1
|
Khalil NY, Bakheit AH, Alkahtani HM, Al-Muhanna T. Vinpocetine (A comprehensive profile). PROFILES OF DRUG SUBSTANCES, EXCIPIENTS, AND RELATED METHODOLOGY 2022; 47:1-54. [PMID: 35396012 DOI: 10.1016/bs.podrm.2021.10.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Vinpocetine (VIN) is a herbal supplement extracted from the periwinkle plant. It is a multi-action agent, which is used to treat various neurological disorders such as Alzheimer's and Parkinson's disease. Vinpocetine has also anti-inflammatory, analgesic, antioxidant property and treats various thinking and memory problems. Currently, vinpocetine is also available in the market as a dietary supplement to enhance cognition and memory. This profile explains the physicochemical properties, methods of preparation, content of related impurities and different spectroscopical behavior of vinpocetine. It also discusses the reported methods of analysis of the drug, which include Compendial Methods, Electrochemical Methods, Spectrophotometric Methods and Chromatographic Methods of analysis. Furthermore, this profile explains the stability of the drug subjected to stress conditions of acid, alkaline and photolytic degradation. In addition, the clinical applications of the drug, its uses, side effects, dosing information, pharmacokinetics and mechanism of action are also discussed.
Collapse
Affiliation(s)
- Nasr Y Khalil
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia.
| | - Ahmed H Bakheit
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia; Department of Chemistry, Faculty of Science and Technology, Al-Neelain University, Khartoum, Sudan
| | - Hamad M Alkahtani
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Turki Al-Muhanna
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
2
|
Meador KJ, Leeman-Markowski B, Medina AE, Illamola SM, Seliger J, Novak G, Lin C, Ivanisevic M, Razavi B, Marino S, Boyd A, Loring DW. Vinpocetine, cognition, and epilepsy. Epilepsy Behav 2021; 119:107988. [PMID: 33957389 DOI: 10.1016/j.yebeh.2021.107988] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 04/07/2021] [Accepted: 04/07/2021] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Vinpocetine has been shown to enhance memory in animal models, with possible cognitive benefit in humans. The present study sought to demonstrate if vinpocetine can enhance cognition in healthy volunteers or patients with epilepsy. In addition, we compare blood levels of vinpocetine and its active metabolite (apovincaminic acid; AVA) in humans and animals to further characterize factors related to possible therapeutic benefit. METHODS The cognitive effects of vinpocetine were assessed in healthy adult volunteers (n = 8) using a double-blind, randomized, crossover design at single doses (placebo, 10, 20, and 60 mg oral). Cognitive effects of vinpocetine in patients with focal epilepsy (n = 8) were tested using a double-blind, randomized, crossover design at single doses (placebo, 20 mg oral) followed by one-month open label at 20 mg oral three times a day. The neuropsychological battery included both computerized and non-computerized tests. Levels of vinpocetine and AVA in the human studies were compared to levels in 45 mice across time dosed at 5-20 mg/kg intraperitoneal of vinpocetine. RESULTS No significant cognitive benefits were seen in healthy volunteers or patients with epilepsy. No appreciable side effects occurred. Vinpocetine and AVA levels were lower in humans than animals. CONCLUSIONS Vinpocetine was well tolerated, but was not associated with positive cognitive effects. However, blood levels obtained in humans were substantially less than levels in animals obtained from dosages known to be effective in one model. This suggests that higher dosages are needed in humans to assess vinpocetine's cognitive efficacy.
Collapse
Affiliation(s)
- Kimford J Meador
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA.
| | - Beth Leeman-Markowski
- Department of Neurology, New York University and VA New York Harbor Healthcare System, NY, NY USA.
| | | | - Sílvia M Illamola
- Department of Experimental & Clinical Pharmacology, University of Minnesota, Minneapolis, MN, USA.
| | - Jordan Seliger
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA.
| | - Gloria Novak
- Department of Neurology, Emory University, Atlanta, GA, USA.
| | - Christine Lin
- School of Medicine, University of California, San Diego, CA, USA.
| | | | - Babak Razavi
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA.
| | - Susan Marino
- Department of Experimental & Clinical Pharmacology, University of Minnesota, Minneapolis, MN, USA.
| | | | - David W Loring
- Department of Neurology, Emory University, Atlanta, GA, USA.
| |
Collapse
|
3
|
Zhang L, Hu K, Shao T, Hou L, Zhang S, Ye W, Josephson L, Meyer JH, Zhang MR, Vasdev N, Wang J, Xu H, Wang L, Liang SH. Recent developments on PET radiotracers for TSPO and their applications in neuroimaging. Acta Pharm Sin B 2021; 11:373-393. [PMID: 33643818 PMCID: PMC7893127 DOI: 10.1016/j.apsb.2020.08.006] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 07/15/2020] [Accepted: 07/29/2020] [Indexed: 12/12/2022] Open
Abstract
The 18 kDa translocator protein (TSPO), previously known as the peripheral benzodiazepine receptor, is predominately localized to the outer mitochondrial membrane in steroidogenic cells. Brain TSPO expression is relatively low under physiological conditions, but is upregulated in response to glial cell activation. As the primary index of neuroinflammation, TSPO is implicated in the pathogenesis and progression of numerous neuropsychiatric disorders and neurodegenerative diseases, including Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), Parkinson's disease (PD), multiple sclerosis (MS), major depressive disorder (MDD) and obsessive compulsive disorder (OCD). In this context, numerous TSPO-targeted positron emission tomography (PET) tracers have been developed. Among them, several radioligands have advanced to clinical research studies. In this review, we will overview the recent development of TSPO PET tracers, focusing on the radioligand design, radioisotope labeling, pharmacokinetics, and PET imaging evaluation. Additionally, we will consider current limitations, as well as translational potential for future application of TSPO radiopharmaceuticals. This review aims to not only present the challenges in current TSPO PET imaging, but to also provide a new perspective on TSPO targeted PET tracer discovery efforts. Addressing these challenges will facilitate the translation of TSPO in clinical studies of neuroinflammation associated with central nervous system diseases.
Collapse
Key Words
- AD, Alzheimer's disease
- ALS, amyotrophic lateral sclerosis
- AMPA, α-amino-3-hydroxy-5-methyl-4-isoxazole propionic acid
- ANT, adenine nucleotide transporter
- Am, molar activities
- BBB, blood‒brain barrier
- BMSC, bone marrow stromal cells
- BP, binding potential
- BPND, non-displaceable binding potential
- BcTSPO, Bacillus cereus TSPO
- CBD, corticobasal degeneration
- CNS disorders
- CNS, central nervous system
- CRAC, cholesterol recognition amino acid consensus sequence
- DLB, Lewy body dementias
- EP, epilepsy
- FTD, frontotemporal dementia
- HAB, high-affinity binding
- HD, Huntington's disease
- HSE, herpes simplex encephalitis
- IMM, inner mitochondrial membrane
- KA, kainic acid
- LAB, low-affinity binding
- LPS, lipopolysaccharide
- MAB, mixed-affinity binding
- MAO-B, monoamine oxidase B
- MCI, mild cognitive impairment
- MDD, major depressive disorder
- MMSE, mini-mental state examination
- MRI, magnetic resonance imaging
- MS, multiple sclerosis
- MSA, multiple system atrophy
- Microglial activation
- NAA/Cr, N-acetylaspartate/creatine
- Neuroinflammation
- OCD, obsessive compulsive disorder
- OMM, outer mitochondrial membrane
- P2X7R, purinergic receptor P2X7
- PAP7, RIa-associated protein
- PBR, peripheral benzodiazepine receptor
- PCA, posterior cortical atrophy
- PD, Parkinson's disease
- PDD, PD dementia
- PET, positron emission tomography
- PKA, protein kinase A
- PRAX-1, PBR-associated protein 1
- PSP, progressive supranuclear palsy
- Positron emission tomography (PET)
- PpIX, protoporphyrin IX
- QA, quinolinic acid
- RCYs, radiochemical yields
- ROS, reactive oxygen species
- RRMS, relapsing remitting multiple sclerosis
- SA, specific activity
- SAH, subarachnoid hemorrhage
- SAR, structure–activity relationship
- SCIDY, spirocyclic iodonium ylide
- SNL, selective neuronal loss
- SNR, signal to noise ratio
- SUV, standard uptake volume
- SUVR, standard uptake volume ratio
- TBAH, tetrabutyl ammonium hydroxide
- TBI, traumatic brain injury
- TLE, temporal lobe epilepsy
- TSPO
- TSPO, translocator protein
- VDAC, voltage-dependent anion channel
- VT, distribution volume
- d.c. RCYs, decay-corrected radiochemical yields
- dMCAO, distal middle cerebral artery occlusion
- fP, plasma free fraction
- n.d.c. RCYs, non-decay-corrected radiochemical yields
- p.i., post-injection
Collapse
|
4
|
Nag S, Krasikova R, Airaksinen AJ, Arakawa R, Petukhovd M, Gulyas B. Synthesis and biological evaluation of [ 18F]fluorovinpocetine, a potential PET radioligand for TSPO imaging. Bioorg Med Chem Lett 2019; 29:2270-2274. [PMID: 31257082 DOI: 10.1016/j.bmcl.2019.06.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/18/2019] [Accepted: 06/19/2019] [Indexed: 01/31/2023]
Abstract
Despite of various PET radioligands targeting the translocator protein TSPO 18-KDa are used for the investigations of neuroinflammatory conditions associated with neurological disorders, development of new TSPO radiotracers is still an active area of the researches with a major focus on the 18F-labelled radiotracers. Here, we report the radiochemical synthesis of [18F]vinpocetine, fluorinated analogue of previously reported TSPO radioligand, [11C]vinpocetine. Radiolabeling was achieved by [18F]fluoroethylation of apovincaminic acid with [18F]fluoroethyl bromide. [18F]vinpocetine was obtained in quantities >2.7 GBq in RCY of 13% (non-decay corrected), and molar activity >60 GBq/µmol within 95 min synthesis time. Preliminary PET studies in a cynomolgus monkey and metabolite studies by HPLC demonstrated similar results by [18F]vinpocetine as for [11C]vinpocetine, including high blood-brain barrier permeability, regional uptake pattern and fast washout from the NHP brain. These results demonstrate that [18F]fluorovinpocetine warrants further evaluation as an easier accessible alternative to [11C]vinpocetine.
Collapse
Affiliation(s)
- S Nag
- Karolinska Institutet, Department of Clinical Neuroscience, Stockholm, Sweden.
| | - R Krasikova
- Karolinska Institutet, Department of Clinical Neuroscience, Stockholm, Sweden; N.P. Bechtereva Institute of Human Brain Russian Academy of Sciences, St.-Petersburg, Russia
| | - A J Airaksinen
- Department of Chemistry - Radiochemistry, University of Helsinki, Finland
| | - R Arakawa
- Karolinska Institutet, Department of Clinical Neuroscience, Stockholm, Sweden
| | - M Petukhovd
- Petersburg Nuclear Physics Institute named after B.P. Konstantinov, NRC "Kurchatov Institute", Gatchina, Russia; Peter the Great St.-Petersburg Polytechnic University, St.-Petersburg, Russia
| | - B Gulyas
- Karolinska Institutet, Department of Clinical Neuroscience, Stockholm, Sweden; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| |
Collapse
|
5
|
Ayoub S, Melzig MF. Induction Effects of Apigenin, Luteolin and Vinpocetin on Neutral Endopeptidase (NEP) and Angiotensin-Converting Enzyme Activity (ACE) of SK-N-SH Cells. Nat Prod Commun 2019. [DOI: 10.1177/1934578x0600100807] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The long-term effects of a number of flavonoids (such as apigenin, luteolin and amentoflavone) and vinpocetine on the neutral endopeptidase (NEP) and angiotensin-converting enzyme (ACE) were investigated. It was shown that apigenin, luteolin and vinpocetin are able to induce the activity of both NEP and ACE associated with the inhibition of the proliferation of the neuroblastoma cell line SK-N-SH. Amentoflavone has no effect on either NEP or ACE activity. An additional enhancement of cellular NEP activity could be detected after the treatment of the cells with a combination of both arabinosylcytosine and either apigenin or luteolin. This effect supports the assumption that apigenin and luteolin influence directly the gene expression of NEP. Taking into account the significant role of NEP and ACE in the degradation of amyloid beta peptides, the induction of both enzymes by long-term treatment with apigenin, luteolin and vinpocetine may have a beneficial effect regarding the prevention of the formation of amyloid plaques and the effect of these substances may be discussed as neuroprotective.
Collapse
Affiliation(s)
- Shereen Ayoub
- Institut für Pharmazie, Freie Universität Berlin, Königin-Luise-Str. 2-4, 14195 Berlin, Germany
| | - Matthias F. Melzig
- Institut für Pharmazie, Freie Universität Berlin, Königin-Luise-Str. 2-4, 14195 Berlin, Germany
| |
Collapse
|
6
|
Vinpocetine protects inner retinal neurons with functional NMDA glutamate receptors against retinal ischemia. Exp Eye Res 2018; 167:1-13. [DOI: 10.1016/j.exer.2017.10.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 07/31/2017] [Accepted: 10/08/2017] [Indexed: 11/21/2022]
|
7
|
Keller T, Krzyczmonik A, Forsback S, Picón FRL, Kirjavainen AK, Takkinen J, Rajander J, Cacheux F, Damont A, Dollé F, Rinne JO, Haaparanta-Solin M, Solin O. Radiosynthesis and Preclinical Evaluation of [ 18F]F-DPA, A Novel Pyrazolo[1,5a]pyrimidine Acetamide TSPO Radioligand, in Healthy Sprague Dawley Rats. Mol Imaging Biol 2017; 19:736-745. [PMID: 28083825 PMCID: PMC5574958 DOI: 10.1007/s11307-016-1040-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
PURPOSE Many neurological conditions result in the overexpression of the translocator protein 18 kDa (TSPO), today recognized as a biomarker for microglial activation and neuroinflammation imaging. The pyrazolo[1,5-a]pyrimidine acetamides are a particularly attractive class of TSPO-specific ligands, prompting the development of several positron emission tomography (PET) radiotracers. This includes F-DPA, a recently reported fluorinated ligand (K i = 1.7 nM), wherein the fluorine atom is directly linked to the phenyl moiety without the presence of an alkyl or alkoxy spacer chain. Reported here is the preparation of [18F]F-DPA using [18F]Selectfluor bis(triflate) and the preliminary evaluation of [18F]F-DPA in healthy rats. Its metabolic profile and biodistribution in rats are compared with that of [18F]DPA-714, a closely related structure. PROCEDURES [18F]F-DPA was synthesized by electrophilic fluorination using [18F]Selectfluor bis(triflate), [18F]DPA-714 was synthesized by conventional nucleophilic fluorination. The biodistribution of both radiotracers was compared in Sprague Dawley rats. Radiometabolites of both radiotracers in plasma and brain homogenates were analyzed by radioTLC. RESULTS The radiochemical yield of [18F]F-DPA was 15 ± 3 % and the specific activity was 7.8 ± 0.4 GBq/μmol. The radiochemical purity exceeded 99 %. The in vivo time activity curves of [18F]F-DPA demonstrate rapid entry into the brain and a concentration equilibrium at 20-30 min after injection. The metabolic profiles at 90 min after radiotracer injection in the plasma show that unchanged [18F]F-DPA and [18F]DPA-714 account for 28.3 ± 6.4 and 11.1 ± 2.6 % of the remaining radioactivity, respectively. In the brain, unchanged [18F]F-DPA accounts for 93.5 ± 2.8 % of the radioactivity; whereas for [18F]DPA-714, this value is 53.6 ± 1.6 %. CONCLUSIONS [18F]Selectfluor bis(triflate) was successfully used to label F-DPA with fluorine-18. The labeling position on the aromatic moiety imparts a higher stability compared to [18F]DPA-714 with regard to in vivo metabolism. [18F]F-DPA is a promising new radiotracer and warrants further investigation in animal models of disease.
Collapse
Affiliation(s)
- Thomas Keller
- Radiopharmaceutical Chemistry Laboratory, Turku PET Centre, University of Turku, Turku, Finland
| | - Anna Krzyczmonik
- Radiopharmaceutical Chemistry Laboratory, Turku PET Centre, University of Turku, Turku, Finland
| | - Sarita Forsback
- Radiopharmaceutical Chemistry Laboratory, Turku PET Centre, University of Turku, Turku, Finland
- Department of Chemistry, University of Turku, Turku, Finland
| | - Francisco R López Picón
- PET Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, Turku, Finland
- MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Anna K Kirjavainen
- Radiopharmaceutical Chemistry Laboratory, Turku PET Centre, University of Turku, Turku, Finland
| | - Jatta Takkinen
- PET Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, Turku, Finland
- MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Johan Rajander
- Accelerator Laboratory, Turku PET Centre, Åbo Akademi University, Turku, Finland
| | - Fanny Cacheux
- CEA, I2BM, Service Hospitalier Frédéric Joliot, Orsay, France
| | | | - Frédéric Dollé
- CEA, I2BM, Service Hospitalier Frédéric Joliot, Orsay, France
| | - Juha O Rinne
- Turku PET Centre, Division of Clinical Neurosciences, Turku University Hospital, Turku, Finland
| | - Merja Haaparanta-Solin
- PET Preclinical Imaging Laboratory, Turku PET Centre, University of Turku, Turku, Finland
- MediCity Research Laboratory, University of Turku, Turku, Finland
| | - Olof Solin
- Radiopharmaceutical Chemistry Laboratory, Turku PET Centre, University of Turku, Turku, Finland.
- Department of Chemistry, University of Turku, Turku, Finland.
- Accelerator Laboratory, Turku PET Centre, Åbo Akademi University, Turku, Finland.
- Turku PET Centre, University of Turku, Kiinamyllynkatu 4-8, 20520, Turku, Finland.
| |
Collapse
|
8
|
Nivison-Smith L, Khoo P, Acosta ML, Kalloniatis M. Pre-treatment with vinpocetine protects against retinal ischemia. Exp Eye Res 2016; 154:126-138. [PMID: 27899287 DOI: 10.1016/j.exer.2016.11.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 10/17/2016] [Accepted: 11/22/2016] [Indexed: 01/15/2023]
Abstract
Vinpocetine has been shown to have beneficial effects for tissues of the central nervous system subjected to ischemia and other related metabolic insults. We recently showed vinpocetine promotes glucose availability, prevents unregulated cation channel permeability and regulates glial reactivity when present during retinal ischemia. Less is known however about the ability of vinpocetine to protect against future ischemic insults. This study explores the effect of vinpocetine when used as a pre-treatment in an ex vivo model for retinal ischemia using cation channel permeability of agmatine (AGB) combined with immunohistochemistry as a measure for cell functionality. We found that vinpocetine pre-treatment reduced cation channel permeability and apoptotic marker immunoreactivity in the GCL and increased parvalbumin immunoreactivity of inner retinal neurons in the inner nuclear layer following ischemic insult. Vinpocetine pre-treatment also reduced Müller cell reactivity following ischemic insults of up to 120 min compared to untreated controls. Many of vinpocetine's effects however were transient in nature suggesting the drug can protect retinal neurons against future ischemic damage but may have limited long-term applications.
Collapse
Affiliation(s)
- Lisa Nivison-Smith
- Centre for Eye Health, University of New South Wales, Sydney, 2052, Australia; School of Optometry and Vision Science, University of New South Wales, Sydney, 2052, Australia.
| | - Pauline Khoo
- School of Optometry and Vision Science, University of New South Wales, Sydney, 2052, Australia
| | - Monica L Acosta
- School of Optometry and Vision Science, University of Auckland, New Zealand; New Zealand National Eye Centre, University of Auckland, New Zealand
| | - Michael Kalloniatis
- Centre for Eye Health, University of New South Wales, Sydney, 2052, Australia; School of Optometry and Vision Science, University of New South Wales, Sydney, 2052, Australia; School of Optometry and Vision Science, University of Auckland, New Zealand; New Zealand National Eye Centre, University of Auckland, New Zealand
| |
Collapse
|
9
|
Nivison-Smith L, O'Brien BJ, Truong M, Guo CX, Kalloniatis M, Acosta ML. Vinpocetine modulates metabolic activity and function during retinal ischemia. Am J Physiol Cell Physiol 2015; 308:C737-49. [PMID: 25696811 DOI: 10.1152/ajpcell.00291.2014] [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: 08/26/2014] [Accepted: 02/17/2015] [Indexed: 12/18/2022]
Abstract
Vinpocetine protects against a range of degenerative conditions and insults of the central nervous system via multiple modes of action. Little is known, however, of its effects on metabolism. This may be highly relevant, as vinpocetine is highly protective against ischemia, a process that inhibits normal metabolic function. This study uses the ischemic retina as a model to characterize vinpocetine's effects on metabolism. Vinpocetine reduced the metabolic demand of the retina following ex vivo hypoxia and ischemia to normal levels based on lactate dehydrogenase activity. Vinpocetine delivered similar effects in an in vivo model of retinal ischemia-reperfusion, possibly through increasing glucose availability. Vinpocetine's effects on glucose also appeared to improve glutamate homeostasis in ischemic Müller cells. Other actions of vinpocetine following ischemia-reperfusion, such as reduced cell death and improved retinal function, were possibly a combination of the drug's actions on metabolism and other retinal pathways. Vinpocetine's metabolic effects appeared independent of its other known actions in ischemia, as it recovered retinal function in a separate metabolic model where the glutamate-to-glutamine metabolic pathway was inhibited in Müller cells. The results of this study indicate that vinpocetine mediates ischemic damage partly through altered metabolism and has potential beneficial effects as a treatment for ischemia of neuronal tissues.
Collapse
Affiliation(s)
- Lisa Nivison-Smith
- School of Optometry and Vision Science, University of New South Wales, Sydney, Australia
| | - Brendan J O'Brien
- Department of Optometry and Vision Science, University of Auckland, Auckland, New Zealand
| | - Mai Truong
- Department of Optometry and Vision Science, University of Auckland, Auckland, New Zealand
| | - Cindy X Guo
- Department of Optometry and Vision Science, University of Auckland, Auckland, New Zealand
| | - Michael Kalloniatis
- School of Optometry and Vision Science, University of New South Wales, Sydney, Australia; Department of Optometry and Vision Science, University of Auckland, Auckland, New Zealand; Centre for Eye Health, University of New South Wales, Sydney, Australia; and
| | - Monica L Acosta
- Department of Optometry and Vision Science, University of Auckland, Auckland, New Zealand; New Zealand National Eye Centre, University of Auckland, Auckland, New Zealand
| |
Collapse
|
10
|
Vinpocetine regulates cation channel permeability of inner retinal neurons in the ischaemic retina. Neurochem Int 2014; 66:1-14. [DOI: 10.1016/j.neuint.2014.01.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 12/09/2013] [Accepted: 01/04/2014] [Indexed: 11/23/2022]
|
11
|
Gulyás B, Tóth M, Schain M, Airaksinen A, Vas Á, Kostulas K, Lindström P, Hillert J, Halldin C. Evolution of microglial activation in ischaemic core and peri-infarct regions after stroke: A PET study with the TSPO molecular imaging biomarker [ C]vinpocetine. J Neurol Sci 2012; 320:110-7. [DOI: 10.1016/j.jns.2012.06.026] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Revised: 05/03/2012] [Accepted: 06/23/2012] [Indexed: 01/17/2023]
|
12
|
Gulyás B, Vas Á, Tóth M, Takano A, Varrone A, Cselényi Z, Schain M, Mattsson P, Halldin C. Age and disease related changes in the translocator protein (TSPO) system in the human brain: Positron emission tomography measurements with [11C]vinpocetine. Neuroimage 2011; 56:1111-21. [DOI: 10.1016/j.neuroimage.2011.02.020] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Revised: 02/02/2011] [Accepted: 02/05/2011] [Indexed: 01/06/2023] Open
|
13
|
Yanamoto K, Yamasaki T, Kumata K, Yui J, Odawara C, Kawamura K, Hatori A, Inoue O, Yamaguchi M, Suzuki K, Zhang MR. Evaluation of N-benzyl-N-[11C]methyl-2- (7-methyl-8-oxo-2-phenyl-7,8-dihydro-9H-purin-9-yl)acetamide ([11C]DAC) as a novel translocator protein (18 kDa) radioligand in kainic acid-lesioned rat. Synapse 2009; 63:961-71. [PMID: 19593823 DOI: 10.1002/syn.20678] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The aim of this study was to evaluate N-benzyl-N-[11C]methyl-2-(7-methyl-8-oxo-2-phenyl-7,8-dihydro-9H-purin-9-yl)acetamide ([11C]DAC) as a new translocator protein (18 kDa) [TSPO, formerly known as the peripheral-type benzodiazepine receptor (PBR)] positron emission tomography (PET) ligand in normal mice and unilateral kainic acid (KA)-lesioned rats. DAC is a derivative of AC-5216, which is a potent and selective PET ligand for the clinical investigation of TSPO. The binding affinity and selectivity of DAC for TSPO were similar to those of AC-5216, and DAC was less lipophilic than AC-5216. The distribution pattern of [11C]DAC was in agreement with TSPO distribution in rodents. No radioactive metabolite of [11C]DAC was found in the mouse brain, although it was metabolized rapidly in mouse plasma. Using small-animal PET, we examined the in vivo binding of [11C]DAC for TSPO in KA-lesioned rats. [11C]DAC and [11C]AC-5216 exhibited similar brain uptake in the lesioned and nonlesioned striatum, respectively. The binding of [11C]DAC to TSPO was increased significantly in the lesioned striatum, and [(11)C]DAC showed good contrast between the lesioned and nonlesioned striatum (the maximum ratio was about threefold). In displacement experiments, the uptake of [11C]DAC in the lesioned striatum was eventually blocked using an excess of either unlabeled DAC or PK11195 injected. [11C]DAC had high in vivo specific binding to TSPO in the injured rat brain. Therefore, [11C]DAC is a useful PET ligand for TSPO imaging, and its specific binding to TSPO is suitable as a new biomarker for brain injury.
Collapse
Affiliation(s)
- Kazuhiko Yanamoto
- Department of Molecular Probes, Molecular Imaging Center, National Institute of Radiological Sciences, 4-9-1 Anagawa, Inage-ku, Chiba, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Nuclear imaging of neuroinflammation: a comprehensive review of [11C]PK11195 challengers. Eur J Nucl Med Mol Imaging 2008; 35:2304-19. [DOI: 10.1007/s00259-008-0908-9] [Citation(s) in RCA: 324] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2008] [Accepted: 07/17/2008] [Indexed: 12/22/2022]
|
15
|
Abstract
Stroke is the second most common cause of death and major cause of disability worldwide. Because of the ageing population, the burden will increase greatly during the next 20 years, especially in developing countries. Advances have occurred in the prevention and treatment of stroke during the past decade. For patients with acute stroke, management in a stroke care unit, intravenous tissue plasminogen activator within 3 h or aspirin within 48 h of stroke onset, and decompressive surgery for supratentorial malignant hemispheric cerebral infarction are interventions of proven benefit; several other interventions are being assessed. Proven secondary prevention strategies are warfarin for patients with atrial fibrillation, endarterectomy for symptomatic carotid stenosis, antiplatelet agents, and cholesterol reduction. The most important intervention is the management of patients in stroke care units because these provide a framework within which further study might be undertaken. These advances have exposed a worldwide shortage of stroke health-care workers, especially in developing countries.
Collapse
Affiliation(s)
- Geoffrey A Donnan
- National Stroke Research Institute, Austin Hospital, University of Melbourne, Melbourne, Victoria, Australia.
| | | | | | | |
Collapse
|
16
|
Vas A, Shchukin Y, Karrenbauer VD, Cselényi Z, Kostulas K, Hillert J, Savic I, Takano A, Halldin C, Gulyás B. Functional neuroimaging in multiple sclerosis with radiolabelled glia markers: preliminary comparative PET studies with [11C]vinpocetine and [11C]PK11195 in patients. J Neurol Sci 2007; 264:9-17. [PMID: 17727889 DOI: 10.1016/j.jns.2007.07.018] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2006] [Revised: 07/05/2007] [Accepted: 07/09/2007] [Indexed: 12/11/2022]
Abstract
With the purpose of demonstrating the use of positron emission tomography (PET) and radiolabelled glia markers to indicate regional cerebral damage, we measured with PET in four young multiplex sclerosis (MS) patients in two consecutive measurements the global and regional brain uptake as well as regional distribution and binding potential (BP) of [(11)C]vinpocetine and [(11)C]PK11195. Both ligands showed increased uptake and BP in the regions of local brain damage. However, regional BP values for [(11)C]vinpocetine were markedly higher than those for [(11)C]PK11195. This feature of the former radioligand may be related to its high brain uptake and marked affinity to the peripheral benzodiazepine receptor binding sites (PBBS), characteristic for glia cells. As local brain traumas entail reactive glia accumulation in and around the site of the damage, the present findings may indicate that [(11)C]vinpocetine marks the place or boundaries of local brain damage by binding to the PBBS present in glia cells, which, in turn, accumulate in the region of the damage. The present findings (i) confirm earlier observations with [(11)C]PK11195 as a potential glia marker in PET studies and (ii) support the working hypothesis that [(11)C]vinpocetine is a potentially useful PET marker of regional and global brain damage resulting in glia accumulation locally or globally in the human brain. The comparative analysis of the two ligands indicate that [(11)C]vinpocetine shows a number of characteristics favourable in comparison with [(11)C]PK11195.
Collapse
Affiliation(s)
- Adám Vas
- Chemical Works of Gedeon Richter Ltd., Gyomroi ut 19/21, H-1103 Budapest, Hungary.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
The Apocynaceae plant family contains a great number of so called eburnamine-vincamine alkaloids. Quite a few of these alkaloids exert varied pharmacological activities on the cell multiplication, cardiovascular system, and brain functions. Many derivatives were also synthesized to find pharmacologically active compounds better characterized and safer to be administered than the natural plant alkaloids themselves. We concentrate on the eburnamine structures with cerebral activities in this review. Vincamine, vinburnine, vindeburnol, apovincaminate, and vinpocetine (cis-ethyl-apovincaminate) all share modulatory effects on brain circulation and neuronal homeostasis, bear antihypoxic and neuroprotective potencies to various degrees. The most eminent compound of this class of alkaloids is vinpocetine. Since its introduction to the market as a neuroprotective agent many non clinical and clinical studies proved vinpocetine's effects on calmodulin dependent phosphodiesterase E1, on sodium, calcium channels, peripheral benzodiazepine receptor, and glutamate receptors as well as its clinical usefulness in the treatment of post-ischaemic stroke disease states and various disorders of cerebrovascular origin. Lately, positron emission tomography studies proved that vinpocetine has a rapid uptake in the primate and human brain with a heterogeneous distribution pattern (preference areas: thalamus, basal ganglia, and visual cortex) both after intravenous and oral administration. Vinpocetine exerts beneficial effects in cerebral glucose metabolism and regional cerebral blood flow in chronic post-stroke patients.
Collapse
Affiliation(s)
- Adám Vas
- Chemical Works of Gedeon Richter Ltd., Budapest, Hungary.
| | | |
Collapse
|
18
|
Bergström M, Långström B. Pharmacokinetic studies with PET. PROGRESS IN DRUG RESEARCH. FORTSCHRITTE DER ARZNEIMITTELFORSCHUNG. PROGRES DES RECHERCHES PHARMACEUTIQUES 2006; 62:279-317. [PMID: 16329260 DOI: 10.1007/3-7643-7426-8_8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Affiliation(s)
- Mats Bergström
- Uppsala Imanet, GE Health Care, Box 967, SE-751 09 Uppsala, Sweden.
| | | |
Collapse
|
19
|
Gulyás B, Halldin C, Vas A, Banati RB, Shchukin E, Finnema S, Tarkainen J, Tihanyi K, Szilágyi G, Farde L. [11C]Vinpocetine: a prospective peripheral benzodiazepine receptor ligand for primate PET studies. J Neurol Sci 2005; 229-230:219-23. [PMID: 15760643 DOI: 10.1016/j.jns.2004.11.032] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Vinpocetine, a synthetic derivative of the Vinca minor alkaloid vincamine, is a widely used drug in neurological practice. We tested the hypothesis that vinpocetine binds to peripheral benzodiazepine binding sites (PBBS) and is therefore a potential ligand of PBBS. Positron emission tomography (PET) measurements in two cynomolgous monkeys showed that pretreatment with vinpocetine markedly reduced the brain uptake of [11C]PK11195, a known PBBS radioligand. On the other hand, whereas pretreatment with PK11195 increased the brain uptake of [11C]vinpocetine due to the blockade of PBBS in the periphery, it significantly reduced the binding potential (BP) values of [11C]vinpocetine in the whole brain and in individual brain structures to PK11195. These findings indicate that, whereas the two ligands have different affinities to PBBS, vinpocetine is a potent ligand of PBBS, which in turn suggests that the pharmacological activity of vinpocetine may involve the regulation of glial functions.
Collapse
Affiliation(s)
- Balázs Gulyás
- Karolinska Institute, Psychiatry Section, Department of Clinical Neuroscience, Karolinska Hospital, S-17176 Stockholm, Sweden.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Szilágyi G, Nagy Z, Balkay L, Boros I, Emri M, Lehel S, Márián T, Molnár T, Szakáll S, Trón L, Bereczki D, Csiba L, Fekete I, Kerényi L, Galuska L, Varga J, Bönöczk P, Vas A, Gulyás B. Effects of vinpocetine on the redistribution of cerebral blood flow and glucose metabolism in chronic ischemic stroke patients: a PET study. J Neurol Sci 2005; 229-230:275-84. [PMID: 15760651 DOI: 10.1016/j.jns.2004.11.053] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The pharmacological effects of the neuroprotective drug vinpocetine, administered intravenously in a 14-day long treatment regime, on the cerebral blood flow and cerebral glucose metabolism in chronic ischemic stroke patients (n=13) were studied with positron emission tomography in a double-blind design. The regional and global cerebral metabolic rates of glucose (CMRglc) and cerebral blood flow (CBF) as well as vital physiological parameters, clinical performance scales, and transcranial Doppler parameters were measured before and after the treatment period in patient groups treated with daily intravenous infusion with or without vinpocetine. While the global CMRglc values did not change markedly as a result of the infusion treatment with (n=6) or without (n=7) vinpocetine, the global CBF increased and regional CMRglc and CBF values showed marked changes in several brain structures in both cases, with more accentuated changes when the infusion contained vinpocetine. In the latter case the highest rCBF changes were observed in those structures in which the highest regional uptake of labelled vinpocetine was measured in other PET studies (thalamus and caudate nucleus: increases amounting to 36% and 37%, respectively). The findings indicate that a 2-week long intravenous vinpocetine treatment can contribute effectively to the redistribution of rCBF in chronic ischemic stroke patients. The effects are most pronounced in those brain regions with the highest uptake of the drug.
Collapse
Affiliation(s)
- Géza Szilágyi
- National Stroke Center, Department of Vascular Neurology, Semmelweis University, H-1085 Budapest, Hungary
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Scott Mason N, Mathis CA. Positron Emission Tomography Agents for Central Nervous System Drug Development Applications. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2005. [DOI: 10.1016/s0065-7743(05)40004-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2023]
|
22
|
Zhou X, Dong XW, Crona J, Maguire M, Priestley T. Vinpocetine is a potent blocker of rat NaV1.8 tetrodotoxin-resistant sodium channels. J Pharmacol Exp Ther 2003; 306:498-504. [PMID: 12730276 DOI: 10.1124/jpet.103.051086] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Vinpocetine is a clinically used synthetic vincamine derivative with a diverse pharmacological profile that includes action at several ion channels, principally "generic" populations of sodium channels that give rise to tetrodotoxin-sensitive conductances. A number of cell types are known to express tetrodotoxin-resistant (TTXr) sodium conductances, the molecular bases of which have remained elusive until recently. One such TTXr channel, termed NaV1.8, is of particular interest because of its prominent and selective expression in peripheral afferent nerves. The effects of vinpocetine on TTXr channels specifically, are unknown. We have assessed the effects of the drug on cloned rat NaV1.8 channels expressed in a dorsal root ganglion-derived cell line, ND7/23. Vinpocetine produced a concentration- and state-dependent inhibition of NaV1.8 sodium channel activity. Voltage-clamp experiments revealed an approximately 3-fold increase in vinpocetine potency when whole-cell NaV1.8 conductances were elicited from relatively depolarized potentials (-35 mV; IC50 = 3.5 microM) compared with hyperpolarized holding potentials (-90 mV; IC50 = 10.4 microM). Vinpocetine also produced an approximately 22 mV leftward shift in the voltage dependence of NaV1.8 channel inactivation but did not affect the voltage range of channel activation. These properties are reminiscent of several other known sodium channel blockers and suggested that vinpocetine may exhibit frequency-dependent block. Accordingly, tonic block of NaV1.8 channels by vinpocetine (3 microM) increased proportionally with increasing depolarizing commands over the frequency range 0.1 to 1Hz. In summary, the present data demonstrate that vinpocetine is capable of blocking NaV1.8 sodium channel activity and suggest a potential additional utility in various sensory abnormalities arising from abnormal peripheral nerve activity.
Collapse
Affiliation(s)
- Xiaoping Zhou
- CNS/CV Biological Research, Schering-Plough Research Institute, Kenilworth, NJ 07033, USA
| | | | | | | | | |
Collapse
|
23
|
Hall H, Varnäs K, Sandell J, Halldin C, Farde L, Vas A, Kárpáti E, Gulyás B. Autoradiographic evaluation of [11C]vinpocetine binding in the human postmortem brain. ACTA BIOLOGICA HUNGARICA 2002; 53:59-66. [PMID: 12064779 DOI: 10.1556/abiol.53.2002.1-2.7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The main objective ofthe study was to evaluate with autoradiographic technique whether or not [11C]vinpocetine, a compound widely used in the prevention and treatment of cerebrovascular diseases (Cavinton, Gedeon Richter Ltd., Budapest), binds to specific sites in the human brain in post mortem human brain sections. Binding was assessed under four conditions: the incubation was performed using Tris-HCl buffer with or without the addition of salts (0.1% (weight/vol) ascorbic acid, 120 mM NaCl, 5 mM KCl, 2 mM CaCl2 and 1 mM MgCl2), with or without the addition of excess (10 microM) unlabelled vinpocetine. Measurements on digitized autoradiograms indicated that [11C]vinpocetine labelled all grey matter areas in the human brain to a similar extent and no significantly heterogeneous binding could be demonstrated among cortical or subcortical regions. The addition of excess unlabelled vinpocetine lowered the binding slightly in all regions. Although these results indicate that [11C]vinpocetine does not bind to human brain transmitter receptors or transporters with a high affinity (Ki < 10 nM), it cannot be ruled out that the compound binds to receptors and/or transporters with lower affinity.
Collapse
Affiliation(s)
- H Hall
- Karolinska Institutet, Department of Clinical Neuroscience, Karolinska Hospital, Stockholm, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Gulyás B, Halldin C, Sandell J, Karlsson P, Sóvágó J, Kárpáti E, Kiss B, Vas A, Cselényi Z, Farde L. PET studies on the brain uptake and regional distribution of [11C]vinpocetine in human subjects. Acta Neurol Scand 2002; 106:325-32. [PMID: 12460136 DOI: 10.1034/j.1600-0404.2002.01302.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
OBJECTIVES Vinpocetine is a compound widely used in the prevention and treatment of cerebrovascular diseases. It is still not clear whether the drug has a direct and specific effect on neurotransmission or its effects are due to extracerebral actions, such as changes in cerebral blood flow. The main objective of the present investigation was to determine the global uptake and regional distribution of radiolabelled vinpocetine in the human brain in order to explore whether it may have direct central nervous system effects. MATERIAL AND METHODS Three healthy subjects were examined with positron emission tomography and [11C]vinpocetine. The regional uptake was determined in anatomically defined volumes-of-interest. The fractions of [11C]vinpocetine and labelled metabolites in plasma were determined using high pressure liquid chromatography. RESULTS The uptake of [11C]vinpocetine in brain was rapid and 3.7% (mean; n = 4) of the total radioactivity injected was in brain 2 min after radioligand administration. The uptake was heterogeneously distributed among brain regions. When compared with the cerebellum, an a priori reference region, the highest regional uptake was in the thalamus, upper brain stem, striatum and cortex. Following an initial peak, the total concentration of radioactivity in blood was relatively stable with time, whereas the concentration of the unchanged compound decreased with time in an exponential manner. CONCLUSION Vinpocetine, administered intravenously in humans, readily passes the blood-brain barrier and enters the brain. Its regional uptake and distribution in the brain is heterogeneous, indicating binding to specific sites. The brain regions showing increased uptake in the human brain correspond to those in which vinpocetine has been shown to induce elevated metabolism and blood flow. These observations support the hypothesis that vinpocetine has direct neuronal actions in the human brain.
Collapse
Affiliation(s)
- B Gulyás
- Department of Clinical Neuroscience, Psychiatry Section, Karolinska Institute, Stockholm, Sweden.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Vas A, Gulyás B, Szabó Z, Bönöczk P, Csiba L, Kiss B, Kárpáti E, Pánczél G, Nagy Z. Clinical and non-clinical investigations using positron emission tomography, near infrared spectroscopy and transcranial Doppler methods on the neuroprotective drug vinpocetine: a summary of evidences. J Neurol Sci 2002; 203-204:259-62. [PMID: 12417394 DOI: 10.1016/s0022-510x(02)00301-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Vinpocetine (Cavinton, Gedeon Richter, Budapest) is widely used as a neuroprotective drug in the prevention and treatment of cerebrovascular diseases. Vinpocetine is a potent inhibitor of the voltage-dependent Na(+) channels and a selective inhibitor of the Ca(2+)/caldmoduline-dependent phosphodiesterase 1. The clinical efficacy has been supported by several previous studies. Positron emission tomography (PET) is a powerful method to evaluate the fate, the site of action, the pharmacological and physiological effects of a drug in the brain and other organs. We have demonstrated in monkey that the [11C]-labelled vinpocetine rapidly enters the brain after intravenous (i.v.) injection, the maximal uptake being approximately 5% of the total injected radioactivity. The distribution pattern of vinpocetine in the brain was heterogenous, with the highest uptake in the thalamus, basal ganglia and visual cortex. These findings were confirmed in healthy humans, where the i.v. administered [11C]-labelled vinpocetine had a similar distribution pattern. The highest uptake in the brain was 3.71% of the total administered radioactivity. Quite recently, we have shown that [11C]-labelled vinpocetine administered orally to healthy human volunteers also rapidly appears in the brain and shows a similar distribution pattern, the highest uptake being 0.71% of the total administered radioactivity. In two separate sets of clinical studies where chronic ischaemic post-stroke patients were either treated with a single infusion (Study 1) or with daily vinpocetine infusion for 2 weeks (Study 2), we have shown that vinpocetine increases the regional cerebral glucose uptake and to a certain extent glucose metabolism in the so-called peri-stroke region as well as in the relatively intact brain tissue. The 2-week-long treatment also increased the regional cerebral blood flow (CBF) especially in the thalamus, basal ganglia and visual cortex of the nonsymptomatic hemisphere. We have demonstrated the cerebral perfusion-enhancing and parenchymal oxygen extraction-increasing effects of vinpocetine in subacute ischaemic stroke patients by near infrared spectroscopy (NIRS) and transcranial Doppler (TCD) methods.
Collapse
Affiliation(s)
- Adám Vas
- Chemical Works of Gedeon Richter Limited, H-1103 Budapest, Gyömrõi út 19/21 P.O.B. 27, Budapest 1103, Hungary.
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Gulyás B, Vas A, Halldin C, Sóvágó J, Sandell J, Olsson H, Fredriksson A, Stone-Elander S, Farde L. Cerebral uptake of [ethyl-11C]vinpocetine and 1-[11C]ethanol in cynomolgous monkeys: a comparative preclinical PET study. Nucl Med Biol 2002; 29:753-9. [PMID: 12381455 DOI: 10.1016/s0969-8051(02)00319-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PET provides the potential to quantify the distribution of radiolabelled drugs in the human body. In cases when radiolabelled compounds undergo metabolic transformation after administration in vivo, it is necessary to examine the kinetics and distribution of both the labeled mother compound and labeled metabolites. The objective of this study was to assess the extent by which 11C-labeled ethanol, the product arising from the de-esterification of the neuroprotective drug vinpocetine (ethyl-apovincaminate), might contribute to the regional cerebral radioactivity measured by PET after the administration of [ethyl-11C]vinpocetine. In three cynomolgous monkeys PET measurements were made after intravenous bolus injection of both [11C]vinpocetine and 1-[11C]ethanol. There was a marked difference between the regional time-activity curves of [11C]ethanol and [11C]vinpocetine. The distribution pattern obtained with [11C]ethanol was similar to that observed with blood flow tracers such as [15O]water and [15O]butanol. The study shows that although [11C]ethanol may moderately contribute to the brain radioactivity distribution pattern of [11C]vinpocetine, the rapid degradation of [11C]ethanol makes it unlikely that the contribution of this metabolite is of importance. The distinct distribution patterns and kinetics of [11C]vinpocetine and [11C]ethanol also support the view, obtained from our previous observations, that vinpocetine may bind to specific sites in the monkey and human brain, especially in the thalamus.
Collapse
Affiliation(s)
- Balázs Gulyás
- Psychiatry Section, Department of Clinical Neuroscience, Karolinska Institutet, S-171 76 Stockholm, Sweden.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Oxidative damage is present within the brains of patients with Alzheimer's disease (AD), and is observed within every class of biomolecule, including nucleic acids, proteins, lipids and carbohydrates. Oxidative injury may develop secondary to excessive oxidative stress resulting from beta-amyloid-induced free radicals, mitochondrial abnormalities, inadequate energy supply, inflammation or altered antioxidant defences. Treatment with antioxidants is a promising approach for slowing disease progression to the extent that oxidative damage may be responsible for the cognitive and functional decline observed in AD. Although not a uniformly consistent observation, a number of epidemiological studies have found a link between antioxidant intake and a reduced incidence of dementia, AD and cognitive decline in elderly populations. In AD clinical trials molecules with antioxidant properties such as vitamin E and Ginkgo biloba extract have shown modest benefit. A clinical trial with vitamin E is currently ongoing to determine if it can delay progression to AD in individuals with mild cognitive impairment. Combinations of antioxidants might be of even greater potential benefit for AD, especially if the agents worked in different cellular compartments or had complementary activity (e.g. vitamins E, C and ubiquinone). Naturally-occurring compounds with antioxidant capacity are available and widely marketed (e.g. vitamin C, ubiquinone, lipoic acid, beta-carotene, creatine, melatonin, curcumin) and synthetic compounds are under development by industry. Nevertheless, the clinical value of these agents for AD prevention and treatment is ambiguous, and will remain so until properly designed human trials have been performed.
Collapse
Affiliation(s)
- Michael Grundman
- Alzheimer's Disease Cooperative Study, University of California, San Diego, 8950 Villa La Jolla Drive, Suite 2200, La Jolla, California 92037, USA.
| | | | | |
Collapse
|
28
|
Bönöczk P, Gulyás B, Adam-Vizi V, Nemes A, Kárpáti E, Kiss B, Kapás M, Szántay C, Koncz I, Zelles T, Vas A. Role of sodium channel inhibition in neuroprotection: effect of vinpocetine. Brain Res Bull 2000; 53:245-54. [PMID: 11113577 DOI: 10.1016/s0361-9230(00)00354-3] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Vinpocetine (ethyl apovincaminate) discovered during the late 1960s has successfully been used in the treatment of central nervous system disorders of cerebrovascular origin for decades. The increase in the regional cerebral blood flow in response to vinpocetine administration is well established and strengthened by new diagnostical techniques (transcranial Doppler, near infrared spectroscopy, positron emission tomography). The latest in vitro studies have revealed the effect of the compound on Ca(2+)/calmodulin dependent cyclic guanosine monophosphate-phosphodiesterase 1, voltage-operated Ca(2+) channels, glutamate receptors and voltage dependent Na(+)-channels; the latest being especially relevant to the neuroprotective action of vinpocetine. The good brain penetration profile and heterogenous brain distribution pattern (mainly in the thalamus, basal ganglia and visual cortex) of labelled vinpocetin were demonstrated by positron emission tomography in primates and man. Multicentric, randomized, placebo-controlled clinical studies proved the efficacy of orally administered vinpocetin in patients with organic psychosyndrome. Recently positron emission tomography studies have proved that vinpocetine is able to redistribute regional cerebral blood flow and enhance glucose supply of brain tissue in ischemic post-stroke patients.
Collapse
Affiliation(s)
- P Bönöczk
- Chemical Works of Gedeon Richter Ltd., Budapest, Hungary
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|