1
|
Naganawa M, Gallezot JD, Li S, Nabulsi NB, Henry S, Cai Z, Matuskey D, Huang Y, Carson RE. Noninvasive quantification of [ 18F]SynVesT-1 binding using simplified reference tissue model 2. Eur J Nucl Med Mol Imaging 2024; 52:113-121. [PMID: 39155309 DOI: 10.1007/s00259-024-06885-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 08/12/2024] [Indexed: 08/20/2024]
Abstract
PURPOSE [18F]SynVesT-1, a positron emission tomography (PET) radiotracer for the synaptic vesicle glycoprotein 2A (SV2A), demonstrates kinetics similar to [11C]UCB-J, with high brain uptake, fast kinetics fitting well with the one-tissue compartment (1TC) model, and excellent test-retest reproducibility. Challenges arise due to the similarity between k2 and [Formula: see text] (efflux rate of the reference region), when applying the simplified reference tissue model (SRTM) and related methods in [11C]UCB-J studies to accurately estimate [Formula: see text]. This study evaluated the suitability of these methods to estimate [18F]SynVesT-1 binding using centrum semiovale (CS) or cerebellum (CER) as reference regions. METHOD Seven healthy participants underwent 120-min PET scans on the HRRT scanner with [18F]SynVesT-1. Six participants underwent test and retest scans. Arterial blood sampling and metabolite analysis provided input functions for the 1TC model, serving as the gold standard for kinetic parameters values. SRTM, coupled SRTM (SRTMC) and SRTM2 estimated were applied to estimate [Formula: see text](ref: CS) and DVRCER(ref: CER) values. For SRTM2, the population average of [Formula: see text] was determined from the 1TC model applied to the reference region. Test-retest variability and minimum scan time were also calculated. RESULTS The 1TC k2 (1/min) values for CS and CER were 0.031 ± 0.004 and 0.021 ± 0.002, respectively. Although SRTMC [Formula: see text] was much higher than 1TC [Formula: see text], SRTMC underestimated BPND(ref: CS) and DVRCER by an average of 3% and 1% across regions, respectively, due to similar bias in k2 and [Formula: see text] estimation. SRTM underestimated BPND(ref: CS) by an average of 3%, but with the CER as reference region, SRTM estimation was unstable and DVRCER underestimation varied by region (mean 10%). Using population average [Formula: see text] values, SRTM2 BPND and DVRCER showed the best agreement with 1TC estimates. CONCLUSION Our findings support the use of population [Formula: see text] value in SRTM2 with [18F]SynVesT-1 for the estimation of [Formula: see text] or DVRCER, regardless of the choice of reference region.
Collapse
Affiliation(s)
- Mika Naganawa
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 801 Howard Avenue, PO Box 208048, New Haven, CT, 06520- 8048, USA.
| | - Jean-Dominique Gallezot
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 801 Howard Avenue, PO Box 208048, New Haven, CT, 06520- 8048, USA.
| | - Songye Li
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 801 Howard Avenue, PO Box 208048, New Haven, CT, 06520- 8048, USA
| | - Nabeel B Nabulsi
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 801 Howard Avenue, PO Box 208048, New Haven, CT, 06520- 8048, USA
| | - Shannan Henry
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 801 Howard Avenue, PO Box 208048, New Haven, CT, 06520- 8048, USA
| | - Zhengxin Cai
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 801 Howard Avenue, PO Box 208048, New Haven, CT, 06520- 8048, USA
| | - David Matuskey
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 801 Howard Avenue, PO Box 208048, New Haven, CT, 06520- 8048, USA
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 801 Howard Avenue, PO Box 208048, New Haven, CT, 06520- 8048, USA
| | - Richard E Carson
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, 801 Howard Avenue, PO Box 208048, New Haven, CT, 06520- 8048, USA
| |
Collapse
|
2
|
Kaur T, Shao X, Horikawa M, Sharninghausen LS, Preshlock S, Brooks AF, Henderson BD, Koeppe RA, DaSilva AF, Sanford MS, Scott PJH. Strategies for the Production of [ 11C]LY2795050 for Clinical Use. Org Process Res Dev 2023; 27:373-381. [PMID: 36874204 PMCID: PMC9983641 DOI: 10.1021/acs.oprd.2c00388] [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] [Indexed: 02/11/2023]
Abstract
This report describes a comparison of four different routes for the clinical-scale radiosynthesis of the κ-opioid receptor antagonist [11C]LY2795050. Palladium-mediated radiocyanation and radiocarbonylation of an aryl iodide precursor as well as copper-mediated radiocyanation of an aryl iodide and an aryl boronate ester have been investigated. Full automation of all four methods is reported, each of which provides [11C]LY2795050 in sufficient radiochemical yield, molar activity, and radiochemical purity for clinical use. The advantages and disadvantages of each radiosynthesis method are compared and contrasted.
Collapse
Affiliation(s)
- Tanpreet Kaur
- Department of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, Michigan 48109, USA
| | - Xia Shao
- Department of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, Michigan 48109, USA
| | - Mami Horikawa
- Department of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, Michigan 48109, USA
| | - Liam S. Sharninghausen
- Department of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, Michigan 48109, USA
| | - Sean Preshlock
- Department of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, Michigan 48109, USA
| | - Allen F. Brooks
- Department of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, Michigan 48109, USA
| | - Bradford D. Henderson
- Department of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, Michigan 48109, USA
| | - Robert A. Koeppe
- Department of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, Michigan 48109, USA
| | - Alexandre F. DaSilva
- Headache Orofacial Pain Effort (H.O.P.E.), Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Melanie S. Sanford
- Department of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, Michigan 48109, USA
| | - Peter J. H. Scott
- Department of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, Michigan 48109, USA
| |
Collapse
|
3
|
The Opioid System in Depression. Neurosci Biobehav Rev 2022; 140:104800. [PMID: 35914624 PMCID: PMC10166717 DOI: 10.1016/j.neubiorev.2022.104800] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 06/20/2022] [Accepted: 07/25/2022] [Indexed: 12/16/2022]
Abstract
Opioid receptors are widely distributed throughout the brain and play an essential role in modulating aspects of human mood, reward, and well-being. Accumulating evidence indicates the endogenous opioid system is dysregulated in depression and that pharmacological modulators of mu, delta, and kappa opioid receptors hold potential for the treatment of depression. Here we review animal and clinical data, highlighting evidence to support: dysregulation of the opioid system in depression, evidence for opioidergic modulation of behavioural processes and brain regions associated with depression, and evidence for opioidergic modulation in antidepressant responses. We evaluate clinical trials that have examined the safety and efficacy of opioidergic agents in depression and consider how the opioid system may be involved in the effects of other treatments, including ketamine, that are currently understood to exert antidepressant effects through non-opioidergic actions. Finally, we explore key neurochemical and molecular mechanisms underlying the potential therapeutic effects of opioid system engagement, that together provides a rationale for further investigation into this relevant target in the treatment of depression.
Collapse
|
4
|
Baynard C, Prisinzano TE, Butelman ER. Rapid-Onset Anti-Stress Effects of a Kappa-Opioid Receptor Antagonist, LY2795050, Against Immobility in an Open Space Swim Paradigm in Male and Female Mice. Front Pharmacol 2021; 12:775317. [PMID: 34880762 PMCID: PMC8645979 DOI: 10.3389/fphar.2021.775317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 11/08/2021] [Indexed: 11/13/2022] Open
Abstract
The kappa-opioid receptor (KOR) / dynorphin system is implicated with behavioral and neurobiological effects of stress exposure (including heavy exposure to drugs of abuse) in translational animal models. Thus some KOR-antagonists can decrease the aversive, depressant-like and anxiety-like effects caused by stress exposure. The first generation of selective KOR-antagonists have slow onsets (hours) and extremely long durations of action (days-weeks), in vivo. A new generation of KOR antagonists with rapid onset and shorter duration of action can potentially decrease the effects of stress exposure in translational models, and may be of interest for medication development. This study examined the rapid onset anti-stress effects of one of the shorter acting novel KOR-antagonists (LY2795050, (3-chloro-4-(4-(((2S)-2-pyridin-3-ylpyrrolidin-1-yl)methyl) phenoxy)benzamide)) in a single-session open space swim (OSS) stress paradigm (15 min duration), in adult male and female C57BL/6 J mice. LY2795050 (0.32 mg/kg, i.p.) had rapid onset (within 15 min) and short duration (<3 h) of KOR-antagonist effects, based on its blockade of the locomotor depressant effects of the KOR-agonist U50,488 (10 mg/kg). LY2795050 (0.32 mg/kg), when administered only 1 min prior to the OSS stress paradigm, decreased immobility in males, but not females. With a slightly longer pretreatment time (15 min), this dose of LY2795050 decreased immobility in both males and females. A 10-fold smaller dose of LY2795050 (0.032 mg/kg) was inactive in the OSS, showing dose-dependence of this anti-stress effect. Overall, these studies show that a novel KOR-antagonist can produce very rapid onset anti-immobility effects in this model of acute stress exposure.
Collapse
Affiliation(s)
- Caroline Baynard
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, NY, United States
| | - Thomas E Prisinzano
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, United States
| | - Eduardo R Butelman
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, NY, United States
| |
Collapse
|
5
|
Fillesoye F, Ibazizène M, Marie N, Noble F, Perrio C. Evaluation of Specific Binding of [ 11C]RTI-97 to Kappa Opioid Receptor by Autoradiography and PET Imaging in Rat. ACS Med Chem Lett 2021; 12:1739-1744. [PMID: 34795862 DOI: 10.1021/acsmedchemlett.1c00369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/15/2021] [Indexed: 11/28/2022] Open
Abstract
Kappa opioid receptor (KOR) PET imaging remains attractive to understand the role of KOR in health and diseases and to help the development of drugs especially for psychiatric disorders such as depression, anxiety, and addiction. The potent and selective KOR antagonist RTI-97 labeled with carbon-11 was previously demonstrated to display specific KOR binding in mouse brain by ex vivo autoradiography studies. Herein, we evaluated [11C]RTI-97 in rat by in vitro autoradiography and by in vivo PET imaging. The radiosynthesis of [11C]RTI-97 was optimized to obtain high molar activities. Despite a low cerebral uptake, the overall results showed a heterogeneous repartition and specific KOR binding of [11C]RTI-97 in brain and a high and specific accumulation of [11C]RTI-97 in pituitary in accordance with KOR expression.
Collapse
Affiliation(s)
- Fabien Fillesoye
- Normandie Univ, UNICAEN, CEA, CNRS, UMR 6030, LDM-TEP, Cyceron, Boulevard Henri, Becquerel, 14074 Caen, France
| | - Méziane Ibazizène
- Normandie Univ, UNICAEN, CEA, CNRS, UMR 6030, LDM-TEP, Cyceron, Boulevard Henri, Becquerel, 14074 Caen, France
| | - Nicolas Marie
- Université de Paris, CNRS, ERL 3649, Inserm, UMR-S 1124, Pharmacologie et thérapies des addictions, 75006 Paris, France
| | - Florence Noble
- Université de Paris, CNRS, ERL 3649, Inserm, UMR-S 1124, Pharmacologie et thérapies des addictions, 75006 Paris, France
| | - Cécile Perrio
- Normandie Univ, UNICAEN, CEA, CNRS, UMR 6030, LDM-TEP, Cyceron, Boulevard Henri, Becquerel, 14074 Caen, France
| |
Collapse
|
6
|
Lee HH, Blumberger DM, Lenze EJ, Anderson SJ, Barch DM, Black KJ, Cristancho P, Daskalakis ZJ, Eisenstein SA, Huang Y, Li S, Lissemore J, McConathy J, Mulsant BH, Rajji TK, Reynolds CF, Su Y, Tu Z, Voineskos D, Karp JF. Low-Dose Augmentation With Buprenorphine for Treatment-Resistant Depression: A Multisite Randomized Controlled Trial With Multimodal Assessment of Target Engagement. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2021; 2:127-135. [PMID: 36325158 PMCID: PMC9616305 DOI: 10.1016/j.bpsgos.2021.09.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 01/31/2023] Open
Abstract
Background The experimental therapeutics approach that combines a placebo-controlled clinical trial with translational neuroscience methods can provide a better understanding of both the clinical and physiological effects of pharmacotherapy. We aimed to test the efficacy and tolerability of low-dose augmentation with buprenorphine (BPN) for treatment-resistant depression, combined with multimodal assessment of target engagement. Methods In this multisite randomized clinical trial, 85 participants ≥50 years of age with a major depressive episode that had not responded to venlafaxine extended release were randomized to augmentation with BPN or placebo for 8 weeks. The primary outcome measure was the Montgomery-Åsberg Depression Rating Scale. In addition, three linked experiments were conducted to test target engagement: 1) functional magnetic resonance imaging using the monetary incentive delay task, 2) brain positron emission tomography of healthy participants using a novel kappa opioid receptor antagonist tracer [11C]LY2795050, and 3) transcranial magnetic stimulation measure of cortical transmission after daily BPN administration. Results The mean ± SD dosage of BPN was 0.59 ± 0.33 mg/day. There were no significant differences between the BPN and placebo groups in Montgomery-Åsberg Depression Rating Scale changes over time or adverse effects. BPN administration had minimal effects on functional magnetic resonance imaging blood oxygen level-dependent responses in regions involved in reward anticipation and response, no significant displacement of kappa opioid receptor radioligand in positron emission tomography imaging, and no significant changes in transcranial magnetic stimulation measures of inhibitory and excitatory cortical transmission. Conclusions Our findings suggest a lack of clinical effect of low-dose BPN augmentation and lack of target engagement with this dosage and physiological probes.
Collapse
Affiliation(s)
- Hyewon H. Lee
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada,Address correspondence to Hyewon H. Lee, M.D.
| | - Daniel M. Blumberger
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada,Temerty Centre for Therapeutic Brain Intervention and Campbell Family Research Institute at the Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Eric J. Lenze
- Department of Psychiatry, Washington University in St. Louis, St. Louis, Missouri
| | - Stewart J. Anderson
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Deanna M. Barch
- Departments of Psychological & Brain Sciences, Psychiatry, and Radiology, Washington University in St. Louis, St. Louis, Missouri
| | - Kevin J. Black
- Departments of Psychiatry, Neurology, Radiology, and Neuroscience, Washington University in St. Louis, St. Louis, Missouri
| | - Pilar Cristancho
- Department of Psychiatry, Washington University in St. Louis, St. Louis, Missouri
| | - Zafiris J. Daskalakis
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada,Temerty Centre for Therapeutic Brain Intervention and Campbell Family Research Institute at the Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Sarah A. Eisenstein
- Department of Psychiatry, Washington University in St. Louis, St. Louis, Missouri
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Songye Li
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut
| | - Jennifer Lissemore
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada,Temerty Centre for Therapeutic Brain Intervention and Campbell Family Research Institute at the Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Jonathan McConathy
- Molecular Imaging and Therapeutics, Department of Radiology, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Benoit H. Mulsant
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada,Temerty Centre for Therapeutic Brain Intervention and Campbell Family Research Institute at the Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Tarek K. Rajji
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada,Temerty Centre for Therapeutic Brain Intervention and Campbell Family Research Institute at the Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Charles F. Reynolds
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yi Su
- Banner Alzheimer’s Institute and Arizona Alzheimer’s Consortium, Phoenix, Arizona
| | - Zhude Tu
- Department of Radiology, Washington University in St. Louis, St. Louis, Missouri
| | - Daphne Voineskos
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada,Temerty Centre for Therapeutic Brain Intervention and Campbell Family Research Institute at the Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Jordan F. Karp
- Department of Psychiatry, University of Arizona College of Medicine, Tucson, Arizona
| |
Collapse
|
7
|
Affiliation(s)
- Youwen Xu
- Independent Consultant/Contractor 3900 Ford Road, Unit 18O Philadelphia PA USA
| | - Wenchao Qu
- Departments of Psychiatry and Chemistry Stony Brook University New York NY USA
| |
Collapse
|
8
|
Butelman ER, Baynard C, McElroy BD, Prisinzano TE, Kreek MJ. Profile of a short-acting κ-antagonist, LY2795050, on self-grooming behaviors, forced swim test and locomotor activity: sex comparison in mice. J Psychopharmacol 2021; 35:579-590. [PMID: 33769112 DOI: 10.1177/0269881121996883] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Novel short-acting κ(kappa)-opioid receptor selective antagonists are translational tools to examine the impact of the κ-receptor/dynorphin system in assays related to central nervous system dysfunction (e.g., substance use disorders, anhedonia and depression). The effects of such compounds have been compared in males and females under very limited conditions. AIMS The goal of this study was to examine potential sex differences in the effects of a κ-agonist and a short-acting κ-antagonist in an ethologically relevant test of anhedonia, the "splash test" of self-grooming, and also in the forced swim test and in locomotor activity. METHODS We examined the dose-dependence of grooming deficits caused by the κ-agonist U50,488 (0.1-3.2 mg/kg intraperitoneal (i.p.)) in gonadally intact adult male and female C57BL/6J mice. We then compared the effects of the short-acting κ-antagonist LY2795050 ((3-chloro-4-(4-(((2S)-2-pyridin-3-ylpyrrolidin-1-yl)methyl) phenoxy)benzamide)); 0.032-0.1 mg/kg i.p.) in blocking grooming deficits caused by U50,488 (3.2 mg/kg). The effects of LY2795050 were also studied in the forced swim test (FST). The effects of LY2795050 in blocking the locomotor depressant effects of U50,488 (10 mg/kg) were also studied. RESULTS U50,488 produced dose-dependent grooming deficits in male and female mice, and LY2795050 prevented these effects. In contrast, LY2795050 decreased immobility in the FST in males at a dose of 0.1 mg/kg, but not in females, up to a dose of 0.32 mg/kg. Also, LY2795050 (0.32 mg/kg) prevented and also reversed the locomotor-depressant effects of U50,488 (10 mg/kg), in males and females. CONCLUSIONS This study further implicates the κ-receptor system in ethologically relevant aspects of anhedonia, and confirms sexual dimorphism in some behavioral effects of novel κ-antagonists.
Collapse
Affiliation(s)
- Eduardo R Butelman
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, USA
| | - Caroline Baynard
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, USA
| | - Bryan D McElroy
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, USA
| | | | - Mary Jeanne Kreek
- Laboratory on the Biology of Addictive Diseases, The Rockefeller University, New York, USA
| |
Collapse
|
9
|
Blackwood CA, Cadet JL. The molecular neurobiology and neuropathology of opioid use disorder. CURRENT RESEARCH IN NEUROBIOLOGY 2021; 2. [PMID: 35548327 PMCID: PMC9090195 DOI: 10.1016/j.crneur.2021.100023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The number of people diagnosed with opioid use disorder has skyrocketed as a consequence of the opioid epidemic and the increased prescribing of opioid drugs for chronic pain relief. Opioid use disorder is characterized by loss of control of drug taking, continued drug use in the presence of adverse consequences, and repeated relapses to drug taking even after long periods of abstinence. Patients who suffer from opioid use disorder often present with cognitive deficits that are potentially secondary to structural brain abnormalities that vary according to the chemical composition of the abused opioid. This review details the neurobiological effects of oxycodone, morphine, heroin, methadone, and fentanyl on brain neurocircuitries by presenting the acute and chronic effects of these drugs on the human brain. In addition, we review results of neuroimaging in opioid use disorder patients and/or histological studies from brains of patients who had expired after acute intoxication following long-term use of these drugs. Moreover, we include relevant discussions of the neurobiological mechanisms involved in promoting abnormalities in the brains of opioid-exposed patients. Finally, we discuss how novel strategies could be used to provide pharmacological treatment against opioid use disorder. Brain abnormalities caused by opioid intoxication. Intoxication of opioids leads to defects in brain neurocircuitries. Insight into the molecular mechanisms associated with craving in heroin addicts.
Collapse
Affiliation(s)
| | - Jean Lud Cadet
- Corresponding author.Molecular Neuropsychiatry Research Branch NIH/NIDA Intramural Research Program 251 Bayview Boulevard Baltimore, MD, USA
| |
Collapse
|
10
|
Jackson IM, Lee SJ, Sowa AR, Rodnick ME, Bruton L, Clark M, Preshlock S, Rothley J, Rogers VE, Botti LE, Henderson BD, Hockley BG, Torres J, Raffel DM, Brooks AF, Frey KA, Kilbourn MR, Koeppe RA, Shao X, Scott PJH. Use of 55 PET radiotracers under approval of a Radioactive Drug Research Committee (RDRC). EJNMMI Radiopharm Chem 2020; 5:24. [PMID: 33175263 PMCID: PMC7658275 DOI: 10.1186/s41181-020-00110-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/19/2020] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND In the US, EU and elsewhere, basic clinical research studies with positron emission tomography (PET) radiotracers that are generally recognized as safe and effective (GRASE) can often be conducted under institutional approval. For example, in the United States, such research is conducted under the oversight of a Radioactive Drug Research Committee (RDRC) as long as certain requirements are met. Firstly, the research must be for basic science and cannot be intended for immediate therapeutic or diagnostic purposes, or to determine the safety and effectiveness of the PET radiotracer. Secondly, the PET radiotracer must be generally recognized as safe and effective. Specifically, the mass dose to be administered must not cause any clinically detectable pharmacological effect in humans, and the radiation dose to be administered must be the smallest dose practical to perform the study and not exceed regulatory dose limits within a 1-year period. In our experience, the main barrier to using a PET radiotracer under RDRC approval is accessing the required information about mass and radioactive dosing. RESULTS The University of Michigan (UM) has a long history of using PET radiotracers in clinical research studies. Herein we provide dosing information for 55 radiotracers that will enable other PET Centers to use them under the approval of their own RDRC committees. CONCLUSIONS The data provided herein will streamline future RDRC approval, and facilitate further basic science investigation of 55 PET radiotracers that target functionally relevant biomarkers in high impact disease states.
Collapse
Affiliation(s)
- Isaac M Jackson
- Department of Radiology, University of Michigan, 2276 Medical Science Bldg I, SPC 5610, Ann Arbor, MI, 48109, USA
- Present Address: Stanford University, Stanford, CA, USA
| | - So Jeong Lee
- Department of Radiology, University of Michigan, 2276 Medical Science Bldg I, SPC 5610, Ann Arbor, MI, 48109, USA
- Present Address: Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alexandra R Sowa
- Department of Radiology, University of Michigan, 2276 Medical Science Bldg I, SPC 5610, Ann Arbor, MI, 48109, USA
| | - Melissa E Rodnick
- Department of Radiology, University of Michigan, 2276 Medical Science Bldg I, SPC 5610, Ann Arbor, MI, 48109, USA
| | - Laura Bruton
- Department of Radiology, University of Michigan, 2276 Medical Science Bldg I, SPC 5610, Ann Arbor, MI, 48109, USA
| | - Mara Clark
- Department of Radiology, University of Michigan, 2276 Medical Science Bldg I, SPC 5610, Ann Arbor, MI, 48109, USA
| | - Sean Preshlock
- Department of Radiology, University of Michigan, 2276 Medical Science Bldg I, SPC 5610, Ann Arbor, MI, 48109, USA
| | - Jill Rothley
- Department of Radiology, University of Michigan, 2276 Medical Science Bldg I, SPC 5610, Ann Arbor, MI, 48109, USA
| | - Virginia E Rogers
- Department of Radiology, University of Michigan, 2276 Medical Science Bldg I, SPC 5610, Ann Arbor, MI, 48109, USA
| | - Leslie E Botti
- Department of Radiology, University of Michigan, 2276 Medical Science Bldg I, SPC 5610, Ann Arbor, MI, 48109, USA
| | - Bradford D Henderson
- Department of Radiology, University of Michigan, 2276 Medical Science Bldg I, SPC 5610, Ann Arbor, MI, 48109, USA
| | - Brian G Hockley
- Department of Radiology, University of Michigan, 2276 Medical Science Bldg I, SPC 5610, Ann Arbor, MI, 48109, USA
| | - Jovany Torres
- Department of Radiology, University of Michigan, 2276 Medical Science Bldg I, SPC 5610, Ann Arbor, MI, 48109, USA
| | - David M Raffel
- Department of Radiology, University of Michigan, 2276 Medical Science Bldg I, SPC 5610, Ann Arbor, MI, 48109, USA
| | - Allen F Brooks
- Department of Radiology, University of Michigan, 2276 Medical Science Bldg I, SPC 5610, Ann Arbor, MI, 48109, USA
| | - Kirk A Frey
- Department of Radiology, University of Michigan, 2276 Medical Science Bldg I, SPC 5610, Ann Arbor, MI, 48109, USA
| | - Michael R Kilbourn
- Department of Radiology, University of Michigan, 2276 Medical Science Bldg I, SPC 5610, Ann Arbor, MI, 48109, USA
| | - Robert A Koeppe
- Department of Radiology, University of Michigan, 2276 Medical Science Bldg I, SPC 5610, Ann Arbor, MI, 48109, USA
| | - Xia Shao
- Department of Radiology, University of Michigan, 2276 Medical Science Bldg I, SPC 5610, Ann Arbor, MI, 48109, USA
| | - Peter J H Scott
- Department of Radiology, University of Michigan, 2276 Medical Science Bldg I, SPC 5610, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
11
|
Tangherlini G, Börgel F, Schepmann D, Slocum S, Che T, Wagner S, Schwegmann K, Hermann S, Mykicki N, Loser K, Wünsch B. Synthesis and Pharmacological Evaluation of Fluorinated Quinoxaline-Based κ-Opioid Receptor (KOR) Agonists Designed for PET Studies. ChemMedChem 2020; 15:1834-1853. [PMID: 33448685 PMCID: PMC7589326 DOI: 10.1002/cmdc.202000502] [Citation(s) in RCA: 5] [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: 07/09/2020] [Indexed: 12/19/2022]
Abstract
κ-Opioid receptors (KORs) play a predominant role in pain alleviation, itching skin diseases, depression and neurodegenerative disorders such as multiple sclerosis. Therefore, imaging of KOR by a fluorinated PET tracer was envisaged. Two strategies were followed to introduce a F atom into the very potent class of cis,trans-configured perhydroquinoxalines. Whereas the synthesis of fluoroethyltriazole 2 has already been reported, fluoropyrrolidines 14 (1-[2-(3,4-dichlorophenyl)acetyl]-8-[(R)-3-fluoropyrrolidin-1-yl]-perhydroquinoxalines) were prepared by SN2 substitution of a cyclic sulfuric acid derivative with hydroxypyrrolidine and subsequent transformation of the OH moiety into a F substituent. Fluoropyrrolidines 14 showed similar low-nanomolar KOR affinity and selectivity to the corresponding pyrrolidines, but the corresponding alcohols were slightly less active. In the cAMP and β-arrestin assay, 14b (proton at the 4-position) exhibited similar KOR agonistic activity as U-50,488. The fluoro derivatives 14b and 14c (CO2CH3 at the 4-position) revealed KOR-mediated anti-inflammatory activity as CD11c and the IFN-γ production were reduced significantly in mouse and human dendritic cells. Compounds 14b and 14-c also displayed anti-inflammatory and immunomodulatory activity in mouse and human T cells. The PET tracer [18F]-2 was prepared by 1,3-dipolar cycloaddition. In vivo, [18F]-2 did not label KOR due to very fast elimination kinetics. Nucleophilic substitution of a mesylate precursor provided [18F]-14c. Unfortunately, defluorination of [18F]-14c occurred in vivo, which was analyzed in detail by in vitro studies.
Collapse
Affiliation(s)
- Giovanni Tangherlini
- Institut für Pharmazeutische und Medizinische ChemieUniversität MünsterCorrensstraße 4848149MünsterGermany
- Cells-in-Motion Cluster of Excellence (EXC 1003-CiM)Westfälische Wilhelms-Universität Münster48149MünsterGermany
| | - Frederik Börgel
- Institut für Pharmazeutische und Medizinische ChemieUniversität MünsterCorrensstraße 4848149MünsterGermany
| | - Dirk Schepmann
- Institut für Pharmazeutische und Medizinische ChemieUniversität MünsterCorrensstraße 4848149MünsterGermany
| | - Samuel Slocum
- Department of PharmacologyUniversity of North Carolina at Chapel Hill School of MedicineChapel HillNC 27599USA
| | - Tao Che
- Department of AnesthesiologyWashington University School of Medicine660 S. Euclid Ave.St. LouisMO 63110USA
| | - Stefan Wagner
- Department of Nuclear MedicineUniversity Hospital MünsterAlbert-Schweitzer-Campus 1, Building A148149MünsterGermany
| | - Katrin Schwegmann
- European Institute for Molecular Imaging (EIMI)University of MünsterWaldeyerstraße 1548149MünsterGermany
| | - Sven Hermann
- European Institute for Molecular Imaging (EIMI)University of MünsterWaldeyerstraße 1548149MünsterGermany
| | - Nadine Mykicki
- Department of DermatologyUniversity of Münstervon-Esmarch-Str. 5848149MünsterGermany
| | - Karin Loser
- Cells-in-Motion Cluster of Excellence (EXC 1003-CiM)Westfälische Wilhelms-Universität Münster48149MünsterGermany
- Department of DermatologyUniversity of Münstervon-Esmarch-Str. 5848149MünsterGermany
- CRC1009 Breaking Barriers and CRC-TR 128 Multiple SclerosisUniversity of Münstervon-Esmarch-Str. 5848149MünsterGermany
| | - Bernhard Wünsch
- Institut für Pharmazeutische und Medizinische ChemieUniversität MünsterCorrensstraße 4848149MünsterGermany
- Cells-in-Motion Cluster of Excellence (EXC 1003-CiM)Westfälische Wilhelms-Universität Münster48149MünsterGermany
| |
Collapse
|
12
|
Naganawa M, Nabulsi N, Henry S, Matuskey D, Lin SF, Slieker L, Schwarz AJ, Kant N, Jesudason C, Ruley K, Navarro A, Gao H, Ropchan J, Labaree D, Carson RE, Huang Y. First-in-Human Assessment of 11C-LSN3172176, an M1 Muscarinic Acetylcholine Receptor PET Radiotracer. J Nucl Med 2020; 62:553-560. [PMID: 32859711 DOI: 10.2967/jnumed.120.246967] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 07/09/2020] [Indexed: 01/25/2023] Open
Abstract
This was a first-in-human study of the PET radiotracer 11C-LSN3172176 for the muscarinic acetylcholine receptor subtype M1. The objectives of the study were to determine the appropriate kinetic model to quantify binding of the tracer to M1 receptors, and the reliability of the chosen quantification method. Methods: Six healthy subjects completed the test-retest protocol, and 5 healthy subjects completed the baseline-scopolamine blocking protocol. Multiple modeling methods were applied to calculate total distribution volume (V T) and nondisplaceable binding potential (BP ND) in various brain regions. The reference region was selected from the blocking study. The occupancy plot was applied to compute receptor occupancy by scopolamine and nondisplaceable distribution volume. Results: Tracer uptake was highest in the striatum, followed by neocortical regions and white matter, and lowest in the cerebellum. Regional time-activity curves were fitted well by all models. The 2-tissue-compartment (2TC) model fits were good, but the 2TC parameters often could not be reliably estimated. Because V T correlated well between the 2TC and 1-tissue-compartment (1TC) models after exclusion of unreliable estimates, the 1TC model was chosen as the most appropriate. The cerebellum showed the lowest V T, consistent with preclinical studies showing little to no specific binding in the region. Further, cerebellar V T did not change between baseline and blocking scans, indicating that the cerebellum is a suitable reference region. The simplified reference tissue model (SRTM) slightly underestimated 1TC BP ND, and the simplified reference tissue model 2 (SRTM2) improved BP ND estimation. An 80-min scan was sufficient to quantify V T and BP ND The test-retest study showed excellent absolute test-retest variability for 1TC V T (≤5%) and BP ND (≤10%). In the baseline and blocking studies, occupancy values were lower in the striatum than in nonstriatal regions, as may be attributed to differences in regional acetylcholine concentrations. Conclusion: The 1TC and SRTM2 models are appropriate for quantitative analysis of 11C-LSN3172176 imaging data. 11C-LSN3172176 displayed excellent test-retest reproducibility and is a highly promising ligand to quantify M1 receptors in the human brain.
Collapse
Affiliation(s)
- Mika Naganawa
- PET Center, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut; and
| | - Nabeel Nabulsi
- PET Center, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut; and
| | - Shannan Henry
- PET Center, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut; and
| | - David Matuskey
- PET Center, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut; and
| | - Shu-Fei Lin
- PET Center, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut; and
| | | | | | - Nancy Kant
- Eli Lilly and Company, Indianapolis, Indiana
| | | | - Kevin Ruley
- Eli Lilly and Company, Indianapolis, Indiana
| | | | - Hong Gao
- PET Center, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut; and
| | - Jim Ropchan
- PET Center, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut; and
| | - David Labaree
- PET Center, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut; and
| | - Richard E Carson
- PET Center, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut; and
| | - Yiyun Huang
- PET Center, Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, Connecticut; and
| |
Collapse
|
13
|
Zhang YH, Chen FF, Li BB, Zhou XY, Chen Q, Xu JH, Zheng GW. Stereocomplementary Synthesis of Pharmaceutically Relevant Chiral 2-Aryl-Substituted Pyrrolidines Using Imine Reductases. Org Lett 2020; 22:3367-3372. [DOI: 10.1021/acs.orglett.0c00802] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Yu-Hui Zhang
- State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Fei-Fei Chen
- State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Bo-Bo Li
- State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Xin-Yi Zhou
- State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Qi Chen
- State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Jian-He Xu
- State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| | - Gao-Wei Zheng
- State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing, East China University of Science and Technology, 130 Meilong Road, Shanghai 200237, China
| |
Collapse
|
14
|
Naganawa M, Li S, Nabulsi N, Lin SF, Labaree D, Ropchan J, Gao H, Mei M, Henry S, Matuskey D, Carson RE, Huang Y. Kinetic Modeling and Test-Retest Reproducibility of 11C-EKAP and 11C-FEKAP, Novel Agonist Radiotracers for PET Imaging of the κ-Opioid Receptor in Humans. J Nucl Med 2020; 61:1636-1642. [PMID: 32169917 DOI: 10.2967/jnumed.119.227694] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/09/2020] [Indexed: 12/13/2022] Open
Abstract
The κ-opioid receptor (KOR) is implicated in various neuropsychiatric disorders. We previously evaluated an agonist tracer, 11C-GR103545, for PET imaging of KOR in humans. Although 11C-GR103545 showed high brain uptake, good binding specificity, and selectivity for KOR, it displayed slow kinetics and relatively large test-retest variability of total distribution volume (V T) estimates (15%). Therefore, we set out to develop 2 novel KOR agonist radiotracers, 11C-EKAP and 11C-FEKAP. In nonhuman primates, both tracers exhibited faster kinetics than 11C-GR103545 and comparable binding parameters to 11C-GR103545. The aim of this study was to assess their kinetic and binding properties in humans. Methods: Six healthy subjects underwent 120-min test-retest PET scans with both 11C-EKAP and 11C-FEKAP. Metabolite-corrected arterial input functions were measured. Regional time-activity curves were generated for 14 regions of interest. One-tissue-compartment and 2-tissue-compartment (2TC) models and the multilinear analysis-1 (MA1) method were applied to the regional time-activity curves to calculate V T The time stability of V T and test-retest reproducibility were evaluated. Levels of specific binding, as measured by the nondisplaceable binding potential (BP ND) for the 3 tracers (11C-EKAP, 11C-FEKAP, and 11C-GR103545), were compared using a graphical method. Results: For both tracers, regional time-activity curves were fitted well with the 2TC model and MA1 method (t* = 20 min) but not with the 1-tissue-compartment model. Given the unreliably estimated parameters in several fits with the 2TC model and a good V T match between MA1 and 2TC, MA1 was chosen as the appropriate model for both tracers. Mean MA1 V T was highest for 11C-GR103545, followed by 11C-EKAP and then 11C-FEKAP. The minimum scan time for stable V T measurement was 90 and 110 min for 11C-EKAP and 11C-FEKAP, respectively, compared with 140 min for 11C-GR103545. The mean absolute test-retest variability in MA1 V T estimates was 7% and 18% for 11C-EKAP and 11C-FEKAP, respectively. BP ND levels were similar for 11C-FEKAP and 11C-GR103545 but were about 25% lower for 11C-EKAP. Conclusion: The 2 novel KOR agonist tracers showed faster tissue kinetics than 11C-GR103545. Even with a slightly lower BP ND, 11C-EKAP is judged to be a better tracer for imaging and quantification of KOR in humans, on the basis of the shorter minimum scan time and the excellent test-retest reproducibility of regional V T.
Collapse
Affiliation(s)
- Mika Naganawa
- Yale PET Center, Yale University, New Haven, Connecticut
| | - Songye Li
- Yale PET Center, Yale University, New Haven, Connecticut
| | - Nabeel Nabulsi
- Yale PET Center, Yale University, New Haven, Connecticut
| | - Shu-Fei Lin
- Yale PET Center, Yale University, New Haven, Connecticut
| | - David Labaree
- Yale PET Center, Yale University, New Haven, Connecticut
| | - Jim Ropchan
- Yale PET Center, Yale University, New Haven, Connecticut
| | - Hong Gao
- Yale PET Center, Yale University, New Haven, Connecticut
| | - Michael Mei
- Yale PET Center, Yale University, New Haven, Connecticut
| | - Shannan Henry
- Yale PET Center, Yale University, New Haven, Connecticut
| | - David Matuskey
- Yale PET Center, Yale University, New Haven, Connecticut
| | | | - Yiyun Huang
- Yale PET Center, Yale University, New Haven, Connecticut
| |
Collapse
|
15
|
McCluskey SP, Plisson C, Rabiner EA, Howes O. Advances in CNS PET: the state-of-the-art for new imaging targets for pathophysiology and drug development. Eur J Nucl Med Mol Imaging 2020; 47:451-489. [PMID: 31541283 PMCID: PMC6974496 DOI: 10.1007/s00259-019-04488-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/15/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE A limit on developing new treatments for a number of central nervous system (CNS) disorders has been the inadequate understanding of the in vivo pathophysiology underlying neurological and psychiatric disorders and the lack of in vivo tools to determine brain penetrance, target engagement, and relevant molecular activity of novel drugs. Molecular neuroimaging provides the tools to address this. This article aims to provide a state-of-the-art review of new PET tracers for CNS targets, focusing on developments in the last 5 years for targets recently available for in-human imaging. METHODS We provide an overview of the criteria used to evaluate PET tracers. We then used the National Institute of Mental Health Research Priorities list to identify the key CNS targets. We conducted a PubMed search (search period 1st of January 2013 to 31st of December 2018), which yielded 40 new PET tracers across 16 CNS targets which met our selectivity criteria. For each tracer, we summarised the evidence of its properties and potential for use in studies of CNS pathophysiology and drug evaluation, including its target selectivity and affinity, inter and intra-subject variability, and pharmacokinetic parameters. We also consider its potential limitations and missing characterisation data, but not specific applications in drug development. Where multiple tracers were present for a target, we provide a comparison of their properties. RESULTS AND CONCLUSIONS Our review shows that multiple new tracers have been developed for proteinopathy targets, particularly tau, as well as the purinoceptor P2X7, phosphodiesterase enzyme PDE10A, and synaptic vesicle glycoprotein 2A (SV2A), amongst others. Some of the most promising of these include 18F-MK-6240 for tau imaging, 11C-UCB-J for imaging SV2A, 11C-CURB and 11C-MK-3168 for characterisation of fatty acid amide hydrolase, 18F-FIMX for metabotropic glutamate receptor 1, and 18F-MNI-444 for imaging adenosine 2A. Our review also identifies recurrent issues within the field. Many of the tracers discussed lack in vivo blocking data, reducing confidence in selectivity. Additionally, late-stage identification of substantial off-target sites for multiple tracers highlights incomplete pre-clinical characterisation prior to translation, as well as human disease state studies carried out without confirmation of test-retest reproducibility.
Collapse
Affiliation(s)
- Stuart P McCluskey
- Invicro LLC, A Konica Minolta Company, Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK.
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK.
| | - Christophe Plisson
- Invicro LLC, A Konica Minolta Company, Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Eugenii A Rabiner
- Invicro LLC, A Konica Minolta Company, Burlington Danes Building, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0NN, UK
| | - Oliver Howes
- Psychiatric Imaging Group, MRC London Institute of Medical Sciences, Imperial College London, Hammersmith Hospital, London, UK
| |
Collapse
|
16
|
A Survey of Molecular Imaging of Opioid Receptors. Molecules 2019; 24:molecules24224190. [PMID: 31752279 PMCID: PMC6891617 DOI: 10.3390/molecules24224190] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/11/2019] [Accepted: 11/13/2019] [Indexed: 01/09/2023] Open
Abstract
The discovery of endogenous peptide ligands for morphine binding sites occurred in parallel with the identification of three subclasses of opioid receptor (OR), traditionally designated as μ, δ, and κ, along with the more recently defined opioid-receptor-like (ORL1) receptor. Early efforts in opioid receptor radiochemistry focused on the structure of the prototype agonist ligand, morphine, although N-[methyl-11C]morphine, -codeine and -heroin did not show significant binding in vivo. [11C]Diprenorphine ([11C]DPN), an orvinol type, non-selective OR antagonist ligand, was among the first successful PET tracers for molecular brain imaging, but has been largely supplanted in research studies by the μ-preferring agonist [11C]carfentanil ([11C]Caf). These two tracers have the property of being displaceable by endogenous opioid peptides in living brain, thus potentially serving in a competition-binding model. Indeed, many clinical PET studies with [11C]DPN or [11C]Caf affirm the release of endogenous opioids in response to painful stimuli. Numerous other PET studies implicate μ-OR signaling in aspects of human personality and vulnerability to drug dependence, but there have been very few clinical PET studies of μORs in neurological disorders. Tracers based on naltrindole, a non-peptide antagonist of the δ-preferring endogenous opioid enkephalin, have been used in PET studies of δORs, and [11C]GR103545 is validated for studies of κORs. Structures such as [11C]NOP-1A show selective binding at ORL-1 receptors in living brain. However, there is scant documentation of δ-, κ-, or ORL1 receptors in healthy human brain or in neurological and psychiatric disorders; here, clinical PET research must catch up with recent progress in radiopharmaceutical chemistry.
Collapse
|
17
|
Butelman ER, McElroy BD, Prisinzano TE, Kreek MJ. Impact of Pharmacological Manipulation of the κ-Opioid Receptor System on Self-grooming and Anhedonic-like Behaviors in Male Mice. J Pharmacol Exp Ther 2019; 370:1-8. [PMID: 30975792 PMCID: PMC6538891 DOI: 10.1124/jpet.119.256354] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 04/09/2019] [Indexed: 12/11/2022] Open
Abstract
The kappa (κ) opioid receptor/dynorphin system modulates depression-like states and anhedonia, as well adaptations to stress and exposure to drugs of abuse. Several relatively short-acting small molecule κ-receptor antagonists have been synthesized, and their behavioral profile has been examined under some conditions. The hypothesis of this study is that pharmacological manipulations of the κ-receptor system will result in changes in ethologically relevant anhedonic-like behaviors in mice. Adult male C57BL/6j mice (n = 6-8) were examined for self-grooming behavior in the splash test (in which robust self-grooming is elicited by spraying the dorsum of the mouse with a sucrose solution). The κ-agonist salvinorin A (0.56-1.8 mg/kg) produced dose-dependent decreases in self-grooming, a marker of anhedonia. The selectivity, potency, and duration of action of two relatively short-acting κ-antagonists, LY2444296 [(S)-3-fluoro-4-(4-((2-(3-fluorophenyl) pyrrolidin-1-yl)methyl)phenoxy)benzamide] and LY2795050 [3-chloro-4-(4-(((2S)-2-pyridin-3-ylpyrrolidin-1-yl)methyl) phenoxy)benzamide], were studied for their effectiveness in preventing grooming deficits caused by salvinorin A (1.8 mg/kg). κ-selective doses of both LY2444296 (0.032-1 mg/kg) and LY2795050 (0.032-0.32 mg/kg) dose- and time-dependently prevented the grooming deficits caused by salvinorin A (1.8 m/kg). We also found that a κ-selective dose of each of these antagonists decreased immobility in the forced swim test, a common test of anti-anhedonia effects. This study shows that the κ-receptor system is involved in an ethologically relevant measure of anhedonia, and that κ-selective doses of these antagonists can produce effects consistent with rapid anti-anhedonia. SIGNIFICANCE STATEMENT: Activation of the κ-opioid receptor system results in grooming deficits in mice, an ethologically relevant marker of anhedonia. Shorter acting κ-antagonists are able to cause effects consistent with rapid antianhedonia.
Collapse
Affiliation(s)
- Eduardo R Butelman
- Laboratory on the Biology of Addictive Diseases, the Rockefeller University, New York, New York (E.R.B., B.D.M., M.J.K.), and Department of Medicinal Chemistry, School of Pharmacy, University of Kansas, Lawrence, Kansas (T.E.P.)
| | - Bryan D McElroy
- Laboratory on the Biology of Addictive Diseases, the Rockefeller University, New York, New York (E.R.B., B.D.M., M.J.K.), and Department of Medicinal Chemistry, School of Pharmacy, University of Kansas, Lawrence, Kansas (T.E.P.)
| | - Thomas E Prisinzano
- Laboratory on the Biology of Addictive Diseases, the Rockefeller University, New York, New York (E.R.B., B.D.M., M.J.K.), and Department of Medicinal Chemistry, School of Pharmacy, University of Kansas, Lawrence, Kansas (T.E.P.)
| | - Mary Jeanne Kreek
- Laboratory on the Biology of Addictive Diseases, the Rockefeller University, New York, New York (E.R.B., B.D.M., M.J.K.), and Department of Medicinal Chemistry, School of Pharmacy, University of Kansas, Lawrence, Kansas (T.E.P.)
| |
Collapse
|
18
|
Li S, Zheng MQ, Naganawa M, Gao H, Pracitto R, Shirali A, Lin SF, Teng JK, Ropchan J, Huang Y. Novel Kappa Opioid Receptor Agonist as Improved PET Radiotracer: Development and in Vivo Evaluation. Mol Pharm 2019; 16:1523-1531. [PMID: 30726092 DOI: 10.1021/acs.molpharmaceut.8b01209] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The kappa opioid receptor (KOR) is involved in depression, alcoholism, and drug abuse. The current agonist radiotracer 11C-GR103545 is not ideal for imaging KOR due to its slow tissue kinetics in human. The aim of our project was to develop novel KOR agonist radiotracers with improved imaging properties. A novel compound FEKAP ((( R))-4-(2-(3,4-dichlorophenyl)acetyl)-3-((ethyl(2-fluoroethyl)amino)methyl) piperazine-1-carboxylate) was designed, synthesized, and assayed for in vitro binding affinities. It was then radiolabeled and evaluated in rhesus monkeys. Baseline and blocking scans were conducted on a Focus-220 scanner to assess binding specificity and selectivity. Metabolite-corrected arterial activities over time were measured and used as input functions to analyze the brain regional time-activity curves and derive kinetic and binding parameters with kinetic modeling. FEKAP displayed high KOR binding affinity ( Ki = 0.43 nM) and selectivity (17-fold over mu opioid receptor and 323-fold over delta opioid receptor) in vitro. 11C-FEKAP was prepared in high molar activity (mean of 718 GBq/μmol, n = 19) and >99% radiochemical purity. In monkeys, 11C-FEKAP metabolized fairly fast, with ∼31% of intact parent fraction at 30 min post-injection. In the brain, it exhibited fast and reversible kinetics with good uptake. Pretreatment with the nonselective opioid receptor antagonist naloxone (1 mg/kg) decreased uptake in high binding regions to the level in the cerebellum, and the selective KOR antagonist LY2456302 (0.02 and 0.1 mg/kg) reduced 11C-FEKAP specific binding in a dose-dependent manner. As a measure of specific binding signals, the mean binding potential ( BPND) values of 11C-FEKAP derived from the multilinear analysis-1 (MA1) method were greater than 0.5 for all regions, except for the thalamus. The novel KOR agonist tracer 11C-FEKAP demonstrated binding specificity and selectivity in vivo and exhibited attractive properties of fast tissue kinetics and high specific binding.
Collapse
Affiliation(s)
- Songye Li
- PET Center, Department of Radiology and Biomedical Imaging , Yale University School of Medicine , New Haven , Connecticut 06520 , United States
| | - Ming-Qiang Zheng
- PET Center, Department of Radiology and Biomedical Imaging , Yale University School of Medicine , New Haven , Connecticut 06520 , United States
| | - Mika Naganawa
- PET Center, Department of Radiology and Biomedical Imaging , Yale University School of Medicine , New Haven , Connecticut 06520 , United States
| | - Hong Gao
- PET Center, Department of Radiology and Biomedical Imaging , Yale University School of Medicine , New Haven , Connecticut 06520 , United States
| | - Richard Pracitto
- PET Center, Department of Radiology and Biomedical Imaging , Yale University School of Medicine , New Haven , Connecticut 06520 , United States
| | - Anupama Shirali
- PET Center, Department of Radiology and Biomedical Imaging , Yale University School of Medicine , New Haven , Connecticut 06520 , United States
| | - Shu-Fei Lin
- PET Center, Department of Radiology and Biomedical Imaging , Yale University School of Medicine , New Haven , Connecticut 06520 , United States
| | - Jo-Ku Teng
- PET Center, Department of Radiology and Biomedical Imaging , Yale University School of Medicine , New Haven , Connecticut 06520 , United States
| | - Jim Ropchan
- PET Center, Department of Radiology and Biomedical Imaging , Yale University School of Medicine , New Haven , Connecticut 06520 , United States
| | - Yiyun Huang
- PET Center, Department of Radiology and Biomedical Imaging , Yale University School of Medicine , New Haven , Connecticut 06520 , United States
| |
Collapse
|
19
|
Burns JA, Kroll DS, Feldman DE, Kure Liu C, Manza P, Wiers CE, Volkow ND, Wang GJ. Molecular Imaging of Opioid and Dopamine Systems: Insights Into the Pharmacogenetics of Opioid Use Disorders. Front Psychiatry 2019; 10:626. [PMID: 31620026 PMCID: PMC6759955 DOI: 10.3389/fpsyt.2019.00626] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 08/05/2019] [Indexed: 12/21/2022] Open
Abstract
Opioid use in the United States has steadily risen since the 1990s, along with staggering increases in addiction and overdose fatalities. With this surge in prescription and illicit opioid abuse, it is paramount to understand the genetic risk factors and neuropsychological effects of opioid use disorder (OUD). Polymorphisms disrupting the opioid and dopamine systems have been associated with increased risk for developing substance use disorders. Molecular imaging studies have revealed how these polymorphisms impact the brain and contribute to cognitive and behavioral differences across individuals. Here, we review the current molecular imaging literature to assess how genetic variations in the opioid and dopamine systems affect function in the brain's reward, cognition, and stress pathways, potentially resulting in vulnerabilities to OUD. Continued research of the functional consequences of genetic variants and corresponding alterations in neural mechanisms will inform prevention and treatment of OUD.
Collapse
Affiliation(s)
- Jamie A Burns
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Danielle S Kroll
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Dana E Feldman
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | | | - Peter Manza
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Corinde E Wiers
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| | - Nora D Volkow
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States.,National Institute on Drug Abuse, Bethesda, MD, United States
| | - Gene-Jack Wang
- National Institute on Alcohol Abuse and Alcoholism, Bethesda, MD, United States
| |
Collapse
|
20
|
Peciña M, Karp JF, Mathew S, Todtenkopf MS, Ehrich EW, Zubieta JK. Endogenous opioid system dysregulation in depression: implications for new therapeutic approaches. Mol Psychiatry 2019; 24:576-587. [PMID: 29955162 PMCID: PMC6310672 DOI: 10.1038/s41380-018-0117-2] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 04/11/2018] [Accepted: 05/25/2018] [Indexed: 12/12/2022]
Abstract
The United States is in the midst of an opioid addiction and overdose crisis precipitated and exacerbated by use of prescription opioid medicines. The majority of opioid prescriptions are dispensed to patients with comorbid mood disorders including major depressive disorder (MDD). A growing body of research indicates that the endogenous opioid system is directly involved in the regulation of mood and is dysregulated in MDD. This involvement of the endogenous opioid system may underlie the disproportionate use of opioids among patients with mood disorders. Emerging approaches to address endogenous opioid dysregulation in MDD may yield novel therapeutics that have a low or absent risk of abuse and addiction relative to µ-opioid agonists. Moreover, agents targeting the endogenous opioid system would be expected to yield clinical benefits qualitatively different from conventional monaminergic antidepressants. The development of safe and effective agents to treat MDD-associated endogenous opioid dysregulation may represent a distinct and currently underappreciated means of addressing treatment resistant depression with the potential to attenuate the on-going opioid crisis.
Collapse
Affiliation(s)
- Marta Peciña
- 0000 0004 1936 9000grid.21925.3dDepartment of Psychiatry, University of Pittsburgh, Pittsburgh, PA USA
| | - Jordan F. Karp
- 0000 0004 1936 9000grid.21925.3dDepartment of Psychiatry, University of Pittsburgh, Pittsburgh, PA USA
| | - Sanjay Mathew
- 0000 0001 2160 926Xgrid.39382.33Menninger Department of Psychiatry & Behavioral Sciences, Baylor College of Medicine, Houston, TX USA
| | | | | | - Jon-Kar Zubieta
- Department of Psychiatry, University of Utah Health Sciences Center, Salt Lake City, UT, USA.
| |
Collapse
|
21
|
Yang L, Brooks AF, Makaravage KJ, Zhang H, Sanford MS, Scott PJH, Shao X. Radiosynthesis of [ 11C]LY2795050 for Preclinical and Clinical PET Imaging Using Cu(II)-Mediated Cyanation. ACS Med Chem Lett 2018; 9:1274-1279. [PMID: 30613339 DOI: 10.1021/acsmedchemlett.8b00460] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 11/13/2018] [Indexed: 01/05/2023] Open
Abstract
Copper-mediated 11C-cyanation reactions have enabled the synthesis of PET radiotracers from a range of readily available precursors and avoid the need to use more toxic Pd catalysts. In this work we adapt our recently developed 11C-cyanation of arylpinacolboronate (BPin) esters for the cGMP synthesis of [11C]LY2795050, a selective antagonist radiotracer for the kappa opioid receptor (KOR). [11C]LY2795050 was synthesized in 6 ± 1% noncorrected radiochemical yield (based on [11C]HCN, n = 3) using an automated synthesis module. Quality control testing confirmed the suitability of doses for preclinical and clinical PET imaging (radiochemical purity >99%; specific activity >900 mCi/μmol; residual Cu < 0.1 μg/mL). PET imaging was conducted in rodent and nonhuman primates, showing good brain uptake of [11C]LY2795050 and the expected distribution of KOR. Analogous imaging with [11C]carfentanil (a selective mu opioid receptor (MOR) radiotracer) revealed the anticipated regional differences in MOR and KOR distribution in the primate brain.
Collapse
Affiliation(s)
- Lingyun Yang
- Department of Radiology, University of Michigan, Ann Arbor, Michigan 48109, United States
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Allen F. Brooks
- Department of Radiology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Katarina J. Makaravage
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Huibin Zhang
- Center of Drug Discovery, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Melanie S. Sanford
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Peter J. H. Scott
- Department of Radiology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Xia Shao
- Department of Radiology, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
22
|
Vijay A, Cavallo D, Goldberg A, de Laat B, Nabulsi N, Huang Y, Krishnan-Sarin S, Morris ED. PET imaging reveals lower kappa opioid receptor availability in alcoholics but no effect of age. Neuropsychopharmacology 2018; 43:2539-2547. [PMID: 30188515 PMCID: PMC6224533 DOI: 10.1038/s41386-018-0199-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 08/01/2018] [Accepted: 08/08/2018] [Indexed: 11/24/2022]
Abstract
Opioid receptors are implicated in alcoholism, other addictions, withdrawal, and depression, and are considered potential pharmacological targets for treatment. Our goal in the present study was to compare the availability of kappa opioid receptors (KOR) between an alcohol-dependent cohort (AD) and a healthy control cohort (HC). Sixty-four participants-36 AD and 28 HC-underwent PET scans with [11C]LY2795050, a selective kappa antagonist tracer. Partial-volume correction was applied to all PET data to correct for atrophy. Volume of distribution (VT) of the tracer was estimated regionally as a measure of KOR availability. VT values of AD versus HC were compared for 15 defined ROIs. Multivariate analysis showed a main effect of group on VT across these 15 ROIs. Post hoc tests showed that AD had significantly lower VT and thus a lower KOR availability than HC in amygdala and pallidum (corrected for multiple comparisons). Exploratory analysis of change in VT with age was conducted; VT was not found to vary significantly with age in any region. Our findings of lower VT in AD versus HC in multiple regions are in contrast to findings in the mu and delta opioid receptor systems of higher VT in AD versus HC. Although age-related decline in receptors has previously been observed in the mu opioid receptor system, we found that KOR availability does not change with age.
Collapse
Affiliation(s)
- Aishwarya Vijay
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Dana Cavallo
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Alissa Goldberg
- Department of Psychiatry, Yale University, New Haven, CT, USA
| | - Bart de Laat
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Nabeel Nabulsi
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA
| | | | - Evan D Morris
- Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT, USA.
- Department of Psychiatry, Yale University, New Haven, CT, USA.
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA.
| |
Collapse
|
23
|
Finnema SJ, Nabulsi NB, Mercier J, Lin SF, Chen MK, Matuskey D, Gallezot JD, Henry S, Hannestad J, Huang Y, Carson RE. Kinetic evaluation and test-retest reproducibility of [ 11C]UCB-J, a novel radioligand for positron emission tomography imaging of synaptic vesicle glycoprotein 2A in humans. J Cereb Blood Flow Metab 2018; 38:2041-2052. [PMID: 28792356 PMCID: PMC6259313 DOI: 10.1177/0271678x17724947] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2016] [Revised: 07/07/2017] [Accepted: 07/09/2017] [Indexed: 11/15/2022]
Abstract
Synaptic vesicle glycoprotein 2A (SV2A) is ubiquitously present in presynaptic terminals. Here we report kinetic modeling and test-retest reproducibility assessment of the SV2A positron emission tomography (PET) radioligand [11C]UCB-J in humans. Five volunteers were examined twice on the HRRT after bolus injection of [11C]UCB-J. Arterial blood samples were collected for measurements of radiometabolites and free fraction. Regional time-activity curves were analyzed with 1-tissue (1T) and 2-tissue (2T) compartment models to estimate volumes of distribution ( VT). Parametric maps were generated using the 1T model. [11C]UCB-J metabolized fairly quickly, with parent fraction of 36 ± 13% at 15 min after injection. Plasma free fraction was 32 ± 1%. Regional time-activity curves displayed rapid kinetics and were well described by the 1T model, except for the cerebellum and hippocampus. VT values estimated with the 2T model were similar to 1T values. Parametric maps were of high quality and VT values correlated well with time activity curve (TAC)-based estimates. Shortening of acquisition time from 120 min to 60 min had a negligible effect on VT values. The mean absolute test-retest reproducibility for VT was 3-9% across regions. In conclusion, [11C]UCB-J exhibited excellent PET tracer characteristics and has potential as a general purpose tool for measuring synaptic density in neurodegenerative disorders.
Collapse
Affiliation(s)
- Sjoerd J Finnema
- Department of Radiology and Biomedical
Imaging, Yale Positron Emission Tomography Center,
Yale
University, New Haven, CT, USA
| | - Nabeel B Nabulsi
- Department of Radiology and Biomedical
Imaging, Yale Positron Emission Tomography Center,
Yale
University, New Haven, CT, USA
| | | | - Shu-fei Lin
- Department of Radiology and Biomedical
Imaging, Yale Positron Emission Tomography Center,
Yale
University, New Haven, CT, USA
| | - Ming-Kai Chen
- Department of Radiology and Biomedical
Imaging, Yale Positron Emission Tomography Center,
Yale
University, New Haven, CT, USA
| | - David Matuskey
- Department of Radiology and Biomedical
Imaging, Yale Positron Emission Tomography Center,
Yale
University, New Haven, CT, USA
| | - Jean-Dominique Gallezot
- Department of Radiology and Biomedical
Imaging, Yale Positron Emission Tomography Center,
Yale
University, New Haven, CT, USA
| | - Shannan Henry
- Department of Radiology and Biomedical
Imaging, Yale Positron Emission Tomography Center,
Yale
University, New Haven, CT, USA
| | | | - Yiyun Huang
- Department of Radiology and Biomedical
Imaging, Yale Positron Emission Tomography Center,
Yale
University, New Haven, CT, USA
| | - Richard E Carson
- Department of Radiology and Biomedical
Imaging, Yale Positron Emission Tomography Center,
Yale
University, New Haven, CT, USA
- Department of Biomedical Engineering,
Yale
University, New Haven, CT, USA
| |
Collapse
|
24
|
Zanderigo F, D’Agostino AE, Joshi N, Schain M, Kumar D, Parsey RV, DeLorenzo C, Mann JJ. [11C]Harmine Binding to Brain Monoamine Oxidase A: Test-Retest Properties and Noninvasive Quantification. Mol Imaging Biol 2018; 20:667-681. [DOI: 10.1007/s11307-018-1165-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
25
|
Quantitative PET Imaging in Drug Development: Estimation of Target Occupancy. Bull Math Biol 2017; 81:3508-3541. [PMID: 29230702 DOI: 10.1007/s11538-017-0374-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 11/27/2017] [Indexed: 01/13/2023]
Abstract
Positron emission tomography, an imaging tool using radiolabeled tracers in humans and preclinical species, has been widely used in recent years in drug development, particularly in the central nervous system. One important goal of PET in drug development is assessing the occupancy of various molecular targets (e.g., receptors, transporters, enzymes) by exogenous drugs. The current linear mathematical approaches used to determine occupancy using PET imaging experiments are presented. These algorithms use results from multiple regions with different target content in two scans, a baseline (pre-drug) scan and a post-drug scan. New mathematical estimation approaches to determine target occupancy, using maximum likelihood, are presented. A major challenge in these methods is the proper definition of the covariance matrix of the regional binding measures, accounting for different variance of the individual regional measures and their nonzero covariance, factors that have been ignored by conventional methods. The novel methods are compared to standard methods using simulation and real human occupancy data. The simulation data showed the expected reduction in variance and bias using the proper maximum likelihood methods, when the assumptions of the estimation method matched those in simulation. Between-method differences for data from human occupancy studies were less obvious, in part due to small dataset sizes. These maximum likelihood methods form the basis for development of improved PET covariance models, in order to minimize bias and variance in PET occupancy studies.
Collapse
|
26
|
Zanderigo F, Kang Y, Kumar D, Nikolopoulou A, Mozley PD, Kothari PJ, He B, Schlyer D, Rapoport SI, Oquendo MA, Vallabhajosula S, Mann JJ, Sublette ME. [ 11 C]arachidonic acid incorporation measurement in human brain: Optimization for clinical use. Synapse 2017; 72. [PMID: 29144569 DOI: 10.1002/syn.22018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 11/10/2017] [Accepted: 11/12/2017] [Indexed: 01/06/2023]
Abstract
Arachidonic acid (AA) is involved in signal transduction, neuroinflammation, and production of eicosanoid metabolites. The AA brain incorporation coefficient (K*) is quantifiable in vivo using [11 C]AA positron emission tomography, although repeatability remains undetermined. We evaluated K* estimates obtained with population-based metabolite correction (PBMC) and image-derived input function (IDIF) in comparison to arterial blood-based estimates, and compared repeatability. Eleven healthy volunteers underwent a [11 C]AA scan; five repeated the scan 6 weeks later, simulating a pre- and post-treatment study design. For all scans, arterial blood was sampled to measure [11 C]AA plasma radioactivity. Plasma [11 C]AA parent fraction was measured in 5 scans. K* was quantified using both blood data and IDIF, corrected for [11 C]AA parent fraction using both PBMC (from published values) and individually measured values (when available). K* repeatability was calculated in the test-retest subset. K* estimates based on blood and individual metabolites were highly correlated with estimates using PBMC with arterial input function (r = 0.943) or IDIF (r = 0.918) in the subset with measured metabolites. In the total dataset, using PBMC, IDIF-based estimates were moderately correlated with arterial input function-based estimates (r = 0.712). PBMC and IDIF-based K* estimates were ∼6.4% to ∼11.9% higher, on average, than blood-based estimates. Average K* test-retest absolute percent difference values obtained using blood data or IDIF, assuming PBMC for both, were between 6.7% and 13.9%, comparable to other radiotracers. Our results support the possibility of simplified [11 C]AA data acquisition through eliminating arterial blood sampling and metabolite analysis, while retaining comparable repeatability and validity.
Collapse
Affiliation(s)
- Francesca Zanderigo
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York.,Department of Psychiatry, Columbia University, New York, New York
| | - Yeona Kang
- Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Dileep Kumar
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York
| | | | - P David Mozley
- Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Paresh J Kothari
- Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Bin He
- Department of Radiology, Weill Cornell Medicine, New York, New York
| | - David Schlyer
- Department of Radiology, Weill Cornell Medicine, New York, New York
| | | | - Maria A Oquendo
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York.,Department of Psychiatry, Columbia University, New York, New York
| | | | - J John Mann
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York.,Department of Psychiatry, Columbia University, New York, New York.,Department of Radiology, Columbia University, New York, New York
| | - M Elizabeth Sublette
- Molecular Imaging and Neuropathology Division, New York State Psychiatric Institute, New York, New York.,Department of Psychiatry, Columbia University, New York, New York
| |
Collapse
|
27
|
Li S, Cai Z, Zheng MQ, Holden D, Naganawa M, Lin SF, Ropchan J, Labaree D, Kapinos M, Lara-Jaime T, Navarro A, Huang Y. Novel 18F-Labeled κ-Opioid Receptor Antagonist as PET Radiotracer: Synthesis and In Vivo Evaluation of 18F-LY2459989 in Nonhuman Primates. J Nucl Med 2017; 59:140-146. [PMID: 28747521 DOI: 10.2967/jnumed.117.195586] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 06/28/2017] [Indexed: 01/02/2023] Open
Abstract
The κ-opioid receptor (KOR) has been implicated in depression, addictions, and other central nervous system disorders and, thus, is an important target for drug development. We previously developed several 11C-labeled PET radiotracers for KOR imaging in humans. Here we report the synthesis and evaluation of 18F-LY2459989 as the first 18F-labeled KOR antagonist radiotracer in nonhuman primates and its comparison with 11C-LY2459989. Methods: The novel radioligand 18F-LY2459989 was synthesized by 18F displacement of a nitro group or an iodonium ylide. PET scans in rhesus monkeys were obtained on a small-animal scanner to assess the pharmacokinetic and in vivo binding properties of the ligand. Metabolite-corrected arterial activity curves were measured and used as input functions in the analysis of brain time-activity curves and the calculation of binding parameters. Results: With the iodonium ylide precursor, 18F-LY2459989 was prepared at high radiochemical yield (36% ± 7% [mean ± SD]), radiochemical purity (>99%), and mean molar activity (1,175 GBq/μmol; n = 6). In monkeys, 18F-LY2459989 was metabolized at a moderate rate, with a parent fraction of approximately 35% at 30 min after injection. Fast and reversible kinetics were observed, with a regional peak uptake time of less than 20 min. Pretreatment with the selective KOR antagonist LY2456302 (0.1 mg/kg) decreased the activity level in regions with high levels of binding to that in the cerebellum, thus demonstrating the binding specificity and selectivity of 18F-LY2459989 in vivo. Regional time-activity curves were well fitted by the multilinear analysis 1 kinetic model to derive reliable estimates of regional distribution volumes. With the cerebellum as the reference region, regional binding potentials were calculated and ranked as follows: cingulate cortex > insula > caudate/putamen > frontal cortex > temporal cortex > thalamus, consistent with the reported KOR distribution in the monkey brain. Conclusion: The evaluation of 18F-LY2459989 in nonhuman primates demonstrated many attractive imaging properties: fast tissue kinetics, specific and selective binding to the KOR, and high specific binding signals. A side-by-side comparison of 18F-LY2459989 and 11C-LY2459989 indicated similar kinetic and binding profiles for the 2 radiotracers. Taken together, the results indicated that 18F-LY2459989 appears to be an excellent PET radiotracer for the imaging and quantification of the KOR in vivo.
Collapse
Affiliation(s)
- Songye Li
- PET Center, Yale University School of Medicine, New Haven, Connecticut; and
| | - Zhengxin Cai
- PET Center, Yale University School of Medicine, New Haven, Connecticut; and
| | - Ming-Qiang Zheng
- PET Center, Yale University School of Medicine, New Haven, Connecticut; and
| | - Daniel Holden
- PET Center, Yale University School of Medicine, New Haven, Connecticut; and
| | - Mika Naganawa
- PET Center, Yale University School of Medicine, New Haven, Connecticut; and
| | - Shu-Fei Lin
- PET Center, Yale University School of Medicine, New Haven, Connecticut; and
| | - Jim Ropchan
- PET Center, Yale University School of Medicine, New Haven, Connecticut; and
| | - David Labaree
- PET Center, Yale University School of Medicine, New Haven, Connecticut; and
| | - Michael Kapinos
- PET Center, Yale University School of Medicine, New Haven, Connecticut; and
| | - Teresa Lara-Jaime
- PET Center, Yale University School of Medicine, New Haven, Connecticut; and
| | | | - Yiyun Huang
- PET Center, Yale University School of Medicine, New Haven, Connecticut; and
| |
Collapse
|
28
|
Abstract
This paper is the thirty-eighth consecutive installment of the annual review of research concerning the endogenous opioid system. It summarizes papers published during 2015 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides, opioid receptors, opioid agonists and opioid antagonists. The particular topics that continue to be covered include the molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors related to behavior, and the roles of these opioid peptides and receptors in pain and analgesia, stress and social status, tolerance and dependence, learning and memory, eating and drinking, drug abuse and alcohol, sexual activity and hormones, pregnancy, development and endocrinology, mental illness and mood, seizures and neurologic disorders, electrical-related activity and neurophysiology, general activity and locomotion, gastrointestinal, renal and hepatic functions, cardiovascular responses, respiration and thermoregulation, and immunological responses.
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, Flushing, NY 11367, United States.
| |
Collapse
|
29
|
Cai Z, Li S, Pracitto R, Navarro A, Shirali A, Ropchan J, Huang Y. Fluorine-18-Labeled Antagonist for PET Imaging of Kappa Opioid Receptors. ACS Chem Neurosci 2017; 8:12-16. [PMID: 27741398 DOI: 10.1021/acschemneuro.6b00268] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Kappa opioid receptor (KOR) antagonists are potential drug candidates for diseases such as treatment-refractory depression, anxiety, and addictive disorders. PET imaging radiotracers for KOR can be used in occupancy study to facilitate drug development, and to investigate the roles of KOR in health and diseases. We have previously developed two 11C-labeled antagonist radiotracers with high affinity and selectivity toward KOR. What is limiting their wide applications is the short half-life of 11C. Herein, we report the synthesis of a first 18F-labeled KOR antagonist radiotracer and the initial PET imaging study in a nonhuman primate.
Collapse
Affiliation(s)
- Zhengxin Cai
- PET
Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Songye Li
- PET
Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Richard Pracitto
- PET
Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Antonio Navarro
- Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Anupama Shirali
- PET
Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Jim Ropchan
- PET
Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| | - Yiyun Huang
- PET
Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, Connecticut 06520, United States
| |
Collapse
|
30
|
Schmitt S, Delamare J, Tirel O, Fillesoye F, Dhilly M, Perrio C. N-[ 18F]-FluoropropylJDTic for κ-opioid receptor PET imaging: Radiosynthesis, pre-clinical evaluation, and metabolic investigation in comparison with parent JDTic. Nucl Med Biol 2016; 44:50-61. [PMID: 27821345 DOI: 10.1016/j.nucmedbio.2016.09.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 09/07/2016] [Accepted: 09/17/2016] [Indexed: 02/08/2023]
Abstract
INTRODUCTION To image kappa opioid receptor (KOR) for preclinical studies, N-fluoropropylJDTic 9 derived from the best-established KOR antagonist JDTic, was labeled with fluorine-18. METHODS Radiosynthesis of [18F]9 was achieved according to an automated two-step procedure from [18F]-fluoride. Peripheral and cerebral distributions were determined by ex vivo experiments and by PET imaging in mouse. Radiometabolism studies were performed both in vivo in mice and in vitro in mouse and human liver microsomes. Identification of the major metabolic fragmentations was carried out by UPLC-MS analysis of enzymatic cleavage of non-radioactive ligand 9. Microsomal metabolic degradation of parent JDTic was also achieved for comparison. RESULTS The radiotracer [18F]9 was produced after 140±5min total synthesis time (2.2±0.4% not decay corrected radiochemical yield) with a specific activity of 41-89GBq/μmol (1.1-2.4Ci/μmol). Peripheral and regional brain distributions of [18F]9 were consistent with known KOR locations but no significant specific binding in brain was shown. [18F]9 presented a typical hepatobiliary and renal elimination, and was rapidly metabolized. The in vivo and in vitro radiometabolic profiles of [18F]9 were similar. Piperidine 12 was identified as the major metabolic fragment of the non-radioactive ligand 9. JDTic 7 was found to be much more stable than 9. CONCLUSION Although the newly proposed radioligand [18F]9 was concluded to be not suitable for KOR PET imaging due to the formation of brain penetrating radiometabolites, our findings highlight the metabolic stability of JDTic and may help in the design of novel JDTic derivatives for in vivo applications.
Collapse
Affiliation(s)
- Sébastien Schmitt
- Normandie Univ, UNICAEN, CEA, CNRS, UMR6301-ISTCT, LDM-TEP, Cyceron, Boulevard Henri Becquerel, 14000, Caen, France
| | - Jérôme Delamare
- Normandie Univ, UNICAEN, CEA, CNRS, UMR6301-ISTCT, LDM-TEP, Cyceron, Boulevard Henri Becquerel, 14000, Caen, France
| | - Olivier Tirel
- Normandie Univ, UNICAEN, CEA, CNRS, UMR6301-ISTCT, LDM-TEP, Cyceron, Boulevard Henri Becquerel, 14000, Caen, France
| | - Fabien Fillesoye
- Normandie Univ, UNICAEN, CEA, CNRS, UMR6301-ISTCT, LDM-TEP, Cyceron, Boulevard Henri Becquerel, 14000, Caen, France
| | - Martine Dhilly
- Normandie Univ, UNICAEN, CEA, CNRS, UMR6301-ISTCT, LDM-TEP, Cyceron, Boulevard Henri Becquerel, 14000, Caen, France
| | - Cécile Perrio
- Normandie Univ, UNICAEN, CEA, CNRS, UMR6301-ISTCT, LDM-TEP, Cyceron, Boulevard Henri Becquerel, 14000, Caen, France.
| |
Collapse
|
31
|
Rotstein BH, Liang SH, Placzek MS, Hooker JM, Gee AD, Dollé F, Wilson AA, Vasdev N. (11)C[double bond, length as m-dash]O bonds made easily for positron emission tomography radiopharmaceuticals. Chem Soc Rev 2016; 45:4708-26. [PMID: 27276357 PMCID: PMC5000859 DOI: 10.1039/c6cs00310a] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The positron-emitting radionuclide carbon-11 ((11)C, t1/2 = 20.3 min) possesses the unique potential for radiolabeling of any biological, naturally occurring, or synthetic organic molecule for in vivo positron emission tomography (PET) imaging. Carbon-11 is most often incorporated into small molecules by methylation of alcohol, thiol, amine or carboxylic acid precursors using [(11)C]methyl iodide or [(11)C]methyl triflate (generated from [(11)C]carbon dioxide or [(11)C]methane). Consequently, small molecules that lack an easily substituted (11)C-methyl group are often considered to have non-obvious strategies for radiolabeling and require a more customized approach. [(11)C]Carbon dioxide itself, [(11)C]carbon monoxide, [(11)C]cyanide, and [(11)C]phosgene represent alternative reactants to enable (11)C-carbonylation. Methodologies developed for preparation of (11)C-carbonyl groups have had a tremendous impact on the development of novel PET tracers and provided key tools for clinical research. (11)C-Carbonyl radiopharmaceuticals based on labeled carboxylic acids, amides, carbamates and ureas now account for a substantial number of important imaging agents that have seen translation to higher species and clinical research of previously inaccessible targets, which is a testament to the creativity, utility and practicality of the underlying radiochemistry.
Collapse
Affiliation(s)
| | - Steven H Liang
- Massachusetts General Hospital, Harvard Medical School, Boston, USA.
| | - Michael S Placzek
- Athinoula A. Martinos Center for Biomedical Imaging, MGH, HMS, Charlestown, USA and McLean Hospital, Belmont, USA
| | - Jacob M Hooker
- Athinoula A. Martinos Center for Biomedical Imaging, MGH, HMS, Charlestown, USA
| | | | - Frédéric Dollé
- CEA - Institut d'imagerie biomédicale, Service hospitalier Frédéric Joliot, Université Paris-Saclay, Orsay, France
| | - Alan A Wilson
- Centre for Addiction and Mental Health, Toronto, Canada
| | - Neil Vasdev
- Massachusetts General Hospital, Harvard Medical School, Boston, USA.
| |
Collapse
|
32
|
Vijay A, Wang S, Worhunsky P, Zheng MQ, Nabulsi N, Ropchan J, Krishnan-Sarin S, Huang Y, Morris ED. PET imaging reveals sex differences in kappa opioid receptor availability in humans, in vivo. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2016; 6:205-214. [PMID: 27648372 PMCID: PMC5004062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 05/17/2016] [Indexed: 06/06/2023]
Abstract
Opioid receptors may play critical roles in alcoholism and other addictions, addiction withdrawal, and depression and are considered pharmacological targets for treatment of these conditions. Sex differences have been demonstrated in mu (MOR) and delta (DOR) opioid receptors in humans, in vivo. In addition, sex differences have been observed in efficacy of treatment targeting kappa opioid receptors (KOR). Our goal in the present study was to compare the availability of KOR (1) between healthy control (HC) men and women. Twenty-seven subjects-18 males (M) and 9 females (F)-underwent PET scans with [(11)C] LY2795050, a selective kappa antagonist tracer. Partial volume correction was applied to all PET data. Volume of distribution (V T) of the tracer was estimated regionally as well as at the voxel level. V T values of males versus females were compared for 19 defined ROIs. Results at the regional and voxel levels were consistent. Males had significantly higher V T and thus a higher KOR availability than women in multiple brain regions. To our knowledge, this is the first report of sex differences in the KOR system in humans, in vivo. These findings could have implications for the treatment of pain with kappa opioid analgesics. The results may also have an impact on the diagnosis and treatment of addictive and other disorders.
Collapse
Affiliation(s)
- Aishwarya Vijay
- Department of Radiology and Biomedical Imaging, Yale Universit New Haven, CT, USA
| | - Shuo Wang
- Department of Radiology and Biomedical Imaging, Yale UniversitNew Haven, CT, USA; Department of Biomedical Engineering, Yale UniversityNew Haven, CT, USA
| | - Patrick Worhunsky
- Department of Radiology and Biomedical Imaging, Yale Universit New Haven, CT, USA
| | - Ming-Qiang Zheng
- Department of Radiology and Biomedical Imaging, Yale Universit New Haven, CT, USA
| | - Nabeel Nabulsi
- Department of Radiology and Biomedical Imaging, Yale Universit New Haven, CT, USA
| | - Jim Ropchan
- Department of Radiology and Biomedical Imaging, Yale Universit New Haven, CT, USA
| | | | - Yiyun Huang
- Department of Radiology and Biomedical Imaging, Yale Universit New Haven, CT, USA
| | - Evan D Morris
- Department of Radiology and Biomedical Imaging, Yale UniversitNew Haven, CT, USA; Department of Biomedical Engineering, Yale UniversityNew Haven, CT, USA; Department of Psychiatry, Yale UniversityNew Haven, CT, USA
| |
Collapse
|
33
|
Naganawa M, Dickinson GL, Zheng MQ, Henry S, Vandenhende F, Witcher J, Bell R, Nabulsi N, Lin SF, Ropchan J, Neumeister A, Ranganathan M, Tauscher J, Huang Y, Carson RE. Receptor Occupancy of the κ-Opioid Antagonist LY2456302 Measured with Positron Emission Tomography and the Novel Radiotracer 11C-LY2795050. J Pharmacol Exp Ther 2016; 356:260-6. [PMID: 26628406 PMCID: PMC4727157 DOI: 10.1124/jpet.115.229278] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 11/30/2015] [Indexed: 11/22/2022] Open
Abstract
The κ-opioid receptor (KOR) is thought to play an important therapeutic role in a wide range of neuropsychiatric and substance abuse disorders, including alcohol dependence. LY2456302 is a recently developed KOR antagonist with high affinity and selectivity and showed efficacy in the suppression of ethanol consumption in rats. This study investigated brain penetration and KOR target engagement after single oral doses (0.5-25 mg) of LY2456302 in 13 healthy human subjects. Three positron emission tomography scans with the KOR antagonist radiotracer (11)C-LY2795050 were conducted at baseline, 2.5 hours postdose, and 24 hours postdose. LY2456302 was well tolerated in all subjects without serious adverse events. Distribution volume was estimated using the multilinear analysis 1 method for each scan. Receptor occupancy (RO) was derived from a graphical occupancy plot and related to LY2456302 plasma concentration to determine maximum occupancy (rmax) and IC50. LY2456302 dose dependently blocked the binding of (11)C-LY2795050 and nearly saturated the receptors at 10 mg, 2.5 hours postdose. Thus, a dose of 10 mg of LY2456302 appears well suited for further clinical testing. Based on the pharmacokinetic (PK)-RO model, the rmax and IC50 of LY2456302 were estimated as 93% and 0.58 ng/ml to 0.65 ng/ml, respectively. Assuming that rmax is 100%, IC50 was estimated as 0.83 ng/ml.
Collapse
Affiliation(s)
- Mika Naganawa
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| | - Gemma L Dickinson
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| | - Ming-Qiang Zheng
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| | - Shannan Henry
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| | - Francois Vandenhende
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| | - Jennifer Witcher
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| | - Robert Bell
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| | - Nabeel Nabulsi
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| | - Shu-Fei Lin
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| | - Jim Ropchan
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| | - Alexander Neumeister
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| | - Mohini Ranganathan
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| | - Johannes Tauscher
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| | - Yiyun Huang
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| | - Richard E Carson
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut (M.N., M.Z., S.H., N.N., S.L., J.R., Y.H., R.C.); Eli Lilly and Company, Indianapolis, Indiana (G.D., J.W., R.B., J.T.); ClinBAY, Belgium (F.V.); and Department of Psychiatry, Yale University School of Medicine, New Haven, Connecticut (A.N., M.R.)
| |
Collapse
|
34
|
Immediate and Persistent Effects of Salvinorin A on the Kappa Opioid Receptor in Rodents, Monitored In Vivo with PET. Neuropsychopharmacology 2015; 40:2865-72. [PMID: 26058662 PMCID: PMC4864638 DOI: 10.1038/npp.2015.159] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 05/18/2015] [Accepted: 06/02/2015] [Indexed: 11/08/2022]
Abstract
Monitoring changes in opioid receptor binding with positron emission tomography (PET) could lead to a better understanding of tolerance and addiction because altered opioid receptor dynamics following agonist exposure has been linked to tolerance mechanisms. We have studied changes in kappa opioid receptor (KOR) binding availability in vivo with PET following kappa opioid agonist administration. Male Sprague-Dawley rats (n=31) were anesthetized and treated with the (KOR) agonist salvinorin A (0.01-1.8 mg/kg, i.v.) before administration of the KOR selective radiotracer [(11)C]GR103545. When salvinorin A was administered 1 min prior to injection of the radiotracer, [(11)C]GR103545 binding potential (BPND) was decreased in a dose-dependent manner, indicating receptor binding competition. In addition, the unique pharmacokinetics of salvinorin A (half-life ~8 min in non-human primates) allowed us to study the residual impact on KOR after the drug had eliminated from the brain. Salvinorin A was administered up to 5 h prior to [(11)C]GR103545, and the changes in BPND were compared with baseline, 2.5 h, 1 h, and 1 min pretreatment times. At lower doses (0.18 mg/kg and 0.32 mg/kg) we observed no prolonged effect on KOR binding but at 0.60 mg/kg salvinorin A induced a sustained decrease in KOR binding (BPND decreased by 40-49%) which persisted up to 2.5 h post administration, long after salvinorin A had been eliminated from the brain. These data point towards an agonist-induced adaptive response by KOR, the dynamics of which have not been previously studied in vivo with PET.
Collapse
|