was purified by distillation. Subsequently, [ 18 F] 5 was reacted with 2-nitroimidazole ( 6 ) or PBB3 ( 7 ) as a precursor for 18 F-labeling, and each reaction mixture was purified by preparative high-performance liquid chromatography and formulated to obtain the [ 18 F]FMISO or [ 18 F]PM-PBB3 injection. All synthetic sequences were performed using an automated 18 F-labeling synthesizer. The obtained [ 18 F]FMISO showed sufficient radioactivity (0.83 ± 0.20 GBq at the end of synthesis (EOS); n = 8) with appropriate radiochemical yield based on [ 18F]F - (26 ± 7.5% at EOS, decay-corrected; n = 8). The obtained [ 18 F]PM-PBB3 also showed sufficient radioactivity (0.79 ± 0.10 GBq at EOS; n = 11) with appropriate radiochemical yield based on [ 18 F]F - (16 ± 3.2% at EOS, decay-corrected; n = 11). Conclusions Both [ 18 F]FMISO and [ 18 F]PM-PBB3 injections were successfully synthesized with sufficient radioactivity by 18 F-fluoroalkylation using [ 18 F] 5 .
- Published
- 2021
42. In vivo visualization of propagating α-synuclein pathologies in mouse and marmoset models by a bimodal imaging probe, C05-05
- Author
-
Shoko Uchida, Yoshiki Yamaguchi, Wakana Mori, Maiko Ono, Yuji Nagai, Takeharu Minamihisamatsu, Akiyoshi Kakita, Aki Shimozawa, Masayuki Fujinaga, Koki Mimura, Masato Hasegawa, Naruhiko Sahara, Ming-Rong Zhang, Hironobu Endo, Masafumi Shimojo, Katsushi Kumata, Makoto Higuchi, Hiroshi Shimizu, Takafumi Minamimoto, Manami Takahashi, Hiroyuki Takuwa, Tatsuya Kikuchi, Sushil K. Mishra, and Yuhei Takado
- Subjects
biology ,Chemistry ,Neurodegeneration ,In vitro toxicology ,Marmoset ,Fibrillogenesis ,Human brain ,medicine.disease ,medicine.anatomical_structure ,In vivo ,biology.animal ,medicine ,biology.protein ,Neuroscience ,Intracellular ,Dopamine transporter - Abstract
Deposition of intracellular α-synuclein fibrils is implicated in neurodegenerative parkinsonian disorders, while high-contrast in vivo detection of α-synuclein depositions has been unsuccessful in animal models and humans. Here, we have developed a bimodal imaging probe, C05-05, for visualizing α-synuclein inclusions in the brains of living animals modeling α-synuclein propagation. In vivo optical and PET imaging of a mouse model demonstrated visualization of α-synuclein aggregates by C05-05, revealing a dynamic propagation of fibrillogenesis along neural pathways followed by disruptions of these structures. Moreover, longitudinal 18F-C05-05-PET of a marmoset model captured widespread dissemination of fibrillary pathologies accompanied by neurodegeneration detected by dopamine transporter PET. In addition, in vitro assays demonstrated the high-affinity binding of 18F-C05-05 to α-synuclein versus other protein pathologies in human brain tissues. Collectively, we propose a new imaging technology enabling etiological and therapeutic assessments of α-synuclein pathogenesis at non-clinical levels, highlighting the applicability of C05-05 to clinical PET.
- Published
- 2020
- Full Text
- View/download PDF
43. Development of a highly-specific
- Author
-
Zhen, Chen, Wakana, Mori, Jian, Rong, Michael A, Schafroth, Tuo, Shao, Richard S, Van, Daisuke, Ogasawara, Tomoteru, Yamasaki, Atsuto, Hiraishi, Akiko, Hatori, Jiahui, Chen, Yiding, Zhang, Kuan, Hu, Masayuki, Fujinaga, Jiyun, Sun, Qingzhen, Yu, Thomas L, Collier, Yihan, Shao, Benjamin F, Cravatt, Lee, Josephson, Ming-Rong, Zhang, and Steven H, Liang
- Subjects
Central nervous system (CNS) ,Monoacylglycerol lipase (MAGL) ,Short Communication ,Arachidonic acid (AA) ,Positron emission tomography (PET) ,Fluorine-18 ,2-Arachidonylglycerol (2-AG) - Abstract
As a serine hydrolase, monoacylglycerol lipase (MAGL) is principally responsible for the metabolism of 2-arachidonoylglycerol (2-AG) in the central nervous system (CNS), leading to the formation of arachidonic acid (AA). Dysfunction of MAGL has been associated with multiple CNS disorders and symptoms, including neuroinflammation, cognitive impairment, epileptogenesis, nociception and neurodegenerative diseases. Inhibition of MAGL provides a promising therapeutic direction for the treatment of these conditions, and a MAGL positron emission tomography (PET) probe would greatly facilitate preclinical and clinical development of MAGL inhibitors. Herein, we design and synthesize a small library of fluoropyridyl-containing MAGL inhibitor candidates. Pharmacological evaluation of these candidates by activity-based protein profiling identified 14 as a lead compound, which was then radiolabeled with fluorine-18 via a facile SNAr reaction to form 2-[18F]fluoropyridine scaffold. Good blood–brain barrier permeability and high in vivo specific binding was demonstrated for radioligand [18F]14 (also named as [18F]MAGL-1902). This work may serve as a roadmap for clinical translation and further design of potent 18F-labeled MAGL PET tracers., Graphical abstract A highly potent irreversible MAGL PET tracer [18F]14 was described, which exhibited favorable in vitro and in vivo characteristics, including excellent affinity, high brain uptake, and good binding specificity.Image 1
- Published
- 2020
44. Radiosynthesis of
- Author
-
Kazunori, Kawamura, Katsushi, Kumata, Wakana, Mori, Masayuki, Fujinaga, Yusuke, Kurihara, Masanao, Ogawa, Takayuki, Ohkubo, Kenji, Furutsuka, Hiroki, Hashimoto, Nobuki, Nengaki, and Ming-Rong, Zhang
- Abstract
Recently, a straightforward one-pot method for
- Published
- 2020
45. Identification and Development of a New Positron Emission Tomography Ligand 4-(2-Fluoro-4-[
- Author
-
Tomoteru, Yamasaki, Xiaofei, Zhang, Katsushi, Kumata, Yiding, Zhang, Xiaoyun, Deng, Masayuki, Fujinaga, Zhen, Chen, Wakana, Mori, Kuan, Hu, Hidekatsu, Wakizaka, Akiko, Hatori, Lin, Xie, Masanao, Ogawa, Nobuki, Nengaki, Richard, Van, Yihan, Shao, Douglas J, Sheffler, Nicholas D P, Cosford, Steven H, Liang, and Ming-Rong, Zhang
- Subjects
Male ,Mice, Knockout ,Molecular Structure ,Brain ,Ligands ,Receptors, Metabotropic Glutamate ,Article ,Cell Line ,Molecular Imaging ,Rats, Sprague-Dawley ,Mice ,Organ Specificity ,Positron-Emission Tomography ,ATP Binding Cassette Transporter, Subfamily G, Member 2 ,Animals ,Autoradiography ,Humans ,Tissue Distribution ,Carbon Radioisotopes - Abstract
Metabotropic glutamate receptor 2 (mGlu(2)) is a known target for treating several central nervous system (CNS) disorders. To develop a viable PET ligand for mGlu(2), we identified new candidates 5a–5i as potent negative allosteric modulators (NAMs) of mGlu(2). Among these candidates, 4-(2-fluoro-4-methoxyphenyl)-5-((1-methyl-1H-pyrazol-3-yl)methoxy)picolinamide (5i, also named as [(11)C]MG2-1812) exhibited high potency, high subtype selectivity, and favorable lipophilicity. Compound 5i was labeled with positron-emitting carbon-11 ((11)C) to obtain [(11)C]5i in high radiochemical yield and high molar activity by O-[(11)C]methylation of the phenol precursor 12 with [(11)C]CH(3)I. In vitro autoradiography with [(11)C]5i showed heterogeneous radioactive accumulation in the brain tissue sections, ranked in the order: cortex > striatum > hippocampus > cerebellum ≫ thalamus > pons. PET study of [(11)C]5i indicated in vivo specific binding of mGlu(2) in the rat brain. Based on the [(11)C]5i scaffold, further optimization for new candidates is underway to identify a more suitable ligand for imaging mGlu(2).
- Published
- 2020
46. 3-(Cyclopropylmethyl)-7-((4-(4-[
- Author
-
Katsushi, Kumata, Yiding, Zhang, Masanao, Ogawa, Yusuke, Kurihara, Wakana, Mori, Kuan, Hu, Masayuki, Fujinaga, Nobuki, Nengaki, and Ming-Rong, Zhang
- Subjects
Male ,Rats, Sprague-Dawley ,Pyridines ,Positron-Emission Tomography ,Animals ,Brain ,Contrast Media ,Carbon Radioisotopes ,Radiopharmaceuticals ,Triazoles ,Receptors, Metabotropic Glutamate - Abstract
Selective metabotropic glutamate receptor 2 (mGluR2) inhibitors have been demonstrated to show therapeutic effects by improving alleviating symptoms of schizophrenic patients in clinical studies. Herein we report the synthesis and preliminary evaluation of a
- Published
- 2020
47. Synthesis and preliminary evaluation of 4-hydroxy-6-(3-[11C]methoxyphenethyl)pyridazin-3(2H)-one, a 11C-labeled D-amino acid oxidase (DAAO) inhibitor for PET imaging
- Author
-
Tuo Shao, Masayuki Fujinaga, Qingzhen Yu, Hsiao-Ying Wey, Richard Van, Steven H. Liang, Loredano Pollegioni, Giulia Murtas, Xiaoyun Deng, Yiding Zhang, Kuan Hu, Wakana Mori, Lee Josephson, Ming-Rong Zhang, Zhen Chen, Yihan Shao, Katsushi Kumata, Akiko Hatori, and Jian Rong
- Subjects
Clinical Biochemistry ,D-amino acid oxidase ,Pharmaceutical Science ,Carbon-11 ,01 natural sciences ,Biochemistry ,In vivo ,Drug Discovery ,medicine ,Molecular Biology ,D-Amino acid oxidase (DAAO) ,Cerebellum function ,Positron emission tomography (PET) ,Schizophrenia ,medicine.diagnostic_test ,010405 organic chemistry ,Chemistry ,Organic Chemistry ,Pet imaging ,Ligand (biochemistry) ,In vitro ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,Positron emission tomography ,Molecular Medicine ,Efflux ,Early phase - Abstract
Selective DAAO inhibitors have demonstrated promising therapeutic effects in clinical studies, including clinically alleviating symptoms of schizophrenic patients and ameliorating cognitive function in Alzheimer’s patients with early phase. Herein we report the synthesis and preliminary evaluation of a 11C-labeled positron emission tomography ligand based on a DAAO inhibitor, DAO-1903 (8). 11C-Isotopologue of 8 was prepared in high radiochemical yield with high radiochemical purity (>99%) and high molar activity (>37 GBq/µmol). In vitro autoradiography studies indicated that the ligand possessed high in vitro specific binding to DAAO, while in vivo dynamic PET studies demonstrated that [11C]8 failed to cross the blood–brain barrier possibly due to moderate brain efflux mechanism. Further chemical scaffold optimization is necessary to overcome limited brain permeability and improve specific binding.
- Published
- 2020
48. Development of 2-(2-(3-(4-([18F]Fluoromethoxy-d2)phenyl)-7-methyl-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)-4-isopropoxyisoindoline-1,3-dione for Positron-Emission-Tomography Imaging of Phosphodiesterase 10A in the Brain
- Author
-
Lin Xie, Nobuki Nengaki, Wakana Mori, Akiko Hatori, Masayuki Fujinaga, Yusuke Kurihara, Masanao Ogawa, Katsushi Kumata, Tomoteru Yamasaki, Yiding Zhang, Takayuki Ohkubo, Hidekatsu Wakizaka, and Ming-Rong Zhang
- Subjects
0303 health sciences ,Stereochemistry ,Ligand ,Metabolite ,Phosphodiesterase ,Binding potential ,Human brain ,01 natural sciences ,0104 chemical sciences ,010404 medicinal & biomolecular chemistry ,03 medical and health sciences ,chemistry.chemical_compound ,medicine.anatomical_structure ,chemistry ,Drug Discovery ,Lipophilicity ,medicine ,Molecular Medicine ,PDE10A ,Lead compound ,030304 developmental biology - Abstract
Phosphodiesterase 10A (PDE10A) is a newly identified therapeutic target for central-nervous-system disorders. 2-(2-(3-(4-([18F]Fluoroethoxy)phenyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)-4-isopropoxyisoindoline-1,3-dione ([18F]MNI-659, [18F]5) is a useful positron-emission-tomography (PET) ligand for imaging of PDE10A in the human brain. However, the radiolabeled metabolite of [18F]5 can accumulate in the brain. In this study, using [18F]5 as a lead compound, we designed four new 18F-labeled ligands ([18F]6–9) to find one more suitable than [18F]5. Of these, 2-(2-(3-(4-([18F]fluoromethoxy-d2)phenyl)-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)-4-isopropoxyisoindoline-1,3-dione ([18F]9) exhibited high in vitro binding affinity (Ki = 2.9 nM) to PDE10A and suitable lipophilicity (log D = 2.2). In PET studies, the binding potential (BPND) of [18F]9 (5.8) to PDE10A in the striatum of rat brains was significantly higher than that of [18F]5 (4.6). Furthermore, metabolite analysis showed much lower levels of contamin...
- Published
- 2018
- Full Text
- View/download PDF
49. Synthesis, pharmacology and preclinical evaluation of 11C-labeled 1,3-dihydro-2H-benzo[d]imidazole-2-ones for imaging γ8-dependent transmembrane AMPA receptor regulatory protein
- Author
-
Akiko Hatori, Hualong Fu, Wakana Mori, Jian Rong, Xiaofei Zhang, Patrick Dunn, Zhen Chen, Tuo Shao, Yihan Shao, Xiaoyun Deng, Lin Xie, Jun-An Ma, Steven H. Liang, Lee Josephson, Qingzhen Yu, Genwei Zhang, Yiding Zhang, Hua Li, Tomoteru Yamasaki, Longle Ma, Masayuki Fujinaga, Susumu Tomita, and Ming-Rong Zhang
- Subjects
0301 basic medicine ,Pharmacology ,Regulation of gene expression ,Drug discovery ,Chemistry ,Organic Chemistry ,Hippocampus ,General Medicine ,AMPA receptor ,Transmembrane protein ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,nervous system ,Drug Discovery ,Distribution (pharmacology) ,Ionotropic glutamate receptor ,Receptor ,030217 neurology & neurosurgery - Abstract
a-Amino-3-hydroxyl-5-methyl-4-isoxazolepropionic acid (AMPA) receptors are implicated in the pathology of neurological diseases such as epilepsy and schizophrenia. As pan antagonists for this target are often accompanied with undesired effects at high doses, one of the recent drug discovery approaches has shifted to subtype-selective AMPA receptor (AMPAR) antagonists, specifically, via modulating transmembrane AMPAR regulatory proteins (TARPs). The quantification of AMPARs by positron emission tomography (PET) would help obtain insights into disease conditions in the living brain and advance the translational development of AMPAR antagonists. Herein we report the design, synthesis and preclinical evaluation of a series of TARP γ-8 antagonists, amenable for radiolabeling, for the development of subtype-selective AMPAR PET imaging agents. Based on the pharmacology evaluation, molecular docking studies and physiochemical properties, we have identified several promising lead compounds 3, 17–19 and 21 for in vivo PET studies. All candidate compounds were labeled with [11C]COCl2 in high radiochemical yields (13–31% RCY) and high molar activities (35–196 GBq/μmol). While tracers 30 ([11C]17) & 32 ([11C]21) crossed the blood-brain barrier and showed heterogeneous distribution in PET studies, consistent with TARP γ-8 expression, high nonspecific binding prevented further evaluation. To our delight, tracer 31 ([11C]3) showed good in vitro specific binding and characteristic high uptake in the hippocampus in rat brain tissues, which provides the guideline for further development of a new generation subtype selective TARP γ-8 dependent AMPAR tracers.
- Published
- 2018
- Full Text
- View/download PDF
50. [18F]DAA1106: Automated radiosynthesis using spirocyclic iodonium ylide and preclinical evaluation for positron emission tomography imaging of translocator protein (18 kDa)
- Author
-
Yiding Zhang, Tomoteru Yamasaki, Lin Xie, Ming-Rong Zhang, Katsushi Kumata, Masayuki Hanyu, Akiko Hatori, Takayuki Ohkubo, Wakana Mori, and Masayuki Fujinaga
- Subjects
0301 basic medicine ,Biodistribution ,Clinical Biochemistry ,Pharmaceutical Science ,Biochemistry ,High-performance liquid chromatography ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Drug Discovery ,medicine ,Translocator protein ,Molecular Biology ,chemistry.chemical_classification ,medicine.diagnostic_test ,biology ,Organic Chemistry ,Radiochemistry ,Automated radiosynthesis ,Ligand (biochemistry) ,030104 developmental biology ,chemistry ,Positron emission tomography ,Ylide ,biology.protein ,Molecular Medicine ,030217 neurology & neurosurgery ,Acetamide - Abstract
DAA1106 (N-(2,5-dimethoxybenzyl)-N-(5-fluoro-2-phenoxyphenyl)acetamide), is a potent and selective ligand for the translocator protein (18 kDa, TSPO) in brain mitochondrial fractions of rats and monkey (Ki = 0.043 and 0.188 nM, respectively). In this study, to translate [18F]DAA1106 for clinical studies, we performed automated syntheses of [18F]DAA1106 using the spirocyclic iodonium ylide (1) as a radiolabelling precursor and conducted preclinical studies including positron emission tomography (PET) imaging of TSPO in ischemic rat brains. Radiofluorination of the ylide precursor 1 with [18F]F−, followed by HPLC separation and formulation, produced the [18F]DAA1106 solution for injection in 6% average (n = 10) radiochemical yield (based on [18F]F−) with >98% radiochemical purity and molar activity of 60–100 GBq/μmol at the end of synthesis. The synthesis time was 87 min from the end of bombardment. The automated synthesis achieved [18F]DAA1106 with sufficient radioactivity available for preclinical and clinical use. Biodistribution study of [18F]DAA1106 showed a low uptake of radioactivity in the mouse bones. Metabolite analysis showed that >96% of total radioactivity in the mouse brain at 60 min after the radiotracer injection was unmetabolized [18F]DAA1106. PET study of ischemic rat brains visualized ischemic areas with a high uptake ratio (1.9 ± 0.3) compared with the contralateral side. We have provided evidence that [18F]DAA1106 could be routinely produced for clinical studies.
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.