1
|
Dubroff JG, Hsieh CJ, Wiers CE, Lee H, Schmitz A, Li EJ, Schubert EK, Mach RH, Kranzler HR. [ 11C]Carfentanil PET Whole-Body Imaging of μ-Opioid Receptors: A First in-Human Study. J Nucl Med 2025:jnumed.124.269413. [PMID: 40341091 DOI: 10.2967/jnumed.124.269413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 04/08/2025] [Indexed: 05/10/2025] Open
Abstract
μ-opioid receptors (MORs) are G-coupled receptors widely expressed in the brain and body. MORs have a high affinity for both endogenous opioids such as β-endorphins and exogenous opioids such as fentanyl. They mediate pain and reward and have been implicated in the pathophysiology of opioid, cocaine, and other substance use disorders. Using an instrument with a long axial field of view and the MOR-selective radioligand [11C]carfentanil, we measured the whole-body distribution of MORs in 13 healthy humans. We also examined sex differences in MOR distribution at baseline and after pretreatment with the MOR antagonist naloxone. Methods: Six female and 7 male healthy subjects underwent 2 [11C]carfentanil PET imaging sessions-one at baseline and one immediately after pretreatment with the MOR antagonist naloxone (13 μg/kg). Whole-body PET imaging was performed on an instrument with a 142-cm axial bore. [11C]carfentanil brain distribution volume ratios were determined using the occipital cortex and the visual cortex within it as reference regions. For peripheral organ distribution volume ratios, the descending aorta and proximal-extremity muscle (biceps/triceps) were used as reference regions. Results: Naloxone blockade reduced MOR availability by 40%-50% in the caudate, putamen, thalamus, amygdala, and ventral tegmentum, brain regions known to express high levels of MORs. Women showed greater receptor occupancy in the thalamus, amygdala, hippocampus, and frontal and temporal lobes and a greater naloxone-induced reduction in thalamic MOR availability than men (P < 0.05). For determining brain MOR availability, there was less variance in the visual cortex than in the occipital cortex reference region. For peripheral MOR determination, the descending aorta reference region showed less variance than the extremity muscle, but both showed blocking effects of naloxone. Conclusion: [11C]carfentanil whole-body PET scans are useful for understanding MOR physiology under both baseline and blocking conditions. Extra-central nervous system reference regions may be useful for quantifying radiotracers when a region devoid of binding in the central nervous system is unavailable. The long axial field of view was useful for measuring changes in the short-lived radiotracer [11C]carfentanil, with and without naloxone blocking. Further research is needed to evaluate the behavioral and clinical relevance of sex differences in naloxone-MOR interactions.
Collapse
Affiliation(s)
- Jacob G Dubroff
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania;
| | - Chia-Ju Hsieh
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Corinde E Wiers
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Hsiaoju Lee
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Alexander Schmitz
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Elizabeth J Li
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Erin K Schubert
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Robert H Mach
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Henry R Kranzler
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
- Mental Illness Research, Education and Clinical Center, Crescenz VAMC, Philadelphia, Pennsylvania
| |
Collapse
|
2
|
Plakas K, Hsieh CJ, Saturnino Guarino D, Hou C, Chia WK, Young A, Schmitz A, Ho YP, Weng CC, Lee H, Li S, Graham TJA, Mach RH. Toward a Small-Molecule Antagonist Radioligand for Positron Emission Tomography Imaging of the Mu Opioid Receptor. ACS Chem Neurosci 2025; 16:1592-1603. [PMID: 40156839 DOI: 10.1021/acschemneuro.5c00140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2025] Open
Abstract
The opioid crisis is a catastrophic health emergency catalyzed by the misuse of opioids that target and activate the mu opioid receptor. Many traditional radioligands used to study the mu opioid receptor are often tightly regulated owing to their abuse and respiratory depression potential. Of those that are not regulated, a lack of opioid receptor subtype selectivity can cause confounding in interpreting results. In the present study, we sought to design and characterize a library of 24 antagonist ligands for the mu opioid receptor. Ligands were evaluated for the binding affinity, intrinsic activity, and predicted blood-brain barrier permeability. Several ligands demonstrated single-digit nM binding affinity for the mu opioid receptor while also demonstrating selectivity over the delta and kappa opioid receptors. The antagonist behavior of 1A and 3A at the mu opioid receptor indicate that these ligands would likely not induce opioid-dependent respiratory depression. Therefore, these ligands can enable a safer means to interrogate the endogenous opioid system. Based on binding affinity, selectivity, and potential off-target binding, [11C]1A was prepared via metallophotoredox of the aryl-bromide functional group to [11C]methyl iodide. The nascent radioligand demonstrated brain uptake in a rhesus macaque model and accumulation in the caudate and putamen. Naloxone was able to reduce [11C]1A binding, though the interactions were not as pronounced as naloxone's ability to displace [11C]carfentanil. These results suggest that GSK1521498 and related congeners are amenable to radioligand design and can offer a safer way to query opioid neurobiology.
Collapse
Affiliation(s)
- Konstantinos Plakas
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Chia-Ju Hsieh
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Dinahlee Saturnino Guarino
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Catherine Hou
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Wai-Kit Chia
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Anthony Young
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Alexander Schmitz
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Yi-Pei Ho
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan 33302, Taiwan
- Healthy Aging Research Center, Chang Gung University, Taoyuan 33302, Taiwan
| | - Chi-Chang Weng
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan 33302, Taiwan
- Healthy Aging Research Center, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Medical Research and Development Research Division, Chang Gung Memorial Hospital Linkou, Taoyuan, Taoyuan 33305, Taiwan
| | - Hsiaoju Lee
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Shihong Li
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Thomas J A Graham
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Robert H Mach
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
3
|
Dubroff JG, Hsieh CJ, Wiers CE, Lee H, Li EJ, Schubert EK, Mach RH, Kranzler HR. [ 11 C]Carfentanil PET Whole-Body Imaging of Mu-Opioid Receptors: A First In-Human Study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2024.12.31.24319819. [PMID: 39802787 PMCID: PMC11722493 DOI: 10.1101/2024.12.31.24319819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
Introduction Mu-opioid receptors (MORs) are G-coupled protein receptors with a high affinity for both endogenous and exogenous opioids. MORs are widely expressed in the central nervous system (CNS), peripheral organs, and the immune system. They mediate pain and reward and have been implicated in the pathophysiology of opioid, cocaine, and other substance use disorders. Using the long axial field-of-view (LAFOV) PennPET Explorer instrument and the MOR selective radioligand [ 11 C]carfentanil ([ 11 C]CFN), we measured the whole-body distribution of MORs in 13 healthy humans. We also examined sex differences in MOR distribution at baseline and after pretreatment with the MOR antagonist naloxone. Methods Six female and seven male healthy subjects underwent two [ 11 C]CFN PET imaging sessions-one at baseline and one immediately following pre-treatment with the MOR antagonist naloxone (13 mcg/kg). Whole-body PET imaging was performed on the PennPET Explorer, a 142-cm axial bore instrument. [ 11 C]CFN brain distribution volume ratios (DVRs) were determined using the occipital cortex and the visual cortex within it as reference regions. For peripheral organ DVRs, the descending aorta and proximal extremity muscle (biceps/triceps) were used as reference regions. Results Naloxone blockade reduced MOR availability by 40-50% in the caudate, putamen, thalamus, amygdala, and ventral tegmentum, brain regions known to express high levels of MORs. Women showed greater receptor occupancy in the thalamus, amygdala, hippocampus and frontal and temporal lobes and a greater naloxone-induced reduction in thalamic MOR availability than men (p's <0.05). For determining brain MOR availability, there was less variance in the visual cortex than the occipital cortex reference region. For peripheral MOR determination, the descending aorta reference region showed less variance than the extremity muscle, but both showed blocking effects of naloxone. Conclusions [ 11 C]CFN whole- body PET scans are useful for understanding MOR physiology under both baseline and blocking conditions. Extra-CNS reference regions may be useful for quantifying radiotracers when a region devoid of binding in the CNS is unavailable. The LAFOV PET instrument was useful for measuring changes in the short-lived radiotracer [ 11 C]CFN, with and without naloxone blocking. Further research is needed to evaluate the behavioral and clinical relevance of sex differences in naloxone-MOR interactions.
Collapse
|
4
|
Hsieh CJ, Hou C, Lee H, Tomita C, Schmitz A, Plakas K, Dubroff JG, Mach RH. Total-body imaging of mu-opioid receptors with [ 11C]carfentanil in non-human primates. Eur J Nucl Med Mol Imaging 2024; 51:3273-3283. [PMID: 38722383 PMCID: PMC11368985 DOI: 10.1007/s00259-024-06746-2] [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: 04/29/2024] [Indexed: 06/02/2024]
Abstract
PURPOSE Mu-opioid receptors (MORs) are widely expressed in the central nervous system (CNS), peripheral organs, and immune system. This study measured the whole body distribution of MORs in rhesus macaques using the MOR selective radioligand [11C]carfentanil ([11C]CFN) on the PennPET Explorer. Both baseline and blocking studies were conducted using either naloxone or GSK1521498 to measure the effect of the antagonists on MOR binding in both CNS and peripheral organs. METHODS The PennPET Explorer was used for MOR total-body PET imaging in four rhesus macaques using [11C]CFN under baseline, naloxone pretreatment, and naloxone or GSK1521498 displacement conditions. Logan distribution volume ratio (DVR) was calculated by using a reference model to quantitate brain regions, and the standard uptake value ratios (SUVRs) were calculated for peripheral organs. The percent receptor occupancy (%RO) was calculated to establish the blocking effect of 0.14 mg/kg naloxone or GSK1521498. RESULTS The %RO in MOR-abundant brain regions was 75-90% for naloxone and 72-84% for GSK1521498 in blocking studies. A higher than 90% of %RO were observed in cervical spinal cord for both naloxone and GSK1521498. It took approximately 4-6 min for naloxone or GSK1521498 to distribute to CNS and displace [11C]CFN from the MOR. A smaller effect was observed in heart wall in the naloxone and GSK1521498 blocking studies. CONCLUSION [11C]CFN total-body PET scans could be a useful approach for studying mechanism of action of MOR drugs used in the treatment of acute and chronic opioid use disorder and their effect on the biodistribution of synthetic opioids such as CFN. GSK1521498 could be a potential naloxone alternative to reverse opioid overdose.
Collapse
Affiliation(s)
- Chia-Ju Hsieh
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - Catherine Hou
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - Hsiaoju Lee
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - Cosette Tomita
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - Alexander Schmitz
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - Konstantinos Plakas
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - Jacob G Dubroff
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA
| | - Robert H Mach
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, 19104, USA.
| |
Collapse
|
5
|
Wong CYJ, Baldelli A, Tietz O, van der Hoven J, Suman J, Ong HX, Traini D. An overview of in vitro and in vivo techniques for characterization of intranasal protein and peptide formulations for brain targeting. Int J Pharm 2024; 654:123922. [PMID: 38401871 DOI: 10.1016/j.ijpharm.2024.123922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 02/15/2024] [Accepted: 02/18/2024] [Indexed: 02/26/2024]
Abstract
The surge in neurological disorders necessitates innovative strategies for delivering active pharmaceutical ingredients to the brain. The non-invasive intranasal route has emerged as a promising approach to optimize drug delivery to the central nervous system by circumventing the blood-brain barrier. While the intranasal approach offers numerous advantages, the lack of a standardized protocol for drug testing poses challenges to both in vitro and in vivo studies, limiting the accurate interpretation of nasal drug delivery and pharmacokinetic data. This review explores the in vitro experimental assays employed by the pharmaceutical industry to test intranasal formulation. The focus lies on understanding the diverse techniques used to characterize the intranasal delivery of drugs targeting the brain. Parameters such as drug release, droplet size measurement, plume geometry, deposition in the nasal cavity, aerodynamic performance and mucoadhesiveness are scrutinized for their role in evaluating the performance of nasal drug products. The review further discusses the methodology for in vivo characterization in detail, which is essential in evaluating and refining drug efficacy through the nose-to-brain pathway. Animal models are indispensable for pre-clinical drug testing, offering valuable insights into absorption efficacy and potential variables affecting formulation safety. The insights presented aim to guide future research in intranasal drug delivery for neurological disorders, ensuring more accurate predictions of therapeutic efficacy in clinical contexts.
Collapse
Affiliation(s)
- Chun Yuen Jerry Wong
- Respiratory Technology, Woolcock Institute of Medical Research, Sydney, NSW 2037, Australia; Faculty of Medicine and Health Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia
| | - Alberto Baldelli
- Faculty of Food and Land Systems, The University of British Columbia, 2357 Main Mall, Vancouver, BC V6T 1Z4, Canada
| | - Ole Tietz
- Dementia Research Centre, Faculty of Medicine and Health Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia
| | - Julia van der Hoven
- Dementia Research Centre, Faculty of Medicine and Health Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia
| | - Julie Suman
- Next Breath, an Aptar Pharma Company, Baltimore, MD 21227, USA
| | - Hui Xin Ong
- Respiratory Technology, Woolcock Institute of Medical Research, Sydney, NSW 2037, Australia; Faculty of Medicine and Health Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia.
| | - Daniela Traini
- Respiratory Technology, Woolcock Institute of Medical Research, Sydney, NSW 2037, Australia; Faculty of Medicine and Health Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia.
| |
Collapse
|
6
|
Zheng L, Zhang W, Olkkola KT, Dallmann A, Ni L, Zhao Y, Wang L, Zhang Q, Hu W. Physiologically based pharmacokinetic modeling of ritonavir-oxycodone drug interactions and its implication for dosing strategy. Eur J Pharm Sci 2024; 194:106697. [PMID: 38199444 DOI: 10.1016/j.ejps.2024.106697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 11/13/2023] [Accepted: 01/07/2024] [Indexed: 01/12/2024]
Abstract
The concomitant administration of ritonavir and oxycodone may significantly increase the plasma concentrations of oxycodone. This study was aimed to simulate DDI between ritonavir and oxycodone, a widely used opioid, and to formulate dosing protocols for oxycodone by using physiologically based pharmacokinetic (PBPK) modeling. We developed a ritonavir PBPK model incorporating induction and competitive and time-dependent inhibition of CYP3A4 and competitive inhibition of CYP2D6. The ritonavir model was evaluated with observed clinical pharmacokinetic data and validated for its CYP3A4 inhibition potency. We then used the model to simulate drug interactions between oxycodone and ritonavir under various dosing scenarios. The developed model captured the pharmacokinetic characteristics of ritonavir from clinical studies. The model also accurately predicts exposure changes of midazolam, triazolam, and oxycodone in the presence of ritonavir. According to model simulations, the steady-state maximum, minimum and average concentrations of oxycodone increased by up to 166% after co-administration with ritonavir, and the total exposure increased by approximately 120%. To achieve similar steady-state concentrations, halving the dose with an unchanged dosing interval or doubling the dosing interval with an unaltered single dose should be practical for oxycodone, whether formulated in uncoated or controlled-release tablets during long-term co-medication with ritonavir. The results revealed exposure-related risks of oxycodone-ritonavir interactions that have not been studied clinically and emphasized PBPK as a workable method to direct judicious dosage.
Collapse
Affiliation(s)
- Liang Zheng
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - Wei Zhang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Klaus T Olkkola
- Department of Anaesthesiology and Intensive Care Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.
| | - André Dallmann
- Pharmacometrics/Modeling and Simulation, Research and Development, Pharmaceuticals, Bayer AG, Leverkusen, Germany.
| | - Liang Ni
- Clinical Pharmacokinetics Laboratory, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yingjie Zhao
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Ling Wang
- Department of Clinical Pharmacy and Pharmacy Administration, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Qian Zhang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, China.
| |
Collapse
|
7
|
Smith K, Fan J, Marriner GA, Gerdes J, Kessler R, Zinn KR. Distribution of insulin in primate brain following nose-to-brain transport. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2024; 10:e12459. [PMID: 38469552 PMCID: PMC10925727 DOI: 10.1002/trc2.12459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/24/2024] [Accepted: 02/02/2024] [Indexed: 03/13/2024]
Abstract
Introduction Nose-to-brain (N2B) insulin delivery has potential for Alzheimer's disease (AD) therapy. However, clinical implementation has been challenging without methods to follow N2B delivery non-invasively. Positron emission tomography (PET) was applied to measure F-18-labeled insulin ([18F]FB-insulin) from intranasal dosing to brain uptake in non-human primates following N2B delivery. Methods [18F]FB-insulin was prepared by reacting A1,B29-di(tert-butyloxycarbonyl)insulin with [18F]-N-succinimidyl-4-fluorobenzoate. Three methods of N2B delivery for [18F]FB-insulin were compared - delivery as aerosol via tubing (rhesus macaque, n = 2), as aerosol via preplaced catheter (rhesus macaque, n = 3), and as solution via preplaced catheter (cynomolgus macaque, n = 3). Following dosing, dynamic PET imaging (120 min) quantified delivery efficiency to the nasal cavity and whole brain. Area under the time-activity curve was calculated for 46 regions of the cynomolgus macaque brain to determine regional [18F]FB-insulin levels. Results Liquid instillation of [18F]FB-insulin by catheter outperformed aerosol methods for delivery to the subject (39.89% injected dose vs 10.03% for aerosol via tubing, 0.17% for aerosol by catheter) and subsequently to brain (0.34% injected dose vs 0.00020% for aerosol via tubing, 0.05% for aerosol by catheter). [18F]FB-insulin was rapidly transferred across the cribriform plate to limbic and frontotemporal areas responsible for emotional and memory processing. [18F]FB-insulin half-life was longer in olfactory nerve projection sites with high insulin receptor density compared to the whole brain. Discussion The catheter-based liquid delivery approach combined with PET imaging successfully tracked the fate of N2B [18F]FB-insulin and is thought to be broadly applicable for assessments of other therapeutic agents. This method can be rapidly applied in humans to advance clinical evaluation of N2B insulin as an AD therapeutic. Highlights for [18F]FB-insulin passage across the cribriform plate was detected by PET.Intranasal [18F]FB-insulin reached the brain within 13 min.[18F]FB-insulin activity was highest in emotional and memory processing regions.Aerosol delivery was less efficient than liquid instillation by preplaced catheter.Insulin delivery to the cribriform plate was critical for arrival in the brain.
Collapse
Affiliation(s)
- Kylie Smith
- Department of Biomedical EngineeringMichigan State UniversityEast LansingMichiganUSA
- Institute for Quantitative Health Science and Engineering, Michigan State UniversityEast LansingMichiganUSA
| | - Jinda Fan
- Institute for Quantitative Health Science and Engineering, Michigan State UniversityEast LansingMichiganUSA
- Department of ChemistryMichigan State UniversityEast LansingMichiganUSA
- Department of RadiologyMichigan State UniversityEast LansingMichiganUSA
| | | | - John Gerdes
- Department of Biomedical and Pharmaceutical SciencesUniversity of MontanaMissoulaMontanaUSA
| | - Robert Kessler
- Department of RadiologyMichigan State UniversityEast LansingMichiganUSA
| | - Kurt R. Zinn
- Department of Biomedical EngineeringMichigan State UniversityEast LansingMichiganUSA
- Institute for Quantitative Health Science and Engineering, Michigan State UniversityEast LansingMichiganUSA
- Department of RadiologyMichigan State UniversityEast LansingMichiganUSA
- Department of Small Animal Clinical SciencesMichigan State UniversityEast LansingMichiganUSA
| |
Collapse
|
8
|
Mizutani A, Kobayashi M, Ohuchi M, Sasaki K, Muranaka Y, Torikai Y, Fukakusa S, Suzuki C, Nishii R, Haruta S, Magata Y, Kawai K. Indirect SPECT Imaging Evaluation for Possible Nose-to-Brain Drug Delivery Using a Compound with Poor Blood–Brain Barrier Permeability in Mice. Pharmaceutics 2022; 14:pharmaceutics14051026. [PMID: 35631611 PMCID: PMC9145277 DOI: 10.3390/pharmaceutics14051026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/22/2022] [Accepted: 05/07/2022] [Indexed: 02/01/2023] Open
Abstract
Single-photon emission computed tomography (SPECT) imaging using intravenous radioactive ligand administration to indirectly evaluate the time-dependent effect of intranasal drugs with poor blood-brain barrier permeability on brain drug distributions in mice was evaluated. The biodistribution was examined using domperidone, a dopamine D2 receptor ligand, as the model drug, with intranasal administration at 0, 15, or 30 min before intravenous [123I]IBZM administration. In the striatum, [123I]IBZM accumulation was significantly lower after intranasal (IN) domperidone administration than in controls 15 min after intravenous [125I]IBZM administration. [123I]IBZM SPECT was acquired with intravenous (IV) or IN domperidone administration 15 min before [123I]IBZM, and time–activity curves were obtained. In the striatum, [123I]IBZM accumulation was clearly lower in the IN group than in the control and IV groups. Time–activity curves showed no significant difference between the control and IV groups in the striatum, and values were significantly lowest during the first 10 min in the IN group. In the IN group, binding potential and % of receptor occupancy were significantly lower and higher, respectively, compared to the control and IV groups. Thus, brain-migrated domperidone inhibited D2R binding of [123I]IBZM. SPECT imaging is suitable for research to indirectly explore nose-to-brain drug delivery and locus-specific biological distribution.
Collapse
Affiliation(s)
- Asuka Mizutani
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa 920-1192, Japan; (A.M.); (M.K.); (M.O.); (Y.M.)
| | - Masato Kobayashi
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa 920-1192, Japan; (A.M.); (M.K.); (M.O.); (Y.M.)
| | - Makoto Ohuchi
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa 920-1192, Japan; (A.M.); (M.K.); (M.O.); (Y.M.)
| | - Keita Sasaki
- R&D Department, TR Company, Shin Nippon Biomedical Laboratories, Ltd., 2438 Miyanoura, Kagoshima 891-1394, Japan; (K.S.); (Y.T.); (S.F.); (S.H.)
| | - Yuka Muranaka
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa 920-1192, Japan; (A.M.); (M.K.); (M.O.); (Y.M.)
| | - Yusuke Torikai
- R&D Department, TR Company, Shin Nippon Biomedical Laboratories, Ltd., 2438 Miyanoura, Kagoshima 891-1394, Japan; (K.S.); (Y.T.); (S.F.); (S.H.)
| | - Shota Fukakusa
- R&D Department, TR Company, Shin Nippon Biomedical Laboratories, Ltd., 2438 Miyanoura, Kagoshima 891-1394, Japan; (K.S.); (Y.T.); (S.F.); (S.H.)
- Department of Molecular Imaging, Institute for Medical Photonics Research, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu 431-3192, Japan; (C.S.); (Y.M.)
| | - Chie Suzuki
- Department of Molecular Imaging, Institute for Medical Photonics Research, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu 431-3192, Japan; (C.S.); (Y.M.)
| | - Ryuichi Nishii
- Department of Molecular Imaging and Theranostics, Institute for Quantum Medical Science, Quantum Life and Medical Science Directorate, National Institutes for Quantum Science and Technology, 4-9-1 Anagawa, Inage, Chiba 263-8555, Japan;
| | - Shunji Haruta
- R&D Department, TR Company, Shin Nippon Biomedical Laboratories, Ltd., 2438 Miyanoura, Kagoshima 891-1394, Japan; (K.S.); (Y.T.); (S.F.); (S.H.)
| | - Yasuhiro Magata
- Department of Molecular Imaging, Institute for Medical Photonics Research, Preeminent Medical Photonics Education & Research Center, Hamamatsu University School of Medicine, 1-20-1 Handayama, Higashi-ku, Hamamatsu 431-3192, Japan; (C.S.); (Y.M.)
| | - Keiichi Kawai
- Faculty of Health Sciences, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, 5-11-80 Kodatsuno, Kanazawa 920-1192, Japan; (A.M.); (M.K.); (M.O.); (Y.M.)
- Biomedical Imaging Research Center, University of Fukui, 23-3 Matsuokashimoaizuki, Eiheiji, Yoshida-gun, Fukui 910-1193, Japan
- Correspondence: ; Tel.: +81-76-265-2527; Fax: +81-76-234-4366
| |
Collapse
|
9
|
Kang Y, O'Conor KA, Kelleher AC, Ramsey J, Bakhoda A, Eisenberg SM, Zhao W, Stodden T, Pearson TD, Guo M, Brown N, Liow JS, Fowler JS, Kim SW, Volkow ND. Naloxone's dose-dependent displacement of [ 11C]carfentanil and duration of receptor occupancy in the rat brain. Sci Rep 2022; 12:6429. [PMID: 35440607 PMCID: PMC9018944 DOI: 10.1038/s41598-022-09601-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 03/23/2022] [Indexed: 11/09/2022] Open
Abstract
The continuous rise in opioid overdoses in the United States is predominantly driven by very potent synthetic opioids, mostly fentanyl and its derivatives (fentanyls). Although naloxone (NLX) has been shown to effectively reverse overdoses by conventional opioids, there may be a need for higher or repeated doses of NLX to revert overdoses from highly potent fentanyls. Here, we used positron emission tomography (PET) to assess NLX's dose-dependence on both its rate of displacement of [11C]carfentanil ([11C]CFN) binding and its duration of mu opioid receptor (MOR) occupancy in the male rat brain. We showed that clinically relevant doses of intravenously (IV) administered NLX (0.035 mg/kg, Human Equivalent Dose (HED) 0.4 mg; 0.17 mg/kg, HED 2 mg) rapidly displaced the specific binding of [11C]CFN in the thalamus in a dose-dependent manner. Brain MOR occupancy by IV NLX was greater than 90% at 5 min after NLX administration for both doses, but at 27.3 min after 0.035 mg/kg dose and at 85 min after 0.17 mg/kg NLX, only 50% occupancy remained. This indicates that the duration of NLX occupancy at MORs is short-lived. Overall, these results show that clinically relevant doses of IV NLX can promptly displace fentanyls at brain MORs, but repeated or higher NLX doses may be required to prevent re-narcotization following overdoses with long-acting fentanyls.
Collapse
Affiliation(s)
- Yeona Kang
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892-1013, USA.,Department of Mathematics, Howard University, Washington, DC, 20059, USA
| | - Kelly A O'Conor
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892-1013, USA
| | - Andrew C Kelleher
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892-1013, USA
| | - Joseph Ramsey
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892-1013, USA
| | - Abolghasem Bakhoda
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892-1013, USA
| | - Seth M Eisenberg
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892-1013, USA
| | - Wenjing Zhao
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892-1013, USA
| | - Tyler Stodden
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892-1013, USA
| | - Torben D Pearson
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892-1013, USA
| | - Min Guo
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892-1013, USA
| | - Nina Brown
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892-1013, USA.,Department of Mathematics, Howard University, Washington, DC, 20059, USA
| | - Jeih-San Liow
- Molecular Imaging Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Joanna S Fowler
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892-1013, USA
| | - Sung Won Kim
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892-1013, USA.
| | - Nora D Volkow
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, 20892-1013, USA. .,National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD, 20892-1013, USA.
| |
Collapse
|
10
|
Li Y, Wu H, Jiang X, Dong Y, Zheng J, Gao J. New idea to promote the clinical applications of stem cells or their extracellular vesicles in central nervous system disorders: combining with intranasal delivery. Acta Pharm Sin B 2022; 12:3215-3232. [PMID: 35967290 PMCID: PMC9366301 DOI: 10.1016/j.apsb.2022.04.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/27/2022] [Accepted: 02/14/2022] [Indexed: 12/25/2022] Open
Abstract
The clinical translation of stem cells and their extracellular vesicles (EVs)-based therapy for central nervous system (CNS) diseases is booming. Nevertheless, the insufficient CNS delivery and retention together with the invasiveness of current administration routes prevent stem cells or EVs from fully exerting their clinical therapeutic potential. Intranasal (IN) delivery is a possible strategy to solve problems as IN route could circumvent the brain‒blood barrier non-invasively and fit repeated dosage regimens. Herein, we gave an overview of studies and clinical trials involved with IN route and discussed the possibility of employing IN delivery to solve problems in stem cells or EVs-based therapy. We reviewed relevant researches that combining stem cells or EVs-based therapy with IN administration and analyzed benefits brought by IN route. Finally, we proposed possible suggestions to facilitate the development of IN delivery of stem cells or EVs.
Collapse
Affiliation(s)
- Yaosheng Li
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Honghui Wu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xinchi Jiang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
| | - Yunfei Dong
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Juanjuan Zheng
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianqing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou 310058, China
- Hangzhou Institute of Innovative Medicine, Zhejiang University, Hangzhou 310058, China
- Corresponding author. Tel.: +86 571 88208436.
| |
Collapse
|
11
|
Goel H, Kalra V, Verma SK, Dubey SK, Tiwary AK. Convolutions in the rendition of nose to brain therapeutics from bench to bedside: Feats & fallacies. J Control Release 2021; 341:782-811. [PMID: 34906605 DOI: 10.1016/j.jconrel.2021.12.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 12/05/2021] [Accepted: 12/06/2021] [Indexed: 12/24/2022]
Abstract
Brain, a subtle organ of multifarious nature presents plethora of physiological, metabolic and bio-chemical convolutions that impede the delivery of biomolecules and thereby resulting in truncated therapeutic outcome in pathological conditions of central nervous system (CNS). The absolute bottleneck in the therapeutic management of such devastating CNS ailments is the BBB. Another pitfall is the lack of efficient technological platforms (due to high cost and low approval rates) as well as limited clinical trials (due to failures of neuro‑leads in late-stage pipelines) for CNS disorders which has become a literal brain drain with poorest success rates compared to other therapeutic areas, owing to time consuming processes, tremendous convolutions and conceivable adverse effects. With the advent of intranasal delivery (via direct N2B or indirect nose to blood to brain), several novel drug delivery carriers viz. unmodified or surface modified nanoparticle based carriers, lipid based colloidal nanocarriers and drysolid/liquid/semisolid nanoformulations or delivery platforms have been designed as a means to deliver therapeutic agents (small and large molecules, peptides and proteins, genes) to brain, bypassing BBB for disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), epilepsy, schizophrenia and CNS malignancies primarily glioblastomas. Intranasal application offers drug delivery through both direct and indirect pathways for the peripherally administered psychopharmacological agents to CNS. This route could also be exploited for the repurposing of conventional drugs for new therapeutic uses. The limited clinical translation of intranasal formulations has been primarily due to existence of barriers of mucociliary clearance in the nasal cavity, enzyme degradation and low permeability of the nasal epithelium. The present review literature aims to decipher the new paradigms of nano therapeutic systems employed for specific N2B drug delivery of CNS drugs through in silico complexation studies using rationally chosen mucoadhesive polymers (exhibiting unique physicochemical properties of nanocarrier's i.e. surface modification, prolonging retention time in the nasal cavity, improving penetration ability, and promoting brain specific delivery with biorecognitive ligands) via molecular docking simulations. Further, the review intends to delineate the feats and fallacies associated with N2B delivery approaches by understanding the physiological/anatomical considerations via decoding the intranasal drug delivery pathways or critical factors such as rationale and mechanism of excipients, affecting the permeability of CNS drugs through nasal mucosa as well as better efficacy in terms of brain targeting, brain bioavailability and time to reach the brain. Additionally, extensive emphasis has also been laid on the innovative formulations under preclinical investigation along with their assessment by means of in vitro /ex vivo/in vivo N2B models and current characterization techniques predisposing an efficient intranasal delivery of therapeutics. A critical appraisal of novel technologies, intranasal products or medical devices available commercially has also been presented. Finally, it could be warranted that more reminiscent pharmacokinetic/pharmacodynamic relationships or validated computational models are mandated to obtain effective screening of molecular architecture of drug-polymer-mucin complexes for clinical translation of N2B therapeutic systems from bench to bedside.
Collapse
Affiliation(s)
- Honey Goel
- Department of Pharmaceutics, University Institute of Pharmaceutical Sciences and Research, Baba Farid University of Health Sciences, Faridkot, Punjab, India.
| | - Vinni Kalra
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India
| | - Sant Kumar Verma
- Department of Pharmaceutical Chemistry, Indo-Soviet Friendship College of Pharmacy, Moga, Punjab, India
| | | | - Ashok Kumar Tiwary
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, India.
| |
Collapse
|
12
|
Pharmacokinetic neuroimaging to study the dose-related brain kinetics and target engagement of buprenorphine in vivo. Neuropsychopharmacology 2021; 46:1220-1228. [PMID: 33603137 PMCID: PMC8115308 DOI: 10.1038/s41386-021-00976-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/18/2020] [Accepted: 01/24/2021] [Indexed: 12/31/2022]
Abstract
A wide range of buprenorphine doses are used for either pain management or maintenance therapy in opioid addiction. The complex in vitro profile of buprenorphine, with affinity for µ-, δ-, and κ-opioid receptors (OR), makes it difficult to predict its dose-related neuropharmacology in vivo. In rats, microPET imaging and pretreatment by OR antagonists were performed to assess the binding of radiolabeled buprenorphine (microdose 11C-buprenorphine) to OR subtypes in vivo (n = 4 per condition). The µ-selective antagonist naloxonazine (10 mg/kg) and the non-selective OR antagonist naloxone (1 mg/kg) blocked the binding of 11C-buprenorphine, while pretreatment by the δ-selective (naltrindole, 3 mg/kg) or the κ-selective antagonist (norbinaltorphimine, 10 mg/kg) did not. In four macaques, PET imaging and kinetic modeling enabled description of the regional brain kinetics of 11C-buprenorphine, co-injected with increasing doses of unlabeled buprenorphine. No saturation of the brain penetration of buprenorphine was observed for doses up to 0.11 mg/kg. Regional differences in buprenorphine-associated receptor occupancy were observed. Analgesic doses of buprenorphine (0.003 and 0.006 mg/kg), respectively, occupied 20% and 49% of receptors in the thalamus while saturating the low but significant binding observed in cerebellum and occipital cortex. Occupancy >90% was achieved in most brain regions with plasma concentrations >7 µg/L. PET data obtained after co-injection of an analgesic dose of buprenorphine (0.003 mg/kg) predicted the binding potential of microdose 11C-buprenorphine. This strategy could be further combined with pharmacodynamic exploration or pharmacological MRI to investigate the neuropharmacokinetics and neuroreceptor correlate, at least at µ-OR, of the acute effects of buprenorphine in humans.
Collapse
|
13
|
O'Doherty J, Mangini CD, Hamby DM, Boozer D, Singh N, Hippeläinen E. Estimation of absorbed radiation doses to skin and S-values for organs at risk due to nasal administration of PET agents using Monte Carlo simulations. Med Phys 2021; 48:871-880. [PMID: 33330987 DOI: 10.1002/mp.14669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/29/2020] [Accepted: 12/08/2020] [Indexed: 11/11/2022] Open
Abstract
PURPOSE The intranasal (IN) administration of radiopharmaceuticals is of interest in being a viable route for the delivery of radiopharmaceuticals that do not ordinarily cross the blood-brain barrier (BBB). However, to be viable in a patient population, good image quality as well as safety of the administration should be demonstrated. This work provides radiation dosimetry calculations and simulations related to the radiation safety of performing such experiments in a human cohort. METHODS We performed Monte Carlo (MC) simulations to estimate radiation dose to the skin inside a cylindrical model of the nasal cavity assuming a homogenous distribution layer of 11 C and 18 F and calculated a geometry conversion factor (FP-C ) which can be used to convert from a planar geometry to a cylindrical geometry using more widely available software tools. We compared radiation doses from our simulated cylindrical geometry with the planar dose estimates employing our geometry conversion factor from VARSKIN 6.1 software and also from an analytical equation. Furthermore, in order to estimate radiation dosimetry to surrounding organs of interest, we performed a voxelized MC simulation of a fixed radioactivity inside the nasal cavity and calculated S-values to organs such as the eyes, thyroid, and brain. RESULTS MC simulations of contamination scenarios using planar absorbed doses of 15.50 and 8.60 mGy/MBq for 18 F and 11 C, respectively, and 35.70 and 19.80 mGy/MBq per hour for cylindrical geometries, leading to determination of an FP-C of 2.3. Planar absorbed doses (also in units of mGy/MBq) determined by the analytical equation were 16.96 and 8.68 (18 F and 11 C) and using VARSKIN were 16.60 and 9.26 (18 F and 11 C), respectively. Application of FP-C to these results demonstrates values with a maximum difference of 9.41% from the cylindrical geometry MC calculation, demonstrating that when accounting for geometry, more simplistic techniques can be utilized to estimate IN dosimetry. Voxelized MC simulations of radiation dosimetry from a fixed source of 1 MBq of activity confined to the nasal cavity resulted in S-values to the thyroid, eyes, and brain of 1.72 x 10-6 , 1.93 x 10-5 , and 3.51 x 10-6 mGy/MBq·s, respectively, for 18 F and 1.80 × 10-6 , 1.95 × 10-5 , and 3.54 × 10-6 mGy/MBq·s for 11 C. CONCLUSION Dosimetry concerns about IN administrations of PET radiotracers should be considered before clinical use. Values presented in the simulations such as the S-values can be further used for assessment of absorbed doses in cases of IN administration, and can be used to develop and adapt specific study protocols. All three presented methods provided similar results when considering the use of a geometry conversion factor for planar to cylindrical geometry, demonstrating that standard tools rather than dedicate MC simulations may be used to perform dose calculations in nasal administrations.
Collapse
Affiliation(s)
- Jim O'Doherty
- Clinical Imaging Research Centre, Centre for Translational Medicine, National University of Singapore, Singapore
| | | | - David M Hamby
- Renaissance Code Development LLC, Oregon, USA.,Department of Nuclear Science and Engineering, Oregon State University, Corvallis, USA
| | - David Boozer
- Department of Nuclear Engineering and Radiation Health Physics, Oregon State University, Corvallis, USA
| | - Nisha Singh
- Department of Psychiatry, University of Oxford, UK
| | - Eero Hippeläinen
- Department of Physics, University of Helsinki, Helsinki, Finland.,HUS Medical imaging Center, Clinical Physiology and Nuclear Medicine, University of Helsinki and Helsinki University Hospital, Finland
| |
Collapse
|
14
|
Ott J, Spilhaug MM, Maschauer S, Rafique W, Jakobsson JE, Hartvig K, Hübner H, Gmeiner P, Prante O, Riss PJ. Pharmacological Characterization of Low-to-Moderate Affinity Opioid Receptor Agonists and Brain Imaging with 18F-Labeled Derivatives in Rats. J Med Chem 2020; 63:9484-9499. [PMID: 32787100 PMCID: PMC7497404 DOI: 10.1021/acs.jmedchem.0c00683] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The 3,4-dichloro-N-(1-(dimethylamino)cyclohexyl)methyl benzamide scaffold was studied as a template for 18F-positron emission tomography (18F-PET) radiotracer development emphasizing sensitivity to changes in opioid receptor (OR) occupancy over high affinity. Agonist potency, binding affinity, and relevant pharmacological parameters of 15 candidates were investigated. Two promising compounds 3b and 3e with μ-OR (MOR) selective agonist activity in the moderate range (EC50 = 1-100 nM) were subjected to 18F-fluorination, autoradiography, and small-animal PET imaging. Radioligands [18F]3b and [18F]3e were obtained in activity yields of 21 ± 5 and 23 ± 4% and molar activities of 25-40 and 200-300 GBq/μmol, respectively. Displaceable binding matching MOR distribution in the brain was confirmed by imaging. Radioligands showed a rapid pharmacokinetic profile; however, metabolite-corrected, blood-based modeling was required for data analysis. Observed BPND was low, although treatment with naloxone leads to a marked decrease in specific binding, confirming the discovery of a new template for 18F-labeled OR-agonist PET ligands.
Collapse
Affiliation(s)
- Julian Ott
- Department of Nuclear Medicine, Molecular Imaging and Radiochemistry, Translational Research Center, Friedrich Alexander University Erlangen-Nürnberg (FAU), Schwabachanlage 12, D-91054 Erlangen, Germany
| | - Mona M Spilhaug
- Realomics SRI, Kjemisk Institutt, Universitetet i Oslo, N-0376 Oslo, Norway
| | - Simone Maschauer
- Department of Nuclear Medicine, Molecular Imaging and Radiochemistry, Translational Research Center, Friedrich Alexander University Erlangen-Nürnberg (FAU), Schwabachanlage 12, D-91054 Erlangen, Germany
| | - Waqas Rafique
- Realomics SRI, Kjemisk Institutt, Universitetet i Oslo, N-0376 Oslo, Norway
| | - Jimmy E Jakobsson
- Realomics SRI, Kjemisk Institutt, Universitetet i Oslo, N-0376 Oslo, Norway
| | - Karoline Hartvig
- Realomics SRI, Kjemisk Institutt, Universitetet i Oslo, N-0376 Oslo, Norway
| | - Harald Hübner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University Erlangen-Nürnberg (FAU), Nikolaus-Fiebiger-Straße 10, D-91058 Erlangen, Germany
| | - Peter Gmeiner
- Department of Chemistry and Pharmacy, Medicinal Chemistry, Emil Fischer Center, Friedrich Alexander University Erlangen-Nürnberg (FAU), Nikolaus-Fiebiger-Straße 10, D-91058 Erlangen, Germany
| | - Olaf Prante
- Department of Nuclear Medicine, Molecular Imaging and Radiochemistry, Translational Research Center, Friedrich Alexander University Erlangen-Nürnberg (FAU), Schwabachanlage 12, D-91054 Erlangen, Germany
| | - Patrick J Riss
- Realomics SRI, Kjemisk Institutt, Universitetet i Oslo, N-0376 Oslo, Norway.,Division of Clinical Neuroscience, Neuroscience Research Unit, OUS-UllevÅl, N-0450 Oslo, Norway
| |
Collapse
|
15
|
Moss RB, Pryor MM, Baillie R, Kudrycki K, Friedrich C, Reed M, Carlo DJ. Higher naloxone dosing in a quantitative systems pharmacology model that predicts naloxone-fentanyl competition at the opioid mu receptor level. PLoS One 2020; 15:e0234683. [PMID: 32544184 PMCID: PMC7297366 DOI: 10.1371/journal.pone.0234683] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 06/01/2020] [Indexed: 12/19/2022] Open
Abstract
Rapid resuscitation of an opioid overdose with naloxone, an opioid antagonist, is critical. We developed an opioid receptor quantitative systems pharmacology (QSP) model for evaluation of naloxone dosing. In this model we examined three opioid exposure levels that have been reported in the literature (25 ng/ml, 50 ng/ml, and 75 ng/ml of fentanyl). The model predicted naloxone-fentanyl interaction at the mu opioid receptor over a range of three naloxone doses. For a 2 mg intramuscular (IM) dose of naloxone at lower fentanyl exposure levels (25 ng/ml and 50 ng/ml), the time to decreasing mu receptor occupancy by fentanyl to 50% was 3 and 10 minutes, respectively. However, at a higher fentanyl exposure level (75 ng/ml), a dose of 2 mg IM of the naloxone failed to reduce mu receptor occupancy by fentanyl to 50%. In contrast, naloxone doses of 5 mg and 10 mg IM reduced mu receptor occupancy by fentanyl to 50% in 5.5 and 4 minutes respectively. These results suggest that the current doses of naloxone (2 mg IM or 4 mg intranasal (IN)) may be inadequate for rapid reversal of toxicity due to fentanyl exposure and that increasing the dose of naloxone is likely to improve outcomes.
Collapse
Affiliation(s)
- Ronald B. Moss
- Adamis Pharmaceuticals Corp, San Diego, CA, United States of America
- * E-mail:
| | | | | | | | | | - Mike Reed
- Rosa & Co. LLC, San Carlos, CA, United States of America
| | - Dennis J. Carlo
- Adamis Pharmaceuticals Corp, San Diego, CA, United States of America
| |
Collapse
|
16
|
Teklezgi B, Pamreddy A, Ntshangase S, Mdanda S, Singh SD, Gopal ND, Naicker T, Kruger HG, Govender T, Baijnath S. Mass Spectrometric Imaging of the Brain Demonstrates the Regional Displacement of 6-Monoacetylmorphine by Naloxone. ACS OMEGA 2020; 5:12596-12602. [PMID: 32548443 PMCID: PMC7288357 DOI: 10.1021/acsomega.9b03570] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 05/04/2020] [Indexed: 06/11/2023]
Abstract
Overdose is the main cause of mortality among heroin users. Many of these overdose-induced deaths can be prevented through the timely administration of naloxone (NLX), a nonselective mu (μ)-, kappa (κ)-, and delta (δ)-opioid receptor antagonist. NLX competitively inhibits opioid-overdose-induced respiratory depression without eliciting any narcotic effect itself. The aim of this study was to investigate the antagonistic action of NLX by comparing its distribution to that of 6-monacetylmorphine (6-MAM), heroin's major metabolite, in a rodent model using mass spectrometric imaging (MSI) in combination with liquid chromatography-tandem mass spectrometry (LC-MS/MS). Male Sprague-Dawley rats (n = 5) received heroin (10 mg kg-1) intraperitoneally, NLX (10 mg kg-1) intranasally, and NLX injected intranasally 5 min after heroin administration. The animals were sacrificed 15 min after dose and brain tissues were harvested. The MSI image analysis showed a region-specific distribution of 6-MAM in the brain regions including the corpus callosum, hippocampal formation, cerebral cortex, corticospinal tracts, caudate putamen, thalamus, globus pallidus, hypothalamus, and basal forebrain regions of the brain. The antagonist had a similar biodistribution throughout the brain in both groups of animals that received NLX or NLX after heroin administration. The MSI analysis demonstrated that the intensity of 6-MAM in these brain regions was reduced following NLX treatment. The decrease in 6-MAM intensity was caused by its displacement by the antagonist and its binding to these receptors in these specific brain regions, consequently enhancing the opioid elimination. These findings will contribute to the evaluation of other narcotic antagonists that might be considered for use in the treatment of drug overdose via MSI.
Collapse
Affiliation(s)
- Belin
G. Teklezgi
- Catalysis
and Peptide Research Unit, University of
KwaZulu-Natal, Westville Campus, Durban 3629, South Africa
| | - Annapurna Pamreddy
- Catalysis
and Peptide Research Unit, University of
KwaZulu-Natal, Westville Campus, Durban 3629, South Africa
| | - Sphamandla Ntshangase
- Catalysis
and Peptide Research Unit, University of
KwaZulu-Natal, Westville Campus, Durban 3629, South Africa
| | - Sipho Mdanda
- Catalysis
and Peptide Research Unit, University of
KwaZulu-Natal, Westville Campus, Durban 3629, South Africa
| | - Sanil D. Singh
- Department
of Pharmaceutical Science, University of
KwaZulu-Natal, Westville
Campus, Durban 3629, South Africa
| | - Nirmala D. Gopal
- Department
of Criminology, University of KwaZulu-Natal, Durban 4041, South Africa
| | - Tricia Naicker
- Catalysis
and Peptide Research Unit, University of
KwaZulu-Natal, Westville Campus, Durban 3629, South Africa
| | - Hendrik G. Kruger
- Catalysis
and Peptide Research Unit, University of
KwaZulu-Natal, Westville Campus, Durban 3629, South Africa
| | - Thavendran Govender
- Department
of Chemistry, University of Zululand, Richards Bay 3900, South Africa
| | - Sooraj Baijnath
- Catalysis
and Peptide Research Unit, University of
KwaZulu-Natal, Westville Campus, Durban 3629, South Africa
| |
Collapse
|
17
|
Scott PJH, Koeppe RA, Shao X, Rodnick ME, Sowa AR, Henderson BD, Stauff J, Sherman PS, Arteaga J, Carlo DJ, Moss RB. The Effects of Intramuscular Naloxone Dose on Mu Receptor Displacement of Carfentanil in Rhesus Monkeys. Molecules 2020; 25:molecules25061360. [PMID: 32192089 PMCID: PMC7144122 DOI: 10.3390/molecules25061360] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 03/13/2020] [Accepted: 03/15/2020] [Indexed: 11/16/2022] Open
Abstract
Naloxone (NLX) is a mu receptor antagonist used to treat acute opioid overdoses. Currently approved doses of naloxone to treat opioid overdoses are 4 mg intranasal (IN) and 2 mg intramuscular (IM). However, higher mu receptor occupancy (RO) may be required to treat overdoses due to more potent synthetic opioids such as fentanyl and carfentanil that have entered the illicit drug market recently. To address this need, a higher dose of NLX has been investigated in a 5 mg IM formulation called ZIMHI but, while the effects of intravenous (IV) and IN administration of NLX on the opioid mu receptor occupancy (RO) have been studied, comparatively little is known about RO for IM administration of NLX. The goal of this study was to examine the effect of IM dosing of NLX on mu RO in rhesus macaques using [11C]carfentanil positron emission tomography (PET) imaging. The lowest dose of NLX (0.06 mg/kg) approximated 51% RO. Higher doses of NLX (0.14 mg/kg, 0.28 mg/kg) resulted in higher mu RO of 70% and 75%, respectively. Plasma levels were 4.6 ng/mL, 16.8 ng/mL, and 43.4 ng/mL for the three IM doses, and a significant correlation between percent RO and plasma NLX level was observed (r = 0.80). These results suggest that higher doses of IM NLX result in higher mu RO and could be useful in combating overdoses resulting from potent synthetic opioids.
Collapse
Affiliation(s)
- Peter J. H. Scott
- Department of Radiology, University of Michigan, Ann Arbor, MI 48105, USA; (R.A.K.); (X.S.); (M.E.R.); (A.R.S.); (B.D.H.); (J.S.); (P.S.S.); (J.A.)
- Correspondence: (P.J.H.S.); (R.B.M.)
| | - Robert A. Koeppe
- Department of Radiology, University of Michigan, Ann Arbor, MI 48105, USA; (R.A.K.); (X.S.); (M.E.R.); (A.R.S.); (B.D.H.); (J.S.); (P.S.S.); (J.A.)
| | - Xia Shao
- Department of Radiology, University of Michigan, Ann Arbor, MI 48105, USA; (R.A.K.); (X.S.); (M.E.R.); (A.R.S.); (B.D.H.); (J.S.); (P.S.S.); (J.A.)
| | - Melissa E. Rodnick
- Department of Radiology, University of Michigan, Ann Arbor, MI 48105, USA; (R.A.K.); (X.S.); (M.E.R.); (A.R.S.); (B.D.H.); (J.S.); (P.S.S.); (J.A.)
| | - Alexandra R. Sowa
- Department of Radiology, University of Michigan, Ann Arbor, MI 48105, USA; (R.A.K.); (X.S.); (M.E.R.); (A.R.S.); (B.D.H.); (J.S.); (P.S.S.); (J.A.)
| | - Bradford D. Henderson
- Department of Radiology, University of Michigan, Ann Arbor, MI 48105, USA; (R.A.K.); (X.S.); (M.E.R.); (A.R.S.); (B.D.H.); (J.S.); (P.S.S.); (J.A.)
| | - Jenelle Stauff
- Department of Radiology, University of Michigan, Ann Arbor, MI 48105, USA; (R.A.K.); (X.S.); (M.E.R.); (A.R.S.); (B.D.H.); (J.S.); (P.S.S.); (J.A.)
| | - Phillip S. Sherman
- Department of Radiology, University of Michigan, Ann Arbor, MI 48105, USA; (R.A.K.); (X.S.); (M.E.R.); (A.R.S.); (B.D.H.); (J.S.); (P.S.S.); (J.A.)
| | - Janna Arteaga
- Department of Radiology, University of Michigan, Ann Arbor, MI 48105, USA; (R.A.K.); (X.S.); (M.E.R.); (A.R.S.); (B.D.H.); (J.S.); (P.S.S.); (J.A.)
| | - Dennis J. Carlo
- Adamis Pharmaceuticals, 11682 El Camino Real, Suite # 300, San Diego, CA 92130, USA;
| | - Ronald B. Moss
- Adamis Pharmaceuticals, 11682 El Camino Real, Suite # 300, San Diego, CA 92130, USA;
- Correspondence: (P.J.H.S.); (R.B.M.)
| |
Collapse
|
18
|
Abstract
This paper is the fortieth consecutive installment of the annual anthological review of research concerning the endogenous opioid system, summarizing articles published during 2017 that studied the behavioral effects of molecular, pharmacological and genetic manipulation of opioid peptides and receptors as well as effects of opioid/opiate agonists and antagonists. The review is subdivided into the following specific topics: molecular-biochemical effects and neurochemical localization studies of endogenous opioids and their receptors (1), the roles of these opioid peptides and receptors in pain and analgesia in animals (2) and humans (3), opioid-sensitive and opioid-insensitive effects of nonopioid analgesics (4), opioid peptide and receptor involvement in tolerance and dependence (5), stress and social status (6), learning and memory (7), eating and drinking (8), drug abuse and alcohol (9), sexual activity and hormones, pregnancy, development and endocrinology (10), mental illness and mood (11), seizures and neurologic disorders (12), electrical-related activity and neurophysiology (13), general activity and locomotion (14), gastrointestinal, renal and hepatic functions (15), cardiovascular responses (16), respiration and thermoregulation (17), and immunological responses (18).
Collapse
Affiliation(s)
- Richard J Bodnar
- Department of Psychology and Neuropsychology Doctoral Sub-Program, Queens College, City University of New York, CUNY, 65-30 Kissena Blvd., Flushing, NY, 11367, United States.
| |
Collapse
|
19
|
Intranasal naloxone rapidly occupies brain mu-opioid receptors in human subjects. Neuropsychopharmacology 2019; 44:1667-1673. [PMID: 30867551 PMCID: PMC6785104 DOI: 10.1038/s41386-019-0368-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 03/03/2019] [Accepted: 03/06/2019] [Indexed: 12/14/2022]
Abstract
Nasal spray formulations of naloxone, a mu-opioid receptor (MOR) antagonist, are currently used for the treatment of opioid overdose. They may have additional therapeutic utility also in the absence of opioid agonist drugs, but the onset and duration of action at brain MORs have been inadequately characterized to allow such projections. This study provides initial characterization of brain MOR availability at high temporal resolution following intranasal (IN) naloxone administration to healthy volunteers in the absence of a competing opioid agonist. Fourteen participants were scanned twice using positron emission tomography (PET) and [11C]carfentanil, a selective MOR agonist radioligand. Concentrations of naloxone in plasma and MOR availability (relative to placebo) were monitored from 0 to 60 min and at 300-360 min post naloxone. Naloxone plasma concentrations peaked at ~20 min post naloxone, associated with slightly delayed development of brain MOR occupancy (half of peak occupancy reached at ~10 min). Estimated peak occupancies were 67 and 85% following 2 and 4 mg IN doses, respectively. The estimated half-life of occupancy disappearance was ~100 min. The rapid onset of brain MOR occupancy by IN naloxone, evidenced by the rapid onset of its action in opioid overdose victims, was directly documented in humans for the first time. The employed high temporal-resolution PET method establishes a model that can be used to predict brain MOR occupancy from plasma naloxone concentrations. IN naloxone may have therapeutic utility in various addictions where brain opioid receptors are implicated, such as gambling disorder and alcohol use disorder.
Collapse
|
20
|
Mackiewicz AL, Salyards GW, Knych HK, Hill AE, Christe KL. Pharmacokinetics of a Long-lasting, Highly Concentrated Buprenorphine Solution after Subcutaneous Administration in Rhesus Macaques ( Macaca mulatta). JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE : JAALAS 2019; 58:501-509. [PMID: 31227029 PMCID: PMC6643088 DOI: 10.30802/aalas-jaalas-18-000115] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/05/2018] [Accepted: 12/13/2018] [Indexed: 11/05/2022]
Abstract
Opioids are essential for use in rhesus macaques (Macaca mulatta) that require multimodal analgesia or those unable to receive NSAID as part of their pain management plan. The current opioid epidemic has universally limited the availability of these vital analgesics, compelling clinicians to investigate other options including novel opioid formulations. A commercially available injectable, long-lasting, highly concentrated buprenorphine solution (HCBS) provides therapeutic plasma concentrations lasting 24 h after a single dose in cats ( Felis catus). We hypothesized that this same HCBS would achieve therapeutic concentrations (≥0.1 ng/mL) for at least 24 h in rhesus macaques. In the current study, 6 healthy, adult rhesus macaques were included in a randomized, 2-period, 2-treatment crossover study. The low dose (0.24 mg/kg SC) achieved a peak plasma concentration of 19.1 ± 5.68 ng/mL at 0.308 ± 0.077 h, with an AUC of 236.4 ± 22.5 h/ng/mL and terminal elimination half-life of 19.6 ± 4.02 h; for the high dose (0.72 mg/kg SC), these parameters were 65.2 ± 14.7 ng/mL, 0.034 ± 0.004 h, 641.3 ± 79.4 h/ng/mL, and 20.6 ± 2.30 h, respectively. The mean plasma concentrations for the low and high doses in rhesus macaques significantly exceeded the therapeutic threshold for 48 and 72 h, respectively. One macaque showed mild somnolence at both doses, and another showed mild pruritus at both doses. These findings show that subcutaneous administration of HCBS provides prolonged and long-lasting therapeutic plasma levels for 48 to 72 h dosing without problematic adverse effects and thus represents a potential new analgesic alternative.
Collapse
Affiliation(s)
- Alexis L Mackiewicz
- Department of Primate Medicine, California National Primate Research Center, University of California, Davis, Davis, California;,
| | - Gregory W Salyards
- Department of Primate Medicine, California National Primate Research Center, University of California, Davis, Davis, California
| | - Heather K Knych
- KL Maddy Equine Analytical Chemistry Laboratory, University of California, Davis, Davis, California
| | - Ashley E Hill
- California Animal Health and Food Safety Laboratory, University of California, Davis, Davis, California; Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, Davis, California
| | - Kari L Christe
- Department of Primate Medicine, California National Primate Research Center, University of California, Davis, Davis, California; Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, Davis, California
| |
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
|
Evaluation of intranasal delivery route of drug administration for brain targeting. Brain Res Bull 2018; 143:155-170. [PMID: 30449731 DOI: 10.1016/j.brainresbull.2018.10.009] [Citation(s) in RCA: 459] [Impact Index Per Article: 65.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 10/20/2018] [Accepted: 10/23/2018] [Indexed: 12/22/2022]
Abstract
The acute or chronic drug treatments for different neurodegenerative and psychiatric disorders are challenging from several aspects. The low bioavailability and limited brain exposure of oral drugs, the rapid metabolism, elimination, the unwanted side effects and also the high dose to be added mean both inconvenience for the patients and high costs for the patients, their family and the society. The reason of low brain penetration of the compounds is that they have to overcome the blood-brain barrier which protects the brain against xenobiotics. Intranasal drug administration is one of the promising options to bypass blood-brain barrier, to reduce the systemic adverse effects of the drugs and to lower the doses to be administered. Furthermore, the drugs administered using nasal route have usually higher bioavailability, less side effects and result in higher brain exposure at similar dosage than the oral drugs. In this review the focus is on giving an overview on the anatomical and cellular structure of nasal cavity and absorption surface. It presents some possibilities to enhance the drug penetration through the nasal barrier and summarizes some in vitro, ex vivo and in vivo technologies to test the drug delivery across the nasal epithelium into the brain. Finally, the authors give a critical evaluation of the nasal route of administration showing its main advantages and limitations of this delivery route for CNS drug targeting.
Collapse
|
23
|
Takai N, Miyajima N, Tonomura M, Abe K. Relationship between receptor occupancy and the antinociceptive effect of mu opioid receptor agonists in male rats. Brain Res 2017; 1680:105-109. [PMID: 29269051 DOI: 10.1016/j.brainres.2017.12.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 11/13/2017] [Accepted: 12/12/2017] [Indexed: 11/24/2022]
Abstract
The analgesic mechanisms of mu opioid receptor (MOR) agonists, including receptor occupancy at the site of action, are not completely understood. The aims of the present study were to evaluate: (i) receptor occupancy in the rat brain after administration of MOR agonists; (ii) the relationship between occupancy and the antinociceptive effect. Morphine (2 or 4 mg/kg) or oxycodone (1 or 3 mg/kg) was subcutaneously administered to rats. The antinociceptive effect of these drugs was measured by the hot-plate test. MOR occupancy in the thalamus was assessed by conducting an ex vivo receptor binding assay using [3H] [D-Ala2, N-MePhe4, Gly-ol]-enkephalin, followed by autoradiographic analysis. Both drugs produced antinociception in a dose-dependent manner, and these effects disappeared after the time point at which the maximal effect was elicited. Thalamic MOR occupancy was observed in a dose-dependent manner at the time point at which maximal antinociception was elicited, and relatively low occupancy was observed when the antinociceptive effect was decreasing. Good correlation between thalamic MOR occupancy and the antinociceptive effect was observed. These findings provide direct evidence for the receptor occupancy of MOR agonists at the site of action and its relationship with the analgesic effect.
Collapse
Affiliation(s)
- Nozomi Takai
- Biomarker R&D Department, Shionogi & Co., Ltd., Osaka, Japan.
| | | | - Misato Tonomura
- Biomarker R&D Department, Shionogi & Co., Ltd., Osaka, Japan.
| | - Kohji Abe
- Biomarker R&D Department, Shionogi & Co., Ltd., Osaka, Japan.
| |
Collapse
|
24
|
Fentanyls continue to replace heroin in the drug arena: the cases of ocfentanil and carfentanil. Forensic Toxicol 2017; 36:12-32. [PMID: 29367860 PMCID: PMC5754389 DOI: 10.1007/s11419-017-0379-4] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 07/23/2017] [Indexed: 01/25/2023]
Abstract
Purpose Ocfentanil and carfentanil are two potent synthetic opioids that are analogues of fentanyl and are actively involved in the recent fentanyl crisis. The aim of this review is to provide all the available information on these two fentanyl analogues. Methods All reviewed information was gathered through a detailed search of PubMed and the World Wide Web using relevant keywords. Results Like most of the members of the family of fentanyls, they are either sold as heroin to unsuspecting users or used extensively to lace heroin street samples. Despite the fact that ocfentanil was studied clinically in the early 1990s, it did not manage to find its place in clinical practice. On the other hand, carfentanil is mainly used today as an anesthetic agent in large animals. Ocfentanil and carfentanil are used and abused extensively, mainly in Europe and in the United States. As a result, they are the cause of some verified intoxication cases and deaths worldwide. This review provides information concerning chemistry, synthesis, prevalence, pharmacology, and toxicology, as well as the current legal status of these two fentanyl analogues. Analytical methods developed for the determination of ocfentanil and carfentanil in biological specimens and seized materials, as well as related intoxication and lethal cases are also presented. Conclusions Ocfentanil and carfentanil are undeniably very dangerous opioid drugs and a very serious matter of concern for public safety. The authorities should take the appropriate actions to avoid the expansion of this threat by taking proper and prompt measures.
Collapse
|
25
|
Blecha JE, Henderson BD, Hockley BG, VanBrocklin HF, Zubieta JK, DaSilva AF, Kilbourn MR, Koeppe RA, Scott PJ, Shao X. An updated synthesis of [ 11 C]carfentanil for positron emission tomography (PET) imaging of the μ-opioid receptor. J Labelled Comp Radiopharm 2017; 60:375-380. [PMID: 28419528 PMCID: PMC9886010 DOI: 10.1002/jlcr.3513] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 04/10/2017] [Accepted: 04/11/2017] [Indexed: 02/01/2023]
Abstract
[11 C]Carfentanil ([11 C]CFN) is a selective radiotracer for in vivo positron emission tomography imaging studies of the μ-opioid system that, in our laboratories, is synthesized by methylation of the corresponding carboxylate precursor with [11 C]MeOTf, and purified using a C2 solid-phase extraction cartridge. Changes in the commercial availability of common C2 cartridges have necessitated future proofing the synthesis of [11 C]CFN to maintain reliable delivery of the radiotracer for clinical imaging studies. An updated synthesis of [11 C]CFN is reported that replaces a now obsolete purification cartridge with a new commercially available version and also substitutes the organic solvents used in traditional production methods with ethanol.
Collapse
Affiliation(s)
- Joseph E. Blecha
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | | | - Brian G. Hockley
- Department of Radiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Henry F. VanBrocklin
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, USA
| | - Jon-Kar Zubieta
- Department of Psychiatry, University Neuropsychiatric Institute, University of Utah Health Center, Salt Lake City, UT, USA
| | - Alexandre F. DaSilva
- Headache and Orofacial Pain Effort, Biologic and Materials Sciences Department, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - Michael R. Kilbourn
- Department of Radiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Robert A. Koeppe
- Department of Radiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Peter J.H. Scott
- Department of Radiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Xia Shao
- Department of Radiology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|