1
|
Zhou YP, Zhang LL, Sun Y, Brugarolas P. Imaging of Pain using Positron Emission Tomography. IRADIOLOGY 2024; 2:339-361. [PMID: 39440326 PMCID: PMC11493400 DOI: 10.1002/ird3.73] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 03/06/2024] [Indexed: 10/25/2024]
Abstract
Positron emission tomography (PET) is a noninvasive molecular imaging technique that utilizes biologically active radiolabeled compounds to image biochemical processes. As such, PET can provide important pathophysiological information associated with pain of different etiologies. As such, the information obtained using PET often combined with MRI or CT can provide useful information for diagnosing and monitoring changes associated with pain. This review covers the most important PET tracers that have been used to image pain including tracers for fundamental biological processes such as glucose metabolism and cerebral blood flow to receptor-specific tracers such as ion channels and neurotransmitters. For tracer type, we describe the structure and radiochemical synthesis of the tracer followed by a brief summary of the available preclinical and clinical studies. By providing a summary of the PET tracers that have been employed for PET imaging of pain, this review aims to serve as a reference for preclinical, translational and clinical investigators interested in molecular imaging of pain. Finally, the review ends with an outlook of the needs and opportunities in this area.
Collapse
Affiliation(s)
- Yu-Peng Zhou
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Lauren L Zhang
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Yang Sun
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Pedro Brugarolas
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
2
|
van der Heijden RA, Biswal S. Up-and-coming Radiotracers for Imaging Pain Generators. Semin Musculoskelet Radiol 2023; 27:661-675. [PMID: 37935213 PMCID: PMC10629993 DOI: 10.1055/s-0043-1775745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2023]
Abstract
Chronic musculoskeletal pain is among the most highly prevalent diseases worldwide. Managing patients with chronic pain remains very challenging because current imaging techniques focus on morphological causes of pain that can be inaccurate and misleading. Moving away from anatomical constructs of disease, molecular imaging has emerged as a method to identify diseases according to their molecular, physiologic, or cellular signatures that can be applied to the variety of biomolecular changes that occur in nociception and pain processing and therefore have tremendous potential for precisely pinpointing the source of a patient's pain. Several molecular imaging approaches to image the painful process are now available, including imaging of voltage-gated sodium channels, calcium channels, hypermetabolic processes, the substance P receptor, the sigma-1 receptor, and imaging of macrophage trafficking. This article provides an overview of promising molecular imaging approaches for the imaging of musculoskeletal pain with a focus on preclinical methods.
Collapse
Affiliation(s)
- Rianne A. van der Heijden
- Department of Radiology and Nuclear Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Sandip Biswal
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
3
|
Cai L, Liow JS, Morse CL, Telu S, Davies R, Manly LS, Zoghbi SS, Chin FT, Innis RB, Pike VW. Candidate 3-benzazepine-1-ol type GluN2B receptor radioligands ( 11C-NR2B-Me enantiomers) have high binding in cerebellum but not to σ1 receptors. EJNMMI Res 2023; 13:28. [PMID: 37017827 PMCID: PMC10076467 DOI: 10.1186/s13550-023-00975-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 03/12/2023] [Indexed: 04/06/2023] Open
Abstract
INTRODUCTION We recently reported 11C-NR2B-SMe ([S-methyl-11C](R,S)-7-thiomethoxy-3-(4-(4-methyl-phenyl)butyl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-ol) and its enantiomers as candidate radioligands for imaging the GluN2B subunit within rat N-methyl-D-aspartate receptors. However, these radioligands gave unexpectedly high and displaceable binding in rat cerebellum, possibly due to cross-reactivity with sigma-1 (σ1) receptors. This study investigated 11C-labeled enantiomers of a close analogue (7-methoxy-3-(4-(p-tolyl)butyl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-ol; NR2B-Me) of 11C-NR2B-SMe as new candidate GluN2B radioligands. PET was used to evaluate these radioligands in rats and to assess potential cross-reactivity to σ1 receptors. METHODS NR2B-Me was assayed for binding affinity and selectivity to GluN2B in vitro. 11C-NR2B-Me and its enantiomers were prepared by Pd-mediated treatment of boronic ester precursors with 11C-iodomethane. Brain PET scans were conducted after radioligand intravenous injection into rats. Various ligands for GluN2B receptors or σ1 receptors were administered at set doses in pre-blocking or displacement experiments to assess their impact on imaging data. 18F-FTC146 and enantiomers of 11C-NR2B-SMe were used for comparison. Radiometabolites from brain and plasma were measured ex vivo and in vitro. RESULTS NR2B-Me enantiomers showed high GluN2B affinity and selectivity in vitro. 11C-NR2B-Me enantiomers gave high early whole rat brain uptake of radioactivity, including high uptake in cerebellum, followed by slower decline. Radioactivity in brain at 30 min ex vivo was virtually all unchanged radioligand. Only less lipophilic radiometabolites appeared in plasma. When 11C-(R)-NR2B-Me was used, three high-affinity GluN2B ligands-NR2B-SMe, Ro25-6981, and CO101,244-showed increasing pre-block of whole brain radioactivity retention with increasing dose. Two σ1 receptor antagonists, FTC146 and BD1407, were ineffective pre-blocking agents. Together, these results strongly resemble those obtained with 11C-NR2B-SMe enantiomers, except that 11C-NR2B-Me enantiomers showed faster reversibility of binding. When 18F-FTC146 was used as a radioligand, FTC146 and BD1407 showed strong pre-blocking effects whereas GluN2B ligands showed only weak blocking effects. CONCLUSION 11C-NR2B-Me enantiomers showed specific binding to GluN2B receptors in rat brain in vivo. High unexpected specific binding in cerebellum was not due to σ1 receptors. Additional investigation is needed to identify the source of the high specific binding.
Collapse
Affiliation(s)
- Lisheng Cai
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr, Bldg 10, Room B3 C346, Bethesda, MD, 20892, USA.
| | - Jeih-San Liow
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr, Bldg 10, Room B3 C346, Bethesda, MD, 20892, USA
| | - Cheryl L Morse
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr, Bldg 10, Room B3 C346, Bethesda, MD, 20892, USA
| | - Sanjay Telu
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr, Bldg 10, Room B3 C346, Bethesda, MD, 20892, USA
| | - Riley Davies
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr, Bldg 10, Room B3 C346, Bethesda, MD, 20892, USA
| | - Lester S Manly
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr, Bldg 10, Room B3 C346, Bethesda, MD, 20892, USA
| | - Sami S Zoghbi
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr, Bldg 10, Room B3 C346, Bethesda, MD, 20892, USA
| | - Frederick T Chin
- Molecular Imaging Program at Stanford, Department of Radiology, Stanford University, 1201 Welch Road, Rm. PS049, Stanford, CA, 94305-584, USA
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr, Bldg 10, Room B3 C346, Bethesda, MD, 20892, USA
| | - Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, 10 Center Dr, Bldg 10, Room B3 C346, Bethesda, MD, 20892, USA
| |
Collapse
|
4
|
Shidahara M, Funaki Y, Watabe H. Noninvasive estimation of human radiation dosimetry of 18F-FDG by whole-body small animal PET imaging in rats. Appl Radiat Isot 2022; 181:110071. [PMID: 34952332 DOI: 10.1016/j.apradiso.2021.110071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 12/02/2021] [Accepted: 12/15/2021] [Indexed: 11/02/2022]
Abstract
PURPOSE Small animal PET provides the biodistribution of administrated radiotracer in vivo and have a potential to contribute on dosimetry study. The aim of this study is to investigate the effect of region-of-interest (ROI)-delineation in whole-body rat PET image toward non-invasive estimation of human dosimetry of 18F-FDG. METHOD After administration of 18F-FDG (averaged 11.7 MBq), 3.5-h PET and 20-min CT scans were sequentially performed for three rats by Clairvivo PET/CT system. Seven source organs, and the remainder of the body, were studied to extrapolate %ID(t) and estimate time-integrated activity coefficients [kBq-h/MBq] in human. The mean absorbed dose in each target organ and the effective dose were estimated by MIRD method. Effects of ROI-definitions on both extrapolated %ID(t) in human and estimated doses were also investigated by using (i) small ROIs of high uptake region and (ii) whole organ ROIs. RESULTS Averaged effective doses of 18F-FDG in human by using high-uptake and whole-organ ROIs were 27.8 ± 6.54 and 19.3 ± 2.72 μSv/MBq, respectively. CONCLUSION The use of small animal PET scanner, which allows repeatedly PET scans, have a potential to contribute on the reduction of the number of experimental animals. However, the ways of ROI drawing influences on the estimated effective dose and safe-side ROI definition may be preferred.
Collapse
Affiliation(s)
- Miho Shidahara
- Division of Applied Quantum Medical Engineering, Department of Quantum Science and Energy Engineering, Graduate School of Engineering, Tohoku University, Japan.
| | - Yoshihito Funaki
- Division of Radiopharmaceutical Chemistry, Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Hiroshi Watabe
- Division of Radiation Protection and Safety Control, Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| |
Collapse
|
5
|
Wilson LL, Eans SO, Ramadan-Siraj I, Modica MN, Romeo G, Intagliata S, McLaughlin JP. Examination of the Novel Sigma-1 Receptor Antagonist, SI 1/28, for Antinociceptive and Anti-allodynic Efficacy against Multiple Types of Nociception with Fewer Liabilities of Use. Int J Mol Sci 2022; 23:615. [PMID: 35054797 PMCID: PMC8775934 DOI: 10.3390/ijms23020615] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 12/29/2021] [Indexed: 02/05/2023] Open
Abstract
Neuropathic pain is a significant problem with few effective treatments lacking adverse effects. The sigma-1 receptor (S1R) is a potential therapeutic target for neuropathic pain, as antagonists for this receptor effectively ameliorate pain in both preclinical and clinical studies. The current research examines the antinociceptive and anti-allodynic efficacy of SI 1/28, a recently reported benzylpiperazine derivative and analog of the S1R antagonist SI 1/13, that was 423-fold more selective for S1R over the sigma-2 receptor (S2R). In addition, possible liabilities of respiration, sedation, and drug reinforcement caused by SI 1/28 have been evaluated. Inflammatory and chemical nociception, chronic nerve constriction injury (CCI) induced mechanical allodynia, and adverse effects of sedation in a rotarod assay, conditioned place preference (CPP), and changes in breath rate and locomotor activity were assessed after i.p. administration of SI 1/28. Pretreatment with SI 1/28 produced dose-dependent antinociception in the formalin test, with an ED50 (and 95% C.I.) value of 13.2 (7.42-28.3) mg/kg, i.p. Likewise, SI 1/28 produced dose-dependent antinociception against visceral nociception and anti-allodynia against CCI-induced neuropathic pain. SI 1/28 demonstrated no impairment of locomotor activity, conditioned place preference, or respiratory depression. In summary, SI 1/28 proved efficacious in the treatment of acute inflammatory pain and chronic neuropathy without liabilities at therapeutic doses, supporting the development of S1R antagonists as therapeutics for chronic pain.
Collapse
Affiliation(s)
- Lisa L. Wilson
- Department of Pharmacodynamics, The University of Florida, Gainesville, FL 32610, USA; (L.L.W.); (S.O.E.); (I.R.-S.)
| | - Shainnel O. Eans
- Department of Pharmacodynamics, The University of Florida, Gainesville, FL 32610, USA; (L.L.W.); (S.O.E.); (I.R.-S.)
| | - Insitar Ramadan-Siraj
- Department of Pharmacodynamics, The University of Florida, Gainesville, FL 32610, USA; (L.L.W.); (S.O.E.); (I.R.-S.)
| | - Maria N. Modica
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy; (M.N.M.); (G.R.)
| | - Giuseppe Romeo
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy; (M.N.M.); (G.R.)
| | - Sebastiano Intagliata
- Department of Drug and Health Sciences, University of Catania, 95125 Catania, Italy; (M.N.M.); (G.R.)
| | - Jay P. McLaughlin
- Department of Pharmacodynamics, The University of Florida, Gainesville, FL 32610, USA; (L.L.W.); (S.O.E.); (I.R.-S.)
| |
Collapse
|
6
|
Reyes ST, Deacon RMJ, Guo SG, Altimiras FJ, Castillo JB, van der Wildt B, Morales AP, Park JH, Klamer D, Rosenberg J, Oberman LM, Rebowe N, Sprouse J, Missling CU, McCurdy CR, Cogram P, Kaufmann WE, Chin FT. Effects of the sigma-1 receptor agonist blarcamesine in a murine model of fragile X syndrome: neurobehavioral phenotypes and receptor occupancy. Sci Rep 2021; 11:17150. [PMID: 34433831 PMCID: PMC8387417 DOI: 10.1038/s41598-021-94079-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 06/21/2021] [Indexed: 11/08/2022] Open
Abstract
Fragile X syndrome (FXS), a disorder of synaptic development and function, is the most prevalent genetic form of intellectual disability and autism spectrum disorder. FXS mouse models display clinically-relevant phenotypes, such as increased anxiety and hyperactivity. Despite their availability, so far advances in drug development have not yielded new treatments. Therefore, testing novel drugs that can ameliorate FXS' cognitive and behavioral impairments is imperative. ANAVEX2-73 (blarcamesine) is a sigma-1 receptor (S1R) agonist with a strong safety record and preliminary efficacy evidence in patients with Alzheimer's disease and Rett syndrome, other synaptic neurodegenerative and neurodevelopmental disorders. S1R's role in calcium homeostasis and mitochondrial function, cellular functions related to synaptic function, makes blarcamesine a potential drug candidate for FXS. Administration of blarcamesine in 2-month-old FXS and wild type mice for 2 weeks led to normalization in two key neurobehavioral phenotypes: open field test (hyperactivity) and contextual fear conditioning (associative learning). Furthermore, there was improvement in marble-burying (anxiety, perseverative behavior). It also restored levels of BDNF, a converging point of many synaptic regulators, in the hippocampus. Positron emission tomography (PET) and ex vivo autoradiographic studies, using the highly selective S1R PET ligand [18F]FTC-146, demonstrated the drug's dose-dependent receptor occupancy. Subsequent analyses also showed a wide but variable brain regional distribution of S1Rs, which was preserved in FXS mice. Altogether, these neurobehavioral, biochemical, and imaging data demonstrates doses that yield measurable receptor occupancy are effective for improving the synaptic and behavioral phenotype in FXS mice. The present findings support the viability of S1R as a therapeutic target in FXS, and the clinical potential of blarcamesine in FXS and other neurodevelopmental disorders.
Collapse
Affiliation(s)
- Samantha T Reyes
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | - Robert M J Deacon
- FRAXA-DVI, FRAXA, Santiago, Chile
- IEB, Faculty of Science, University of Chile, Santiago, Chile
- Fraunhofer Chile Research, Center for Systems Biotechnology, Santiago, Chile
| | - Scarlett G Guo
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | - Francisco J Altimiras
- FRAXA-DVI, FRAXA, Santiago, Chile
- Faculty of Engineering and Business, Universidad de las Américas, Santiago, Chile
| | - Jessa B Castillo
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | | | - Aimara P Morales
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | - Jun Hyung Park
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | - Daniel Klamer
- Anavex Life Sciences Corp., New York, NY, 10019, USA
| | - Jarrett Rosenberg
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | - Lindsay M Oberman
- Center for Neuroscience & Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Nell Rebowe
- Anavex Life Sciences Corp., New York, NY, 10019, USA
| | | | | | - Christopher R McCurdy
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Patricia Cogram
- FRAXA-DVI, FRAXA, Santiago, Chile
- IEB, Faculty of Science, University of Chile, Santiago, Chile
- Fraunhofer Chile Research, Center for Systems Biotechnology, Santiago, Chile
| | - Walter E Kaufmann
- Anavex Life Sciences Corp., New York, NY, 10019, USA.
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA, 30322, USA.
| | - Frederick T Chin
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
7
|
Shigeno T, Kozaka T, Kitamura Y, Ogawa K, Taki J, Kinuya S, Shiba K. In vitro and in vivo evaluation of [ 125/123I]-2-[4-(2-iodophenyl)piperidino]cyclopentanol([ 125/123I]-OI5V) as a potential sigma-1 receptor ligand for SPECT. Ann Nucl Med 2021; 35:167-175. [PMID: 33417152 DOI: 10.1007/s12149-020-01552-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/28/2020] [Indexed: 11/26/2022]
Abstract
INTRODUCTION We investigated the characteristics of radio-iodinated 2-[4-(2-iodophenyl)piperidino]cyclopentanol (OI5V) as a single photon emission computed tomography (SPECT) ligand for mapping sigma-1 receptor (σ-1R), which plays an important role in stress remission in many organs. METHODS OI5V was synthesized from o-bromobenzaldehyde in three steps. OI5V was evaluated for its affinity to VAChT, σ-1 and σ-2 receptor by in vitro competitive binding assays using rat tissues and radioligands, [3H]vesamicol, ( +)-[3H]pentazocine and [3H]DTG, respectively. [125/123I]OI5V was prepared from o-trimethylstannyl-cyclopentanevesamicol (OT5V) by the iododestannylation reaction under no-carrier-added conditions. In vivo biodistribution study of [125I]OI5V in blood, brain regions and major organs of rats was performed at 2, 10, 30 and 60 min post-injection. In vivo blocking study and ex vivo autoradiography were performed to assess the binding selectivity of [125I]OI5V for σ-1 receptor. SPECT-CT imaging study was performed using [123I]OI5V. RESULTS OI5V demonstrated high selective binding affinity for σ-1R in vitro. In the biodistribution study, the blood-brain barrier (BBB) permeability of [125I]OI5V was high and the accumulation of [125I]OI5V in the rat cortex at 2 min post-injection exceeded 2.00%ID/g. In the in vivo blocking study, the accumulation of [125I]OI5V in the brain was significantly blocked by co-administration of 0.5 μmol of SA4503 and 1.0 μmol of pentazocine. Ex vivo autoradiography revealed that the regional brain accumulation of [125I]OI5V was similar to σ-1R-rich regions of the rat brain. SPECT images of [123I]OI5V in the rat brain reflected the distribution of sigma receptors in the brain. CONCLUSIONS This study confirmed that [125/123I]OI5V selectively binds σ-1R in the rat brain in vivo. [123I]OI5V was suggested to be useful as a σ-1R ligand for SPECT.
Collapse
Affiliation(s)
- Taiki Shigeno
- Division of Tracer Kinetics, Advanced Science Research Center, Kanazawa, Ishikawa, Japan
| | - Takashi Kozaka
- Division of Tracer Kinetics, Advanced Science Research Center, Kanazawa, Ishikawa, Japan
- Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa , Ishikawa, Japan
| | - Yoji Kitamura
- Division of Tracer Kinetics, Advanced Science Research Center, Kanazawa, Ishikawa, Japan
- Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa , Ishikawa, Japan
| | - Kazuma Ogawa
- Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa , Ishikawa, Japan
| | - Junichi Taki
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Seigo Kinuya
- Division of Tracer Kinetics, Advanced Science Research Center, Kanazawa, Ishikawa, Japan
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Kazuhiro Shiba
- Division of Tracer Kinetics, Advanced Science Research Center, Kanazawa, Ishikawa, Japan.
- Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa , Ishikawa, Japan.
| |
Collapse
|
8
|
Toussaint M, Deuther-Conrad W, Kranz M, Fischer S, Ludwig FA, Juratli TA, Patt M, Wünsch B, Schackert G, Sabri O, Brust P. Sigma-1 Receptor Positron Emission Tomography: A New Molecular Imaging Approach Using ( S)-(-)-[ 18F]Fluspidine in Glioblastoma. Molecules 2020; 25:E2170. [PMID: 32384802 PMCID: PMC7248975 DOI: 10.3390/molecules25092170] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 04/30/2020] [Accepted: 05/02/2020] [Indexed: 11/16/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most devastating primary brain tumour characterised by infiltrative growth and resistance to therapies. According to recent research, the sigma-1 receptor (sig1R), an endoplasmic reticulum chaperone protein, is involved in signaling pathways assumed to control the proliferation of cancer cells and thus could serve as candidate for molecular characterisation of GBM. To test this hypothesis, we used the clinically applied sig1R-ligand (S)-(-)-[18F]fluspidine in imaging studies in an orthotopic mouse model of GBM (U87-MG) as well as in human GBM tissue. A tumour-specific overexpression of sig1R in the U87-MG model was revealed in vitro by autoradiography. The binding parameters demonstrated target-selective binding according to identical KD values in the tumour area and the contralateral side, but a higher density of sig1R in the tumour. Different kinetic profiles were observed in both areas, with a slower washout in the tumour tissue compared to the contralateral side. The translational relevance of sig1R imaging in oncology is reflected by the autoradiographic detection of tumour-specific expression of sig1R in samples obtained from patients with glioblastoma. Thus, the herein presented data support further research on sig1R in neuro-oncology.
Collapse
Affiliation(s)
- Magali Toussaint
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, Research site Leipzig, 04318 Leipzig, Germany; (W.D.-C.); (M.K.); (S.F.); (F.-A.L.); (P.B.)
| | - Winnie Deuther-Conrad
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, Research site Leipzig, 04318 Leipzig, Germany; (W.D.-C.); (M.K.); (S.F.); (F.-A.L.); (P.B.)
| | - Mathias Kranz
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, Research site Leipzig, 04318 Leipzig, Germany; (W.D.-C.); (M.K.); (S.F.); (F.-A.L.); (P.B.)
- PET Imaging Center, University Hospital of North Norway (UNN), 9009 Tromsø, Norway
- Nuclear Medicine and Radiation Biology Research Group, The Arctic University of Norway, 9009 Tromsø, Norway
| | - Steffen Fischer
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, Research site Leipzig, 04318 Leipzig, Germany; (W.D.-C.); (M.K.); (S.F.); (F.-A.L.); (P.B.)
| | - Friedrich-Alexander Ludwig
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, Research site Leipzig, 04318 Leipzig, Germany; (W.D.-C.); (M.K.); (S.F.); (F.-A.L.); (P.B.)
| | - Tareq A. Juratli
- Department of Neurosurgery, Technische Universität Dresden (TUD), University Hospital Carl Gustav Carus, 01307 Dresden, Germany; (T.A.J.); (G.S.)
| | - Marianne Patt
- Department of Nuclear Medicine, University Hospital Leipzig, 04318 Leipzig, Germany; (M.P.); (O.S.)
| | - Bernhard Wünsch
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, 48149 Münster, Germany;
| | - Gabriele Schackert
- Department of Neurosurgery, Technische Universität Dresden (TUD), University Hospital Carl Gustav Carus, 01307 Dresden, Germany; (T.A.J.); (G.S.)
| | - Osama Sabri
- Department of Nuclear Medicine, University Hospital Leipzig, 04318 Leipzig, Germany; (M.P.); (O.S.)
| | - Peter Brust
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, Research site Leipzig, 04318 Leipzig, Germany; (W.D.-C.); (M.K.); (S.F.); (F.-A.L.); (P.B.)
| |
Collapse
|
9
|
Cai L, Liow JS, Morse CL, Telu S, Davies R, Frankland MP, Zoghbi SS, Cheng K, Hall MD, Innis RB, Pike VW. Evaluation of 11C-NR2B-SMe and Its Enantiomers as PET Radioligands for Imaging the NR2B Subunit Within the NMDA Receptor Complex in Rats. J Nucl Med 2020; 61:1212-1220. [PMID: 31924728 DOI: 10.2967/jnumed.119.235143] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 12/19/2019] [Indexed: 01/01/2023] Open
Abstract
[S-methyl-11C](±)-7-methoxy-3-(4-(4-(methylthio)phenyl)butyl)-2,3,4,5-tetrahydro-1H-benzo[d]azepin-1-ol (11C-NR2B-SMe) and its enantiomers were synthesized as candidates for imaging the NR2B subunit within the N-methyl-d-aspartate receptor with PET. Methods: Brains were scanned with PET for 90 min after intravenous injection of one of the candidate radioligands into rats. To detect any NR2B-specific binding of radioligand in brain, various preblocking or displacing agents were evaluated for their impact on the PET brain imaging data. Radiometabolites from brain and other tissues were measured ex vivo and in vitro. Results: Each radioligand gave high early whole-brain uptake of radioactivity, followed by a brief fast decline and then a slow final decline. 11C-(S)-NR2B-SMe was studied extensively. Ex vivo measurements showed that radioactivity in rat brain at 30 min after radioligand injection was virtually unchanged radioligand. Only less lipophilic radiometabolites appeared in plasma. High-affinity NR2B ligands, Ro-25-6981, ifenprodil, and CO101244, showed increasing preblocking of whole-brain radioactivity retention with increasing dose (0.01-3.00 mg/kg, intravenously). Five σ1 antagonists (FTC146, BD1407, F3, F4, and NE100) and 4 σ1 agonists ((+)-pentazocine, (±)-PPCC, PRE-084, and (+)-SKF10047) were ineffective preblocking agents, except FTC146 and F4 at a high dose. Two potent σ1 receptor agonists, TC1 and SA4503, showed dose-dependent preblocking effects in the presence or absence of pharmacologic σ1 receptor blockade with FTC146. Conclusion: 11C-(S)-NR2B-SMe has adequate NR2B-specific PET signal in rat brain to warrant further evaluation in higher species. TC1 and SA4503 likely have off-target binding to NR2B in vivo.
Collapse
Affiliation(s)
- Lisheng Cai
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland; and
| | - Jeih-San Liow
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland; and
| | - Cheryl L Morse
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland; and
| | - Sanjay Telu
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland; and
| | - Riley Davies
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland; and
| | - Michael P Frankland
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland; and
| | - Sami S Zoghbi
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland; and
| | - Ken Cheng
- NCATS Chemical Genomics Center, National Institutes of Health, Rockville, Maryland
| | - Matthew D Hall
- NCATS Chemical Genomics Center, National Institutes of Health, Rockville, Maryland
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland; and
| | - Victor W Pike
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland; and
| |
Collapse
|
10
|
Lan Y, Bai P, Chen Z, Neelamegam R, Placzek MS, Wang H, Fiedler SA, Yang J, Yuan G, Qu X, Schmidt HR, Song J, Normandin MD, Ran C, Wang C. Novel radioligands for imaging sigma-1 receptor in brain using positron emission tomography (PET). Acta Pharm Sin B 2019; 9:1204-1215. [PMID: 31867166 PMCID: PMC6900558 DOI: 10.1016/j.apsb.2019.07.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 06/28/2019] [Accepted: 07/04/2019] [Indexed: 12/18/2022] Open
Abstract
The sigma-1 receptor (σ 1R) is a unique intracellular protein. σ 1R plays a major role in various pathological conditions in the central nervous system (CNS), implicated in several neuropsychiatric disorders. Imaging of σ 1R in the brain using positron emission tomography (PET) could serve as a noninvasively tool for enhancing the understanding of the disease's pathophysiology. Moreover, σ 1R PET tracers can be used for target validation and quantification in diagnosis. Herein, we describe the radiosynthesis, in vivo PET/CT imaging of novel σ 1R 11C-labeled radioligands based on 6-hydroxypyridazinone, [11C]HCC0923 and [11C]HCC0929. Two radioligands have high affinities to σ 1R, with good selectivity. In mice PET/CT imaging, both radioligands showed appropriate kinetics and distributions. Additionally, the specific interactions of two radioligands were reduced by compounds 13 and 15 (self-blocking). Of the two, [11C]HCC0929 was further investigated in positive ligands blocking studies, using classic σ 1R agonist SA 4503 and σ 1R antagonist PD 144418. Both σ 1R ligands could extensively decreased the uptake of [11C]HCC0929 in mice brain. Besides, the biodistribution of major brain regions and organs of mice were determined in vivo. These studies demonstrated that two radioligands, especially [11C]HCC0929, possessed ideal imaging properties and might be valuable tools for non-invasive quantification of σ 1R in brain.
Collapse
Key Words
- 11C-labeled radioligand
- 3D, three-dimensional
- 6-Hydroxypyridazinone
- AF, ammonium formate
- BBB, brain blood barrier
- BP, binding potential
- Brain imaging
- CNS, center nervous systems
- CRPS, complex regional pain syndrome
- DMF, dimethyl formamide
- DMSO, dimethylsulfoxide
- ER, endoplasmic reticulum
- LCP, lipidic cubic phase
- MAM, mitochondria-associated ER membrane
- PCP, phencyclidine
- PET
- PET, positron emission tomography
- TFA, trifluoroacetic acid
- σ1R
- σ1R, sigma-1 receptor
- σ2R, sigma-2 receptor
Collapse
Affiliation(s)
- Yu Lan
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ping Bai
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Zude Chen
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Ramesh Neelamegam
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Michael S. Placzek
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Hao Wang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Stephanie A. Fiedler
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Jing Yang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Gengyang Yuan
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Xiying Qu
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Hayden R. Schmidt
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02129, USA
| | - Jinchun Song
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Marc D. Normandin
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Chongzhao Ran
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Changning Wang
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| |
Collapse
|
11
|
Cirino TJ, Eans SO, Medina JM, Wilson LL, Mottinelli M, Intagliata S, McCurdy CR, McLaughlin JP. Characterization of Sigma 1 Receptor Antagonist CM-304 and Its Analog, AZ-66: Novel Therapeutics Against Allodynia and Induced Pain. Front Pharmacol 2019; 10:678. [PMID: 31258480 PMCID: PMC6586922 DOI: 10.3389/fphar.2019.00678] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/24/2019] [Indexed: 12/16/2022] Open
Abstract
Sigma-1 receptors (S1R) and sigma-2 receptors (S2R) may modulate nociception without the liabilities of opioids, offering a promising therapeutic target to treat pain. The purpose of this study was to investigate the in vivo analgesic and anti-allodynic activity of two novel sigma receptor antagonists, the S1R-selective CM-304 and its analog the non-selective S1R/S2R antagonist AZ-66. Inhibition of thermal, induced chemical or inflammatory pain, as well as the allodynia resulting from chronic nerve constriction injury (CCI) and cisplatin exposure as models of neuropathic pain were assessed in male mice. Both sigma receptor antagonists dose-dependently (10–45 mg/kg, i.p.) reduced allodynia in the CCI and cisplatin neuropathic pain models, equivalent at the higher dose to the effect of the control analgesic gabapentin (50 mg/kg, i.p.), although AZ-66 demonstrated a much longer duration of action. Both CM-304 and AZ-66 produced antinociception in the writhing test [0.48 (0.09–1.82) and 2.31 (1.02–4.81) mg/kg, i.p., respectively] equivalent to morphine [1.75 (0.31–7.55) mg/kg, i.p.]. Likewise, pretreatment (i.p.) with either sigma-receptor antagonist dose-dependently produced antinociception in the formalin paw assay of inflammatory pain. However, CM-304 [17.5 (12.7–25.2) mg/kg, i.p.) and AZ-66 [11.6 (8.29–15.6) mg/kg, i.p.) were less efficacious than morphine [3.87 (2.85–5.18) mg/kg, i.p.] in the 55°C warm-water tail-withdrawal assay. While AZ-66 exhibited modest sedative effects in a rotarod assay and conditioned place aversion, CM-304 did not produce significant effects in the place conditioning assay. Overall, these results demonstrate the S1R selective antagonist CM-304 produces antinociception and anti-allodynia with fewer liabilities than established therapeutics, supporting the use of S1R antagonists as potential treatments for chronic pain.
Collapse
Affiliation(s)
- Thomas J Cirino
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, United States
| | - Shainnel O Eans
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, United States
| | - Jessica M Medina
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, United States
| | - Lisa L Wilson
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, United States
| | - Marco Mottinelli
- Department of Medicinal Chemistry, University of Florida, Gainesville, FL, United States
| | - Sebastiano Intagliata
- Department of Medicinal Chemistry, University of Florida, Gainesville, FL, United States
| | - Christopher R McCurdy
- Department of Medicinal Chemistry, University of Florida, Gainesville, FL, United States
| | - Jay P McLaughlin
- Department of Pharmacodynamics, University of Florida, Gainesville, FL, United States
| |
Collapse
|
12
|
Cipriano PW, Lee SW, Yoon D, Shen B, Tawfik VL, Curtin CM, Dragoo JL, James ML, McCurdy CR, Chin FTN, Biswal S. Successful treatment of chronic knee pain following localization by a sigma-1 receptor radioligand and PET/MRI: a case report. J Pain Res 2018; 11:2353-2357. [PMID: 30349360 PMCID: PMC6190812 DOI: 10.2147/jpr.s167839] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background The ability to accurately diagnose and objectively localize pain generators in chronic pain sufferers remains a major clinical challenge since assessment relies on subjective patient complaints and relatively non-specific diagnostic tools. Developments in clinical molecular imaging, including advances in imaging technology and radiotracer design, have afforded the opportunity to identify tissues involved in pain generation based on their pro-nociceptive condition. The sigma-1 receptor (S1R) is a pro-nociceptive receptor upregulated in painful, inflamed tissues, and it can be imaged using the highly specific radioligand 18F-FTC-146 with PET. Case presentation A 50-year-old woman with a 7-year history of refractory, left-knee pain of unknown origin was referred to our pain management team. Over the past several years, she had undergone multiple treatments, including a lateral retinacular release, radiofrequency ablation of a peripheral nerve, and physical therapy. While certain treatments provided partial relief, her pain would inevitably return to its original state. Using simultaneous positron emission tomography/magnetic resonance imaging (PET/MRI) with the novel radiotracer 18F-FTC-146, imaging showed increased focal uptake of 18F-FTC-146 in the intercondylar notch, corresponding to an irregular but equivocal lesion identified in the simultaneously acquired MRI. These imaging results prompted surgical removal of the lesion, which upon resection was identified as an inflamed, intraarticular synovial lipoma. Removal of the lesion relieved the patient's pain, and to date the pain has not recurred. Conclusion We present a case of chronic, debilitating knee pain that resolved with surgery following identification of the pathology with a novel clinical molecular imaging approach that detects chronic pain generators at the molecular and cellular level. This approach has the potential to identify and localize pain-associated pathology in a variety of chronic pain syndromes.
Collapse
Affiliation(s)
| | - Sheen-Woo Lee
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA, , .,Department of Radiology, Gachon University Gil Hospital, Incheon, South Korea
| | - Daehyun Yoon
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA, ,
| | - Bin Shen
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA, ,
| | - Vivianne Lily Tawfik
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Catherine Mills Curtin
- Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Jason L Dragoo
- Department of Orthopedic Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michelle Louise James
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA, ,
| | - Christopher Robert McCurdy
- Clinical and Translational Science Institute, Translational Drug Development Core, University of Florida, Gainesville, FL, USA
| | | | - Sandip Biswal
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA, ,
| |
Collapse
|
13
|
Bhyrapuneni G, Thentu JB, Mohammed AR, Aleti RR, Padala NP, Ajjala DR, Nirogi R. Assessment of sigma-1 receptor occupancy in mice with non-radiolabelled FTC-146 as a tracer. J Recept Signal Transduct Res 2018; 38:290-298. [PMID: 29912606 DOI: 10.1080/10799893.2018.1478855] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/09/2018] [Accepted: 05/10/2018] [Indexed: 12/21/2022]
Abstract
The use of liquid chromatography coupled with mass spectrometry (LC-MS/MS) is advantageous in in-vivo receptor occupancy assays at pre-clinical drug developmental stages. Relatively, its application is effective in terms of high throughput, data reproducibility, sensitivity, and sample processing. In this perspective, we have evaluated the use of FTC-146 as a non-radiolabelled tracer to determine the sigma-1 receptor occupancy of test drugs in mice brain. Further, the brain and plasma exposures of test drug were determined at their corresponding occupancies. In this occupancy method, the optimized tracer treatment (sacrification) time after intravenous administration was 30 min. The tracer dose was 3 µg/kg and specific brain regions of interest were frontal cortex, pons and midbrain. Mice were pretreated orally with SA4503, fluspidine, haloperidol, and donepezil followed by tracer treatment. Among the test drugs, SA4503 was used as positive control group at its highest test dose (7 mg/kg, intraperitoneal). There was a dose-dependent decrease in brain regional FTC-146 binding in pretreated mice. From the occupancy curves of SA4503, fluspidine, haloperidol, and donepezil the effective dose (ED50) value ranges are 0.74-1.45, 0.09-0.11, 0.11-0.12, and 0.07-0.09 mg/kg, respectively. Their corresponding brain effective concentration (EC50) values are 74.3-132.5, 3.4-3.7, 122.5-139.5, and 8.8-11.0 ng/g and plasma EC50 values are 34.3-53.7, 0.08-0.10, 7.8-9.5, and 0.6-0.7 ng/mL. Brain regional distribution and binding inhibition upon pretreatment were comparable with data reported with labeled [18F]FTC-146. Drug exposures were simultaneously determined and correlated with sigma-1 occupancy from the same experiment. Wide category drugs can be assayed for sigma-1 receptor engagement and their correlation with exposures aid in clinical development.
Collapse
|
14
|
Shen B, Park JH, Hjørnevik T, Cipriano PW, Yoon D, Gulaka PK, Holly D, Behera D, Avery BA, Gambhir SS, McCurdy CR, Biswal S, Chin FT. Radiosynthesis and First-In-Human PET/MRI Evaluation with Clinical-Grade [ 18F]FTC-146. Mol Imaging Biol 2018; 19:779-786. [PMID: 28280965 DOI: 10.1007/s11307-017-1064-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE Sigma-1 receptors (S1Rs) play an important role in many neurological disorders. Simultaneous positron emission tomography (PET)/magnetic resonance imaging (MRI) with S1R radioligands may provide valuable information for diagnosing and guiding treatment for these diseases. Our previously reported S1R radioligand, [18F]FTC-146, demonstrated high affinity for the S1R (K i = 0.0025 nM) and excellent selectivity for the S1R over the sigma-2 receptor (S2Rs; K i = 364 nM) across several species (from mouse to non-human primate). Herein, we report the clinical-grade radiochemistry filed with exploratory Investigational New Drug (eIND) and first-in-human PET/MRI evaluation of [18F]FTC-146. PROCEDURES [18F]FTC-146 is prepared via a direct [18F] fluoride nucleophilic radiolabeling reaction and formulated in 0.9 % NaCl containing no more than 10 % ethanol through sterile filtration. Quality control (QC) was performed based on USP 823 before doses were released for clinical use. The safety and whole body biodistribution of [18F]FTC-146 were evaluated using a simultaneous PET/MR scanner in two representative healthy human subjects. RESULTS [18F]FTC-146 was synthesized with a radiochemical yield of 3.3 ± 0.7 % and specific radioactivity of 8.3 ± 3.3 Ci/μmol (n = 10, decay corrected to EOB). Both radiochemical and chemical purities were >95 %; the prepared doses were stable for 4 h at ambient temperature. All QC test results met specified clinical criteria. The in vivo PET/MRI investigations showed that [18F]FTC-146 rapidly crossed the blood brain barrier and accumulated in S1R-rich regions of the brain. There were also radioactivity distributed in the peripheral organs, i.e., the lungs, spleen, pancreas, and thyroid. Furthermore, insignificant uptake of [18F]FTC-146 was observed in cortical bone and muscle. CONCLUSION A reliable and automated radiosynthesis for providing routine clinical-grade [18F]FTC-146 for human studies was established in a modified GE TRACERlab FXFN. PET/MRI demonstrated the initial tracer biodistribution in humans, and clinical studies investigating different S1R-related diseases are in progress.
Collapse
Affiliation(s)
- Bin Shen
- Molecular Imaging Program at Stanford (MIPS), Departments of Radiology and Bioengineering, Bio-X Program, Stanford University School of Medicine, 1201 Welch Road, PS049, Stanford, CA, 94305-5484, USA
| | - Jun Hyung Park
- Molecular Imaging Program at Stanford (MIPS), Departments of Radiology and Bioengineering, Bio-X Program, Stanford University School of Medicine, 1201 Welch Road, PS049, Stanford, CA, 94305-5484, USA
| | - Trine Hjørnevik
- Molecular Imaging Program at Stanford (MIPS), Departments of Radiology and Bioengineering, Bio-X Program, Stanford University School of Medicine, 1201 Welch Road, PS049, Stanford, CA, 94305-5484, USA.,Department of Diagnostic Physics, Oslo University Hospital, Oslo, Norway.,The Norwegian Medical Cyclotron Centre, Oslo, Norway
| | - Peter W Cipriano
- Molecular Imaging Program at Stanford (MIPS), Departments of Radiology and Bioengineering, Bio-X Program, Stanford University School of Medicine, 1201 Welch Road, PS049, Stanford, CA, 94305-5484, USA
| | - Daehyun Yoon
- Radiological Sciences Laboratory, Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | - Praveen K Gulaka
- Molecular Imaging Program at Stanford (MIPS), Departments of Radiology and Bioengineering, Bio-X Program, Stanford University School of Medicine, 1201 Welch Road, PS049, Stanford, CA, 94305-5484, USA
| | - Dawn Holly
- Molecular Imaging Program at Stanford (MIPS), Departments of Radiology and Bioengineering, Bio-X Program, Stanford University School of Medicine, 1201 Welch Road, PS049, Stanford, CA, 94305-5484, USA
| | - Deepak Behera
- Molecular Imaging Program at Stanford (MIPS), Departments of Radiology and Bioengineering, Bio-X Program, Stanford University School of Medicine, 1201 Welch Road, PS049, Stanford, CA, 94305-5484, USA
| | - Bonnie A Avery
- Department of Pharmaceutics, P1-27, University of Florida, Gainesville, FL, 32610, USA
| | - Sanjiv S Gambhir
- Molecular Imaging Program at Stanford (MIPS), Departments of Radiology and Bioengineering, Bio-X Program, Stanford University School of Medicine, 1201 Welch Road, PS049, Stanford, CA, 94305-5484, USA
| | - Christopher R McCurdy
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Sandip Biswal
- Department of Radiology and Molecular Imaging Program at Stanford (MIPS), Stanford University School of Medicine, 300 Pasteur Drive S-068B, Stanford, CA, 94305, USA.
| | - Frederick T Chin
- Molecular Imaging Program at Stanford (MIPS), Departments of Radiology and Bioengineering, Bio-X Program, Stanford University School of Medicine, 1201 Welch Road, PS049, Stanford, CA, 94305-5484, USA.
| |
Collapse
|
15
|
Hjørnevik T, Cipriano PW, Shen B, Park JH, Gulaka P, Holley D, Gandhi H, Yoon D, Mittra ES, Zaharchuk G, Gambhir SS, McCurdy CR, Chin FT, Biswal S. Biodistribution and Radiation Dosimetry of 18F-FTC-146 in Humans. J Nucl Med 2017; 58:2004-2009. [PMID: 28572487 PMCID: PMC6944163 DOI: 10.2967/jnumed.117.192641] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 05/16/2017] [Indexed: 01/27/2023] Open
Abstract
The purpose of this study was to assess safety, biodistribution, and radiation dosimetry in humans for the highly selective σ-1 receptor PET agent 18F-6-(3-fluoropropyl)-3-(2-(azepan-1-yl)ethyl)benzo[d]thiazol-2(3H)-one (18F-FTC-146). Methods: Ten healthy volunteers (5 women, 5 men; age ± SD, 34.3 ± 6.5 y) were recruited, and written informed consent was obtained from all participants. Series of whole-body PET/MRI examinations were acquired for up to 3 h after injection (357.2 ± 48.8 MBq). Blood samples were collected, and standard vital signs (heart rate, pulse oximetry, and body temperature) were monitored at regular intervals. Regions of interest were delineated, time-activity curves were calculated, and organ uptake and dosimetry were estimated. Results: All subjects tolerated the PET/MRI examination well, and no adverse reactions to 18F-FTC-146 were reported. High accumulation of 18F-FTC-146 was observed in σ-1 receptor-dense organs such as the pancreas and spleen, moderate uptake in the brain and myocardium, and low uptake in bone and muscle. High uptake was also observed in the kidneys and bladder, indicating renal tracer clearance. The effective dose of 18F-FTC-146 was 0.0259 ± 0.0034 mSv/MBq (range, 0.0215-0.0301 mSv/MBq). Conclusion: First-in-human studies with clinical-grade 18F-FTC-146 were successful. Injection of 18F-FTC-146 is safe, and absorbed doses are acceptable. The potential of 18F-FTC-146 as an imaging agent for a variety of neuroinflammatory diseases is currently under investigation.
Collapse
Affiliation(s)
- Trine Hjørnevik
- Department of Radiology, Stanford University, Stanford, California
- Department of Diagnostic Physics, Oslo University Hospital, Oslo, Norway
- The Norwegian Medical Cyclotron Centre, Oslo, Norway
| | - Peter W Cipriano
- Department of Radiology, Stanford University, Stanford, California
| | - Bin Shen
- Department of Radiology, Stanford University, Stanford, California
| | - Jun Hyung Park
- Department of Radiology, Stanford University, Stanford, California
| | - Praveen Gulaka
- Department of Radiology, Stanford University, Stanford, California
| | - Dawn Holley
- Department of Radiology, Stanford University, Stanford, California
| | - Harsh Gandhi
- Department of Radiology, Stanford University, Stanford, California
| | - Daehyun Yoon
- Department of Radiology, Stanford University, Stanford, California
| | - Erik S Mittra
- Department of Radiology, Stanford University, Stanford, California
| | - Greg Zaharchuk
- Department of Radiology, Stanford University, Stanford, California
| | - Sanjiv S Gambhir
- Department of Radiology, Stanford University, Stanford, California
| | - Christopher R McCurdy
- Department of Medicinal Chemistry, University of Florida, Gainesville, Florida; and
- UF Translational Drug Development Core, University of Florida, Gainesville, Florida
| | - Frederick T Chin
- Department of Radiology, Stanford University, Stanford, California
| | - Sandip Biswal
- Department of Radiology, Stanford University, Stanford, California
| |
Collapse
|
16
|
Shen B, Behera D, James ML, Reyes ST, Andrews L, Cipriano PW, Klukinov M, Lutz AB, Mavlyutov T, Rosenberg J, Ruoho AE, McCurdy CR, Gambhir SS, Yeomans DC, Biswal S, Chin FT. Visualizing Nerve Injury in a Neuropathic Pain Model with [ 18F]FTC-146 PET/MRI. Theranostics 2017; 7:2794-2805. [PMID: 28824716 PMCID: PMC5562216 DOI: 10.7150/thno.19378] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Accepted: 03/31/2017] [Indexed: 12/18/2022] Open
Abstract
The ability to locate nerve injury and ensuing neuroinflammation would have tremendous clinical value for improving both the diagnosis and subsequent management of patients suffering from pain, weakness, and other neurologic phenomena associated with peripheral nerve injury. Although several non-invasive techniques exist for assessing the clinical manifestations and morphological aspects of nerve injury, they often fail to provide accurate diagnoses due to limited specificity and/or sensitivity. Herein, we describe a new imaging strategy for visualizing a molecular biomarker of nerve injury/neuroinflammation, i.e., the sigma-1 receptor (S1R), in a rat model of nerve injury and neuropathic pain. The two-fold higher increase of S1Rs was shown in the injured compared to the uninjured nerve by Western blotting analyses. With our novel S1R-selective radioligand, [18F]FTC-146 (6-(3-[18F]fluoropropyl)-3-(2-(azepan-1-yl)ethyl)benzo[d]thiazol-2(3H)-one), and positron emission tomography-magnetic resonance imaging (PET/MRI), we could accurately locate the site of nerve injury created in the rat model. We verified the accuracy of this technique by ex vivo autoradiography and immunostaining, which demonstrated a strong correlation between accumulation of [18F]FTC-146 and S1R staining. Finally, pain relief could also be achieved by blocking S1Rs in the neuroma with local administration of non-radioactive [19F]FTC-146. In summary, [18F]FTC-146 S1R PET/MR imaging has the potential to impact how we diagnose, manage and treat patients with nerve injury, and thus warrants further investigation.
Collapse
Affiliation(s)
- Bin Shen
- Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Deepak Behera
- Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michelle L. James
- Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Samantha T. Reyes
- Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Lauren Andrews
- Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Peter W. Cipriano
- Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael Klukinov
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Amanda Brosius Lutz
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Timur Mavlyutov
- Department of Neuroscience, University of Wisconsin, Madison, WI 53726, USA
| | - Jarrett Rosenberg
- Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Arnold E. Ruoho
- Department of Neuroscience, University of Wisconsin, Madison, WI 53726, USA
| | - Christopher R. McCurdy
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Sanjiv S. Gambhir
- Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA
- Departments of Bioengineering and Materials Sciences & Engineering, Stanford University, Stanford, CA 94305, USA
| | - David C. Yeomans
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sandip Biswal
- Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Frederick T. Chin
- Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
17
|
Baum E, Cai Z, Bois F, Holden D, Lin SF, Lara-Jaime T, Kapinos M, Chen Y, Deuther-Conrad W, Fischer S, Dukic-Stefanovic S, Bunse P, Wünsch B, Brust P, Jia H, Huang Y. PET Imaging Evaluation of Four σ 1 Radiotracers in Nonhuman Primates. J Nucl Med 2017; 58:982-988. [PMID: 28232607 DOI: 10.2967/jnumed.116.188052] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 02/09/2017] [Indexed: 01/24/2023] Open
Abstract
The σ1 receptors (S1Rs) are implicated in a variety of diseases including Alzheimer disease and cancer. Previous PET S1R radiotracers are characterized by slow kinetics or off-target binding that impedes their use in humans. Here, we report the first PET imaging evaluation in rhesus monkeys of 4 18F-labeled spirocyclic piperidine-based PET radiotracers (18F-1 to 18F-4). Methods: Baseline scans for the 4 radiotracers were obtained on an adult male rhesus monkey. Blocking scans were obtained with the S1R-selective agonist SA4503 to assess binding specificity of 18F-2 and 18F-4 Arterial input functions were measured, and binding parameters were determined with kinetic modeling analysis. Results: In the rhesus brain, all 4 radiotracers showed high and fast uptake. Tissue activity washout was rapid for 18F-2 and 18F-4, and much slower for 18F-1 and 18F-3, in line with their respective in vitro S1R-binding affinities. Both the 1-tissue-compartment and multilinear analysis-1 kinetic models provided good fits of time-activity curves and reliable estimates of distribution volume. Regional distribution volume values were highest in the cingulate cortex and lowest in the thalamus for all radiotracers. 18F-4 showed greater differential uptake across brain regions and 3-fold-higher binding potential than 18F-2 SA4503 at the dose of 0.5 mg/kg blocked approximately 85% (18F-2) and 95% (18F-4) of radiotracer binding. Conclusion: Tracers 18F-2 and 18F-4 displayed high brain uptake and fast tissue kinetics, with 18F-4 having higher specific binding signals than 18F-2 in the same monkey. Taken together, these data indicate that both 18F-2 and 18F-4 possess the requisite kinetic and imaging properties as viable PET tracers for imaging S1R in the human brain.
Collapse
Affiliation(s)
- Evan Baum
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut
| | - Zhengxin Cai
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut
| | - Frederic Bois
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut
| | - Daniel Holden
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut
| | - Shu-Fei Lin
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut
| | - Teresa Lara-Jaime
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut
| | - Michael Kapinos
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut
| | - Yuanyuan Chen
- Ministry of Education Key Laboratory of Radiopharmaceuticals, College of Chemistry, Beijing Normal University, Beijing, China
| | - Winnie Deuther-Conrad
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Leipzig, Germany; and
| | - Steffen Fischer
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Leipzig, Germany; and
| | - Sladjana Dukic-Stefanovic
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Leipzig, Germany; and
| | - Paul Bunse
- Department of Pharmaceutical and Medicinal Chemistry, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Bernhard Wünsch
- Department of Pharmaceutical and Medicinal Chemistry, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Peter Brust
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Leipzig, Germany; and
| | - Hongmei Jia
- Ministry of Education Key Laboratory of Radiopharmaceuticals, College of Chemistry, Beijing Normal University, Beijing, China
| | - Yiyun Huang
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut
| |
Collapse
|
18
|
He Y, Xie F, Ye J, Deuther-Conrad W, Cui B, Wang L, Lu J, Steinbach J, Brust P, Huang Y, Lu J, Jia H. 1-(4-[ 18F]Fluorobenzyl)-4-[(tetrahydrofuran-2-yl)methyl]piperazine: A Novel Suitable Radioligand with Low Lipophilicity for Imaging σ 1 Receptors in the Brain. J Med Chem 2017; 60:4161-4172. [PMID: 28409931 DOI: 10.1021/acs.jmedchem.6b01723] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We have designed and synthesized novel piperazine compounds with low lipophilicity as σ1 receptor ligands. 1-(4-Fluorobenzyl)-4-[(tetrahydrofuran-2-yl)methyl]piperazine (10) possessed a low nanomolar σ1 receptor affinity and a high selectivity toward the vesicular acetylcholine transporter (>2000-fold), σ2 receptors (52-fold), and adenosine A2A, adrenergic α2, cannabinoid CB1, dopamine D1, D2L, γ-aminobutyric acid A (GABAA), NMDA, melatonin MT1, MT2, and serotonin 5-HT1 receptors. The corresponding radiotracer [18F]10 demonstrated high brain uptake and extremely high brain-to-blood ratios in biodistribution studies in mice. Pretreatment with the selective σ1 receptor agonist SA4503 significantly reduced the level of accumulation of the radiotracer in the brain. No radiometabolite of [18F]10 was observed to enter the brain. Positron emission tomography and magnetic resonance imaging confirmed suitable kinetics and a high specific binding of [18F]10 to σ1 receptors in rat brain. Ex vivo autoradiography showed a reduced level of binding of [18F]10 in the cortex and hippocampus of the senescence-accelerated prone (SAMP8) compared to that of the senescence-accelerated resistant (SAMR1) mice, indicating the potential dysfunction of σ1 receptors in Alzheimer's disease.
Collapse
Affiliation(s)
- Yingfang He
- Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University , Beijing, China
| | - Fang Xie
- Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University , Beijing, China
| | - Jiajun Ye
- Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University , Beijing, China
| | - Winnie Deuther-Conrad
- Helmholtz-Zentrum Dresden-Rossendorf , Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, 04318 Leipzig, Germany
| | - Bixiao Cui
- Department of Nuclear Medicine, Xuanwu Hospital Capital Medical University , Beijing, China
| | - Liang Wang
- Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University , Beijing, China
| | - Jie Lu
- Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University , Beijing, China
| | - Jörg Steinbach
- Helmholtz-Zentrum Dresden-Rossendorf , Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, 04318 Leipzig, Germany
| | - Peter Brust
- Helmholtz-Zentrum Dresden-Rossendorf , Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, 04318 Leipzig, Germany
| | - Yiyun Huang
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine , New Haven, Connecticut 06520-8048, United States
| | - Jie Lu
- Department of Nuclear Medicine, Xuanwu Hospital Capital Medical University , Beijing, China
| | - Hongmei Jia
- Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University , Beijing, China
| |
Collapse
|
19
|
Chiral resolution of serial potent and selective σ 1 ligands and biological evaluation of (-)-[ 18F]TZ3108 in rodent and the nonhuman primate brain. Bioorg Med Chem 2017; 25:1533-1542. [PMID: 28129990 DOI: 10.1016/j.bmc.2017.01.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 01/11/2017] [Accepted: 01/13/2017] [Indexed: 11/24/2022]
Abstract
Twelve optically pure enantiomers were obtained using either crystallization or chiral high performance liquid chromatography (HPLC) separation methodologies to resolve six racemic sigma-1 (σ1) receptor ligands. The in vitro binding affinities of each enantiomer for σ1, σ2 receptors and vesicular acetylcholine transporter (VAChT) were determined. Out of the 12 optically pure enantiomers, five displayed very high affinities for σ1 (Ki<2nM) and high selectivity for σ1 versus σ2 and VAChT (>100-fold). The minus enantiomer, (-)-14a ((-)-TZ3108) (Ki-σ1=1.8±0.4nM, Ki-σ2=6960±810nM, Ki-VAChT=980±87nM), was chosen for radiolabeling and further in vivo evaluation in rodents and nonhuman primates (NHPs). A biodistribution study in Sprague Dawley rats showed brain uptake (%ID/gram) of (-)-[18F]TZ3108 reached 1.285±0.062 at 5min and 0.802±0.129 at 120min. NHP microPET imaging studies revealed higher brain uptake of (-)-[18F]TZ3108 and more favorable pharmacokinetics compared to its racemic counterpart. Pretreatment of the animal using two structurally different σ1 ligands significantly decreased accumulation of (-)-[18F]TZ3108 in the brain. Together, our in vivo evaluation results suggest that (-)-[18F]TZ3108 is a promising positron emission tomography (PET) tracer for quantifying σ1 receptor in the brain.
Collapse
|
20
|
Lever SZ, Fan KH, Lever JR. Tactics for preclinical validation of receptor-binding radiotracers. Nucl Med Biol 2017; 44:4-30. [PMID: 27755986 PMCID: PMC5161541 DOI: 10.1016/j.nucmedbio.2016.08.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Revised: 08/24/2016] [Accepted: 08/24/2016] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Aspects of radiopharmaceutical development are illustrated through preclinical studies of [125I]-(E)-1-(2-(2,3-dihydrobenzofuran-5-yl)ethyl)-4-(iodoallyl)piperazine ([125I]-E-IA-BF-PE-PIPZE), a radioligand for sigma-1 (σ1) receptors, coupled with examples from the recent literature. Findings are compared to those previously observed for [125I]-(E)-1-(2-(2,3-dimethoxy-5-yl)ethyl)-4-(iodoallyl)piperazine ([125I]-E-IA-DM-PE-PIPZE). METHODS Syntheses of E-IA-BF-PE-PIPZE and [125I]-E-IA-BF-PE-PIPZE were accomplished by standard methods. In vitro receptor binding studies and autoradiography were performed, and binding potential was predicted. Measurements of lipophilicity and protein binding were obtained. In vivo studies were conducted in mice to evaluate radioligand stability, as well as specific binding to σ1 sites in brain, brain regions and peripheral organs in the presence and absence of potential blockers. RESULTS E-IA-BF-PE-PIPZE exhibited high affinity and selectivity for σ1 receptors (Ki = 0.43 ± 0.03 nM, σ2/σ1 = 173). [125I]-E-IA-BF-PE-PIPZE was prepared in good yield and purity, with high specific activity. Radioligand binding provided dissociation (koff) and association (kon) rate constants, along with a measured Kd of 0.24 ± 0.01 nM and Bmax of 472 ± 13 fmol/mg protein. The radioligand proved suitable for quantitative autoradiography in vitro using brain sections. Moderate lipophilicity, Log D7.4 2.69 ± 0.28, was determined, and protein binding was 71 ± 0.3%. In vivo, high initial whole brain uptake, >6% injected dose/g, cleared slowly over 24 h. Specific binding represented 75% to 93% of total binding from 15 min to 24 h. Findings were confirmed and extended by regional brain biodistribution. Radiometabolites were not observed in brain (1%). CONCLUSIONS Substitution of dihydrobenzofuranylethyl for dimethoxyphenethyl increased radioligand affinity for σ1 receptors by 16-fold. While high specific binding to σ1 receptors was observed for both radioligands in vivo, [125I]-E-IA-BF-PE-PIPZE displayed much slower clearance kinetics than [125I]-E-IA-DM-PE-PIPZE. Thus, minor structural modifications of σ1 receptor radioligands lead to major differences in binding properties in vitro and in vivo.
Collapse
Affiliation(s)
- Susan Z Lever
- Department of Chemistry, University of Missouri, Columbia, MO, USA; University of Missouri Research Reactor Center, Columbia, MO, USA.
| | - Kuo-Hsien Fan
- Department of Chemistry, University of Missouri, Columbia, MO, USA
| | - John R Lever
- Department of Radiology, University of Missouri, Columbia, MO, USA; Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, USA.
| |
Collapse
|
21
|
Weber F, Brust P, Laurini E, Pricl S, Wünsch B. Fluorinated PET Tracers for Molecular Imaging of σ 1 Receptors in the Central Nervous System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 964:31-48. [PMID: 28315263 DOI: 10.1007/978-3-319-50174-1_4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
At first the role of σ1 receptors in various neurological, psychiatric and neurodegenerative disorders is discussed. In the second part, the principle of positron emission tomography (PET ) is described and the known fluorinated PET tracers for labeling of σ1 receptors are presented. The third part focuses on fluoroalkyl substituted spirocyclic PET tracers, which represent the most promising class of fluorinated PET tracers reported so far. The homologous fluoroalkyl derivatives 12-15 show high σ1 affinity (K i = 0.59-1.4 nM) and high selectivity over the σ2 subtype (408-1331-fold). The enantiomers of the fluoroethyl derivative fluspidine 13 were prepared and pharmacologically characterized. Whereas the (S)-configured enantiomer (S)-13 (K i = 2.3 nM) is 4-fold less active than the (R)-enantiomer (R)-13 (K i = 0.57 nM), (S)-13 is metabolically more stable. The interactions of (S)-13 and (R)-13 with the σ1 receptor were analyzed at the molecular level using the 3D homology model. In an automated radiosynthesis [18F](S)-13 and [18F](R)-13 were prepared by nucleophilic substitution of the tosylates (S)-17 and (R)-17 with K[18F]F in high radiochemical yield, high radiochemical purity and short reaction time. Application of both enantiomers [18F](S)-13 and [18F](R)-13 to mice and piglets led to fast uptake into the brain, but [18F](R)-13 did not show washout from the brain indicating a quasi-irreversible binding. Both radiotracers [18F](S)-13 and [18F](R)-13 were able to label regions in the mouse and piglet brain with high σ1 receptor density. The specific binding of the enantiomeric tracers [18F](S)-13 and [18F](R)-13 could be replaced by the selective σ1 ligand SA4503.
Collapse
Affiliation(s)
- Frauke Weber
- Institute of Pharmaceutical and Medicinal Chemistry, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149, Münster, Germany
| | - Peter Brust
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Permoserstraße 15, D-04318, Leipzig, Germany
| | - Erik Laurini
- Molecular Simulations Engineering (MOSE) Laboratory, Department of Engineering and Architecture (DEA), University of Trieste, Via Valerio 6, 34127, Trieste, Italy
| | - Sabrina Pricl
- Molecular Simulations Engineering (MOSE) Laboratory, Department of Engineering and Architecture (DEA), University of Trieste, Via Valerio 6, 34127, Trieste, Italy
- National Interuniversity Consortium for Material Science and Technology (INSTM), Research Unit MOSE-DEA, University of Trieste, Via Valerio 6, 32127, Trieste, Italy
| | - Bernhard Wünsch
- Institute of Pharmaceutical and Medicinal Chemistry, Westfälische Wilhelms-Universität Münster, Corrensstraße 48, D-48149, Münster, Germany.
- Cells-in-Motion Cluster of Excellence (EXC 1003 - CiM), University Münster, Münster, Germany.
| |
Collapse
|