1,546
Views
8
CrossRef citations to date
0
Altmetric
Research Article

Synthesis, characterization and in vitro evaluation of substituted N-(2-phenylcyclopropyl)carbamates as acetyl- and butyrylcholinesterase inhibitors

, , , , , , & show all
Pages 173-179 | Received 13 Apr 2016, Accepted 05 Jul 2016, Published online: 01 Aug 2016

Abstract

A serie of O-substituted N-2-phenylcyclopropylcarbamates was prepared and characterized. These carbamates were tested as inhibitors of acetylcholinesterase (AChE) and butyrylcholinesterase (BChE). It was found, that these compounds exhibit moderate inhibition activity with values of IC50 in the range of 54.8–94.4 μM (for AChE) and up to 5.8 μM (for BChE). The AChE/BChE selectivity for each carbamate was calculated. These values varied from 0.50 to 9.46, two carbamate derivatives inhibited only AChE selectively. The most promising derivative was prepared in all optically pure forms (four isomers). It was found that individual stereoisomers differed only slightly in the inhibition ability. The cytotoxicity of all carbamates was evaluated using the standard in vitro test with Jurkat cells. With regard to their inhibition activity and cytotoxicity as well as easy preparation, O-substituted N-2-phenylcyclopropylcarbamates can be considered as promising compounds for potential medicinal applications.

Introduction

Alzheimer’s disease (AD) is known as progressive neurodegenerative brain disorder with characteristic clinical and pathological symptomsCitation1–3. AD belongs among most often causation of formation of dementia in the elderly population and it is supposed that about 6% of the population worldwide aged over 65 is affected by ADCitation1–4. AD represents the complex disease, whose exact cause of attendance is unknown at this timeCitation5. One of clinical manifestation of AD is connected with low acetylcholine (ACh) concentration in cholinergic synapses caused by its excessive degradation; ACh being very important neurotransmitter of the cholinergic central nervous system of human organismCitation6,Citation7. ACh is hydrolyzed by cholinesterases (ChEs) to choline and acetic acid. In vertebrates, there are two enzymes which are usually defined as ChEs: acetylcholinesterase (AChE, EC: 3.1.1.7) and butyrylcholinesterase (BChE, EC: 3.1.1.8). AChE plays a crucial role in ACh breaking down in cholinergic brain synapses and neuromuscular junctionsCitation8. The main function of BChE is still unclear. BChE enables the hydrolysis of ACh as well as other estersCitation9–12 and can also act as a scavenger for some toxins by reacting with them before they reach AChECitation13–15. The AD therapy is based on inhibition of the ChEs in order to maintain the proper level of AChCitation16,Citation17. The ChEs are inhibited by several compounds, which can act as reversible or irreversible inhibitors. In AD therapy only reversible inhibitors are usedCitation8,Citation18,Citation19.

On the other hand, BChE substitutes the function of AChE in the later stages of AD, where its activity is significantly lowerCitation10,20–23. According to this published results, the ratio of BChE/AChE in the normal brain was estimated 0.2, whereas the ratio in the brain of people with AD reaches the value ca. 11Citation24. Moreover, selective BChE inhibitors do not exhibit the adverse cholinergic effects, which are characteristic for AChE inhibitorsCitation25–28. Therefore, the research concerned in the selective BChE inhibitors is currently a promising direction in medicinal chemistry research. Furthermore, the AChE and BChE inhibitors respectively can be applied in the therapy of other diseases, including myasthenia gravis, some other dementias, parasitic infections, glaucoma, obstipation or to antagonize muscle relaxationCitation29–31.

Many of the potential or approved commercially available pharmaceutical substances acting as AChE inhibitors contain carbamate functional group among othersCitation16,Citation32; for instance, human drug RivastigminCitation33,Citation34, which is used in the therapy of patients in the early or middle stage of AD. Besides this, many of differently substituted carbamate derivatives are described in literatureCitation35–46, which embody significant inhibition activity against AChE and/or BChE. Unfortunately, the clinical applicability of these compounds is limited with regard to many side effects or demerits, e.g. general or specific toxicity, periphery side effect, short half-life, or gastrointestinal tract disordersCitation47.

The cyclopropane cycle occurs in many of natural products and biologically active substancesCitation48, e.g. terpenes, pheromones, fatty acids and unusual amino acids. The natural, as well as synthetic compounds containing cyclopropane moiety, possess a wide spectrum of biological effects, including enzyme inhibition, fungicidal, herbicidal, antimicrobial, antibiotic, antiviral and cytostatic activityCitation49. Therefore, they can be considered as an interesting class of the organic chemicals. Well known (±)-trans-2-phenylcyclopropyl-1-amine can be mentioned as such cyclopropane derivative, used in human medicine as the monoamine oxidase inhibitorCitation50.

The aim of this work was the preparation and characterization of a series of N-(2-phenylcyclopropyl)carbamate derivatives as compounds, which would contain both above mentioned types of pharmacophores. Subsequently, the inhibition activity of these compounds against AChE and BChE respectively as well as their cytotoxicity should be studied and evaluated. With respect to the fact, that N-(2-phenylcyclopropyl)carbamate derivatives are chiral compounds, the most promising derivative with the highest inhibition activity should be prepared in all of the configuration forms (four stereoisomers), which would enable verification of the influence of absolute configuration at stereogenic centers on resulting biological activity.

Materials and methods

Lipophilicity

The traditional shake-flask method was used for determination of logPow valuesCitation51,Citation52. First, the two solvents were mutually saturated at the temperature of the experiment in a way described in literatureCitation52. Then, the determination logPow was performed subsequently: n-octanol (1.5 ml) and n-octanol solution of tested compound (10 μl, 0.01 M) were placed into the test tube. This mixture was intensively shaken for 15 min. Then, 1 ml of this mixture was placed into the quartz cuvette and its absorbance at the absorption maximum wavelength was measured. Thereby the value of absorbance corresponding to 100% of the tested compound in n-octanol was obtained. The reference solution was n-octanol. Into the other test tube n-octanol (1.5 ml), water (1.5 ml) and n-octanol solution of tested compound (10 μl, 0.01 M) were placed. This mixture was intensively shaken for 15 min and then centrifuged (3000 rpm, 10 min). Then 1 ml of this mixture was placed into the quartz cuvette and its absorbance at the absorption maximum wavelength was measured. The reference solution was n-octanol again. Thereby the percentage content of tested compound in the n-octanol layer (%) was obtained. Subsequently, the logPow (logPow =log(c1/c2), where c1 and c2 are molar concentrations of tested compound in n-octanol and water) was calculated. For each compound, at least two determinations were performed. Obtained results are shown in .

Table 1. The list of prepared carbamates 3ao and their inhibition activity against AChE and BChE expressed as IC50; lipophilicity logPow and AChE/BChE selectivity.

Biological assay

AChE, BChE, acetylthiocholine (ATCh), 5,5′-dithiobis-2-nitrobenzoic acid (DTNB) and n-octanol were purchased from Sigma-Aldrich (St. Louis, MO). KH2PO4, Na2HPO4·12H2O, KCl, NaCl, dimethyl sulfoxide (DMSO) were purchased from Penta (Prague, Czech Republic).

All tested compounds were dissolved in DMSO (concentration 0.01 M) and diluted in demineralized water (concentration 0.001 M). The ability of tested compounds to inhibit AChE (from electric eel) and BChE (from equine serum) was determined using modified Ellman’s method at 25 °C in the presence of phosphate-buffered saline (PBS, 0.1 M, pH 7.4) in glass cuvette with 1 cm optical path. The enzyme activity in the total reaction mixture (2 ml) was 0.2 U/ml, the concentration of ATCh 40 μM and concentration of DTNB 0.1 mM for all reactions. The IC50 value was obtained from the dependence v0/vi versus concentration of tested compound (inhibitor), where v0 is the reaction rate of uninhibited reaction and vi is the reaction rate of inhibited reaction (for given concentration of inhibitor).

First, v0 was determined. Into the cuvette, PBS (0.1 M, pH 7.4), DTNB and ATCh were placed. The enzymatic reaction was started by adding the enzyme. The dependence of absorbance (λ = 412 nm) versus time was observed for 70 s (reference solution contained PBS, DTNB and ATCh) and then reaction rate (v0) was calculated (v = ΔAt). The measurement was performed in triplicate at least and average v0 was determined.

Then, vi (for given concentration of inhibitor) was determined. Into the cuvette DTNB, ATCh, chosen volume of suitably diluted inhibitor (to achieve required concentration of inhibitor in the total reaction mixture) and certain volume of PBS (to achieve a total volume of reaction mixture 2 ml after adding of enzyme) were placed. The enzymatic reaction was started by adding of the enzyme. The dependence of absorbance (λ = 412 nm) versus time was observed for 70 s (reference solution was the same as for uninhibited reaction) and then reaction rate (vi) was calculated. Five different concentration of inhibitor were used and each measurement was performed in duplicate at least.

Finally, the dependence v0/vi versus concentration of inhibitor was constructed and IC50 was calculated from obtained equation of regression curve for y = 2 (coming out from the definition of IC50). The obtained IC50 values of all tested compounds are shown in . The comparison of both IC50 values (for AChE and BChE) of each compounds 3ao was statistically analyzed with GraphPad Prism (version 5.00; La Jolla, CA) statistical software using unpaired t-test: p ≤ 0.01 or p < 0.05.

In vitro cytotoxicity assay

Cell lines

The human T-cell acute lymphoblastic leukemia cell line Jurkat was purchased from European Collection of Cell Cultures (UK). These cells were cultured in Roswell Park Memorial Institute 1640 medium (Life Technologies, Carlsbad, CA) supplemented with 10% fetal bovine serum, 1% non-essential amino acid, 1% penicillin/streptomycin, 2 mM L-glutamine, 1 mM sodium pyruvate and 10 mM 4-(2-hydroxyethyl)-piperazine-1-ethanesulfonic acid buffer (all supplements from Life Technologies) in a humidified atmosphere containing 5% CO2 at 37 °C.

Cytotoxicity measurement

All compounds were tested using a standard colorimetric method measuring a tetrazolium salt reduction via mitochondrial dehydrogenase activity in Jurkat cells. The cells were seeded in density 133 000 cells per well in a 96-well plate. The cells were treated with each of the tested substances dissolved in DMSO. All compounds were prepared in six incubation concentrations (1–500 μM) in triplicates. Also, the vehiculum controls (0.2% DMSO) were prepared in triplicates. After 48 h incubation, cell survival was determined using Cell Proliferation Kit II (XTT, Roche, Germany) according to manufacturer’s instructions. In brief, XTT reagent (2,3-bis-(2-methoxy-4-nitro-5-sulfophenyl)-2H-tetrazolium-5-carboxanilide) was added to each well and incubated for 3 h at 37 °C; absorbance was then measured at 470 nm using a 96-multiwell microplate reader Tecan Infinite M200 (Tecan Group Ltd., Männedorf, Switzerland). Viability was calculated as described in the paper by Havelek et al. using the following formula: (%) viability = (A470sample − A470blank)/(A470control − A470blank) × 100, where A470 is the absorbance of utilized XTT formazan measured at 470 nmCitation53. Data were statistically analyzed with GraphPad Prism (version 5.00; La Jolla, CA) statistical software using unpaired t-test: p ≤ 0.01.

Results

Chemistry

The serie of N-(2-phenylcyclopropyl)carbamate derivatives 3ao was prepared by three-step synthesis with good yields. All of the newly prepared carbamates 3ao were characterized by means of melting point, 1H and 13C NMR spectroscopy and high-resolution mass spectroscopy. The optical purity of non-racemic derivatives was determined by means of chiral HPLC and their optical rotatory power was measured. The purity of all compounds was verified by means of elemental analysis.

Lipophilicity

The values of partition coefficient were determined using traditional shake-flask methodCitation51,Citation52,Citation54. The obtained results are shown in . All tested compounds could be divided into two groups: Group 1 includes compounds 3ai (R = 9 alkyls) and Group 2 includes compounds 3jo (R = 6 glycosyls). All compounds in the Group 1 fulfill in fact the condition of Lipinski’s rule of five (except trans(±)3c with logPow = 6.63). The lowest value of logPow in the Group 1 was obtained for compound trans(±)3d. But there is no significant trend in increasing/decreasing of logPow values depending on chain length. The compounds in Group 2 containing protecting isopropylidene groups show high lipophilicity, the logPow values are in range from 4.10 for trans(±)3k to 5.87 for (1S,2S)-cis(–)3j.

Inhibition studies

The ability of all prepared carbamates to inhibit AChE from electric eel (Electrophorus electricus) and BChE from equine serum was determined in vitro using modified Ellman’s method. The effectiveness of the inhibitors was expressed as IC50 value representing the concentration of an inhibitor which is necessary for reduction of enzyme activity (or reaction rate) to 50%. The obtained results are shown in .

In vitro cytotoxicity assay

The cytotoxicity of all prepared compounds was screened using a standard tetrazolium salt XTT cytotoxicity assay after 48 h of treatment. As shown in , most of the evaluated compounds of Group 1 exhibited cytotoxic activity against Jurkat cells at concentrations ≥100 μM. Contrary thereto, trans(±)3g at all tested concentrations and trans(±)3d at concentrations ≤ 250 μM do not significantly affect cell viability of Jurkat cells. Similarly, also the majority of the compounds in Group 2 exhibited cytotoxic activity against Jurkat cells at concentrations ≥100 μM (). Exceptions are trans(±)3m with statistically insignificant effect on Jurkat cell viability at concentrations ≤ 250 μM and trans(±)3n together with trans(−)3j with statistically insignificant effect on Jurkat cell survival at concentrations ≤ 100 μM.

Table 2. Cell viability of Jurkat cells measured by using XTT cytotoxicity assay after treatment with different concentrations of the tested compounds. Treatment for 48 h. Viability is referred to cells treated with DMSO 0.2% (control). Negative control (NC) was treated with the vehicle (DMSO 0.2%) used for diluting the tested substance.

Discussion

The key intermediate for the synthesis of carbamate derivatives 3al was 2-phenylcyclopropanecarboxylic acid 2. It was prepared by standard two-step synthesis, including at first cyclopropane cycle formation by the action of ethyl diazoacetate onto styrene afforded cyclopropanecarboxylate 1 and subsequent hydrolysis of ester group under basic condition (Scheme 1).

Scheme 1. Three-step synthesis of carbamates 3a–o.

Scheme 1. Three-step synthesis of carbamates 3a–o.

The cyclopropanation leading to racemic trans/cis (±)-1 was conducted at high temperature (120–130 °C) without the presence of any catalyst with the yield of 62%Citation55. The individual isomers formed trans(±)-1 and cis(±)-1 were not separated, the minor cis isomer was removed from the mixture by recrystallization after its hydrolysis to the corresponding acid trans/cis (±)-2. The optically pure forms of ester 1 were prepared according to the protocol described by Evans et al.Citation56 employing the enantioselective catalyst based on copper(I) complex of commercially available chiral bisoxazoline derivative. This method can be considered as the most convenient for the preparation of all of the isomers of 1, because, according to the original paper, it enables the formation of the esters 1 with excellent optical purity (95–98% ee) and with diastereomeric ratio 77/23 (trans/cis), i.e. relatively favorable ratio for less thermodynamically stable cis form. In our case, we observed optical purity 96% ee, the ratio of trans/cis, we found 75/25 and isolated yields were 79–85%. The asymmetric cyclopropanation with a recoverable and recyclable form of a catalyst based on copper(I) complex of chiral bisoxazoline ligand supported by Amberlite® IR-120 was also studied. Unfortunately, the chemical yields and first of all optical purity of the cyclopropane derivatives 1 obtained in this way were unsatisfactory. The individual cis and trans diastereomers of non-racemic forms of ester 1 were separated by means of column chromatography.

The carboxylic acid 2 was transformed to individual carbamates 3al by the action of diphenylphosphoryl azide in the presence of triethylamine and appropriate alcohol (Scheme 1)Citation57. This one-pot reaction involved at first acylazide formation, which underwent Curtius rearrangement under heating at 80 °C. The corresponding isocyanate formed was attacked by the present hydroxy derivative afforded desired carbamate 3al. The presence of isocyanate derivative as the intermediate of this synthetic process needed to perform the reaction under strictly non-aqueous condition. The reacting alcohol was used as the solvent in the case of preparation of carbamates 3ae (Method A). The carbamates 3fl formed from higher alcohols were prepared in toluene (1,4-dioxane for 3jl) containing slight excess (1.2 eq.) of respective hydroxy derivative (Method B)Citation58. The yields of this transformation (40–80%) depend on the individual alcohol used.

The serie of prepared carbamates also includes three members (3jl) derived from protected monosaccharides (glucose, galactose and inositol) and their corresponding deprotected forms 3mo. The preparation of such designed carbamates was inspired by the hypothesis that they can possess a high level of transport abilityCitation59–61 over blood-brain barrier via GLUT-family sugar/polyol transport facilitatorsCitation62. Better transport of the active molecules can then lead to their enhanced concentration in intracellular space of CNS. The carbamates 3jl were deprotected (deacetalization of monosaccharide moiety) by the treatment of aqueous trifluoroacetic acid (TFA) (1/1)Citation63. It was found out, that both isopropylidene groups were removed completely within 3 h at room temperature, without observation of any significant decomposition of carbamate group as well as ring-opening of cyclopropane cycle (Scheme 2). The mild condition (1% TFA in methanol, 10 h of reflux) led to only partial deprotection, with prevailing 1,2-monoprotected derivative.

Scheme 2. Deprotection of O-glycosyl carbamate derivatives 3j–l; the survey of individual carbamates 3j–o.

Scheme 2. Deprotection of O-glycosyl carbamate derivatives 3j–l; the survey of individual carbamates 3j–o.

Many drugs cross biological membranes through passive transport, which strongly depends on their lipophilicity. The necessary condition for exploitation of chosen compound in vivo as the brain AChE inhibitor is its ability to cross the blood-brain barrier. For assessment of this ability, the partition coefficient in system n-octanol:water (usually expressed as Pow or logPow) can be usedCitation64,Citation65. Experimentally, it is done by partitioning the molecule between two immiscible phases (water and n-octanol) and determining the Pow value as the ratio of the tested compound concentration in n-octanol and in water. The partition coefficient serves as a quantitative descriptor of lipophilicity and is one of the key determinants of pharmacokinetic properties. According to Lipinski’s rule of five, logPow should not exceed 5Citation66. Expectably, the corresponding deprotected compounds 3mo exhibit relatively low lipophilicity. Generally, it is possible to conclude, that all tested compound show adequate lipophilicity and could cross the blood-brain barrier.

Ellman’s methodCitation67 is widely used for measuring of ChE activity and effectiveness of ChE inhibitors. It is a simple, rapid and direct method to determine the SH and −S−S− groupsCitation68. ChE activity is measured indirectly by quantifying the concentration of 2-nitro-5-sulfanylbenzoic acid ion formed in the reaction between disulfide reagent DTNB and thiocholine, a product of substrate (i.e. ATCh) hydrolysis catalyzed by ChE.

Based on the obtained results, it is obvious that all tested compounds in Group 1 show moderate inhibition of AChE from electric ell and BChE from equine serum. Generally, it is possible to conclude, that compound 3ai show higher selectivity to BChE. The IC50 values for AChE inhibition are in the range of 60.45–77.63 μM, whereas the most potent inhibitor is trans(±)3i and the least effective is trans(±)3e. The IC50 values for BChE inhibition are for all compounds lower than for AChE (9.05–40.77 μM), i.e. that compounds 3ai show higher selectivity to BChE. The most effective BChE inhibitor is trans(±)3g with IC50 =9.05 μM.

All tested compounds in Group 2 show moderate inhibition of AChE and BChE, with the exception of quite potent BChE inhibitors trans(±)3j and trans(+)3j. Compounds trans(±)3j, trans(+)3j, trans(–)3j, (1S,2S)-cis(–)3j and (1R,2R)-cis(–)3j show higher selectivity to BChE. The interesting result is, that trans(±)3j does not show significantly lower inhibitory effectiveness than trans(+)3j, trans(–)3j. On the contrary, its anticholinesterase activity is comparable to the activity of trans(+)3j. Compounds 3ko show (if it is possible to determine) higher selectivity to AChE. The IC50 values for AChE inhibition are in the range of 63.3–89.63 μM, and for BChE inhibition in the range of 105.57–152.46 μM, whereas the anti-BChE activity for trans(±)3m and trans(±)3o was undetectable under given conditions.

Among these derivatives, trans(±)3d and trans(+)3j can be considered as the most promising compounds for further study due to their moderate inhibitory activity against AChE, high inhibitory effects on BChE and concurrently low cytotoxicity against blood originated Jurkat cell line.

Conclusion

In this study, the serie of N-(2-phenylcyclopropyl)carbamate derivatives 3ao was prepared by three-step synthesis. These compounds were tested for their inhibition of AChE from electric eel and BChE from equine serum. Generally, it is possible to conclude, that all tested compounds show moderate inhibition activity, where the most effective inhibitor was found the derivative trans(+)3j. Evaluating cytotoxic activity of all derivatives, the derivative trans(±)3g can be considered as the most promising AChE and BChE inhibitor due to its potent activity and concurrently low cytotoxicity against Jurkat cells. In this regard, the further research efforts on these compounds would appear to hold great potential, especially in the light of the recent findings of the important role of BChE in the central cholinergic transmission and during the AD therapy. Overall, the obtained results indicated that substituted N-(2-phenylcyclopropyl)carbamates have the potential to be good candidates for AChE and BChE inhibitors and therefore they deserve further attention with the aim of their possible medicinal applications.

Declaration of interest

The authors report no conflicts of interest.

Supplementary material available online

Supplemental material

IENZ_1212193_Supplementray_Information.pdf

Download PDF (419.5 KB)

Acknowledgements

The authors acknowledge the financial support from the project GAČR 14-00925S.

References

  • Lu LC, Bludau J. Alzheimer’s disease. Santa Barbara: Greenwood Publishing Group; 2011
  • Ballard C, Gauthier S, Corbett A, et al. Alzheimer’s disease. Lancet 2011;377:1019–31
  • Amemori T, Jendelova P, Ruzicka J, et al. Alzheimer’s disease: mechanism and approach to cell therapy. Int J Mol Sci 2015;16:26417–51
  • Sun ZQ, Tu LX, Zhuo FJ, Liu SX. Design and discovery of Novel Thiazole acetamide derivatives as anticholinesterase agent for possible role in the management of Alzheimer’s. Bioorg Med Chem Lett 2016;26:747–50
  • Alzheimer’s Association. Alzheimer’s disease facts and figures. Alzheimers Dement 2015;11:332–84
  • Contestabile A. The history of the cholinergic hypothesis. Behav Brain Res 2011;221:334–40
  • Bartus RT, Dean RL, Beer B, Lippa AS. The cholinergic hypothesis of geriatric memory dysfunction. Science 1982;217:408–14
  • Colović MB, Krstić DZ, Lazarević-Pašti TD, et al. Acetylcholinesterase inhibitors: pharmacology and toxicology. Curr Neuropharmacol 2013;11:315–35
  • Groner E, Ashani Y, Schorer-Apelbaum D, et al. The kinetics of inhibition of human acetylcholinesterase and butyrylcholinesterase by two series of novel carbamates. Mol Pharmacol 2007;71:1610–17
  • Greig NH, Utsuki T, Yu Q, et al. A new therapeutic target in Alzheimer’s disease treatment: attention to butyrylcholinesterase. Curr Med Res Opin 2001;17:159–65
  • Schwarz S, Lucas SD, Sommerwerk S, Csuk R. Amino derivatives of glycyrrhetinic acid as potential inhibitors of cholinesterases. Bioorg Med Chem 2014;22:3370–8
  • Saeed A, Mahesar PA, Zaib S, et al. Synthesis, cytotoxicity and molecular modelling studies of new phenylcinnamide derivatives as potent inhibitors of cholinesterases. Eur J Med Chem 2014;78:43–53
  • Soreq H, Seidman S. Acetylcholinesterase – new roles for an old actor. Nat Rev Neurosci 2001;2:294–302
  • Aurbek N, Thiermann H, Eyer F, et al. Suitability of human butyrylcholinesterase as therapeutic marker and pseudo catalytic scavenger in organophosphate poisoning: a kinetic analysis. Toxicology 2009;259:133–9
  • Kovarik Z, Katalinić M, Sinko G, et al. Pseudo-catalytic scavenging: searching for a suitable reactivator of phosphorylated butyrylcholinesterase. Chem Biol Interact 2010;187:167–71
  • Benzi G, Moretti A. Is there a rationale for the use of acetylcholinesterase inhibitors in the therapy of Alzheimer’s disease? Eur J Pharmacol 1998;346:1–13
  • Kandemirli F, Saraçoglu M, Kovalishyn V. Human acetylcholinesterase inhibitors: electronic-topological and neural network approaches to the structure-activity relationships study. Mini Rev Med Chem 2005;5:479–87
  • Onor ML, Trevisiol M, Aguglia E. Rivastigmine in the treatment of Alzheimer’s disease: an update. Clin Interv Aging 2007;2:17–32
  • Cacabelos R. Donepezil in Alzheimer’s disease: from conventional trials to pharmacogenetics. Neuropsychiatr Dis Treat 2007;3:303–33
  • Giacobini E. Cholinesterase inhibitors: from Calabar bean to Alzheimer therapy. In: Giacobini E, ed. Cholinesterases and cholinesterase inhibitors. London: Martin Dunitz; 2000:181–226
  • Mesulam MM, Guillozet A, Shaw P, et al. Acetylcholinesterase knockouts establish central cholinergic pathways and can use butyrylcholinesterase to hydrolyze acetylcholine. Neuroscience 2002;110:627–39
  • Ballard CG. Advances in the treatment of Alzheimer’s disease: benefits of dual cholinesterase inhibition. Eur Neurol 2002;47:64–70
  • Mushtaq G, Greig NH, Khan JA, Kamal MA. Status of acetylcholinesterase and butyrylcholinesterase in Alzheimer’s disease and type 2 diabetes mellitus. CNS Neurol Disord Drug Targets 2014;13:1432–9
  • Greig NH, Utsuki T, Ingram DK, et al. Selective butyrylcholinesterase inhibition elevates brain acetylcholine, augments learning and lowers Alzheimer beta-amyloid peptide in rodent. Proc Natl Acad Sci U S A 2005;47:17213–18
  • Giacobini E. Cholinesterase inhibitors: new roles and therapeutic alternatives. Pharmacol Res 2004;50:433–40
  • Cherif O, Allouche F, Chabchoub F, et al. Isoxazolotacrines as non-toxic and selective butyrylcholinesterase inhibitors for Alzheimer’s disease. Future Med Chem 2014;6:1883–91
  • Brus B, Košak U, Turk S, et al. Discovery, biological evaluation, and crystal structure of a novel nanomolar selective butyrylcholinesterase inhibitor. J Med Chem 2014;57:8167–79
  • Zeb A, Hameed A, Khan L, et al. Quinoxaline derivatives: novel and selective butyrylcholinesterase inhibitors. Med Chem 2014;10:724–9
  • Pohanka M. Cholinesterases, a target of pharmacology and toxicology. Biomed Pap Med Fac Univ Palacky Olomouc Czech Rep 2011;155:219–29
  • Heinig R, Zimmer D, Yeh S, Krol GJ. Development, validation and application of assays to quantify metrifonate and 2,2-dichlorovinyl dimethylphosphate in human body fluids. J Chromatogr B Biomed Sci Appl 2000;741:257–69
  • Ibach B, Haen E. Acetylcholinesterase inhibition in Alzheimer’s disease. Curr Pharm Des 2004;10:231–51
  • Štěpánková Š, Komers K. Cholinesterases and cholinesterase inhibitors. Curr Enz Inhib 2008;4:160–71
  • O'neil MJ. The Merck index. 13th ed. New Jersey: Merck & Co., Inc.; 2001
  • Jann MW. Rivastigmine, a new-generation cholinesterase inhibitor for the treatment of Alzheimer’s disease. Pharmacotherapy 2000;20:1–12
  • Sawatzky E, Wehle S, Kling B, et al. Discovery of highly selective and nanomolar carbamate-based butyrylcholinesterase inhibitors by rational investigation into their inhibition mode. J Med Chem 2016;59:2067–82
  • Liew KF, Chan KL, Lee CY. Blood-brain barrier permeable anticholinesterase aurones: synthesis, structure-activity relationship, and drug-like properties. Eur J Med Chem 2015;94:195–210
  • Bohn P, Gourand F, Papamicaël C, et al. Dihydroquinoline carbamate derivatives as “bio-oxidizable” prodrugs for brain delivery of acetylcholinesterase inhibitors: [11C] radiosynthesis and biological evaluation. ACS Chem Neurosci 2015;6:737–44
  • Sedlák M, Hanusek J, Drabina P, et al. Substituted benzyl N-phenylcarbamates – their solvolysis and inhibition activity to acetylcholinesterase and butyrylcholinesterase. Arkivoc 2009;vii:1–11
  • Imramovský A, Pejchal V, Štěpánková Š, et al. Synthesis and in vitro evaluation of new derivatives of 2-substituted-6-fluorobenzo[d]thiazoles as cholinesterase inhibitors. Bioorg Med Chem 2013;21:1735–48
  • Vorčáková K, Štěpánková Š, Sedlák M, Vytřas K. Electrochemical sensors for the estimation of the inhibitory effect of phenylcarbamates to cholinesterase. Chemosensors 2015;3:274–83
  • Saxena J, Meloni D, Huang MT, et al. Ethynylphenyl carbonates and carbamates as dual-action acetylcholinesterase inhibitors and anti-inflammatory agents. Bioorg Med Chem Lett 2015;25:5609–12
  • Camerino E, Wong DM, Tong F, et al. Difluoromethyl ketones: potent inhibitors of wild type and carbamate-insensitive G119S mutant Anopheles gambiae acetylcholinesterase. Bioorg Med Chem Lett 2015;25:4405–11
  • Verma A, Wong DM, Islam R, et al. 3-Oxoisoxazole-2(3H)-carboxamides and isoxazol-3-yl carbamates: resistance-breaking acetylcholinesterase inhibitors targeting the malaria mosquito, Anopheles gambiae. Bioorg Med Chem 2015;23:1321–40
  • Anand P, Singh B. Synthesis and evaluation of substituted 4-methyl-2-oxo-2H-chromen-7-yl phenyl carbamates as potent acetylcholinesterase inhibitors and anti-amnestic agents. Med Chem 2013;9:694–702
  • Anand P, Singh B. Synthesis and evaluation of novel carbamate substituted flavanone derivatives as potent acetylcholinesterase inhibitors and anti-amnestic agents. Med Chem Res 2013;22:1648–59
  • Ma H-J, Xie RL, Zhao QF, et al. Synthesis and insecticidal activity of novel carbamate derivatives as potential dual-binding site acetylcholinesterase inhibitors. J Agric Food Chem 2010;58:12817–21
  • León R, Garcia AG, Marco-Contelles J. Recent advances in the multitarget-directed ligands approach for the treatment of Alzheimer’s disease. Med Res Rev 2013;33:139–89
  • Gnad F, Reiser O. Synthesis and applications of beta-aminocarboxylic acids containing a cyclopropane ring. Chem Rev 2003;103:1603–24
  • Rappopot Z, ed. The chemistry of the cyclopropyl group vol. 1–2. New York: Wiley-VCH; 1987
  • Tedeschi RE. Monoamine oxidase inhibition, US Patent 2,997,422; 1961
  • Sangster JJ. Octanol-water partition coefficients of simple organic compounds. Phys Chem Ref Data 1989;18:1111–227
  • OECD Guidelines for the Testing of Chemicals, Section 1, Books / OECD Guidelines for the Testing of Chemicals, Section 1/Test No. 107: Partition Coefficient (n-octanol/water): Shake Flask Method; [cited 2016 Jul 18]. Available from: http://www.oecd-ilibrary.org/environment/test-no-107-partition-coefficient-n-octanol-water-shake-flask-method_9789264069626-en
  • Havelek R, Siman P, Cmielova J, et al. Differences in vanadocene dichloride and cisplatin effect on MOLT-4 leukemia and human peripheral blood mononuclear cells. Med Chem 2012;8:615–21
  • Berthod A, Carda-Broch S. Determination of liquid-liquid partition coefficients by separation methods. J Chromatogr A 2004;1037:3–14
  • Burger A, Yost WL. Arylcycloalkylamines. I. 2-Phenylcyclopropylamine. J Am Chem Soc 1948;70:2198–201
  • Evans DA, Woerpel KA, Hinman MM, Faul MM. Bis(oxazolines) as chiral ligands in metal-catalyzed asymmetric reactions. Catalytic, asymmetric cyclopropanation of olefins. J Am Chem Soc 1991;113:726–8
  • Tsai TY, Hsu T, Chen CT, et al. Novel trans-2-aryl-cyclopropylamine analogues as potent and selective dipeptidyl peptidase IV inhibitors. Bioorg Med Chem 2009;17:2388–99
  • Ninomiya K, Shioiri T, Yamada S. Phosphorus in organic synthesis – VII: diphenyl phosphorazidate (DPPA). A new convenient reagent for a modified curtius reaction. Tetrahedron 1974;30:2151–7
  • Fernández C, Nieto O, Fontenla JA, et al. Synthesis of glycosyl derivatives as dopamine prodrugs: interaction with glucose carrier GLUT-1. Org Biomol Chem 2003;1:767–71
  • Calvaresi EC, Hergenrother PJ. Glucose conjugation for the specific targeting and treatment of cancer. Chem Sci 2013;4:2319–33
  • Uriel C, Egron MJ, Santarromana M, et al. Hexose keto-C-glycoside conjugates: design, synthesis, cytotoxicity, and evaluation of their affinity for the glucose transporter Glut-1. Bioorg Med Chem 1996;4:2081–90
  • Joost HG, Thorens B. The extended GLUT-family of sugar/polyol transport facilitators: nomenclature, sequence characteristics, and potential function of its novel members (review). Mol Membr Biol 2001;18:247–56
  • Shakeel UR, Khursheed AB, Shabir HL, Fayaz AM. Click chemistry inspired facile synthesis and bioevaluation of novel triazolyl analogs of D-(+)-pinitol. Arab J Chem 2015. [Epub ahead of print]. doi:10.1016/j.arabjc.2015.10.009
  • Leo A, Hansch C, Elkins D. Partition coefficients and their uses. Chem Rev 1971;71:525–616
  • Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev 2001;46:3–26
  • Lipinski CA. Lead- and drug-like compounds: the rule-of-five revolution. Drug Discov Today Technol 2004;1:337–41
  • Ellman GL, Courtney KD, Andres V, Feather-Stone RM. A new and rapid colorimetric determination of acetylcholinesterase activity. Biochem Pharmacol 1961;7:88–95
  • Ou S, Kwok KC, Wang Y, Bao H. An improved method to determine SH and -S-S- group content in soymilk protein. Food Chem 2004;88:317–20

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.