1
|
Xiao Z, Wei H, Xu Y, Haider A, Wei J, Yuan S, Rong J, Zhao C, Li G, Zhang W, Chen H, Li Y, Zhang L, Sun J, Zhang S, Luo HB, Yan S, Cai Q, Hou L, Che C, Liang SH, Wang L. Discovery of a highly specific 18F-labeled PET ligand for phosphodiesterase 10A enabled by novel spirocyclic iodonium ylide radiofluorination. Acta Pharm Sin B 2022; 12:1963-1975. [PMID: 35847497 PMCID: PMC9279629 DOI: 10.1016/j.apsb.2021.11.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/30/2021] [Accepted: 10/20/2021] [Indexed: 12/14/2022] Open
Abstract
As a member of cyclic nucleotide phosphodiesterase (PDE) enzyme family, PDE10A is in charge of the degradation of cyclic adenosine (cAMP) and guanosine monophosphates (cGMP). While PDE10A is primarily expressed in the medium spiny neurons of the striatum, it has been implicated in a variety of neurological disorders. Indeed, inhibition of PDE10A has proven to be of potential use for the treatment of central nervous system (CNS) pathologies caused by dysfunction of the basal ganglia–of which the striatum constitutes the largest component. A PDE10A-targeted positron emission tomography (PET) radioligand would enable a better assessment of the pathophysiologic role of PDE10A, as well as confirm the relationship between target occupancy and administrated dose of a given drug candidate, thus accelerating the development of effective PDE10A inhibitors. In this study, we designed and synthesized a novel 18F-aryl PDE10A PET radioligand, codenamed [18F]P10A-1910 ([18F]9), in high radiochemical yield and molar activity via spirocyclic iodonium ylide-mediated radiofluorination. [18F]9 possessed good in vitro binding affinity (IC50 = 2.1 nmol/L) and selectivity towards PDE10A. Further, [18F]9 exhibited reasonable lipophilicity (logD = 3.50) and brain permeability (Papp > 10 × 10−6 cm/s in MDCK-MDR1 cells). PET imaging studies of [18F]9 revealed high striatal uptake and excellent in vivo specificity with reversible tracer kinetics. Preclinical studies in rodents revealed an improved plasma and brain stability of [18F]9 when compared to the current reference standard for PDE10A-targeted PET, [18F]MNI659. Further, dose–response experiments with a series of escalating doses of PDE10A inhibitor 1 in rhesus monkey brains confirmed the utility of [18F]9 for evaluating target occupancy in vivo in higher species. In conclusion, our results indicated that [18F]9 is a promising PDE10A PET radioligand for clinical translation.
Collapse
Affiliation(s)
- Zhiwei Xiao
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
| | - Huiyi Wei
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Yi Xu
- Department of Cardiology, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Ahmed Haider
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
| | - Junjie Wei
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Shiyu Yuan
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Jian Rong
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
| | - Chunyu Zhao
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
| | - Guocong Li
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Weibin Zhang
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Huangcan Chen
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| | - Yuefeng Li
- Guangdong Landau Biotechnology Co. Ltd., Guangzhou 510555, China
| | - Lingling Zhang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Jiyun Sun
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
| | - Shaojuan Zhang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Hai-Bin Luo
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Sen Yan
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou 510632, China
| | - Qijun Cai
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Lu Hou
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
| | - Chao Che
- State Key Laboratory of Chemical Oncogenomics, Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
- Corresponding authors. Tel./fax: +86 755 26032530 (Chao Che), +1 617 7266165 (Steven H. Liang), +86 20 38688692 (Lu Wang).
| | - Steven H. Liang
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital & Department of Radiology, Harvard Medical School, Boston, MA 02114, USA
- Corresponding authors. Tel./fax: +86 755 26032530 (Chao Che), +1 617 7266165 (Steven H. Liang), +86 20 38688692 (Lu Wang).
| | - Lu Wang
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, the First Affiliated Hospital of Jinan University, Guangzhou 510630, China
- Corresponding authors. Tel./fax: +86 755 26032530 (Chao Che), +1 617 7266165 (Steven H. Liang), +86 20 38688692 (Lu Wang).
| |
Collapse
|
2
|
Damuka N, Martin TJ, Bansode AH, Krizan I, Martin CW, Miller M, Whitlow CT, Nader MA, Solingapuram Sai KK. Initial Evaluations of the Microtubule-Based PET Radiotracer, [11C]MPC-6827 in a Rodent Model of Cocaine Abuse. Front Med (Lausanne) 2022; 9:817274. [PMID: 35295607 PMCID: PMC8918945 DOI: 10.3389/fmed.2022.817274] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 02/07/2022] [Indexed: 11/13/2022] Open
Abstract
PurposeMicrotubules (MTs) are structural units made of α and β tubulin subunits in the cytoskeleton responsible for axonal transport, information processing, and signaling mechanisms—critical for healthy brain function. Chronic cocaine exposure affects the function, organization, and stability of MTs in the brain, thereby impairing overall neurochemical and cognitive processes. At present, we have no reliable, non-invasive methods to image MTs for cocaine use disorder (CUD). Recently we reported the effect of cocaine in patient-derived neuroblastoma SH-SY5Y cells. Here we report preliminary results of a potential imaging biomarker of CUD using the brain penetrant MT-based radiotracer, [11C]MPC-6827, in an established rodent model of cocaine self-administration (SA).MethodsCell uptake studies were performed with [11C]MPC-6827 in SH-SY5Y cells, treated with or without cocaine (n = 6/group) at 30 and 60 min incubations. MicroPET/CT brain scans were performed in rats at baseline and 35 days after cocaine self-administration and compared with saline-treated rats as controls (n = 4/sex). Whole-body post-PET biodistribution, plasma metabolite assay, and brain autoradiography were performed in the same rats from imaging.ResultsCocaine-treated SH-SY5Y cells demonstrated a ∼26(±4)% decrease in radioactive uptake compared to non-treated controls. Both microPET/CT imaging and biodistribution results showed lower (∼35 ± 3%) [11C]MPC-6827 brain uptake in rats that had a history of cocaine self-administration compared to the saline-treated controls. Plasma metabolite assays demonstrate the stability (≥95%) of the radiotracer in both groups. In vitro autoradiography also demonstrated lower radioactive uptake in cocaine rats compared to the control rats. [11C]MPC-6827’s in vitro SH-SY5Y neuronal cell uptake, in vivo positron emission tomography (PET) imaging, ex vivo biodistribution, and in vitro autoradiography results corroborated well with each other, demonstrating decreased radioactive brain uptake in cocaine self-administered rats versus controls. There were no significant differences either in cocaine intake or in [11C]MPC-6827 uptake between the male and female rats.ConclusionsThis project is the first to validate in vivo imaging of the MT-associations with CUD in a rodent model. Our initial observations suggest that [11C]MPC-6827 uptake decreases in cocaine self-administered rats and that it may selectively bind to destabilized tubulin units in the brain. Further longitudinal studies correlating cocaine intake with [11C]MPC-6827 PET brain measures could potentially establish the MT scaffold as an imaging biomarker for CUD, providing researchers and clinicians with a sensitive tool to better understand the biological underpinnings of CUD and tailor new treatments.
Collapse
Affiliation(s)
- Naresh Damuka
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Thomas J. Martin
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Avinash H. Bansode
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Ivan Krizan
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Conner W. Martin
- Department of Anesthesiology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Mack Miller
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Christopher T. Whitlow
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Michael A. Nader
- Department of Physiology and Pharmacology, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Kiran Kumar Solingapuram Sai
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC, United States
- *Correspondence: Kiran Kumar Solingapuram Sai,
| |
Collapse
|
3
|
Suppression of Proliferation of Human Glioblastoma Cells by Combined Phosphodiesterase and Multidrug Resistance-Associated Protein 1 Inhibition. Int J Mol Sci 2021; 22:ijms22189665. [PMID: 34575827 PMCID: PMC8471536 DOI: 10.3390/ijms22189665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 08/29/2021] [Accepted: 09/03/2021] [Indexed: 12/17/2022] Open
Abstract
The paucity of currently available therapies for glioblastoma multiforme requires novel approaches to the treatment of this brain tumour. Disrupting cyclic nucleotide-signalling through phosphodiesterase (PDE) inhibition may be a promising way of suppressing glioblastoma growth. Here, we examined the effects of 28 PDE inhibitors, covering all the major PDE classes, on the proliferation of the human U87MG, A172 and T98G glioblastoma cells. The PDE10A inhibitors PF-2545920, PQ10 and papaverine, the PDE3/4 inhibitor trequinsin and the putative PDE5 inhibitor MY-5445 potently decreased glioblastoma cell proliferation. The synergistic suppression of glioblastoma cell proliferation was achieved by combining PF-2545920 and MY-5445. Furthermore, a co-incubation with drugs that block the activity of the multidrug resistance-associated protein 1 (MRP1) augmented these effects. In particular, a combination comprising the MRP1 inhibitor reversan, PF-2545920 and MY-5445, all at low micromolar concentrations, afforded nearly complete inhibition of glioblastoma cell growth. Thus, the potent suppression of glioblastoma cell viability may be achieved by combining MRP1 inhibitors with PDE inhibitors at a lower toxicity than that of the standard chemotherapeutic agents, thereby providing a new combination therapy for this challenging malignancy.
Collapse
|
4
|
Sun J, Xiao Z, Haider A, Gebhard C, Xu H, Luo HB, Zhang HT, Josephson L, Wang L, Liang SH. Advances in Cyclic Nucleotide Phosphodiesterase-Targeted PET Imaging and Drug Discovery. J Med Chem 2021; 64:7083-7109. [PMID: 34042442 DOI: 10.1021/acs.jmedchem.1c00115] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cyclic nucleotide phosphodiesterases (PDEs) control the intracellular concentrations of cAMP and cGMP in virtually all mammalian cells. Accordingly, the PDE family regulates a myriad of physiological functions, including cell proliferation, differentiation and apoptosis, gene expression, central nervous system function, and muscle contraction. Along this line, dysfunction of PDEs has been implicated in neurodegenerative disorders, coronary artery diseases, chronic obstructive pulmonary disease, and cancer development. To date, 11 PDE families have been identified; however, their distinct roles in the various pathologies are largely unexplored and subject to contemporary research efforts. Indeed, there is growing interest for the development of isoform-selective PDE inhibitors as potential therapeutic agents. Similarly, the evolving knowledge on the various PDE isoforms has channeled the identification of new PET probes, allowing isoform-selective imaging. This review highlights recent advances in PDE-targeted PET tracer development, thereby focusing on efforts to assess disease-related PDE pathophysiology and to support isoform-selective drug discovery.
Collapse
Affiliation(s)
- Jiyun Sun
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States
| | - Zhiwei Xiao
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States
| | - Ahmed Haider
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States
| | - Catherine Gebhard
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, Zurich 8006, Switzerland.,Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, Schlieren 8952, Switzerland
| | - Hao Xu
- Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, 613 West Huangpu Road, Tianhe District, Guangzhou 510630, China
| | - Hai-Bin Luo
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China
| | - Han-Ting Zhang
- Departments of Neuroscience, Behavioral Medicine & Psychiatry, and Physiology & Pharmacology, the Rockefeller Neuroscience Institute, West Virginia University Health Sciences Center, Morgantown, West Virginia 26506, United States
| | - Lee Josephson
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States
| | - Lu Wang
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States.,Center of Cyclotron and PET Radiopharmaceuticals, Department of Nuclear Medicine and PET/CT-MRI Center, The First Affiliated Hospital of Jinan University, 613 West Huangpu Road, Tianhe District, Guangzhou 510630, China
| | - Steven H Liang
- Department of Radiology, Division of Nuclear Medicine and Molecular Imaging Massachusetts General Hospital and Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, United States
| |
Collapse
|
5
|
Schröder S, Scheunemann M, Wenzel B, Brust P. Challenges on Cyclic Nucleotide Phosphodiesterases Imaging with Positron Emission Tomography: Novel Radioligands and (Pre-)Clinical Insights since 2016. Int J Mol Sci 2021; 22:ijms22083832. [PMID: 33917199 PMCID: PMC8068090 DOI: 10.3390/ijms22083832] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/21/2022] Open
Abstract
Cyclic nucleotide phosphodiesterases (PDEs) represent one of the key targets in the research field of intracellular signaling related to the second messenger molecules cyclic adenosine monophosphate (cAMP) and/or cyclic guanosine monophosphate (cGMP). Hence, non-invasive imaging of this enzyme class by positron emission tomography (PET) using appropriate isoform-selective PDE radioligands is gaining importance. This methodology enables the in vivo diagnosis and staging of numerous diseases associated with altered PDE density or activity in the periphery and the central nervous system as well as the translational evaluation of novel PDE inhibitors as therapeutics. In this follow-up review, we summarize the efforts in the development of novel PDE radioligands and highlight (pre-)clinical insights from PET studies using already known PDE radioligands since 2016.
Collapse
Affiliation(s)
- Susann Schröder
- Department of Research and Development, ROTOP Pharmaka Ltd., 01328 Dresden, Germany
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Research Site Leipzig, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 04318 Leipzig, Germany; (M.S.); (B.W.); (P.B.)
- Correspondence: ; Tel.: +49-341-234-179-4631
| | - Matthias Scheunemann
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Research Site Leipzig, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 04318 Leipzig, Germany; (M.S.); (B.W.); (P.B.)
| | - Barbara Wenzel
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Research Site Leipzig, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 04318 Leipzig, Germany; (M.S.); (B.W.); (P.B.)
| | - Peter Brust
- Department of Neuroradiopharmaceuticals, Institute of Radiopharmaceutical Cancer Research, Research Site Leipzig, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 04318 Leipzig, Germany; (M.S.); (B.W.); (P.B.)
| |
Collapse
|
6
|
Amin HS, Parikh PK, Ghate MD. Medicinal chemistry strategies for the development of phosphodiesterase 10A (PDE10A) inhibitors - An update of recent progress. Eur J Med Chem 2021; 214:113155. [PMID: 33581555 DOI: 10.1016/j.ejmech.2021.113155] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/27/2020] [Accepted: 01/03/2021] [Indexed: 11/26/2022]
Abstract
Phosphodiesterase 10A is a member of Phosphodiesterase (PDE)-superfamily of the enzyme which is responsible for hydrolysis of cAMP and cGMP to their inactive forms 5'-AMP and 5'-GMP, respectively. PDE10A is highly expressed in the brain, particularly in the putamen and caudate nucleus. PDE10A plays an important role in the regulation of localization, duration, and amplitude of the cyclic nucleotide signalling within the subcellular domain of these regions, and thereby modulation of PDE10A enzyme can give rise to a new therapeutic approach in the treatment of schizophrenia and other neurodegenerative disorders. Limitation of the conventional therapy of schizophrenia forced the pharmaceutical industry to move their efforts to develop a novel treatment approach with reduced side effects. In the past decade, considerable developments have been made in pursuit of PDE10A centric antipsychotic agents by several pharmaceutical industries due to the distribution of PDE10A in the brain and the ability of PDE10A inhibitors to mimic the effect of D2 antagonists and D1 agonists. However, no selective PDE10A inhibitor is currently available in the market for the treatment of schizophrenia. The present compilation concisely describes the role of PDE10A inhibitors in the therapy of neurodegenerative disorders mainly in psychosis, the structure of PDE10A enzyme, key interaction of different PDE10A inhibitors with human PDE10A enzyme and recent medicinal chemistry developments in designing of safe and effective PDE10A inhibitors for the treatment of schizophrenia. The present compilation also provides useful information and future direction to bring further improvements in the discovery of PDE10A inhibitors.
Collapse
Affiliation(s)
- Harsh S Amin
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382 481, Gujarat, India
| | - Palak K Parikh
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382 481, Gujarat, India; Department of Pharmaceutical Chemistry, L. M. College of Pharmacy, Navrangpura, Ahmedabad, 380 009, Gujarat, India.
| | - Manjunath D Ghate
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382 481, Gujarat, India
| |
Collapse
|
7
|
Shaw RC, Tamagnan GD, Tavares AAS. Rapidly (and Successfully) Translating Novel Brain Radiotracers From Animal Research Into Clinical Use. Front Neurosci 2020; 14:871. [PMID: 33117115 PMCID: PMC7559529 DOI: 10.3389/fnins.2020.00871] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 07/27/2020] [Indexed: 12/26/2022] Open
Abstract
The advent of preclinical research scanners for in vivo imaging of small animals has added confidence into the multi-step decision-making process of radiotracer discovery and development. Furthermore, it has expanded the utility of imaging techniques available to dissect clinical questions, fostering a cyclic interaction between the clinical and the preclinical worlds. Significant efforts from medicinal chemistry have also made available several high-affinity and selective compounds amenable for radiolabeling, that target different receptors, transporters and enzymes in vivo. This substantially increased the range of applications of molecular imaging using positron emission tomography (PET) or single photon emission computed tomography (SPECT). However, the process of developing novel radiotracers for in vivo imaging of the human brain is a multi-step process that has several inherent pitfalls and technical difficulties, which often hampers the successful translation of novel imaging agents from preclinical research into clinical use. In this paper, the process of radiotracer development and its relevance in brain research is discussed; as well as, its pitfalls, technical challenges and future promises. Examples of successful and unsuccessful translation of brain radiotracers will be presented.
Collapse
Affiliation(s)
- Robert C. Shaw
- BHF Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Imaging, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Adriana Alexandre S. Tavares
- BHF Centre for Cardiovascular Sciences, University of Edinburgh, Edinburgh, United Kingdom
- Edinburgh Imaging, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
8
|
Gabr M, Rehman AU, Chen HF. Quinoline-Pyrazole Scaffold as a Novel Ligand of Galectin-3 and Suppressor of TREM2 Signaling. ACS Med Chem Lett 2020; 11:1759-1765. [PMID: 32944144 DOI: 10.1021/acsmedchemlett.0c00330] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 08/11/2020] [Indexed: 12/20/2022] Open
Abstract
Galectin-3 has been identified as a critical player in driving the neuroinflammatory responses in Alzheimer's disease (AD). A key feature of this function of galectin-3 is associated with its interaction with the triggering receptor expressed on myeloid cells-2 (TREM2). Herein, we report a high-throughput screening (HTS) platform that can be used for the identification of inhibitors of TREM2 and galectin-3 interaction. We have utilized this HTS assay to screen a focused library of compounds optimized for the central nervous system (CNS)-related diseases. MG-257 was identified from this screen as the first example of a small molecule that can attenuate TREM2 signaling based on its high affinity to galectin-3 (endogenous ligand of TREM2). Remarkably, MG-257 reduced the levels of proinflammatory cytokines in activated microglial cells, which highlights its ability to inhibit the neuroinflammatory response associated with AD.
Collapse
Affiliation(s)
- Moustafa Gabr
- Department of Radiology, Stanford University School of Medicine, Stanford, California 94305, United States
| | - Ashfaq Ur Rehman
- Medicinal Bioinformatics Center, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
- State Key Laboratory of Microbial Metabolism, Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Sciences and Biotechnology, College of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hai-Feng Chen
- State Key Laboratory of Microbial Metabolism, Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Sciences and Biotechnology, College of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
- Shanghai Center for Bioinformation Technology, Shanghai 200235, China
| |
Collapse
|
9
|
Mori W, Yamasaki T, Fujinaga M, Ogawa M, Zhang Y, Hatori A, Xie L, Kumata K, Wakizaka H, Kurihara Y, Ohkubo T, Nengaki N, Zhang MR. Development of 2-(2-(3-(4-([ 18F]Fluoromethoxy- d 2)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. J Med Chem 2018; 62:688-698. [PMID: 30516998 DOI: 10.1021/acs.jmedchem.8b01366] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
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 contamination with radiolabeled metabolites in the brains of rats given [18F]9 than in those given [18F]5. In conclusion, [18F]9 is a useful PET ligand for PDE10A imaging in brain.
Collapse
Affiliation(s)
| | | | | | - Masanao Ogawa
- SHI Accelerator Service, Ltd. , 1-17-6 Osaki , Shinagawa-ku, Tokyo 141-0032 , Japan
| | | | | | | | | | | | - Yusuke Kurihara
- SHI Accelerator Service, Ltd. , 1-17-6 Osaki , Shinagawa-ku, Tokyo 141-0032 , Japan
| | - Takayuki Ohkubo
- SHI Accelerator Service, Ltd. , 1-17-6 Osaki , Shinagawa-ku, Tokyo 141-0032 , Japan
| | - Nobuki Nengaki
- SHI Accelerator Service, Ltd. , 1-17-6 Osaki , Shinagawa-ku, Tokyo 141-0032 , Japan
| | | |
Collapse
|
10
|
Liu H, Jin H, Luo Z, Yue X, Zhang X, Flores H, Su Y, Perlmutter JS, Tu Z. In Vivo Characterization of Two 18F-Labeled PDE10A PET Radioligands in Nonhuman Primate Brains. ACS Chem Neurosci 2018; 9:1066-1073. [PMID: 29400443 PMCID: PMC5955820 DOI: 10.1021/acschemneuro.7b00458] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Positron emission tomography (PET) with phosphodiesterase 10A (PDE10A) specific radioligands provides a noninvasive and quantitative imaging tool to access the expression of this enzyme in vivo under normal and diseased conditions. We recently reported two potent 18F-labeled PDE10A radioligands (18F-TZ19106B and 18F-TZ8110); initial evaluation in rats and nonhuman primates indicated stable metabolic profiles and excellent target-to-nontarget ratio (striatum/cerebellum) for both tracers. Herein, we focused on in vivo characterization of 18F-TZ19106B and 18F-TZ8110 to identify a suitable radioligand for imaging PDE10A in vivo. We directly compared microPET studies of these two radiotracers in adult male Macaca fascicularis nonhuman primates (NHPs). 18F-TZ19106B had higher striatal uptake and tracer retention in NHP brains than 18F-TZ8110, quantified by either standardized uptake values (SUVs) or nondisplaceable binding potential (BPND) estimated using reference-based modeling analysis. Blocking and displacement studies using the PDE10A inhibitor MP-10 indicated the binding of 18F-TZ19106B to PDE10A was specific and reversible. We also demonstrated sensitivity of 18F-TZ19106B binding to varying number of specific binding sites using escalating doses of MP-10 blockade (0.3, 0.5, 1.0, 1.5, and 2.0 mg/kg). Pretreatment with a dopamine D2-like receptor antagonist enhanced the striatal uptake of 18F-TZ19106B. Our results indicate that 18F-TZ19106B is a promising radioligand candidate for imaging PDE10A in vivo and it may be used to determine target engagement of PDE10A inhibitors and serve as a tool to evaluate the effect of novel antipsychotic therapies.
Collapse
Affiliation(s)
- Hui Liu
- Department of Radiology, Physical Therapy and Occupational Therapy, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Hongjun Jin
- Department of Radiology, Physical Therapy and Occupational Therapy, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Zonghua Luo
- Department of Radiology, Physical Therapy and Occupational Therapy, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Xuyi Yue
- Department of Radiology, Physical Therapy and Occupational Therapy, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Xiang Zhang
- Department of Radiology, Physical Therapy and Occupational Therapy, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Hubert Flores
- Department of Neurology, Physical Therapy and Occupational Therapy, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Yi Su
- Department of Radiology, Physical Therapy and Occupational Therapy, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Joel S. Perlmutter
- Department of Radiology, Physical Therapy and Occupational Therapy, Washington University School of Medicine, St. Louis, Missouri 63110, United States
- Department of Neurology, Physical Therapy and Occupational Therapy, Washington University School of Medicine, St. Louis, Missouri 63110, United States
- Department of Neuroscience, Physical Therapy and Occupational Therapy, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Zhude Tu
- Department of Radiology, Physical Therapy and Occupational Therapy, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| |
Collapse
|
11
|
Development of two fluorine-18 labeled PET radioligands targeting PDE10A and in vivo PET evaluation in nonhuman primates. Nucl Med Biol 2017; 57:12-19. [PMID: 29223715 DOI: 10.1016/j.nucmedbio.2017.10.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/02/2017] [Accepted: 10/20/2017] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Phosphodiesterase 10A (PDE10A) is a member of the PDE enzyme family that degrades cyclic adenosine and guanosine monophosphates (cAMP and cGMP). Based on the successful development of [11C]T-773 as PDE10A positron emission tomography (PET) radioligand, in this study our aim was to develop and evaluate fluorine-18 analogs of [11C]T-773. METHODS [18F]FM-T-773-d2 and [18F]FE-T-773-d4 were synthesized from the same precursor used for 11C-labeling of T-773 in a two-step approach via 18F-fluoromethylation and 18F-fluoroethylation, respectively, using corresponding deuterated synthons. A total of 12 PET measurements were performed in seven non-human primates. First, baseline PET measurements were performed using High Resolution Research Tomograph system with both [18F]FM-T-773-d2 and [18F]FE-T-773-d4; the uptake in whole brain and separate brain regions, as well as the specific binding and tissue ratio between putamen and cerebellum, was examined. Second, baseline and pretreatment PET measurements using MP-10 as the blocker were performed for [18F]FM-T-773-d2 including arterial blood sampling with radiometabolite analysis in four NHPs. RESULTS Both [18F]FM-T-773-d2 and [18F]FE-T-773-d4 were successfully radiolabeled with an average molar activity of 293 ± 114 GBq/μmol (n=8) for [18F]FM-T-773-d2 and 209 ± 26 GBq/μmol (n=4) for [18F]FE-T-773-d4, and a radiochemical yield of 10% (EOB, n=12, range 3%-16%). Both radioligands displayed high brain uptake (~5.5% of injected radioactivity for [18F]FM-T-773-d2 and ~3.5% for [18F]FE-T-773-d4 at the peak) and a fast washout. Specific binding reached maximum within 30 min for [18F]FM-T-773-d2 and after approximately 45 min for [18F]FE-T-773-d4. [18F]FM-T-773-d2 data fitted well with kinetic compartment models. BPND values obtained indirectly through compartment models were correlated well with those obtained by SRTM. BPND calculated with SRTM was 1.0-1.7 in the putamen. The occupancy with 1.8 mg/kg of MP-10 was approximately 60%. CONCLUSIONS [18F]FM-T-773-d2 and [18F]FE-T-773-d4 were developed as fluorine-18 PET radioligands for PDE10A, with the [18F]FM-T-773-d2 being the more promising PET radioligand warranting further evaluation.
Collapse
|
12
|
Abstract
PURPOSE A positron emission tomography (PET) tracer for the enzyme phosphodiesterase 10A (PDE10A) is desirable to guide the discovery and development of PDE10A inhibitors as potential therapeutics. The preclinical characterization of the PDE10A PET tracer [(11)C]MK-8193 is described. PROCEDURES In vitro binding studies with [(3)H]MK-8193 were conducted in rat, monkey, and human brain tissue. PET studies with [(11)C]MK-8193 were conducted in rats and rhesus monkeys at baseline and following administration of a PDE10A inhibitor. RESULTS [(3)H]MK-8193 is a high-affinity, selective PDE10A radioligand in rat, monkey, and human brain tissue. In vivo, [(11)C]MK-8193 displays rapid kinetics, low test-retest variability, and a large specific signal that is displaced by a structurally diverse PDE10A inhibitor, enabling the determination of pharmacokinetic/enzyme occupancy relationships. CONCLUSIONS [(11)C]MK-8193 is a useful PET tracer for the preclinical characterization of PDE10A therapeutic candidates in rat and monkey. Further evaluation of [(11)C]MK-8193 in humans is warranted.
Collapse
|
13
|
Mori W, Takei M, Furutsuka K, Fujinaga M, Kumata K, Muto M, Ohkubo T, Hashimoto H, Tamagnan G, Higuchi M, Kawamura K, Zhang MR. Comparison between [ 18F]fluorination and [ 18F]fluoroethylation reactions for the synthesis of the PDE10A PET radiotracer [ 18F]MNI-659. Nucl Med Biol 2017; 55:12-18. [PMID: 28972915 DOI: 10.1016/j.nucmedbio.2017.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 08/04/2017] [Accepted: 08/15/2017] [Indexed: 12/20/2022]
Abstract
INTRODUCTION 2-(2-(3-(4-(2-[18F]Fluoroethoxy)phenyl)-7-methyl-4-oxo-3,4-dihydroquinazolin-2-yl)ethyl)-4-isopropoxyisoindoline-1,3-dione ([18F]MNI-659, [18F]1) is a useful PET radiotracer for imaging phosphodiesterase 10A (PDE10A) in human brain. [18F]1 has been previously prepared by direct [18F]fluorination of a tosylate precursor 2 with [18F]F-. The aim of this study was to determine the conditions for the [18F]fluorination reaction to obtain [18F]1 of high quality and with sufficient radioactivity for clinical use in our institute. Moreover, we synthesized [18F]1 by [18F]fluoroethylation of a phenol precursor 3 with [18F]fluoroethyl bromide ([18F]FEtBr), and the outcomes of [18F]fluorination and [18F]fluoroethylation were compared. METHODS We performed the automated synthesis of [18F]1 by [18F]fluorination and [18F]fluoroethylation using a multi-purpose synthesizer. We determined the amounts of tosylate precursor 2 and potassium carbonate as well as the reaction temperature for direct [18F]fluorination. RESULTS The efficiency of the [18F]fluorination reaction was strongly affected by the amount of 2 and potassium carbonate. Under the determined reaction conditions, [18F]1 with 0.82±0.2GBq was obtained in 13.6%±3.3% radiochemical yield (n=8, decay-corrected to EOB and based on [18F]F-) at EOS, starting from 11.5±0.4GBq of cyclotron-produced [18F]F-. On the other hand, the [18F]fluoroethylation of 3 with [18F]FEtBr produced [18F]1 with 1.0±0.2GBq and in 22.5±2.5 % radiochemical yields (n=7, decay-corrected to EOB and based on [18F]F-) at EOS, starting from 7.4GBq of cyclotron-produced [18F]F-. Clearly, [18F]fluoroethylation resulted in a higher radiochemical yield of [18F]1 than [18F]fluorination. CONCLUSION [18F]1 of high quality and with sufficient radioactivity was successfully radiosynthesized by two methods. [18F]1 synthesized by direct [18F]fluorination has been approved and will be provided for clinical use in our institute.
Collapse
Affiliation(s)
- Wakana Mori
- National Institute of Radiological Science, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Makoto Takei
- National Institute of Radiological Science, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Kenji Furutsuka
- National Institute of Radiological Science, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan; SHI Accelerator Service Ltd., Tokyo 141-0032, Japan
| | - Masayuki Fujinaga
- National Institute of Radiological Science, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Katsushi Kumata
- National Institute of Radiological Science, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Masatoshi Muto
- National Institute of Radiological Science, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan; Tokyo Nuclear Services Ltd., Tokyo 110-0016, Japan
| | - Takayuki Ohkubo
- National Institute of Radiological Science, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan; SHI Accelerator Service Ltd., Tokyo 141-0032, Japan
| | - Hiroki Hashimoto
- National Institute of Radiological Science, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | | | - Makoto Higuchi
- National Institute of Radiological Science, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Kazunori Kawamura
- National Institute of Radiological Science, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan
| | - Ming-Rong Zhang
- National Institute of Radiological Science, National Institutes for Quantum and Radiological Science and Technology, Chiba 263-8555, Japan.
| |
Collapse
|
14
|
Target Engagement Measures in Preclinical Drug Discovery: Theory, Methods, and Case Studies. TRANSLATING MOLECULES INTO MEDICINES 2017. [DOI: 10.1007/978-3-319-50042-3_3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
15
|
Wagner S, Teodoro R, Deuther-Conrad W, Kranz M, Scheunemann M, Fischer S, Wenzel B, Egerland U, Hoefgen N, Steinbach J, Brust P. Radiosynthesis and biological evaluation of the new PDE10A radioligand [ 18 F]AQ28A. J Labelled Comp Radiopharm 2016; 60:36-48. [PMID: 27896836 DOI: 10.1002/jlcr.3471] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 08/19/2016] [Accepted: 10/12/2016] [Indexed: 01/10/2023]
Abstract
Cyclic nucleotide phosphodiesterase 10A (PDE10A) regulates the level of the second messengers cAMP and cGMP in particular in brain regions assumed to be associated with neurodegenerative and psychiatric diseases. A better understanding of the pathophysiological role of the expression of PDE10A could be obtained by quantitative imaging of the enzyme by positron emission tomography (PET). Thus, in this study we developed, radiolabeled, and evaluated a new PDE10A radioligand, 8-bromo-1-(6-[18 F]fluoropyridin-3-yl)-3,4-dimethylimidazo[1,5-a]quinoxaline ([18 F]AQ28A). [18 F]AQ28A was radiolabeled by both nucleophilic bromo-to-fluoro or nitro-to-fluoro exchange using K[18 F]F-K2.2.2 -carbonate complex with different yields. Using the superior nitro precursor, we developed an automated synthesis on a Tracerlab FX F-N module and obtained [18 F]AQ28A with high radiochemical yields (33 ± 6%) and specific activities (96-145 GBq·μmol-1 ) for further evaluation. Initially, we investigated the binding of [18 F]AQ28A to the brain of different species by autoradiography and observed the highest density of binding sites in striatum, the brain region with the highest PDE10A expression. Subsequent dynamic PET studies in mice revealed a region-specific accumulation of [18 F]AQ28A in this region, which could be blocked by preinjection of the selective PDE10A ligand MP-10. In conclusion, the data suggest [18 F]AQ28A is a suitable candidate for imaging of PDE10A in rodent brain by PET.
Collapse
Affiliation(s)
- Sally Wagner
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Research Site Leipzig, Department of Neuroradiopharmaceuticals, Leipzig, Germany
| | - Rodrigo Teodoro
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Research Site Leipzig, Department of Neuroradiopharmaceuticals, Leipzig, Germany
| | - Winnie Deuther-Conrad
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Research Site Leipzig, Department of Neuroradiopharmaceuticals, Leipzig, Germany
| | - Mathias Kranz
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Research Site Leipzig, Department of Neuroradiopharmaceuticals, Leipzig, Germany
| | - Matthias Scheunemann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Research Site Leipzig, Department of Neuroradiopharmaceuticals, Leipzig, Germany
| | - Steffen Fischer
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Research Site Leipzig, Department of Neuroradiopharmaceuticals, Leipzig, Germany
| | - Barbara Wenzel
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Research Site Leipzig, Department of Neuroradiopharmaceuticals, Leipzig, Germany
| | | | | | - Jörg Steinbach
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Research Site Leipzig, Department of Neuroradiopharmaceuticals, Leipzig, Germany
| | - Peter Brust
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Research Site Leipzig, Department of Neuroradiopharmaceuticals, Leipzig, Germany
| |
Collapse
|
16
|
Takano A, Stenkrona P, Stepanov V, Amini N, Martinsson S, Tsai M, Goldsmith P, Xie J, Wu J, Uz T, Halldin C, Macek TA. A human [ 11 C]T-773 PET study of PDE10A binding after oral administration of TAK-063, a PDE10A inhibitor. Neuroimage 2016; 141:10-17. [DOI: 10.1016/j.neuroimage.2016.06.047] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 06/24/2016] [Indexed: 01/22/2023] Open
|
17
|
Liu H, Jin H, Yue X, Han J, Yang H, Flores H, Su Y, Alagille D, Perlmutter JS, Tamagnan G, Tu Z. Comparison of [ 11C]TZ1964B and [ 18F]MNI659 for PET imaging brain PDE10A in nonhuman primates. Pharmacol Res Perspect 2016; 4:e00253. [PMID: 27713824 PMCID: PMC5045939 DOI: 10.1002/prp2.253] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 07/21/2016] [Accepted: 07/24/2016] [Indexed: 12/26/2022] Open
Abstract
Phosphodiesterase 10A (PDE10A) inhibitors show therapeutic effects for diseases with striatal pathology. PET radiotracers have been developed to quantify in vivo PDE10A levels and target engagement for therapeutic interventions. The aim of this study was to compare two potent and selective PDE10A radiotracers, [11C]TZ1964B and [18F]MNI659 in the nonhuman primate (NHP) brain. Double scans in the same cynomolgus monkey on the same day were performed after injection of [11C]TZ1964B and [18F]MNI659. Specific uptake was determined in two ways: nondisplaceable binding potential (BPND) was calculated using cerebellum as the reference region and the PDE‐10A enriched striatum as the target region of interest (ROI); the area under the time–activity curve (AUC) for the striatum to cerebellum ratio was also calculated. High‐performance liquid chromatography (HPLC) analysis of solvent‐extracted NHP plasma identified the percentage of intact tracer versus radiolabeled metabolites samples post injection of each radiotracer. Both radiotracers showed high specific accumulation in NHP striatum. [11C]TZ1964B has higher striatal retention and lower specific striatal uptake than [18F]MNI659. The BPND estimates of [11C]TZ1964B were 3.72 by Logan Reference model (LoganREF) and 4.39 by simplified reference tissue model (SRTM); the BPND estimates for [18F]MNI659 were 5.08 (LoganREF) and 5.33 (SRTM). AUC ratios were 5.87 for [11C]TZ1964B and 7.60 for [18F]MNI659. Based on BPND values in NHP striatum, coefficients of variation were ~10% for [11C]TZ1964B and ~30% for [18F]MNI659. Moreover, the metabolism study showed the percentage of parent compounds were ~70% for [11C]TZ1964B and ~50% for [18F]MNI659 60 min post injection. These data indicate that either [11C]TZ1964B or [18F]MNI659 could serve as suitable PDE10A PET radiotracers with distinguishing features for particular clinical application.
Collapse
Affiliation(s)
- Hui Liu
- Department of Radiology Washington University School of Medicine St. Louis Missouri
| | - Hongjun Jin
- Department of Radiology Washington University School of Medicine St. Louis Missouri
| | - Xuyi Yue
- Department of Radiology Washington University School of Medicine St. Louis Missouri
| | - Junbin Han
- Department of Radiology Washington University School of Medicine St. Louis Missouri
| | - Hao Yang
- Department of Radiology Washington University School of Medicine St. Louis Missouri
| | - Hubert Flores
- Department of Neurology Washington University School of Medicine St. Louis Missouri
| | - Yi Su
- Department of Radiology Washington University School of Medicine St. Louis Missouri
| | | | - Joel S Perlmutter
- Department of Radiology Washington University School of Medicine St. Louis Missouri; Department of Neurology Washington University School of Medicine St. Louis Missouri; Department of Neuroscience Physical Therapy and Occupational Therapy Washington University School of Medicine St. Louis Missouri
| | | | - Zhude Tu
- Department of Radiology Washington University School of Medicine St. Louis Missouri
| |
Collapse
|
18
|
Novel Radioligands for Cyclic Nucleotide Phosphodiesterase Imaging with Positron Emission Tomography: An Update on Developments Since 2012. Molecules 2016; 21:molecules21050650. [PMID: 27213312 PMCID: PMC6273803 DOI: 10.3390/molecules21050650] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 05/09/2016] [Accepted: 05/10/2016] [Indexed: 12/19/2022] Open
Abstract
Cyclic nucleotide phosphodiesterases (PDEs) are a class of intracellular enzymes that inactivate the secondary messenger molecules, cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Thus, PDEs regulate the signaling cascades mediated by these cyclic nucleotides and affect fundamental intracellular processes. Pharmacological inhibition of PDE activity is a promising strategy for treatment of several diseases. However, the role of the different PDEs in related pathologies is not completely clarified yet. PDE-specific radioligands enable non-invasive visualization and quantification of these enzymes by positron emission tomography (PET) in vivo and provide an important translational tool for elucidation of the relationship between altered expression of PDEs and pathophysiological effects as well as (pre-)clinical evaluation of novel PDE inhibitors developed as therapeutics. Herein we present an overview of novel PDE radioligands for PET published since 2012.
Collapse
|
19
|
Li J, Zhang X, Jin H, Fan J, Flores H, Perlmutter JS, Tu Z. Synthesis of Fluorine-Containing Phosphodiesterase 10A (PDE10A) Inhibitors and the In Vivo Evaluation of F-18 Labeled PDE10A PET Tracers in Rodent and Nonhuman Primate. J Med Chem 2015; 58:8584-600. [PMID: 26430878 DOI: 10.1021/acs.jmedchem.5b01205] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A series of fluorine-containing PDE10A inhibitors were designed and synthesized to improve the metabolic stability of [(11)C]MP-10. Twenty of the 22 new analogues had high potency and selectivity for PDE10A: 18a-j, 19d-j, 20a-b, and 21b had IC50 values <5 nM for PDE10A. Seven F-18 labeled compounds [(18)F]18a-e, [(18)F]18g, and [(18)F]20a were radiosynthesized by (18)F-introduction onto the quinoline rather than the pyrazole moiety of the MP-10 pharmacophore and performed in vivo evaluation. Biodistribution studies in rats showed ~2-fold higher activity in the PDE10A-enriched striatum than nontarget brain regions; this ratio increased from 5 to 30 min postinjection, particularly for [(18)F]18a-d and [(18)F]20a. MicroPET studies of [(18)F]18d and [(18)F]20a in nonhuman primates provided clear visualization of striatum with suitable equilibrium kinetics and favorable metabolic stability. These results suggest this strategy may identify a (18)F-labeled PET tracer for quantifying the levels of PDE10A in patients with CNS disorders including Huntington's disease and schizophrenia.
Collapse
Affiliation(s)
- Junfeng Li
- Department of Radiology and ‡Department of Neurology, Washington University School of Medicine , St. Louis, Missouri 63110, United States
| | - Xiang Zhang
- Department of Radiology and ‡Department of Neurology, Washington University School of Medicine , St. Louis, Missouri 63110, United States
| | - Hongjun Jin
- Department of Radiology and ‡Department of Neurology, Washington University School of Medicine , St. Louis, Missouri 63110, United States
| | - Jinda Fan
- Department of Radiology and ‡Department of Neurology, Washington University School of Medicine , St. Louis, Missouri 63110, United States
| | - Hubert Flores
- Department of Radiology and ‡Department of Neurology, Washington University School of Medicine , St. Louis, Missouri 63110, United States
| | - Joel S Perlmutter
- Department of Radiology and ‡Department of Neurology, Washington University School of Medicine , St. Louis, Missouri 63110, United States
| | - Zhude Tu
- Department of Radiology and ‡Department of Neurology, Washington University School of Medicine , St. Louis, Missouri 63110, United States
| |
Collapse
|
20
|
Liu H, Jin H, Yue X, Zhang X, Yang H, Li J, Flores H, Su Y, Perlmutter JS, Tu Z. Preclinical evaluation of a promising C-11 labeled PET tracer for imaging phosphodiesterase 10A in the brain of living subject. Neuroimage 2015. [PMID: 26216275 DOI: 10.1016/j.neuroimage.2015.07.049] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Phosphodiesterase 10A (PDE10A) plays a key role in the regulation of brain striatal signaling. A PET tracer for PDE10A may serve as a tool to evaluate PDE10A expression in vivo in central nervous system disorders with striatal pathology. Here, we further characterized the binding properties of a previously reported radioligand we developed for PDE10A, [(11)C]TZ1964B, in rodents and nonhuman primates (NHPs). The tritiated counterpart [(3)H]TZ1964B was used for in vitro binding characterizations in rat striatum homogenates and in vitro autoradiographic studies in rat brain slices. The carbon-11 labeled [(11)C]TZ1964B was utilized in the ex vivo autoradiography studies for the brain of rats and microPET imaging studies for the brain of NHPs. MicroPET scans of [(11)C]TZ1964B in NHPs were conducted at baseline, as well as with using a selective PDE10A inhibitor MP-10 for either pretreatment or displacement. The in vivo regional target occupancy (Occ) was obtained by pretreating with different doses of MP-10 (0.05-2.00 mg/kg). Both in vitro binding assays and in vitro autoradiographic studies revealed a nanomolar binding affinity of [(3)H]TZ1964B to the rat striatum. The striatal binding of [(3)H]TZ1964B and [(11)C]TZ1964B was either displaced or blocked by MP-10 in rats and NHPs. Autoradiography and microPET imaging confirmed that the specific binding of the radioligand was found in the striatum but not in the cerebellum. Blocking studies also confirmed the suitability of the cerebellum as an appropriate reference region. The binding potentials (BPND) of [(11)C]TZ1964B in the NHP striatum that were calculated using either the Logan reference model (LoganREF, 3.96 ± 0.17) or the simplified reference tissue model (SRTM, 4.64 ± 0.47), with the cerebellum as the reference region, was high and had good reproducibility. The occupancy studies indicated a MP-10 dose of 0.31 ± 0.09 mg/kg (LoganREF)/0.45 ± 0.17mg/kg (SRTM) occupies 50% striatal PDE10A binding sites. Studies in rats and NHPs demonstrated radiolabeled TZ1964B has a high binding affinity and good specificity for PDE10A, as well as favorable in vivo pharmacokinetic properties and binding profiles. Our data suggests that [(11)C]TZ1964B is a promising radioligand for in vivo imaging PDE10A in the brain of living subject.
Collapse
Affiliation(s)
- Hui Liu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hongjun Jin
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Xuyi Yue
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Xiang Zhang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hao Yang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Junfeng Li
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Hubert Flores
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yi Su
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joel S Perlmutter
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Physical Therapy and Occupational Therapy, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Zhude Tu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
21
|
Tu Z, Zhang X, Jin H, Yue X, Padakanti PK, Yu L, Liu H, Flores HP, Kaneshige K, Parsons SM, Perlmutter JS. Synthesis and biological characterization of a promising F-18 PET tracer for vesicular acetylcholine transporter. Bioorg Med Chem 2015; 23:4699-4709. [PMID: 26138195 DOI: 10.1016/j.bmc.2015.05.058] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 05/22/2015] [Accepted: 05/28/2015] [Indexed: 12/14/2022]
Abstract
Nine fluorine-containing vesicular acetylcholine transporter (VAChT) inhibitors were synthesized and screened as potential PET tracers for imaging the VAChT. Compound 18a was one of the most promising carbonyl-containing benzovesamicol analogs; the minus enantiomer, (-)-18a displayed high potency (VAChT Ki=0.59 ± 0.06 nM) and high selectivity for VAChT versus σ receptors (>10,000-fold). The radiosynthesis of (-)-[(18)F]18a was accomplished by a two-step procedure with 30-40% radiochemical yield. Preliminary biodistribution studies of (-)-[(18)F]18a in adult male Sprague-Dawley rats at 5, 30, 60 and 120 min post-injection (p.i.) were promising. The total brain uptake of (-)-[(18)F]18a was 0.684%ID/g at 5 min p.i. and by 120 min p.i. slowly washed out to 0.409 %ID/g; evaluation of regional brain uptake showed stable levels of ∼0.800 %ID/g from 5 to 120 min p.i in the VAChT-enriched striatal tissue of rats, indicating the tracer had crossed the blood brain barrier and was retained in the striatum. Subsequent microPET brain imaging studies of (-)-[(18)F]18a in nonhuman primates (NHPs) showed high striatal accumulation in the NHP brain; the standardized uptake value (SUV) for striatum reached a maximum value of 5.1 at 15 min p.i. The time-activity curve for the target striatal region displayed a slow and gradual decreasing trend 15 min after injection, while clearance of the radioactivity from the cerebellar reference region was much more rapid. Pretreatment of NHPs with 0.25mg/kg of the VAChT inhibitor (-)-vesamicol resulted in a ∼90% decrease of striatal uptake compared to baseline studies. HPLC metabolite analysis of NHP plasma revealed that (-)-[(18)F]18a had a good in vivo stability. Together, these preliminary results suggest (-)-[(18)F]18a is a promising PET tracer candidate for imaging VAChT in the brain of living subjects.
Collapse
Affiliation(s)
- Zhude Tu
- Department of Radiology, Washington University School of Medicine, 510 South Kingshighway Blvd., St. Louis, MO 63110, USA.
| | - Xiang Zhang
- Department of Radiology, Washington University School of Medicine, 510 South Kingshighway Blvd., St. Louis, MO 63110, USA
| | - Hongjun Jin
- Department of Radiology, Washington University School of Medicine, 510 South Kingshighway Blvd., St. Louis, MO 63110, USA
| | - Xuyi Yue
- Department of Radiology, Washington University School of Medicine, 510 South Kingshighway Blvd., St. Louis, MO 63110, USA
| | - Prashanth K Padakanti
- Department of Radiology, Washington University School of Medicine, 510 South Kingshighway Blvd., St. Louis, MO 63110, USA
| | - Lihai Yu
- Department of Radiology, Washington University School of Medicine, 510 South Kingshighway Blvd., St. Louis, MO 63110, USA
| | - Hui Liu
- Department of Radiology, Washington University School of Medicine, 510 South Kingshighway Blvd., St. Louis, MO 63110, USA
| | - Hubert P Flores
- Department of Neurology, Washington University School of Medicine, 510 South Kingshighway Blvd., St. Louis, MO 63110, USA
| | - Kota Kaneshige
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106, USA
| | - Stanley M Parsons
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106, USA
| | - Joel S Perlmutter
- Department of Radiology, Washington University School of Medicine, 510 South Kingshighway Blvd., St. Louis, MO 63110, USA; Department of Neurology, Washington University School of Medicine, 510 South Kingshighway Blvd., St. Louis, MO 63110, USA
| |
Collapse
|
22
|
Cox CD, Hostetler ED, Flores BA, Evelhoch JL, Fan H, Gantert L, Holahan M, Eng W, Joshi A, McGaughey G, Meng X, Purcell M, Raheem IT, Riffel K, Yan Y, Renger JJ, Smith SM, Coleman PJ. Discovery of [¹¹C]MK-8193 as a PET tracer to measure target engagement of phosphodiesterase 10A (PDE10A) inhibitors. Bioorg Med Chem Lett 2015; 25:4893-4898. [PMID: 26077491 DOI: 10.1016/j.bmcl.2015.05.080] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Revised: 05/22/2015] [Accepted: 05/26/2015] [Indexed: 01/30/2023]
Abstract
Phosphodiesterase 10A (PDE10A) inhibition has recently been identified as a potential mechanism to treat multiple symptoms that manifest in schizophrenia. In order to facilitate preclinical development and support key proof-of-concept clinical trials of novel PDE10A inhibitors, it is critical to discover positron emission tomography (PET) tracers that enable plasma concentration/PDE10A occupancy relationships to be established across species with structurally diverse PDE10A inhibitors. In this Letter, we describe how a high-throughput screening hit was optimized to provide [(11)C]MK-8193 (8j), a PET tracer that supports the determination of plasma concentration/PDE10A occupancy relationships for structurally diverse series of PDE10A inhibitors in both rat and rhesus monkey.
Collapse
Affiliation(s)
- Christopher D Cox
- Discovery Chemistry, Merck Research Laboratories, West Point, PA 19486, USA.
| | | | - Broc A Flores
- Discovery Chemistry, Merck Research Laboratories, West Point, PA 19486, USA
| | | | - Hong Fan
- Imaging, Merck Research Laboratories, West Point, PA 19486, USA
| | - Liza Gantert
- Imaging, Merck Research Laboratories, West Point, PA 19486, USA
| | - Marie Holahan
- Imaging, Merck Research Laboratories, West Point, PA 19486, USA
| | - Waisi Eng
- Imaging, Merck Research Laboratories, West Point, PA 19486, USA
| | - Aniket Joshi
- Imaging, Merck Research Laboratories, West Point, PA 19486, USA
| | - Georgia McGaughey
- Chemical Modeling & Informatics, Merck Research Laboratories, West Point, PA 19486, USA
| | - Xiangjun Meng
- Imaging, Merck Research Laboratories, West Point, PA 19486, USA
| | - Mona Purcell
- Imaging, Merck Research Laboratories, West Point, PA 19486, USA
| | - Izzat T Raheem
- Discovery Chemistry, Merck Research Laboratories, West Point, PA 19486, USA
| | - Kerry Riffel
- Imaging, Merck Research Laboratories, West Point, PA 19486, USA
| | - Youwei Yan
- Structural Chemistry, Merck Research Laboratories, West Point, PA 19486, USA
| | - John J Renger
- Neuroscience, Merck Research Laboratories, West Point, PA 19486, USA
| | - Sean M Smith
- Neuroscience, Merck Research Laboratories, West Point, PA 19486, USA
| | - Paul J Coleman
- Discovery Chemistry, Merck Research Laboratories, West Point, PA 19486, USA
| |
Collapse
|
23
|
Takano A, Stepanov V, Gulyás B, Nakao R, Amini N, Miura S, Kimura H, Taniguchi T, Halldin C. Evaluation of a novel PDE10A PET radioligand, [(11) C]T-773, in nonhuman primates: brain and whole body PET and brain autoradiography. Synapse 2015; 69:345-55. [PMID: 25892433 DOI: 10.1002/syn.21821] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 03/30/2015] [Accepted: 04/01/2015] [Indexed: 11/08/2022]
Abstract
Phosphodiesterase 10A (PDE10A) is considered to be a key target for the treatment of several neuropsychiatric diseases. The characteristics of [(11) C]T-773, a novel positron emission tomography (PET) radioligand with high binding affinity and selectivity for PDE10A, were evaluated in autoradiography and in nonhuman primate (NHP) PET. Brain PET measurements were performed under baseline conditions and after administration of a selective PDE10A inhibitor, MP-10. Total distribution volume (VT ) and binding potential (BPND ) were calculated using various kinetic models. Whole body PET measurements were performed to calculate the effective dose of [(11) C]T-773. Autoradiography studies in postmortem human and monkey brain sections showed high accumulation of [(11) C]T-773 in the striatum and substantia nigra which was blocked by MP-10. Brain PET showed high accumulation of [(11) C]T-773 in the striatum, and the data could be fitted using a two tissue compartment model. BPND was approximately 1.8 in the putamen when the cerebellum was used as the reference region. Approximately 70% of PDE10A binding was occupied by 1.8 mg/kg of MP-10. Whole body PET showed high accumulation of [(11) C]T-773 in the liver, kidney, heart, and brain in the initial phase. The radioligand was partly excreted via bile and the gastrointestinal tract, and partly excreted through the urinary tract. The calculated effective dose was 0.007 mSv/MBq. In conclusion, [(11) C]T-773 was demonstrated to be a promising PET radioligand for PDE10A with favorable brain kinetics. Dosimetry results support multiple PET measurements per person in human studies. Further research is required with [(11) C]T-773 in order to test the radioligand's potential clinical applications.
Collapse
Affiliation(s)
- Akihiro Takano
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Vladimir Stepanov
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Balázs Gulyás
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Ryuji Nakao
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Nahid Amini
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Shotaro Miura
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden.,CNS Drug Discovery Unit, Pharmaceutical Research Division, TAKEDA Pharmaceutical Company, Ltd., Fujisawa, Japan
| | - Haruhide Kimura
- CNS Drug Discovery Unit, Pharmaceutical Research Division, TAKEDA Pharmaceutical Company, Ltd., Fujisawa, Japan
| | - Takahiko Taniguchi
- CNS Drug Discovery Unit, Pharmaceutical Research Division, TAKEDA Pharmaceutical Company, Ltd., Fujisawa, Japan
| | - Christer Halldin
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
24
|
Stepanov V, Miura S, Takano A, Amini N, Nakao R, Hasui T, Nakashima K, Taniguchi T, Kimura H, Kuroita T, Halldin C. Development of a series of novel carbon-11 labeled PDE10A inhibitors. J Labelled Comp Radiopharm 2015; 58:202-8. [PMID: 25891816 DOI: 10.1002/jlcr.3284] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 12/22/2014] [Accepted: 03/04/2015] [Indexed: 11/11/2022]
Abstract
Phosphodiesterase 10A (PDE10A) is a member of the PDE family of enzymes that degrades cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Our aim was to label a series of structurally related PDE10A inhibitors with carbon-11 and evaluate them as potential positron emission tomography (PET) radioligands for PDE10A using nonhuman primates. The series consisted of seven compounds based on the 3-(1H-pyrazol-5-yl)pyridazin-4(1H)-one backbone. These compounds were selected from the initial larger library based on a number of parameters such as affinity, selectivity for hPDE10A in in vitro tests, lipophilicity, and on the results of multidrug resistance protein 1 (MDR1)-LLCPK1 and the parallel artificial membrane permeability assays. Seven radioligands (KIT-1, 3, 5, 6, 7, 9, and 12) were radiolabeled with carbon-11 employing O-methylation on the hydroxyl moiety using [(11)C]methyl triflate. In vivo examination of each radioligand was performed using PET in rhesus monkeys; analysis of radiometabolites in plasma also was conducted using HPLC. All seven radioligands were labeled with high (>90%) incorporation of [(11)C]methyl triflate into their appropriate precursors and with high specific radioactivity. Carbon-11 labeled KIT-5 and KIT-6 showed high accumulation in the striatum, consistent with the known anatomical distribution of PDE10A in brain, accompanied by fast washout and high specific binding ratio. In particular [(11)C]KIT-6, named [(11)C]T-773, is a promising PET tool for further examination of PDE10A in human brain.
Collapse
Affiliation(s)
- Vladimir Stepanov
- Department of Clinical Neuroscience, Center for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Durham TB, Blanco MJ. Target Engagement in Lead Generation. Bioorg Med Chem Lett 2015; 25:998-1008. [DOI: 10.1016/j.bmcl.2014.12.076] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Revised: 12/15/2014] [Accepted: 12/23/2014] [Indexed: 12/15/2022]
|
26
|
Yang H, Murigi FN, Wang Z, Li J, Jin H, Tu Z. Synthesis and in vitro characterization of cinnoline and benzimidazole analogues as phosphodiesterase 10A inhibitors. Bioorg Med Chem Lett 2014; 25:919-24. [PMID: 25592707 DOI: 10.1016/j.bmcl.2014.12.054] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 12/12/2014] [Accepted: 12/15/2014] [Indexed: 12/11/2022]
Abstract
Fifteen cinnoline analogues and six benzimidazole phosphodiesterase 10A (PDE10A) inhibitors were synthesized as potential PET radiopharmaceuticals and their in vitro activity as PDE10A inhibitors was determined. Nine out of twenty-one compounds were potent inhibitors of PDE10A with IC50 values ranging from 1.5 to 18.6nM. Notably, the IC50 values of compounds 26a, 26b, and 33c were 1.52±0.18, 2.86±0.10, and 3.73±0.60nM, respectively; these three compounds also showed high in vitro selectivity (>1000-fold) for PDE10A over PDE 3A/3B, PDE4A/4B. The high potency and selectivity of these three compounds suggests that they could be radiolabeled with PET radionuclides for further evaluation of their in vivo pharmacological behavior and ability to quantify PDE10A in the brain.
Collapse
Affiliation(s)
- Hao Yang
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO 63110, United States
| | - Francis N Murigi
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO 63110, United States
| | - Zhijian Wang
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO 63110, United States
| | - Junfeng Li
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO 63110, United States
| | - Hongjun Jin
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO 63110, United States
| | - Zhude Tu
- Department of Radiology, Washington University School of Medicine, Saint Louis, MO 63110, United States.
| |
Collapse
|
27
|
Lin SF, Labaree D, Chen MK, Holden D, Gallezot JD, Kapinos M, Teng JK, Najafzadeh S, Plisson C, Rabiner EA, Gunn RN, Carson RE, Huang Y. Further evaluation of [11C]MP-10 as a radiotracer for phosphodiesterase 10A: PET imaging study in rhesus monkeys and brain tissue metabolite analysis. Synapse 2014; 69:86-95. [PMID: 25450608 DOI: 10.1002/syn.21792] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 10/24/2014] [Accepted: 11/15/2014] [Indexed: 11/06/2022]
Abstract
[(11)C]MP-10 is a potent and specific PET tracer previously shown to be suitable for imaging the phosphodiesterase 10A (PDE10A) in baboons with reversible kinetics and high specific binding. However, another report indicated that [(11)C]MP-10 displayed seemingly irreversible kinetics in rhesus monkeys, potentially due to the presence of a radiolabeled metabolite capable of penetrating the blood-brain-barrier (BBB) into the brain. This study was designed to address the discrepancies between the species by re-evaluating [(11)C]MP-10 in vivo in rhesus monkey with baseline scans to assess tissue uptake kinetics and self-blocking scans with unlabeled MP-10 to determine binding specificity. Ex vivo studies with one rhesus monkey and 4 Sprague-Dawley rats were also performed to investigate the presence of radiolabeled metabolites in the brain. Our results indicated that [(11)C]MP-10 displayed reversible uptake kinetics in rhesus monkeys, albeit slower than in baboons. Administration of unlabeled MP-10 reduced the binding of [(11)C]MP-10 in a dose-dependent manner in all brain regions including the cerebellum. Consequently, the cerebellum appeared not to be a suitable reference tissue in rhesus monkeys. Regional volume of distribution (VT) was mostly reliably derived with the multilinear analysis (MA1) method. In ex vivo studies in the monkey and rats only negligible amount of radiometabolites was seen in the brain of either species. In summary, results from the present study strongly support the suitability of [(11)C]MP-10 as a radiotracer for PET imaging and quantification of PDE10A in nonhuman primates.
Collapse
Affiliation(s)
- Shu-Fei Lin
- Department of Diagnostic Radiology, PET Center, Yale University School of Medicine, New Haven, Connecticut
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Harada A, Suzuki K, Miura S, Hasui T, Kamiguchi N, Ishii T, Taniguchi T, Kuroita T, Takano A, Stepanov V, Halldin C, Kimura H. Characterization of the binding properties of T-773 as a PET radioligand for phosphodiesterase 10A. Nucl Med Biol 2014; 42:146-54. [PMID: 25451212 DOI: 10.1016/j.nucmedbio.2014.09.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Revised: 09/02/2014] [Accepted: 09/04/2014] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Phosphodiesterase 10A (PDE10A) is a dual-substrate PDE that hydrolyzes both cAMP and cGMP and is selectively expressed in striatal medium spiny neurons. Recent studies have suggested that PDE10A inhibition is a novel approach for the treatment of disorders such as schizophrenia and Huntington's disease. A positron emission tomography (PET) occupancy study can provide useful information for the development of PDE10A inhibitors. We discovered T-773 as a candidate PET radioligand for PDE10A and investigated its properties by in vitro autoradiography and a PET study in a monkey. METHODS Profiling of T-773 as a PET radioligand for PDE10A was conducted by in vitro enzyme inhibitory assay, in vitro autoradiography, and PET study in a monkey. RESULTS T-773 showed a high binding affinity and selectivity for human recombinant PDE10A2 in vitro; the IC50 value in an enzyme inhibitory assay was 0.77nmol/L, and selectivity over other PDEs was more than 2500-fold. In autoradiography studies using mouse, rat, monkey, or human brain sections, radiolabeled T-773 selectively accumulated in the striatum. This selective accumulation was not observed in the brain sections of Pde10a-KO mice. The binding of [(3)H]T-773 to PDE10A in rat brain sections was competitively inhibited by MP-10, a selective PDE10A inhibitor. In rat brain sections, [(3)H]T-773 bound to a single high affinity site of PDE10A with Kd values of 12.2±2.2 and 4.7±1.2nmol/L in the caudate-putamen and nucleus accumbens, respectively. In a monkey PET study, [(11)C]T-773 showed good brain penetration and striatum-selective accumulation. CONCLUSION These results suggest that [(11)C]T-773 is a potential PET radioligand for PDE10A.
Collapse
Affiliation(s)
- Akina Harada
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Kazunori Suzuki
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Shotaro Miura
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Tomoaki Hasui
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Naomi Kamiguchi
- Drug Metabolism and Pharmacokinetics Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Tsuyoshi Ishii
- Biomolecular Research Laboratories, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Takahiko Taniguchi
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Takanobu Kuroita
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Akihiro Takano
- Center for Psychiatric Research, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Vladimir Stepanov
- Center for Psychiatric Research, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Christer Halldin
- Center for Psychiatric Research, Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Haruhide Kimura
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Japan.
| |
Collapse
|
29
|
Kehler J, Kilburn JP, Estrada S, Christensen SR, Wall A, Thibblin A, Lubberink M, Bundgaard C, Brennum LT, Steiniger-Brach B, Christoffersen CT, Timmermann S, Kreilgaard M, Antoni G, Bang-Andersen B, Nielsen J. Discovery and development of 11C-Lu AE92686 as a radioligand for PET imaging of phosphodiesterase10A in the brain. J Nucl Med 2014; 55:1513-8. [PMID: 24994928 DOI: 10.2967/jnumed.114.140178] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED Phosphodiesterase 10A (PDE10A) plays a key role in the regulation of brain striatal signaling, and several pharmaceutical companies currently investigate PDE10A inhibitors in clinical trials for various central nervous system diseases. A PDE10A PET ligand may provide evidence that a clinical drug candidate reaches and binds to the target. Here we describe the successful discovery and initial validation of the novel radiolabeled PDE10A ligand 5,8-dimethyl-2-[2-((1-(11)C-methyl)-4-phenyl-1H-imidazol-2-yl)-ethyl]-[1,2,4]triazolo[1,5-a]pyridine ((11)C-Lu AE92686) and its tritiated analog (3)H-Lu AE92686. METHODS Initial in vitro experiments suggested Lu AE92686 as a promising radioligand, and the corresponding tritiated and (11)C-labeled compounds were synthesized. (3)H-Lu AE92686 was evaluated as a ligand for in vivo occupancy studies in mice and rats, and (11)C-Lu AE92686 was evaluated as a PET tracer candidate in cynomolgus monkeys and in humans. RESULTS (11)C-Lu AE92686 displayed high specificity and selectivity for PDE10A-expressing regions in the brain of cynomolgus monkeys and humans. Similar results were found in rodents using (3)H-Lu AE92686. The binding of (11)C-Lu AE92686 and (3)H-Lu AE92686 to striatum was completely and dose-dependently blocked by the structurally different PDE10A inhibitor 2-[4-(1-methyl-4-pyridin-4-yl-1H-pyrazol-3-yl)-phenoxymethyl]-quinoline (MP-10) in rodents and in monkeys. In all species, specific binding of the radioligand was seen in the striatum but not in the cerebellum, supporting the use of the cerebellum as a reference region. The binding potentials (BPND) of (11)C-Lu AE92686 in the striatum of both cynomolgus monkeys and humans were evaluated by the simplified reference tissue model with the cerebellum as the reference tissue, and BPND was found to be high and reproducible-that is, BPNDs were 6.5 ± 0.3 (n = 3) and 7.5 ± 1.0 (n = 12) in monkeys and humans, respectively. CONCLUSION Rodent, monkey, and human tests of labeled Lu AE92686 suggest that (11)C-Lu AE92686 has great potential as a human PET tracer for the PDE10A enzyme.
Collapse
Affiliation(s)
- Jan Kehler
- Division of Discovery Chemistry and DMPK, H. Lundbeck A/S, Valby, Denmark
| | - John Paul Kilburn
- Division of Discovery Chemistry and DMPK, H. Lundbeck A/S, Valby, Denmark
| | - Sergio Estrada
- Preclinical PET Platform, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | | | - Anders Wall
- Nuclear Medicine and PET, Uppsala University and Uppsala University Hospital, Uppsala, Sweden
| | - Alf Thibblin
- Preclinical PET Platform, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Mark Lubberink
- Nuclear Medicine and PET, Uppsala University and Uppsala University Hospital, Uppsala, Sweden
| | | | | | | | | | - Stine Timmermann
- Department of Quantitative Pharmacology, H. Lundbeck A/S, Valby, Denmark; and
| | - Mads Kreilgaard
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Gunnar Antoni
- Preclinical PET Platform, Department of Medicinal Chemistry, Uppsala University, Uppsala, Sweden
| | - Benny Bang-Andersen
- Division of Discovery Chemistry and DMPK, H. Lundbeck A/S, Valby, Denmark Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Jacob Nielsen
- Division of Synaptic Transmission, H. Lundbeck A/S, Valby, Denmark
| |
Collapse
|
30
|
Kulkarni P, Saxena U. Investigational drugs for the management of Huntington's disease: are we there yet? Expert Opin Investig Drugs 2014; 23:1595-603. [PMID: 25084527 DOI: 10.1517/13543784.2014.934807] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
INTRODUCTION Huntington's disease is a hereditary neurodegenerative disease. It is designated as a rare disease in the US, which means there are < 200,000 patients in the country who suffer from it. The drugs that are currently used to treat this disease were not designed specifically for it but developed for other diseases. Presently, two classes of drugs are being developed; those that provide symptomatic relief and those that may modify course of the disease. AREAS COVERED This review is focused on seven selected drugs currently in clinical testing and describes their progress. Five of the seven drugs that are reviewed here, can be categorized as 'symptomatic' drugs, and, selisistat and PBT-2 are amongst the ones that would qualify as 'disease modifying' drugs. EXPERT OPINION The authors believe that the future treatment paradigm for this disease is best met by using a disease-modifying drug that can be administered together with symptomatic drugs. Towards that end, it is important for the industry to focus on disease-modifying drugs by targeting unique pathways and targets. Furthermore, they propose that neuroprotective drugs, that is, drugs that directly work by preserving neuronal health and function is an opportunity for such disease-modifying drugs.
Collapse
Affiliation(s)
- Pushkar Kulkarni
- University of Hyderabad Campus, Dr. Reddy's Institute of Life Sciences , Gachibowli, Hyderabad , India
| | | |
Collapse
|
31
|
Barret O, Thomae D, Tavares A, Alagille D, Papin C, Waterhouse R, McCarthy T, Jennings D, Marek K, Russell D, Seibyl J, Tamagnan G. In Vivo Assessment and Dosimetry of 2 Novel PDE10A PET Radiotracers in Humans: 18F-MNI-659 and 18F-MNI-654. J Nucl Med 2014; 55:1297-304. [DOI: 10.2967/jnumed.113.122895] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 03/27/2014] [Indexed: 11/16/2022] Open
|
32
|
Ooms M, Celen S, Koole M, Langlois X, Schmidt M, De Angelis M, Andrés JI, Verbruggen A, Van Laere K, Bormans G. Synthesis and biological evaluation of carbon-11 and fluorine-18 labeled tracers for in vivo visualization of PDE10A. Nucl Med Biol 2014; 41:695-704. [PMID: 25002365 DOI: 10.1016/j.nucmedbio.2014.05.138] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 05/05/2014] [Accepted: 05/14/2014] [Indexed: 12/31/2022]
Abstract
INTRODUCTION In vivo visualization of PDE10A using PET provides a tool to evaluate the role of PDE10A in various neuropsychiatric diseases and can also be useful in the clinical evaluation of PDE10A inhibitor drug candidates. We evaluated several carbon-11 and fluorine-18 labeled PDE10A inhibitors as potential PDE10A PET radioligands. MATERIALS & METHODS [(11)C]MP10, [(11)C]JNJ42071965 and four other tracers were developed. Their biodistribution was evaluated in rats. Rat plasma and brain radiometabolites were quantified. Baseline microPET imaging was performed in normal rats and PDE10A knockout (KO) and wild-type (WT) mice. Blocking and displacement studies were conducted. The selectivity of the tracer binding was further studied in an ex vivo autoradiography experiment in PDE10A KO and WT mice. RESULTS Biodistribution showed brain uptake for all tracers in the striatum and wash-out from the cerebellum. [(11)C]1 ((11)C-MP10) had the highest specific uptake index (striatum (S) vs. cerebellum (C) ratios (S/C)-1) at 60 min (7.4). [(11)C]5 ([(11)C]JNJ42071965) had a high index at the early time points (1.0 and 3.7 at 2 and 30 min p.i., respectively). The affinity of [(11)C]4, [(18)F]3 and [(18)F]6 was too low to visualize PDE10A using microPET. [(11)C] 2 showed a specific binding, while kinetics of [(11)C]1 were too slow. [(11)C]5 reached equilibrium after 10 min (uptake index=1.2). Blocking and displacement experiments in rats and baseline imaging in PDE10A KO mice showed specific and reversible binding of [(11)C]5 to PDE10A. CONCLUSIONS We successfully radiolabeled and evaluated six radiotracers for their potential to visualize PDE10A in vivo. While [(11)C]1 had the highest striatal specific uptake index, its slow kinetics likely compromise clinical use of this tracer. [(11)C]5 has a relatively high striatum-to-background ratio and fast kinetic profile, which makes it a valuable carbon-11 alternative.
Collapse
Affiliation(s)
- Maarten Ooms
- Laboratory for Radiopharmacy, KU Leuven, Belgium; MoSAIC, Molecular Small Animal Imaging Centre, KU Leuven, Belgium
| | - Sofie Celen
- Laboratory for Radiopharmacy, KU Leuven, Belgium; MoSAIC, Molecular Small Animal Imaging Centre, KU Leuven, Belgium
| | - Michel Koole
- Department of Nuclear Medicine & Molecular Imaging, University Medical Center Groningen, The Netherlands
| | | | | | | | | | - Alfons Verbruggen
- Laboratory for Radiopharmacy, KU Leuven, Belgium; MoSAIC, Molecular Small Animal Imaging Centre, KU Leuven, Belgium
| | - Koen Van Laere
- MoSAIC, Molecular Small Animal Imaging Centre, KU Leuven, Belgium; Division of Nuclear Medicine, KU Leuven and University Hospital Leuven, Belgium
| | - Guy Bormans
- Laboratory for Radiopharmacy, KU Leuven, Belgium; MoSAIC, Molecular Small Animal Imaging Centre, KU Leuven, Belgium.
| |
Collapse
|
33
|
Schwan G, Barbar Asskar G, Höfgen N, Kubicova L, Funke U, Egerland U, Zahn M, Nieber K, Scheunemann M, Sträter N, Brust P, Briel D. Fluorine-containing 6,7-dialkoxybiaryl-based inhibitors for phosphodiesterase 10 A: synthesis and in vitro evaluation of inhibitory potency, selectivity, and metabolism. ChemMedChem 2014; 9:1476-87. [PMID: 24729456 DOI: 10.1002/cmdc.201300522] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 02/25/2014] [Indexed: 11/10/2022]
Abstract
Based on the potent phosphodiesterase 10 A (PDE10A) inhibitor PQ-10, we synthesized 32 derivatives to determine relationships between their molecular structure and binding properties. Their roles as potential positron emission tomography (PET) ligands were evaluated, as well as their inhibitory potency toward PDE10A and other PDEs, and their metabolic stability was determined in vitro. According to our findings, halo-alkyl substituents at position 2 of the quinazoline moiety and/or halo-alkyloxy substituents at positions 6 or 7 affect not only the compounds' affinity, but also their selectivity toward PDE10A. As a result of substituting the methoxy group for a monofluoroethoxy or difluoroethoxy group at position 6 of the quinazoline ring, the selectivity for PDE10A over PDE3A increased. The same result was obtained by 6,7-difluoride substitution on the quinoxaline moiety. Finally, fluorinated compounds (R)-7-(fluoromethoxy)-6-methoxy-4-(3-(quinoxaline-2-yloxy)pyrrolidine-1-yl)quinazoline (16 a), 19 a-d, (R)-tert-butyl-3-(6-fluoroquinoxalin-2-yloxy)pyrrolidine-1-carboxylate (29), and 35 (IC50 PDE10A 11-65 nM) showed the highest inhibitory potential. Further, fluoroethoxy substitution at position 7 of the quinazoline ring improved metabolic stability over that of the lead structure PQ-10.
Collapse
Affiliation(s)
- Gregor Schwan
- Institut für Pharmazie, Universität Leipzig, Brüderstr. 34, 04103 Leipzig (Germany), Fax: (+49) 341 9736889
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Natesan S, Ashworth S, Nielsen J, Tang SP, Salinas C, Kealey S, Lauridsen JB, Stensbøl TB, Gunn RN, Rabiner EA, Kapur S. Effect of chronic antipsychotic treatment on striatal phosphodiesterase 10A levels: a [¹¹C]MP-10 PET rodent imaging study with ex vivo confirmation. Transl Psychiatry 2014; 4:e376. [PMID: 24690597 PMCID: PMC4012281 DOI: 10.1038/tp.2014.17] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Revised: 01/14/2014] [Accepted: 02/23/2014] [Indexed: 12/31/2022] Open
Abstract
A number of phosphodiesterase 10A (PDE10) inhibitors are about to undergo clinical evaluation for their efficacy in treating schizophrenia. As phosphodiesterases are in the same signalling pathway as dopamine D2 receptors, it is possible that prior antipsychotic treatment could influence these enzyme systems in patients. Chronic, in contrast to acute, antipsychotic treatment has been reported to increase brain PDE10A levels in rodents. The aim of this study was to confirm these findings in a manner that can be translated to human imaging studies to understand its consequences. Positron emission tomography (PET) scanning was used to evaluate PDE10A enzyme availability, after chronic haloperidol administration, using a specific PDE10A ligand ([(11)C]MP-10). The binding of [(11)C]MP-10 in the striatum and the cerebellum was measured in rodents and a simplified reference tissue model (SRTM) with cerebellum as the reference region was used to determine the binding potential (BPND). In rats treated chronically with haloperidol (2 mg kg(-1) per day), there was no significant difference in PDE10A levels compared with the vehicle-treated group (BPND±s.d.: 3.57 ± 0.64 versus 2.86 ± 0.71). Following PET scans, ex vivo analysis of striatal brain tissue for PDE10A mRNA (Pde10a) and PDE10A enzyme activity showed no significant difference. Similarly, the PDE10A protein content determined by western blot analysis was similar between the two groups, contrary to an earlier finding. The results of the study indicate that prior exposure to antipsychotic medication in rodents does not alter PDE10A levels.
Collapse
Affiliation(s)
- S Natesan
- Department of Psychosis Studies, Institute of Psychiatry, King's College London, London, UK
| | - S Ashworth
- Imanova Centre for Imaging Sciences, Hammersmith Hospital, London, UK
| | - J Nielsen
- Division of Synaptic Transmission, H. Lundbeck A/S, Copenhagen, Denmark
| | - S-P Tang
- Imanova Centre for Imaging Sciences, Hammersmith Hospital, London, UK
| | - C Salinas
- Imanova Centre for Imaging Sciences, Hammersmith Hospital, London, UK
| | - S Kealey
- Department of Psychosis Studies, Institute of Psychiatry, King's College London, London, UK
| | - J B Lauridsen
- Division of Synaptic Transmission, H. Lundbeck A/S, Copenhagen, Denmark
| | - T B Stensbøl
- Division of Synaptic Transmission, H. Lundbeck A/S, Copenhagen, Denmark
| | - R N Gunn
- Imanova Centre for Imaging Sciences, Hammersmith Hospital, London, UK,Department of Medicine, Imperial College London, London, UK
| | - E A Rabiner
- Imanova Centre for Imaging Sciences, Hammersmith Hospital, London, UK,Department of Neuroimaging, Institute of Psychiatry, King's College London, London, UK
| | - S Kapur
- Department of Psychosis Studies, Institute of Psychiatry, King's College London, London, UK,Department of Psychosis Studies, Institute of Psychiatry, King's College London, De Crespigny Park, Denmark Hill, London SE5 8AF, UK. E-mail:
| |
Collapse
|
35
|
Radiosyntheses and in vivo evaluation of carbon-11 PET tracers for PDE10A in the brain of rodent and nonhuman primate. Bioorg Med Chem 2014; 22:2648-54. [PMID: 24721831 DOI: 10.1016/j.bmc.2014.03.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Revised: 03/06/2014] [Accepted: 03/17/2014] [Indexed: 11/22/2022]
Abstract
The radiosyntheses and in vivo evaluation of four carbon-11 labeled quinoline group-containing radioligands are reported here. Radiolabeling of [(11)C]1-4 was achieved by alkylation of their corresponding desmethyl precursors with [(11)C]CH3I. Preliminary biodistribution evaluation in Sprague-Dawley rats demonstrated that [(11)C]1 and [(11)C]2 had high striatal accumulation (at peak time) for [(11)C]1 and [(11)C]2 were 6.0-fold and 4.5-fold at 60 min, respectively. Following MP-10 pretreatment, striatal uptake in rats of [(11)C]1 and [(11)C]2 was reduced, suggesting that the tracers bind specifically to PDE10A. MicroPET studies of [(11)C]1 and [(11)C]2 in nonhuman primates (NHP) also showed good tracer retention in the striatum with rapid clearance from non-target brain regions. Striatal uptake (SUV) of [(11)C]1 reached 1.8 at 30 min with a 3.5-fold striatum:cerebellum ratio. In addition, HPLC analysis of solvent extracts from NHP plasma samples suggested that [(11)C]1 had a very favorable metabolic stability. Our preclinical investigations suggest that [(11)C]1 is a promising candidate for quantification of PDE10A in vivo using PET.
Collapse
|
36
|
Radiosynthesis and in Vivo Evaluation of Two PET Radioligands for Imaging α-Synuclein. APPLIED SCIENCES-BASEL 2014; 4:66-78. [PMID: 25642331 PMCID: PMC4310556 DOI: 10.3390/app4010066] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Two α-synuclein ligands, 3-methoxy-7-nitro-10H-phenothiazine (2a, Ki = 32.1 ± 1.3 nM) and 3-(2-fluoroethoxy)-7-nitro-10H-phenothiazine (2b, Ki = 49.0 ± 4.9 nM), were radiolabeled as potential PET imaging agents by respectively introducing 11C and 18F. The syntheses of [11C]2a and [18F]2b were accomplished in a good yield with high specific activity. Ex vivo biodistribution studies in rats revealed that both [11C]2a and [18F]2b crossed the blood-brain barrier (BBB) and demonstrated good brain uptake 5 min post-injection. MicroPET imaging of [11C]2a in a non-human primate (NHP) confirmed that the tracer was able to cross the BBB with rapid washout kinetics from brain regions of a healthy macaque. The initial studies suggested that further structural optimization of [11C]2a and [18F]2b is necessary in order to identify a highly specific positron emission tomography (PET) radioligand for in vivo imaging of α-synuclein aggregation in the central nervous system (CNS).
Collapse
|
37
|
Plisson C, Weinzimmer D, Jakobsen S, Natesan S, Salinas C, Lin SF, Labaree D, Zheng MQ, Nabulsi N, Marques TR, Kapur S, Kawanishi E, Saijo T, Gunn RN, Carson RE, Rabiner EA. Phosphodiesterase 10A PET Radioligand Development Program: From Pig to Human. J Nucl Med 2014; 55:595-601. [DOI: 10.2967/jnumed.113.131409] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
38
|
Hwang DR, Hu E, Rumfelt S, Easwaramoorthy B, Castrillon J, Davis C, Allen JR, Chen H, Treanor J, Abi-Dargham A, Slifstein M. Initial characterization of a PDE10A selective positron emission tomography tracer [11C]AMG 7980 in non-human primates. Nucl Med Biol 2014; 41:343-9. [PMID: 24607437 DOI: 10.1016/j.nucmedbio.2014.01.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 12/19/2013] [Accepted: 01/07/2014] [Indexed: 10/25/2022]
Abstract
INTRODUCTION Phosphodiesterase 10A (PDE10A) is an intracellular enzyme responsible for the breakdown of cyclic nucleotides which are important secondary messengers in the central nervous system. Inhibition of PDE10A has been identified as a potential therapeutic target for treatment of various neuropsychiatric disorders. To assist the drug development program, we have identified a selective PDE10A PET tracer, [(11)C]AMG 7980, for imaging PDE10A distribution using positron emission tomography. METHODS [(11)C]AMG 7980 was prepared in a one-pot, two-step reaction. Dynamic PET scans were performed in non-human primates following a bolus or bolus plus constant infusion tracer injection paradigm. Regions-of-interest were defined on individuals' MRIs and transferred to the co-registered PET images. Data were analyzed using Logan graphical analysis with metabolite-corrected input function, the simplified reference tissue model (SRTM) method and occupancy plots. A benchmark PDE10A inhibitor was used to demonstrate PDE10A-specific binding. RESULTS [(11)C]AMG 7980 was prepared with a mean specific activity of 99 ± 74 GBq/μmol (n=10) and a synthesis time of 45 min. Specific binding of the tracer was localized to the striatum and globus pallidus (GP) and low in other brain regions. Thalamus was used as the reference tissue to derive binding potentials (BPND). The BPND for caudate, putamen, and GP were 0.23, 0.65, 0.51, respectively by the graphical method, and 0.42, 0.76, and 0.75 from the SRTM method. A dose dependent decrease of BPND was observed with the pre-treatment of a PDE10A inhibitor. A bolus plus infusion injection paradigm yielded similar results. CONCLUSION [(11)C]AMG 7980 has been successfully used for imaging PDE10A in non-human primate brain. Despite the fast brain kinetics it can be used to measure target occupancy of PDE10A inhibitors in non-human primates and potentially applicable to humans.
Collapse
Affiliation(s)
- Dah-Ren Hwang
- Department of Medical Sciences, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, California 91320-1799, United States.
| | - Essa Hu
- Department of Small Molecule Chemistry, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Shannon Rumfelt
- Department of Small Molecule Chemistry, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Balu Easwaramoorthy
- Department of Psychiatry, Columbia University, New York, NY, USA; New York State Psychiatric Institute, NY, USA
| | | | - Carl Davis
- Department of Pharmacokinetics and Drug Metabolism, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Jennifer R Allen
- Department of Small Molecule Chemistry, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Hang Chen
- Department of Neuroscience, Amgen Inc., South San Francisco, CA
| | - James Treanor
- Department of Neuroscience, Amgen Inc., 1 Amgen Center Drive, Thousand Oaks, California 91320-1799, United States
| | - Anissa Abi-Dargham
- Department of Psychiatry, Columbia University, New York, NY, USA; Department of Radiology, Columbia University, New York, NY, USA; New York State Psychiatric Institute, NY, USA
| | - Mark Slifstein
- Department of Psychiatry, Columbia University, New York, NY, USA; New York State Psychiatric Institute, NY, USA
| |
Collapse
|
39
|
Rachakonda V, Alla M, Kotipalli SS, Ummani R. Design, diversity-oriented synthesis and structure activity relationship studies of quinolinyl heterocycles as antimycobacterial agents. Eur J Med Chem 2013; 70:536-47. [PMID: 24189497 DOI: 10.1016/j.ejmech.2013.10.034] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2013] [Revised: 10/11/2013] [Accepted: 10/12/2013] [Indexed: 11/26/2022]
Abstract
The current study reports design and diversity oriented synthesis of novel bis heterocycles with a common 2-methyl, C-4 unsubstituted quinoline moiety as the central key heterocycle. Employing reagent based skeletal diversity approach; a facile synthesis of bis heterocycles with different heterocyclic rings at C-3 position of the quinoline moiety has been accomplished. A broad range of heterocyclic frameworks thus obtained were evaluated for their antimycobacterial activity. The active scaffolds were further explored by a parallel library generation in order to establish SAR. Further, low cytotoxicity against A549 cell line enhances the potential of the synthesized molecules as promising antimycobacterial agents.
Collapse
Affiliation(s)
- Venkatesham Rachakonda
- Crop Protection Chemicals Division, CSIR-Indian Institute of Chemical Technology, Hyderabad 500 607, Andhra Pradesh, India
| | | | | | | |
Collapse
|
40
|
Van Laere K, Ahmad RU, Hudyana H, Dubois K, Schmidt ME, Celen S, Bormans G, Koole M. Quantification of 18F-JNJ-42259152, a novel phosphodiesterase 10A PET tracer: kinetic modeling and test-retest study in human brain. J Nucl Med 2013; 54:1285-93. [PMID: 23843566 DOI: 10.2967/jnumed.112.118679] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED Phosphodiesterase 10A (PDE10A) plays a central role in striatal signaling and is implicated in several neuropsychiatric disorders, such as movement disorders and schizophrenia. We performed initial brain kinetic modeling of the novel PDE10A tracer (18)F-JNJ-42259152 (2-[[4-[1-(2-(18)F-fluoroethyl)-4-(4-pyridinyl)-1H-pyrazol-3-yl]phenoxy]methyl]-3,5-dimethyl-pyridine) and studied test-retest reproducibility in healthy volunteers. METHODS Twelve healthy volunteers (5 men, 7 women; age range, 42-77 y) were scanned dynamically up to 135 min after bolus injection of 172.5 ± 10.3 MBq of (18)F-JNJ42259152. Four volunteers (2 men, 2 women) underwent retest scanning, with a mean interscan interval of 37 d. Input functions and tracer parent fractions were determined using arterial sampling and high-performance liquid chromatography analysis. Volumes of interest for the putamen, caudate nucleus, ventral striatum, substantia nigra, thalamus, frontal cortex, and cerebellum were delineated using individual volumetric T1 MR imaging scans. One-tissue (1T) and 2-tissue (2T) models were evaluated to calculate total distribution volume (VT). Simplified models were also tested to calculate binding potential (BPND), including the simplified reference tissue model (SRTM) and multilinear reference tissue model, using the frontal cortex as the optimal reference tissue. The stability of VT and BPND was assessed down to a 60-min scan time. RESULTS The average intact tracer half-life in blood was 90 min. The 2T model VT values for the putamen, caudate nucleus, ventral striatum, substantia nigra, thalamus, frontal cortex, and cerebellum were 1.54 ± 0.37, 0.90 ± 0.24, 0.64 ± 0.18, 0.42 ± 0.09, 0.35 ± 0.09, 0.30 ± 0.07, and 0.36 ± 0.12, respectively. The 1T model provided significantly lower VT values, which were well correlated to the 2T VT. SRTM BPND values referenced to the frontal cortex were 3.45 ± 0.43, 1.78 ± 0.35, 1.10 ± 0.31, and 0.44 ± 0.09 for the respective target regions putamen, caudate nucleus, ventral striatum, and substantia nigra, with similar values for the multilinear reference tissue model. Good correlations were found for the target regions putamen, caudate nucleus, ventral striatum, and substantia nigra between the 2T-compartment model BPND and the SRTM BPND (r = 0.57, 0.82, 0.70, and 0.64, respectively). SRTM BPND using a 90- and 60-min acquisition interval showed low bias. Test-retest variability was 5%-19% for 2T VT and 5%-12% for BPND SRTM. CONCLUSION Kinetic modeling of (18)F-JNJ-42259152 shows that PDE10A activity can be reliably quantified and simplified using a reference tissue model with the frontal cortex as reference and a 60-min acquisition period.
Collapse
Affiliation(s)
- Koen Van Laere
- Department of Imaging and Pathology, Nuclear Medicine, University Hospital and KU Leuven, Leuven, Belgium.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Celen S, Koole M, Ooms M, De Angelis M, Sannen I, Cornelis J, Alcazar J, Schmidt M, Verbruggen A, Langlois X, Van Laere K, Andrés JI, Bormans G. Preclinical evaluation of [(18)F]JNJ42259152 as a PET tracer for PDE10A. Neuroimage 2013; 82:13-22. [PMID: 23664955 DOI: 10.1016/j.neuroimage.2013.04.123] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 04/24/2013] [Accepted: 04/27/2013] [Indexed: 01/26/2023] Open
Abstract
Phosphodiesterase-10A (PDE10A) is implicated in several neuropsychiatric disorders involving basal ganglia neurotransmission, such as schizophrenia, obsessive-compulsive disorder and Huntington's disease. To confirm target engagement and exposure-occupancy relationships of clinical candidates for treatment, and to further explore the in vivo biology of PDE10A, non-invasive imaging using a specific PET ligand is warranted. Recently we have reported the in vivo evaluation of [(18)F]JNJ41510417 which showed specific binding to PDE10A in rat striatum, but with relatively slow kinetics. A chemically related derivative JNJ42259152 was found to have a similar in vivo occupancy, but lower lipophilicity and lower PDE10A in vitro inhibitory activity compared to JNJ41510417. (18)F-labeled JNJ42259152 was therefore evaluated as a potential PDE10A PET radiotracer. Baseline PET in rats and monkey showed specific retention in the PDE10A-rich striatum, and fast wash-out, with a good contrast to non-specific binding, in other brain regions. Pretreatment and chase experiments in rats with the selective PDE10A inhibitor MP-10 showed that tracer binding was specific and reversible. Absence of specific binding in PDE10A knock-out (KO) mice further confirmed PDE10A specificity. In vivo radiometabolite analysis using high performance liquid chromatography (HPLC) showed presence of polar radiometabolites in rat plasma and brain. In vivo imaging in rat and monkey further showed faster brain kinetics, and higher striatum-to-cerebellum ratios for [(18)F]JNJ42259152 compared to [(18)F]JNJ41510417. The arterial input function corrected for radiometabolites was determined in rats and basic kinetic modeling was established. For a 60-min acquisition time interval, striatal binding potential of the intact tracer referenced to the cerebellum showed good correlation with corresponding binding potential values of a Simplified Reference Tissue Model and referenced Logan Plot, the latter using a population averaged reference tissue-to-plasma clearance rate and offering the possibility to generate representative parametric binding potential images. In conclusion we can state that in vivo imaging in PDE10A KO mice, rats and monkey demonstrates that [(18)F]JNJ42259152 provides a PDE10A-specific signal in the striatum with good pharmacokinetic properties. Although presence of a polar radiometabolite in rat brain yielded a systematic but reproducible underestimation of the striatal BPND, a Logan reference tissue model approach using 60 min acquisition data is appropriate for quantification.
Collapse
Affiliation(s)
- S Celen
- Laboratory for Radiopharmacy, KU Leuven, Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Holland JP, Cumming P, Vasdev N. PET radiopharmaceuticals for probing enzymes in the brain. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2013; 3:194-216. [PMID: 23638333 PMCID: PMC3627518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Accepted: 03/07/2013] [Indexed: 06/02/2023]
Abstract
Biologically important processes in normal brain function and brain disease involve the action of various protein-based receptors, ion channels, transporters and enzymes. The ability to interrogate the location, abundance and activity of these entities in vivo using non-invasive molecular imaging can provide unprecedented information about the spatio-temporal dynamics of brain function. Indeed, positron emission tomography (PET) imaging is transforming our understanding of the central nervous system and brain disease. Great emphasis has historically been placed on developing radioligands for the non-invasive detection of neuroreceptors. In contrast, relatively few enzymes have been amenable to examination by PET imaging procedures based upon trapping or accumulation of enzymatic products, because only a subset of enzymes have sufficient catalytic rate to produce measureable accumulation within the practical time-limit of PET recordings. However, high affinity inhibitors are now serving as tracers for enzymes, particularly for measuring the abundance of enzymes mediating intracellular signal transduction in the brain, which offer a rich diversity of potential targets for drug discovery. The purpose of this review is to summarize well-known radiotracers for brain enzymes, and draw attention to recent developments in PET radiotracers for imaging signal transduction pathways in the brain. The review is organized by target class and focuses on structural chemistry of the best-established radiotracers identified in each class.
Collapse
Affiliation(s)
- Jason P Holland
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, and Department of Radiology, Harvard Medical School55 Fruit St., White 427, Boston, MA 02114, USA
| | - Paul Cumming
- Department of Nuclear Medicine, Universitätsklinikum ErlangenUlmenweg 18, Erlangen, Germany, 91054
| | - Neil Vasdev
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, and Department of Radiology, Harvard Medical School55 Fruit St., White 427, Boston, MA 02114, USA
| |
Collapse
|
43
|
Li J, Jin H, Zhou H, Rothfuss J, Tu Z. Synthesis and in vitro biological evaluation of pyrazole group-containing analogues for PDE10A. MEDCHEMCOMM 2013; 4:443-449. [PMID: 23585921 DOI: 10.1039/c2md20239e] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Twenty eight new analogues were synthesized by optimizing the structure of MP-10 and their in vitro binding affinities towards PDE10A, PDE3A/B, and PDE4A/B were determined. Among these new analogues, 10a, 10b, 10d, 11a, 11b and 11d are very potent towards PDE10A and have IC50 values of 0.40 ± 0.02, 0.28 ± 0.06, 1.82 ± 0.25, 0.24 ± 0.05, 0.36 ± 0.03 and 1.78 ± 0.03 nM respectively; these six compounds displayed high selectivity for PDE10A versus PDE3A/3B/4A/4B. The promising compounds will be further validated in vivo to identify PDE10A imaging tracers.
Collapse
Affiliation(s)
- Junfeng Li
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA; ; Tel: +1-314-362-8487
| | | | | | | | | |
Collapse
|
44
|
|
45
|
Human biodistribution and dosimetry of 18F-JNJ42259152, a radioligand for phosphodiesterase 10A imaging. Eur J Nucl Med Mol Imaging 2012; 40:254-61. [DOI: 10.1007/s00259-012-2270-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 10/01/2012] [Indexed: 10/27/2022]
|
46
|
Chappie TA, Helal CJ, Hou X. Current landscape of phosphodiesterase 10A (PDE10A) inhibition. J Med Chem 2012; 55:7299-331. [PMID: 22834877 DOI: 10.1021/jm3004976] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Thomas A Chappie
- Neuroscience Medicinal Chemistry, Pfizer, Inc. , 700 Main Street, Cambridge, MA 02139, USA.
| | | | | |
Collapse
|
47
|
Radiosynthesis and Radiotracer Properties of a 7-(2-[18F]Fluoroethoxy)-6-methoxypyrrolidinylquinazoline for Imaging of Phosphodiesterase 10A with PET. Pharmaceuticals (Basel) 2012; 5:169-88. [PMID: 24288087 PMCID: PMC3763632 DOI: 10.3390/ph5020169] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2011] [Revised: 01/18/2012] [Accepted: 01/19/2012] [Indexed: 12/02/2022] Open
Abstract
Phosphodiesterase 10A (PDE10A) is a key enzyme of intracellular signal transduction which is involved in the regulation of neurotransmission. The molecular imaging of PDE10A by PET is expected to allow a better understanding of physiological and pathological processes related to PDE10A expression and function in the brain. The aim of this study was to develop a new 18F-labeled PDE10A ligand based on a 6,7-dimethoxy-4-pyrrolidinylquinazoline and to evaluate its properties in biodistribution studies. Nucleophilic substitution of the 7-tosyloxy-analogue led to the 7-[18F]fluoroethoxy-derivative [18F]IV with radiochemical yields of 25% ± 9% (n = 9), high radiochemical purity of ≥99% and specific activities of 110–1,100 GBq/μmol. [18F]IV showed moderate PDE10A affinity (KD,PDE10A = 14 nM) and high metabolic stability in the brain of female CD-1 mice, wherein the radioligand entered rapidly with a peak uptake of 2.3% ID/g in striatum at 5 min p.i. However, ex vivo autoradiographic and in vivo blocking studies revealed no target specific accumulation and demonstrated [18F]IV to be inapplicable for imaging PDE10A with PET.
Collapse
|
48
|
Recent Advances in the Development of PET and SPECT Tracers for Brain Imaging. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2012. [DOI: 10.1016/b978-0-12-396492-2.00008-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register]
|
49
|
Andrés JI, De Angelis M, Alcázar J, Iturrino L, Langlois X, Dedeurwaerdere S, Lenaerts I, Vanhoof G, Celen S, Bormans G. Synthesis, In Vivo Occupancy, and Radiolabeling of Potent Phosphodiesterase Subtype-10 Inhibitors as Candidates for Positron Emission Tomography Imaging. J Med Chem 2011; 54:5820-35. [DOI: 10.1021/jm200536d] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- José-Ignacio Andrés
- Medicinal Chemistry, Janssen Research & Development, Janssen-Cilag SA, C/Jarama 75, 45007 Toledo, Spain
| | - Meri De Angelis
- Medicinal Chemistry, Janssen Research & Development, Janssen-Cilag SA, C/Jarama 75, 45007 Toledo, Spain
| | - Jesús Alcázar
- Medicinal Chemistry, Janssen Research & Development, Janssen-Cilag SA, C/Jarama 75, 45007 Toledo, Spain
| | - Laura Iturrino
- Medicinal Chemistry, Janssen Research & Development, Janssen-Cilag SA, C/Jarama 75, 45007 Toledo, Spain
| | - Xavier Langlois
- Neuroscience, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Stefanie Dedeurwaerdere
- Neuroscience, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Ilse Lenaerts
- Neuroscience, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Greet Vanhoof
- Neuroscience, Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Sofie Celen
- Laboratory for Radiopharmacy and IMIR, Faculty of Pharmaceutical Sciences, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium
| | - Guy Bormans
- Laboratory for Radiopharmacy and IMIR, Faculty of Pharmaceutical Sciences, KU Leuven, Herestraat 49, B-3000 Leuven, Belgium
| |
Collapse
|