synthesis and pharmacological properties of new gaba ...if-pan.krakow.pl/pjp/pdf/2012/4_817.pdf ·...

17
Synthesis and pharmacological properties of new GABA uptake inhibitors Kinga Sa³at 2 , Anna Wiêckowska 1 , Krzysztof Wiêckowski 1 , Georg C. Höfner 3 , Jacek Kamiñski 1 , Klaus T. Wanner 3 , Barbara Malawska 1 , Barbara Filipek 2 , Katarzyna Kulig 1 Chair of Pharmaceutical Chemistry, Department of Physicochemical Drug Analysis, Department of Pharmacodynamics, Jagiellonian University, Medical College, Medyczna 9, PL 30-688 Kraków, Poland Department of Pharmacy, Centre of Drug Research, Ludwig-Maximilians University, Munich, Butenandtstr. 5-13, 81377 Munich, Germany Correspondence: Katarzyna Kulig, e-mail: [email protected] Abstract: Background: g-Aminobutanoic acid (GABA) is the principal inhibitory neurotransmitter in the mammalian central nervous system. The identification and subsequent development of the GABA transport inhibitors which enhance the GABA-ergic transmission has shown the important role that GABA transporters play in the control of numerous functions of the nervous system. Compounds which inhibit GABA uptake are used as antiepileptic drugs (tiagabine – a selective GAT1 inhibitor), they are also being investigated for other indications, including treatment of psychosis, general anxiety, sleep disorders, drug addiction or acute and chronic pain. Methods: In this paper, the synthesis of 2-substituted-4-(1,3-dioxoisoindolin-2-ylo)-butanamides and 2-substituted-4-amino- butanoic acids derivatives is described. These compounds were tested in vitro for their ability to inhibit GABA uptake. The inhibi- tory potency towards murine plasma membrane GABA transporters (mGAT1-4) was performed as [ H]GABA uptake assay based on stably transfected HEK cells. Compound 18, which demonstrated the highest affinity for mGAT1-4 (pIC ranged from 4.42 for mGAT1 to 5.07 for mGAT3), was additionally investigated in several behavioral tests in mice. Results: Compound 18 increased the locomotor activity (14–38%) and had anxiolytic-like properties in the four-plate test (ED = 9.3 mg/kg). It did not show analgesic activity in acute pain model, namely the hot plate test, however, it was antinociceptive in the acetic acid-induced writhing test (ED = 15.3 mg/kg) and in the formalin model of tonic pain. In the latter assay, it diminished no- cifensive behavior in both phases and in the first (neurogenic) phase of this test the obtained ED value (5.3 mg/kg) was similar to morphine (3.0 mg/kg). Conclusion: Compound 18 exhibited significant anxiolytic-like properties and was antinociceptive in some models of pain in mice. Moreover, it did not impair animals’ motor coordination in the chimney test. Some of the described pharmacological activities of compound 18 can be partly explained based on its affinity for plasma membrane GABA transporters. Key words: GABA uptake inhibitors, mGAT1-4, four-plate test, pain models, motor coordination, butanamides, amino acids Abbreviations: BBB – blood brain barrier, CDI – N,N’- carbonyldiimidazole, CDMT – 2-chloro-4,6-dimethoxy-1,3,5- triazine, CNS – central nervous system, DMAC – N,N-di- methylacetamide, GABA – g-aminobutanoic acid, GAT – GABA transporter, NMM – N-methylmorpholine, TBAB – tetrabu- tylammonium bromide, TLC – thin layer chromatography Introduction 4-Aminobutanoic acid (g-aminobutyric acid; GABA) is the main inhibitory neurotransmitter in the mam- malian central nervous system (CNS). Currently known 817

Upload: vanthuy

Post on 07-Feb-2018

224 views

Category:

Documents


2 download

TRANSCRIPT

Page 1: Synthesis and pharmacological properties of new GABA ...if-pan.krakow.pl/pjp/pdf/2012/4_817.pdf · Synthesis and pharmacological properties of new GABA uptake inhibitors ... a selective

Synthesis and pharmacological properties of new

GABA uptake inhibitors

Kinga Sa³at2, Anna Wiêckowska1, Krzysztof Wiêckowski1,

Georg C. Höfner3, Jacek Kamiñski1, Klaus T. Wanner3,

Barbara Malawska1, Barbara Filipek2, Katarzyna Kulig1

1Chair of Pharmaceutical Chemistry, Department of Physicochemical Drug Analysis,

2Department of

Pharmacodynamics, Jagiellonian University, Medical College, Medyczna 9, PL 30-688 Kraków, Poland

3Department of Pharmacy, Centre of Drug Research, Ludwig-Maximilians University, Munich, Butenandtstr. 5-13,

81377 Munich, Germany

Correspondence: Katarzyna Kulig, e-mail: [email protected]

Abstract:

Background: g-Aminobutanoic acid (GABA) is the principal inhibitory neurotransmitter in the mammalian central nervous system.

The identification and subsequent development of the GABA transport inhibitors which enhance the GABA-ergic transmission has

shown the important role that GABA transporters play in the control of numerous functions of the nervous system. Compounds

which inhibit GABA uptake are used as antiepileptic drugs (tiagabine – a selective GAT1 inhibitor), they are also being investigated

for other indications, including treatment of psychosis, general anxiety, sleep disorders, drug addiction or acute and chronic pain.

Methods: In this paper, the synthesis of 2-substituted-4-(1,3-dioxoisoindolin-2-ylo)-butanamides and 2-substituted-4-amino-

butanoic acids derivatives is described. These compounds were tested in vitro for their ability to inhibit GABA uptake. The inhibi-

tory potency towards murine plasma membrane GABA transporters (mGAT1-4) was performed as [3H]GABA uptake assay based

on stably transfected HEK cells. Compound 18, which demonstrated the highest affinity for mGAT1-4 (pIC50 ranged from 4.42 for

mGAT1 to 5.07 for mGAT3), was additionally investigated in several behavioral tests in mice.

Results: Compound 18 increased the locomotor activity (14–38%) and had anxiolytic-like properties in the four-plate test (ED50 =

9.3 mg/kg). It did not show analgesic activity in acute pain model, namely the hot plate test, however, it was antinociceptive in the

acetic acid-induced writhing test (ED50 = 15.3 mg/kg) and in the formalin model of tonic pain. In the latter assay, it diminished no-

cifensive behavior in both phases and in the first (neurogenic) phase of this test the obtained ED50 value (5.3 mg/kg) was similar to

morphine (3.0 mg/kg).

Conclusion: Compound 18 exhibited significant anxiolytic-like properties and was antinociceptive in some models of pain in mice.

Moreover, it did not impair animals’ motor coordination in the chimney test. Some of the described pharmacological activities of

compound 18 can be partly explained based on its affinity for plasma membrane GABA transporters.

Key words:

GABA uptake inhibitors, mGAT1-4, four-plate test, pain models, motor coordination, butanamides, amino acids

Abbreviations: BBB – blood brain barrier, CDI – N,N’-

carbonyldiimidazole, CDMT – 2-chloro-4,6-dimethoxy-1,3,5-

triazine, CNS – central nervous system, DMAC – N,N-di-

methylacetamide, GABA – g-aminobutanoic acid, GAT – GABA

transporter, NMM – N-methylmorpholine, TBAB – tetrabu-

tylammonium bromide, TLC – thin layer chromatography

Introduction

4-Aminobutanoic acid (g-aminobutyric acid; GABA)

is the main inhibitory neurotransmitter in the mam-

malian central nervous system (CNS). Currently known

Pharmacological Reports, 2012, 64, 817�833 817

Pharmacological Reports2012, 64, 817�833ISSN 1734-1140

Copyright © 2012by Institute of PharmacologyPolish Academy of Sciences

Page 2: Synthesis and pharmacological properties of new GABA ...if-pan.krakow.pl/pjp/pdf/2012/4_817.pdf · Synthesis and pharmacological properties of new GABA uptake inhibitors ... a selective

drugs enhancing the GABAergic transmission can in-

fluence either the GABAergic receptors (benzodiaze-

pines, baclofen), GABA metabolism (vigabatrin) or its

uptake from the synaptic cleft (tiagabine) [17, 46].

The GABA transporters (GAT), which are involved

in the re-uptake of this neurotransmitter from the syn-

aptic cleft, are regarded a very promising biological

target for searching new, therapeutically active mole-

cules. At present, five different transporters for GABA

are distinguished, one of which is the vesicular

GABA transporter (VGAT) belonging to the SLC32

family. Four other ones (GAT1, GAT2, GAT3, GAT4)

are plasma membrane transporters [9] which belong

to the SLC6 superfamily of Na+-dependent transport-

ers, the activity of which is regulated by the extracel-

lular Na+/Cl- levels [46].

The deficiency of GABAergic signaling is in-

volved in several CNS-derived diseases, including

anxiety, sleep disorders, epilepsy or chronic pain. In

view of this, the GABA uptake inhibitors are very

promising agents with their potential application in

the treatment of epilepsy, anxiety, pain, drug abuse,

sleep disorders and others [37]. Until now, only tiaga-

bine – a selective inhibitor of GAT1 transporter is

used in the clinic in the treatment of partial seizures.

Recently, it has been shown to be effective in a variety

of non-epileptic conditions, including psychosis, gen-

eral anxiety and sleep disorders, bruxism, drug addic-

tion, acute and chronic pain, tonic spasm, posttrau-

matic stress, essential tremor and migraine prophy-

laxis [13, 16, 22–24, 27, 31, 34, 42–45, 48, 54]. It was

also used in the treatment of impulsive aggression

[19, 29] and acute mania [18, 24, 49].

The anticonvulsant activity of GAT inhibitors other

to tiagabine seems to be distinct as compared to tiaga-

bine. There are reports suggesting that non-GAT1 in-

hibitors, especially agents acting at GAT2 and GAT4,

are very interesting as potential prospects for the future

epilepsy treatment [36, 37, 57]. Noteworthy, mixed

GAT inhibitors are reported to have much broader

spectrum of anticonvulsant activity as compared to

compounds with affinity only for GAT1 [13, 38].

They can also be pharmacological tools for the re-

search regarding the biological role of GAT, espe-

cially non- GAT1 GABA transporters.

Since the physiological role of GAT other than

GAT1 is poorly established [10, 41, 54, 55], there is

a great need for searching new GABA uptake inhibi-

tors which could be used as potential drugs or tools

for pharmacological research in the future.

The chemical structures of known GABA uptake

inhibitors can be classified into several groups of

compounds, with the most important acyclic and cy-

clic GABA analogues and lipophilic aromatic GABA

analogues able to penetrate the blood brain barrier

(BBB) (Fig. 1). Tiagabine (1) represents the lipophilic

(R)-nipecotic acid analogue. The substitution of the

tiagabine side-chain by another GABA mimicking

amino acid fragment has led to the GAT1/GAT2

semi-selective inhibitor, EF1502 (2) [9]. A large

number of lipophilic GABA uptake inhibitors with the

diaryl or triaryl units have been studied resulting in

very potent compounds 3. However, tri-cyclic struc-

tures 4 led to the compounds with a reduced affinity

for the GABA transporters [28].

818 Pharmacological Reports, 2012, 64, 817�833

Fig. 1. Selected structures of lipophilicGABA uptake inhibitors

Page 3: Synthesis and pharmacological properties of new GABA ...if-pan.krakow.pl/pjp/pdf/2012/4_817.pdf · Synthesis and pharmacological properties of new GABA uptake inhibitors ... a selective

Previously, we found [47, and our unpublished data]

that several compounds with non-selective affinity for

GAT1–4 exert analgesic, anxiolytic-like and anticonvul-

sant activities in rodents. Among the compounds tested,

the most interesting one was 2-(4-benzhydrylpiperazin-

1-yl)-N-(4-chlorobenzyl)-4-hydroxybutanamide (5), for

which the obtained pIC50 values ranged from 4.56 for

mGAT2 to 5.06 for mGAT3 [26]. In this context, the

goal of our research was the development of derivatives

of 2-subtituted-4-(1,3-dioxoisoindolin-2-yl)-butanamides

and 2-substituted-4-aminobutanoic acids as novel

GABA uptake inhibitors.

In this work, we report on the synthesis and in vitro

pharmacological studies of two series of new ana-

logues of compound 5. The first group of compounds

represents analogues of active 2-substituted 4-hydr-

oxybutanamides, where the hydroxyl group was trans-

formed into lipophilic phthalimido residue. In posi-

tion 2 of these phthalimidobutanamides, N-phenyl- or

4-substituted N-phenyl- or N-diphenylmethyl- pipera-

zine was introduced, as a fragment corresponding to

the anticonvulsant active arylpiperazine derivatives of

4-hydroxybutanoic acid and a part mimicking the bi-

aryl moieties of known GAT inhibitors. The second,

small group of compounds consists of derivatives of

2-substituted 4-aminobutanoic acid, containing aryl-

piperazinyl unit, as analogues of 2-substituted phthal-

imidobutanamides. Schematic structures of the com-

pounds designed are presented in Figure 2.

The newly synthesized compounds were tested for

their GABA uptake inhibitory activity. Compound 18,

which had the most beneficial pharmacological pro-

file in vitro, was additionally tested in the behavioral

assays, such as hot plate, writhing, and formalin pain

models, locomotor activity, chimney and four-plate

tests in mice.

Materials and Methods

CHEMICAL PART

Unless otherwise noted, the starting materials were

obtained from commercial suppliers (Merck, Sigma-

Aldrich) and used without purification. Melting

points were determined in open glass capillaries on

the Electrothermal 9300 apparatus and were uncor-

rected. Reactions were monitored by thin layer chro-

matography (TLC) using silica gel 60 F254 plates

(Merck) and following solvent systems: S1 (acetone/

chloroform, 1/1, v/v), S2 (chloroform/methanol/etha-

noic acid, 12/2/1, v/v/v) and S3 (chloroform/metha-

nol/ammonia 25%, 2/2/1, v/v/v). The spots were visu-

alized under UV lamp and by iodine solution (0.05 M

in 10% HCl). 1H NMR spectra were recorded with Var-

ian Mercury 300 spectrometer (300 MHz) in CDCl3,D2O and DMSO-d6, with signal of solvent as an inter-

nal standard. Elemental analyses were carried out on

a Vario EL III Model Elemental Analyzer. Synthesis

of compounds 7, 9, 11 and 15–18 was described else-

where [26, 39].

The synthetic routes leading to the new compounds

are presented in Figure 3.

Pharmacological Reports, 2012, 64, 817�833 819

Pharmacological activity of new GABA uptake inhibitorsKinga Sa³at et al.

Fig. 2. Schematic structure of designed2-substituted-4-(1,3-dioxoisoindolin-2-yl)-butanamides and 2-substituted-4-aminobutanoic acids

Page 4: Synthesis and pharmacological properties of new GABA ...if-pan.krakow.pl/pjp/pdf/2012/4_817.pdf · Synthesis and pharmacological properties of new GABA uptake inhibitors ... a selective

820 Pharmacological Reports, 2012, 64, 817�833

Fig. 3. Synthesis of compounds 15–38

Page 5: Synthesis and pharmacological properties of new GABA ...if-pan.krakow.pl/pjp/pdf/2012/4_817.pdf · Synthesis and pharmacological properties of new GABA uptake inhibitors ... a selective

Synthesis of 3-substituted dihydrofuran-

2(3H)-one derivatives 8, 9 (general procedure)

Anhydrous K2CO3 (1 eq.) was added to the solution of

relevant amine (1 eq.) in 20 ml of solvent (acetonitrile

or Me2CO) and the mixture was stirred at room tem-

perature for 0.5 h. Then, a solution of 3-bromo-

dihydrofuran-2(3H)-one (1 eq.) in 5 ml of appropriate

solvent was added dropwise and stirring was contin-

ued for 24 h. In the case of the synthesis of compound

9, tetrabutylammonium bromide (TBAB) (0.1 eq.)

was added. After the reaction was completed, the pre-

cipitate was filtered off and the filtrate was concen-

trated under vacuum. Crude product (3) obtained was

recrystallized from isopropanol.

3-[4-(4-Fluorophenyl)piperazin-1-yl]dihydrofuran-

2(3H)-one (8)

Reagents and conditions: 3-bromodihydrofuran-2(3H)-

one (16.5 g, 100 mmol), 1-(4-fluorophenyl)piperazine

(18.0 g, 100 mmol), K2CO3 (13.8 g, 100 mmol), ace-

tone (250 ml); yield: 77.3%; m.p. 152–153ºC (isopro-

panol). Analysis: calc. for C14H17N2O2F: C 63.62, H

6.48, N 10.60%; found: C 63.68, H 6.76, N 10.58%. 1HNMR (CDCl3, d, ppm): 2.30–2.46 (m, 2H, CHCH2),2.66–2.74 (m, 4H, piperazine), 2.80–2.94 (m, 4H,

piperazine), 3.43 (t, J = 8.9 Hz, 1H, CH), 4.32–4.47 (m,

2H, CH2O), 7.15–7.40 (m, 4H, Ar-H). TLC: 0.61 (S2).

3-[4-(4-Methoxyphenyl)piperazin-1-yl]dihydrofuran-

2(3H)-one (9)

Reagents and conditions: 3-bromodihydrofuran-2(3H)-

one (16.5 g, 100 mmol), 1-(4-methoxyphenyl)pipera-

zine (19.2 g, 100 mmol), K2CO3 (13.8 g, 100 mmol),

TBAB (10 mmol, 3.22 g), acetonitrile (250 ml); yield:

47.6%; m.p. 192–193ºC (acetone). Analysis: calc. for

C15H20N2O3: C 57.60, H 6.77, N 8.96%; found: C

57.68, H 6.76, N 8.98%. 1H NMR (DMSO-d6, d

ppm): 2.34–2.51 (m, 2H,CHCH2), 2.66–2.74 (m, 4H,

piperazine), 2.80–2.94 (m, 4H, piperazine), 3.43 (t,

J = 8.7 Hz, 1H, CH), 3.83 (s, 3H, CH3) 4.28–4.44 (m,

2H, CH2O), 7.15–7.40 (m, 4H, Ar-H). TLC: 0.80 (S2).

Synthesis of 2-substituted- 4-(1,3-

dioxoisoindolin-2-yl)butanoic acid derivatives

12–14 (general procedure)

A mixture of the relevant 3-substituted lactone 8–10

(50 mmol) and 9.26 g (50 mmol) of potassium phthal-

imide in 100 ml of N,N-dimethylacetamide (DMAC)

was refluxed for 10 h. The reaction mixture was

cooled, and the solution was acidified with NaHSO4.The precipitated product was collected, washed with

water (100 ml) and crystallized from DMF.

4-(1,3-Dioxoisoindolin-2-yl)-2-(4-(4-

fluorophenyl)piperazin-1-yl)butanoic acid (12)

Reagents and conditions: lactone 8 (13.2 g, 50 mmol),

yield: 37.0%; m.p. 235–236ºC. Analysis: calc. for

C22H22N3O4F: C 64.22, H 5.39, N 10.21%; found: C

64.27, H 5.36, N 10.15%. 1H NMR (CDCl3, d ppm):

1.90–2.00 (m, 2H, CHCH2CH2), 2.54–2.67 (m, 4H,

piperazine), 2.75–2.94 (m, 4H, piperazine), 3.49 (t, J =

6.5 Hz, 1H, CH), 4.26 (t, J = 7.2 Hz, 2H, CHCH2CH2),7.15–7.40 (m, 4H, Ar-H), 7.76–7.89 (m, 4H, phtha-

limide), 10.21 (s, 1H, OH)l. TLC: 0.61 (S1).

4-(1,3-Dioxoisoindolin-2-yl)-2-[4-(4-methoxyphenyl)-

piperazin-1-yl]butanoic acid (13)

Reagents and conditions: lactone 9 (13.8 g, 50 mmol),

yield: 46.7%; m.p. 190–191ºC. Analysis: calc. for

C23H25N3O5: C 65.24, H 5.95, N 9.92%; found: C

65.27, H 5.86, N 9.15%. 1H NMR (CDCl3, d ppm):

1.92–2.07 (m, 2H, CHCH2CH2), 2.52–2.67 (m, 4H,

piperazine), 2.72–2.91 (m, 4H, piperazine), 3.54 (t,

J = 6.5 Hz, 1H, CH), 3.83 (s, 3H, CH3), 4.24 (t, J =

7.2 Hz, 2H, CHCH2CH2), 7.05–7.27 (m, 10H, Ar-H),

7.84–7.99 (m, 4H, phthalimide), 10.31 (s, 1H, OH).

TLC: 0.66 (S1).

2-(4-Benzhydrylpiperazin-1-yl)-4-(1,3-

dioxoisoindolin-2-yl)butanoic acid (14)

Reagents and conditions: lactone 10 (16.8 g, 50 mmol),

yield: 46.7%; m.p. 199–200ºC. Analysis: calc. for

C29H29N3O4: C 72.03, H 6.04, N 8.69%; found: C

72.07, H 6.01, N 8.55%. 1H NMR (CDCl3, d, ppm):

1.90–2.14 (m, 2H, CHCH2CH2), 2.50–2.66 (m, 4H,

piperazine), 2.77–2.98 (m, 4H, piperazine), 3.64 (t,

J = 6.5 Hz, 1H, CH), 4.64 (t, J = 7.2 Hz, 2H,

CHCH2CH2), 5.14 (s, 1H, CH), 7.05–7.27 (m, 4H,

Ar-H), 7.84–7.99 (m, 4H, phthalimide), 10.31 (s, 1H,

OH). TLC: 0.36 (S2).

Pharmacological Reports, 2012, 64, 817�833 821

Pharmacological activity of new GABA uptake inhibitorsKinga Sa³at et al.

Page 6: Synthesis and pharmacological properties of new GABA ...if-pan.krakow.pl/pjp/pdf/2012/4_817.pdf · Synthesis and pharmacological properties of new GABA uptake inhibitors ... a selective

Synthesis of 2-substituted 4-(1,3-dioxoisoindolin-

2-yl)butanamides (19–34)

General procedure 1 (GP1)

A solution of N,N’-carbonyldiimidazole (CDI) (1 eq.)

in DMF was added to a solution of 2-substituted

4-(1,3-dioxoisoindolin-2-yl) butanoic acid derivative

(1 eq.) in DMF; after 0.5 h stirring at room tempera-

ture, amine (1.5 eq) in DMF was added to the reaction

mixture. After stirring for another 24 h at room tem-

perature, the reaction mixture was cooled down on ice

bath and dissolved in water. The obtained solid prod-

uct was crystallized from ethanol or isopropanol.

General procedure 2 (GP2)

2-Substituted 4-(1,3-dioxoisoindolin-2-yl)butanoic acid

derivative (1 eq.) and 2-chloro-4,6-dimethoxy-1,3,5-

triazine (CDMT) (1 eq.) were dissolved in DMAC

and the mixture was cooled to 0ºC. Then, after stirring

for 3 h at 0ºC, N-methylmorpholine (NMM) (1 eq.)

and a solution of amine (1 eq.) in DMAC were added

and the reaction mixture was stirred for 1 h at 0ºC and

for another 20 h at room temperature. After that, the

resulting mixture was cooled down on ice bath and

dissolved in water. The obtained solid product was fil-

tered and crystallized from ethanol.

4-(1,3-Dioxoisoindolin-2-yl)-N-(4-methoxybenzyl)-

2-(4-phenylpiperazin-1-yl)butanamide (19)

Procedure GP1, reagents and conditions: compound

11 (3.9 g, 10 mmol), CDI (1.6 g, 10 mmol), 4-

methoxybenzylamine (1.3 g, 10 mmol), yield: 74.5%;

m.p. 142–143ºC. Analysis: calc. for C30H32N4O4: C

70.29, H 6.29, N 10.93%; found: C 70.27, H 6.31, N

10.95%. 1H NMR (CDCl3, d, ppm): 1.84–2.04 (m,

2H, CHCH2CH2), 2.47–2.59 (m, 4H, piperazine),

2.77–2.98 (m, 4H, piperazine), 3.56 (t, J = 6.9 Hz, 1H,

CH), 3.83 (s, 3H, CH3), 4.24 (t, J = 7.0 Hz, 2H,

CHCH2CH2), 4.40 (s, 2H, NHCH2), 6.87–7.27 (m,

9H, Ar-H), 7.84–7.99 (m, 4H, phthalimide), 8.03 (s,

1H, amide). TLC: 0.52 (S2).

N-(2-Chlorobenzyl)-4-(1,3-dioxoisoindolin-2-

yl)-2-(4-phenylpiperazin-1-yl)butanamide (20)

Procedure GP2, reagents and conditions: compound

11 (3.9 g, 10 mmol), CDMT (1.6 g, 10 mmol), NMM

(1.0 g, 10 mmol), 4-methoxybenzylamine (1.3 g, 10

mmol), yield: 74.5%; m.p. 149–150ºC; Analysis: calc.

for C29H29N4O3Cl: C 67.37, H 5.65, N 10.84%; found:

C 67.37, H 5.06, N 10.08%. 1H NMR (CDCl3, d,

ppm): 1.97–2.24 (m, 2H, CHCH2CH2), 2.52–2.69 (m,

4H, piperazine), 2.81–2.98 (m, 4H, piperazine), 3.56

(t, J = 6.7 Hz, 1H, CH), 4.25 (t, J = 7.1 Hz, 2H,

CHCH2CH2), 4.46 (s, 2H, NHCH2), 6.94–7.27 (m,

9H, Ar-H), 7.88–8.04 (m, 4H, phthalimide), 8.13 (s,

1H, amide). TLC: 0.86 (S2).

4-(1,3-Dioxoisoindolin-2-yl)-N-phenyl-2-(4-

phenylpiperazin-1-yl)butanamide (21)

Procedure GP1, reagents and conditions: compound 11

(1.9 g, 5 mmol), CDI (0.8 g, 5 mmol), aniline (0.5 g, 5

mmol), yield: 52.5%; m.p. 132–133ºC; Analysis: calc.

for C28H28N4O3: C 71.78, H 6.02, N 11.96%; found: C

71.77, H 6.03, N 11.95%. 1H NMR (CDCl3, d, ppm):

1.91–2.19 (m, 2H, CHCH2CH2), 2.41–2.55 (m, 4H,

piperazine), 2.79–2.98 (m, 4H, piperazine), 3.56 (t, J =

6.9 Hz, 1H, CH), 4.17 (t, J = 7.4 Hz, 2H, CHCH2CH2),6.87–7.27 (m, 10H, Ar-H), 7.84–7.99 (m, 4H, phtha-

limide), 8.17 (s, 1H, amide). TLC: 0.80 (S2).

2-[4-Oxo-3-(4-phenylpiperazin-1-yl)-4-(pyrrolidin-

1-yl)butyl]isoindoline-1,3-dione (22)

Procedure GP1, reagents and conditions: compound 11

(1.9 g, 5 mmol), CDI (0.8 g, 5 mmol), pyrrolidine (0.4 g,

5 mmol), yield: 65.4%; m.p. 134–135ºC. Analysis:

calc. for C26H30N4O3: C 69.93, H 6.77, N 12.55%;

found: C 70.07, H 6.73, N 12.75%. 1H NMR (CDCl3,d, ppm): 1.74–1.87 (t, J = 5.9 Hz, 4H, CH2CH2 pyrro-

lidine), 1.91–2.19 (m, 2H, CHCH2CH2), 2.45–2.68

(m, 4H, piperazine), 2.81–2.98 (m, 4H, piperazine),

3.52–3.69 (m, 5H, CH,CH2N pyrrolidine), 4.17 (t, J =

7.4 Hz, 2H, CHCH2CH2), 6.93–7.27 (m, 5H, Ar-H),

7.84–7.99 (m, 4H, phthalimide). TLC: 0.90 (S2).

N-Benzyl-4-(1,3-dioxoisoindolin-2-yl)-2-[4-(4-

fluorophenyl)piperazin-1-yl]butanamide (23)

Procedure GP2, reagents and conditions: compound 12

(2.1 g, 5 mmol), CDMT (0.9 g, 5 mmol), NMM (0.5 g,

5 mmol), benzylamine (0.5 g, 5 mmol), yield: 54.4%;

m.p. 156–157ºC. Analysis: calc. for C29H29N4O3F: C

69.58, H 5.84, N 11.19%, found: C 69.53, H 5.85, N

11.95%. 1H NMR (CDCl3, d ppm): 2.08–2.21 (m, 2H,

CHCH2CH2), 2.71–2.81 (m, 4H, piperazine), 3.05–

3.18 (m, 4H, piperazine),3.36 (t, J =7.7 Hz, 1H, CH),

822 Pharmacological Reports, 2012, 64, 817�833

Page 7: Synthesis and pharmacological properties of new GABA ...if-pan.krakow.pl/pjp/pdf/2012/4_817.pdf · Synthesis and pharmacological properties of new GABA uptake inhibitors ... a selective

4.53 (t, J = 7.1Hz, 2H, CHCH2CH2), 4.56 (s, 2H,

NHCH2), 6.87–7.27 (m, 9H, Ar-H), 7.88–8.04 (m,

4H, phthalimide), 8.13 (s, 1H, amide). TLC: 0.88 (S2).

4-(1,3-Dioxoisoindolin-2-yl)-2-[4-(4-fluoro-

phenyl)piperazin-1-yl]-N-4-methylbenzyl-

butanamide (24)

Procedure GP2, reagents and conditions: compound 12

(2.1 g, 5 mmol), CDMT (0.9 g, 5 mmol), NMM (0.5 g,

5 mmol), 4-methylbenzylamine (0.6 g, 5 mmol), yield:

63.9%; m.p. 147–148ºC. Analysis: calc. for C30H31N4O4F:

C 70.02, H 6.07, N 10.08%, found: C 70.33, H 6.05,

N 10.09%. 1H NMR (CDCl3, d ppm): 2.08–2.20 (m,

2H, CHCH2CH2), 2.35 (s, 3H, CH3), 2.70–2.80 (m,

4H, piperazine), 3.06–3.18 (m, 4H, piperazine), 3.20

(t, J =7.0 Hz, 1H, CH), 4.53 (t, J = 6.1Hz, 2H,

CHCH2CH2), 4.56 (s, 2H, NHCH2), 6.87–7.27 (m,

8H, Ar-H), 7.88–8.04 (m, 4H, phthalimide), 8.13 (s,

1H, amide). TLC: 0.94 (S2).

4-(1,3-Dioxoisoindolin-2-yl)-2-[4-(4-

fluorophenyl)piperazin-1-yl]-N-4-fluorobenzyl-

butanamide (25)

Procedure GP2, reagents and conditions: compound

12 (2.1 g, 5 mmol), CDMT (0.9 g, 5 mmol), NMM

(0.5 g, 5 mmol), 4-fluorobenzylamine (0.6 g, 5 mmol),

yield: 58.0%; m.p. 136–148ºC. Analysis: calc. for

C29H28N4O3F2: C 67.17, H 5.44, N 10.80%, found: C

67.13, H 5.45, N 10.89%. 1H NMR (CDCl3, d ppm):

2.08–2.20 (m, 2H, CHCH2CH2), 2.72–2.80 (m, 4H,

piperazine), 3.06–3.17 (m, 4H, piperazine), 3.56 (t,

J = 7.2 Hz, 1H, CH), 4.53 (t, J = 6.4 Hz, 2H,

CHCH2CH2), 4.56 (s, 2H, NHCH2), 6.87–7.27 (m,

8H, Ar-H), 7.88–8.04 (m, 4H, phthalimide), 8.13 (s,

1H, amide). TLC: 0.89 (S2).

N-(4-Chlorobenzyl)-4-(1,3-dioxoisoindolin-2-

yl)-2-[4-(4-fluorophenyl)piperazin-1-yl]butan-

amide (26)

Procedure GP2, reagents and conditions: compound

12 (2.1 g, 5 mmol), CDMT (0.9 g, 5 mmol), NMM

(0.5 g, 5 mmol), 4-chlorobenzylamine (0.7 g, 5 mmol),

yield: 54.5%; m.p. 181–182ºC. Analysis: calc. for

C29H28N4O3ClF: C 65.10, H 5.28, N 10.47%, found: C

65.15, H 5.39, N 10.58%. 1H NMR (CDCl3, d ppm):

2.08–2.20 (m, 2H, CHCH2CH2), 2.72–2.80 (m, 4H,

piperazine), 3.06–3.17 (m, 4H, piperazine), 3.56 (t,

J = 7.2 Hz, 1H, CH), 4.53 (t, J = 6.4 Hz, 2H,

CHCH2CH2), 4.56 (s, 2H, NHCH2), 6.87–7.27 (m,

8H, Ar-H), 7.88–8.04 (m, 4H, phthalimide), 8.13 (s,

1H, amide). TLC: 0.93 (S2).

N-Benzyl-4-(1,3-dioxoisoindolin-2-yl)-2-[4-(4-

methoxyphenyl)piperazin-1-yl]butanamide (27)

Procedure GP2, reagents and conditions: compound 13

(2.1 g, 5 mmol), CDMT (0.9 g, 5 mmol), NMM (0.5 g,

5 mmol), benzylamine (0.5 g, 5 mmol), yield: 56.6%;

m.p. 140–141ºC. Analysis: calc. for C30H32N4O4: C

70.29, H 6.29, N 10.93%, found: C 70.15, H 6.39, N

10.98%. 1H NMR (CDCl3, d ppm): 2.08–2.20 (m, 2H,

CHCH2CH2), 2.72–2.80 (m, 4H, piperazine), 3.06–

3.17 (m, 4H, piperazine), 3.56 (t, J = 7.2 Hz, 1H, CH),

3.83 (s, 3H, CH3), 4.53 (t, J = 6.4 Hz, 2H,

CHCH2CH2), 4.56 (s, 2H, NHCH2), 6.87–7.27 (m,

9H, Ar-H), 7.88–8.04 (m, 4H, phthalimide), 8.13 (s,

1H, amide). TLC: 0.81 (S2).

4-(1,3-dioxoisoindolin-2-yl)-2-[4-(4-

methoxyphenyl)piperazin-1-yl]-N-(4-

methylbenzyl)butanamide (28)

Procedure GP2, reagents and conditions: compound 13

(2.1 g, 5 mmol), CDMT (0.9 g, 5 mmol), NMM (0.5 g,

5 mmol), 4-methylbenzylamine (0.6 g, 5 mmol), yield:

44.1%; m.p. 136–137ºC. Analysis: calc. for C31H34N4O4:C 70.70, H 6.51, N 10.64%, found: C 70.75, H 6.59, N

10.68%. 1H NMR (CDCl3, d ppm): 2.08–2.20 (m, 2H,

CHCH2CH2), 2.34 (s, 3H, CH3), 2.72–2.80 (m, 4H,

piperazine), 3.06–3.17 (m, 4H, piperazine), 3.56 (t, J

=7.2 Hz, 1H, CH), 3.83 (s, 3H, CH3O), 4.53 (t, J = 6.4

Hz, 2H, CHCH2CH2), 4.56 (s, 2H, NHCH2), 6.87–7.27

(m, 8H, Ar-H), 7.88–8.04 (m, 4H, phthalimide), 8.13

(s, 1H, amide). TLC: 0.86 (S2).

4-(1,3-Dioxoisoindolin-2-yl)-N-(4-fluorobenzyl)-

2-[4-(4-methoxyphenyl)piperazin-1-yl]butan-

amide (29)

Procedure GP2, reagents and conditions: compound 13

(2.1 g, 5 mmol), CDMT (0.9 g, 5 mmol), NMM (0.5 g,

5 mmol), 4-fluorobenzylamine (0.6 g, 5 mmol), yield:

64.1%; m.p. 141–142ºC. Analysis: calc. for C30H31N4O4F:

C 67.91, H 5.89, N 10.56%, found: C 67.75, H 5.59, N

10.68%. 1H NMR (CDCl3, d ppm): 2.08–2.20 (m, 2H,

CHCH2CH2), 2.72–2.80 (m, 4H, piperazine),

3.06–3.17 (m, 4H, piperazine), 3.56 (t, J =7.2 Hz, 1H,

CH), 3.83 (s, 3H, CH3O), 4.53 (t, J = 6.4 Hz, 2H,

Pharmacological Reports, 2012, 64, 817�833 823

Pharmacological activity of new GABA uptake inhibitorsKinga Sa³at et al.

Page 8: Synthesis and pharmacological properties of new GABA ...if-pan.krakow.pl/pjp/pdf/2012/4_817.pdf · Synthesis and pharmacological properties of new GABA uptake inhibitors ... a selective

CHCH2CH2), 4.56 (s, 2H, NHCH2), 6.87– 7.27 (m,

8H, Ar-H), 7.88–8.04 (m, 4H, phthalimide), 8.13 (s,

1H, amide). TLC: 0.82 (S2).

N-(4-chlorobenzyl)-4-(1,3-dioxoisoindolin-2-

yl)-2-[4-(4-methoxyphenyl)piperazin-1-yl]bu-

tanamide (30)

Procedure GP2, reagents and conditions: compound 13

(2.1 g, 5 mmol), CDMT (0.9 g, 5 mmol), NMM (0.5 g,

5 mmol), 4-chlorobenzylamine (0.6 g, 5 mmol), yield:

64.1%; m.p. 141–142ºC. Analysis: calc. for C30H31N4O4Cl:

C 65.87, H 5.71, N 10.24%, found: C 65.75, H 5.79,

N 10.28%. 1H NMR (CDCl3, d ppm): 2.08–2.20 (m,

2H, CHCH2CH2), 2.72–2.80 (m, 4H, piperazine),

3.06–3.17 (m, 4H, piperazine), 3.56 (t, J =7.2 Hz, 1H,

CH), 3.83 (s, 3H, CH3O), 4.53 (t, J = 6.4 Hz, 2H,

CHCH2CH2), 4.56 (s, 2H, NHCH2), 6.87–7.27 (m,

8H, Ar-H), 7.88–8.04 (m, 4H, phthalimide), 8.13 (s,

1H, amide). TLC: 0.86 (S2).

2-(4-Benzhydrylpiperazin-1-yl)-N-(4-benzyl)-4-

(1,3-dioxoisoindolin-2-yl)butanamide (31)

Procedure GP1, reagents and conditions: compound

14 (2.4 g, 5 mmol), CDI (0.8 g, 5 mmol), benzylamine

(0.4 g, 5 mmol), yield: 65.4%; m.p. 201–202oC.

Analysis: calc. for C36H36N4O3: C 75.50, H 6.34, N

9.78%, found: C 75.57, H 6.43, N 9.75%. 1H NMR

(CDCl3, d ppm): 1.91–2.19 (m, 2H, CHCH2CH2),2.45–2.68 (m, 4H, piperazine), 2.81–2.98 (m, 4H,

piperazine), 3.52 (t, J =7.2 Hz, 1H, CH), 4.17 (t, J =

7.4 Hz, 2H, CHCH2CH2), 4.49 (s, 2H, NHCH2), 5.14 (s,

1H, CHAr2), 6.93–7.27 (m, 15H, Ar-H), 7.84–7.99 (m,

4H, phthalimide), 8.17 (s, 1H, amide). TLC: 0.75 (S2).

2-(4-Benzhydrylpiperazin-1-yl)-4-(1,3-dioxoiso-

indolin-2-yl)-N-(4-methylbenzyl)butanamide (32)

Procedure GP1, reagents and conditions: compound

14 (2.4 g, 5 mmol), CDI (0.8 g, 5 mmol), 4-methyl-

benzylamine (0.5 g, 5 mmol), yield: 61.4%; m.p.

202–203ºC. Analysis: calc. for C37H38N4O3: C 75.74,

H 6.53, N 9.55%, found: C 75.77, H 6.53, N 9.65%.1H NMR (CDCl3, d ppm): 1.91–2.19 (m, 2H,

CHCH2CH2), 2.34 (s, 3H, CH3), 2.45–2.68 (m, 4H,

piperazine), 2.81–2.98 (m, 4H, piperazine), 3.48 (t, J

=7.2 Hz, 1H, CH), 4.17 (t, J = 7.4 Hz, 2H,

CHCH2CH2), 4.58 (s, 2H, NHCH2), 5.14 (s, 1H,

CHAr2), 6.93–7.27 (m, 14H, Ar-H), 7.84–7.99 (m,

4H, phthalimide), 8.17 (s, 1H, amide). TLC: 0.75 (S2).

2-(4-Benzhydrylpiperazin-1-yl)-4-(1,3-dioxoiso-

indolin-2-yl)-N-(4-fluorobenzyl)butanamide (33)

Procedure GP1, reagents and conditions: compound

14 (2.4 g, 5 mmol), CDI (0.8 g, 5 mmol), 4-fluor-

benzylamine (0.5 g, 5 mmol), yield: 73.6%; m.p.

205–206ºC. Analysis: calc. for C36H35N4O3F: C

73.20, H 5.97, N 9.49%, found: C 73.17, H 5.93, N

9.45%. 1H NMR (CDCl3, d ppm): 1.91–2.19 (m, 2H,

CHCH2CH2), 2.45–2.68 (m, 4H, piperazine), 2.81–

2.98 (m, 4H, piperazine), 3.49 (t, J =7.2 Hz, 1H,

CH),4.17 (t, J = 7.4 Hz, 2H, CHCH2CH2), 4.57 (s, 2H,

NHCH2), 5.14 (s, 1H, CHAr2), 6.93–7.27 (m, 14H,

Ar-H), 7.84–7.99 (m, 4H, phthalimide), 8.17 (s, 1H,

amide). TLC: 0.70 (S2).

2-(4-Benzhydrylpiperazin-1-yl)-N-(4-chlorobenzyl)-

4-(1,3-dioxoisoindolin-2-yl)butanamide (34)

Procedure GP1, reagents and conditions: compound

14 (2.4 g, 5 mmol), CDI (0.8 g, 5 mmol), 4-chloro-

benzylamine (0.5 g, 5 mmol), yield: 70.4%; m.p.

210–211ºC. Analysis: calc. for C36H35N4O3Cl: C

71.22, H 5.81, N 9.23%, found: C 71.17, H 5.93, N

9.25%. 1H NMR (CDCl3, d ppm): 1.91–2.19 (m, 2H,

CHCH2CH2), 2.45–2.68 (m, 4H, piperazine), 2.81–

2.98 (m, 4H, piperazine), 3.50 (t, J =7.2 Hz, 1H, CH),

4.17 (t, J = 7.4 Hz, 2H, CHCH2CH2), 4.58 (s, 2H,

NHCH2), 5.14 (s, 3H, CHAr2), 6.93–7.27 (m, 14H,

Ar-H), 7.84–7.99 (m, 4H, phthalimide), 8.17 (s, 1H,

amide). TLC: 0.67 (S2).

Synthesis of 4-amino-2-substituted-butanoic

acid derivatives (35-38), general procedure

To the solution of relevant acid 11–14 (50 mmol) in an-

hydrous ethanol hydrazine hydrate (50 mmol) was

added dropwise and the reaction mixture was hated at

50°C for 2 h. Then, the solvent was evaporated and ob-

tained oil was dissolved in 25% HCl (25 mL), heated at

50ºC for 1 h and stirred for 16 h at room temperature.

The obtained precipitate was filter off and the filtrate

was neutralized with NaHCO3 and extracted with n-

butanol (3 × 25 ml). The organic layers were collected,

dried with Na2SO4 and evaporated in vacuo.

4-Amino-2-(4-phenylpiperazin-1-yl)butanoic

acid dihydrochloride (35)

Yield: 71.7%, m.p. 192–194ºC. Analysis: calc. for

C14H21N3O2 2HCl: C 50.01, H 6.89, N 12.50%, found:

824 Pharmacological Reports, 2012, 64, 817�833

Page 9: Synthesis and pharmacological properties of new GABA ...if-pan.krakow.pl/pjp/pdf/2012/4_817.pdf · Synthesis and pharmacological properties of new GABA uptake inhibitors ... a selective

C 50.08, H 6.84, N 12.67%; 1H NMR (D2O, d ppm):

1.78–2.11 (m, 2H, CHCH2CH2), 2.45–2.68 (m, 8H,

piperazine), 2.81–2.98 (m, 3H, CHCH2CH2), 6.81–

7.36 (m, 5H, Ar-H). TLC: 0.67 (S3).

4-Amino-2-[4-(4-fluorophenyl)piperazin-1-yl]bu-

tanoic acid dihydrochloride (36)

Yield: 56.7%, m.p. 199–200ºC. Analysis: calc. for

C14H20N3O2F 2HCl: C 47.47, H 6.26, N 11.86%,

found: C 47.48, H 6.24, N 11.87%. 1H NMR (D2O, d

ppm): 1.78–2.11 (m, 2H, CHCH2CH2), 2.45–2.68 (m,

8H, piperazine), 2.80–3.00 (m, 3H, CHCH2CH2),6.79–7.42 (m, 4H, Ar-H). TLC: 0.82 (S3).

4-Amino-2-[4-(4-methoxyphenyl)piperazin-1-

yl]butanoic acid dihydrochloride (37)

Yield: 56.7%, m.p. 189–190ºC. Analysis: calc. for

C15H23N3O32HCl: C 49.19, H 6.26, N 11.86%, found:

C 47.20, H 6.84, N 11.47%. 1H NMR (D2O, d ppm):

1.80–2.17 (m, 2H, CHCH2CH2), 2.45–2.68 (m, 8H,

piperazine), 2.83–3.11 (m, 3H, CHCH2CH2), 3.65 (s,

3H, CH3), 6.81–7.36 (m, 4H, Ar-H). TLC: 0.87 (S3).

4-Amino-2-(4-benzhydrylpiperazin-1-yl)butanoic

acid dihydrochloride (38)

Yield: 71.7%, m.p. 192–193ºC. Analysis: calc. for

C27H30N3O2 2HCl: C 64.54, H 6.22, N 8.36%, found:

C 64.50, H 6.24, N 8.37%. 1H NMR (D2O, d ppm):

1.78–2.11 (m, 2H, CHCH2CH2), 2.48–2.74 (m, 8H,

piperazine), 2.80–3.08 (m, 3H, CHCH2CH2), 4.85 (s,

1H, CH), 6.81–7.36 (m, 10H, Ar-H). TLC: 0.67 (S3).

PHARMACOLOGICAL PART

in vitro activity

[3H]GABA uptake assay

Inhibitory potency of compounds 15–38 was tested at

four murine GABA transporter subtypes mGAT1–

mGAT4. The study was performed as a [3H]GABA

uptake assay based on stably transfected HEK cells,

according to the procedure recently described [60].

Briefly, the affinity for mGAT1 was determined by

MS-binding assay with NO 711 as a non-labeled

marker [20]. Binding assays for mGAT1 based on NO

711 as native marker were performed as described

earlier [59]. NO 711 was analyzed by LC-MS/MS us-

ing an API 3200 triple quadrupole mass spectrometer

according to the method described previously [20].

The compounds were considered active if GABA

uptake or NO 711 binding was reduced at least by

50% at a concentration of 100 µM. For the active

compounds, pIC50 values were assessed.

Behavioral experiments

Animals

The behavioral experiments were carried out at the

Department of Pharmacodynamics, Pharmaceutical

Faculty, Jagiellonian University in Kraków. Adult

male Albino Swiss (Krf: CD-1) mice weighing 18–25 g

were supplied by accredited animal facility localized

at Medical College of the Jagiellonian University. The

animals were kept in groups of 15 mice in cages at

a room temperature of 22 ± 2º C, under light/dark

(12:12 h) cycle and had free access to food and water.

The ambient temperature of the room and the humid-

ity were kept consistent throughout all the tests. For

the experiments, animals were selected randomly and

killed by cervical dislocation immediately after the

assay. Prior to the test, the mice were allowed to accli-

mate to the holding cages for a minimum of 2 h. The

experiments were performed between 8:00 a.m. and

3:00 p.m. Experimental groups consisted of 8–12 ani-

mals/dose and all the animals were used only once.

The number of animals was kept at minimum to ob-

tain definite results. All the procedures were approved

by the Local Ethics Committee of the Jagiellonian

University in Kraków (ZI/329/2006).

Chemicals used in pharmacological assays

For the pharmacological studies, compound 18 was

suspended in 0.5% methylcellulose solution (Loba

Chemie, Germany) and administered intraperitoneally

(ip) 30 min before each assay. Control mice were

given an appropriate amount of vehicle (i.e., the

methylcellulose solution).

Five percent formalin solution and 0.9% ethanoic

acid solution were purchased from POCh (Gliwice, Po-

land). Reference drugs: morphine hydrochloride, diaze-

pam and indomethacin acid were provided by Polfa

Kutno (Poland). They were suspended in methylcellu-

lose and administered ip 30 min before the assay.

Pharmacological Reports, 2012, 64, 817�833 825

Pharmacological activity of new GABA uptake inhibitorsKinga Sa³at et al.

Page 10: Synthesis and pharmacological properties of new GABA ...if-pan.krakow.pl/pjp/pdf/2012/4_817.pdf · Synthesis and pharmacological properties of new GABA uptake inhibitors ... a selective

The behavioral measures were scored by trained

observers blind to the experimental conditions.

Locomotor activity test

The effects of three doses of the investigated com-

pound 18 (3.75; 7.5 and 30 mg/kg; ip) were recorded

with photoresistor actimeters (Multi-Serv, Lublin, Po-

land; 30 cm in diameter, illuminated by two light

beams) connected to a counter for the recording of

light-beam interruptions. The mice were placed indi-

vidually in the actimeters and the number of light-

beam crossings was counted during a 30-min period.

Four-plate test

The anxiolytic-like activity was evaluated in the

four-plate apparatus (Bioseb, France). It consists of

a cage (25 × 18 × 16 cm) floored with four identical

rectangular plates (11 × 8 cm) separated from one an-

other by a gap of 4 mm. The plates are connected to

a device that can generate electric foot shocks

(0.6 mA; 0.5 s). After a 15-s habituation period, each

mouse was subjected to an electric shock when cross-

ing from one plate to another (two limbs on one plate

and two on another). The number of punished cross-

ings was calculated for a period of 60 s. Anxiolytic-

like agents increase the number of punished passages

in this assay [3].

Hot plate test

In the hot plate test mice were treated ip either with

compound 18 or vehicle 30 min before placing the

animal on the hot plate apparatus (Hot Plate 2A Type

Omega, Poland). This apparatus has an electrically

heated surface and is supplied with a temperature-

controller that maintains the temperature at 55–56ºC.

The time until the animal licked its back paws or

jumped was recorded by means of the stopwatch [15].

In this assay the cut-off time (30 s) was established to

avoid tissue damage and mice not responding within

30 s were removed from the apparatus and assigned

a score of 30 s.

Writhing test

In this test the animals were placed individually into

glass beakers and 30 min before the experimentation

were allowed to habituate. Then, each mouse was

weighed, injected with compound 18 or vehicle

(10 ml/kg) and then placed back into the cylinder.

Thirty minutes later, 0.9% ethanoic acid (in saline)

was injected ip (10 ml/kg). Mice were placed in the

beakers once again and observed continuously for

30 min. Stereotypical writhes (lengthwise constric-

tions of the torso with a concomitant concave arching

of the back) were counted over this period [58]. The

percent of antinociception was calculated as: [(meth-

ylcellulose-treated mean writhes – drug-treated mean

writhes)/methylcellulose-treated mean writhes] × 100.

Formalin test

In mice, the injection of diluted formalin produces

a biphasic nocifensive behavioral response (i.e., lick-

ing or biting the injected paw). The acute nociceptive

phase lasts for 5 min, whereas the second (inflamma-

tory) phase occurs between 15–30 min after formalin

injection [21].

The formalin test in mice was performed according

to Laughlin et al. [27]. Briefly, 20 µl of a 5% formalin

solution was injected intraplantarly (ipl) into the right

hind paw using a 26-gauge needle. Immediately after

formalin injection, the animals were placed individu-

ally into glass beakers and were observed for 30 min.

The total time (in s) spent on licking the injected paw

during periods of 0–5 and 15–30 min was measured

and was an indicator of nociceptive behavior.

Chimney test

The chimney test was used for the measurement of

motor coordination. It was performed according to

a method described by Boissier et al. [2]. Thirty min-

utes after ip administration of compound 18, the mice

had to climb backwards up a tube (25 cm length, 3 cm

inner diameter) with a rough inner surface. Motor im-

pairments were expressed as the percentage of ani-

mals unable to perform this test within 60 s. Mean

time necessary to leave the cylinder was also com-

pared between drug-treated and vehicle-treated mice.

Data analysis

The data obtained in the experiments are expressed as

the mean ± SEM (standard error of the mean). To com-

pare the results between groups of animals (the investi-

gated compound group vs. the vehicle-treated group) in

the locomotor activity, hot plate, writhing and four-plate

826 Pharmacological Reports, 2012, 64, 817�833

Page 11: Synthesis and pharmacological properties of new GABA ...if-pan.krakow.pl/pjp/pdf/2012/4_817.pdf · Synthesis and pharmacological properties of new GABA uptake inhibitors ... a selective

tests, one-way ANOVA, followed by Dunnett’s post-

hoc statistics were used. The statistical significance of

the results obtained in the formalin test was evaluated

using one-way ANOVA, followed by Newman-Keuls

test. Qualitative variables from the chimney test were

compared by the use of the Fisher’s exact probability

test and the mean time to leave the cylinder was statisti-

cally evaluated using Student’s t-test. In each assay the

difference of means was statistically significant if p <

0.05.

The log-probit method (Litchfield and Wilcoxon

method) [30] was applied to establish the ED50 values

with their respective 95% confidence limits.

Results

In vitro activity

[3H]GABA uptake assay

Among all the compounds tested, only compound 18

showed affinity for mGAT1–4 (Tab. 1). Its IC50 values for

mGAT1–4 were: 38.02 , 28.84, 8.51 and 18.20 µM,

respectively. This compound has a non-selective affinity

towards mGAT1–4, and for mGAT3 in particular (Tab.

1). However, the affinity for mGAT1 (38.02 µM) was

much lower as compared to tiagabine (0.84 µM; data

from [25]).

Pharmacological Reports, 2012, 64, 817�833 827

Pharmacological activity of new GABA uptake inhibitorsKinga Sa³at et al.

Tab. 1. Results of [3H]GABA uptake and NO711 MS-binding assays

Compound MGAT1uptakea

MGAT2uptakea

MGAT3uptakea

MGAT4uptakea

MGAT1NO 711bindingb

15 84% 81% 72% 63% 91%

16 75% 61% 68% 64% –

17 80% 61% 74% 95% –

18 4.42 ± 0.09(n = 4)

4.54 ± 0.12(n = 4)

5.07 ± 0.16(n = 5)

4.74 ± 0.06 79%

19 85% 56% 67% 85% –

20 66% 59% 79% 61% –

21 81% 79% 73% 95% –

22 102% 72% 65% 96% 100%

23 74% 60% 98% 86% 94%

24 90% 79% 88% 89% –

25 99% 60% 59% 83% –

26 93% 64% 76% 81% –

27 74% 56% 63% 83% 104%

28 79% 58% 63% 60% –

29 87% 72% 59% 86% –

30 87% 59% 58% 59% –

31 85% 82% 70% 81% –

32 84% 78% 81% 84% –

33 69% 36% 78% 81% –

34 79% 82% 81% 93% –

35 105% 104% 83% 86% 90%

36 91% 68% 70% 82% 89%

37 119% 89% 77% 84% 95%

38 74% 91% 76% 92% 94%

TIAGABINE (1)c 6.88 ± 0.12 52% 64% 73% –

a % of remaining GABA uptake at 100 µM concentration of tested compound (the means; n = 2) or pIC50 (the means ± SEM; n = 3 if not speci-fied otherwise). b % of NO711 bound to GAT1 at 100 µM concentration of tested compound. c Data from [22]

Page 12: Synthesis and pharmacological properties of new GABA ...if-pan.krakow.pl/pjp/pdf/2012/4_817.pdf · Synthesis and pharmacological properties of new GABA uptake inhibitors ... a selective

BEHAVIORAL EXPERIMENTS

Locomotor activity test

The investigated compound influenced the spontane-

ous locomotor activity of mice, but only at the dose of

7.5 mg/kg this effect was statistically significant (p <

0.05). The mean number of light-beam interruptions

in the vehicle-treated group was 447. Compound 18

(7.5 mg/kg) increased the number of light-beam

crossings for 38% as compared to the methylcellu-

lose-treated animals (Tab. 2).

Four-plate test

In the four-plate test, compound 18 showed the

anxiolytic-like effect (Tab. 3). It increased the number

of punished crossings in a dose-dependent manner in

this assay. The ED50 value was calculated as 9.3 mg/kg.

Compound 18 was less efficacious than diazepam

used as a reference drug in this assay.

Hot plate test

As demonstrated in Table 4, in the hot plate test the

investigated compound 18 exerted a weak analgesic

activity at doses tested and the results did not reach

statistical significance. Morphine, which was a drug

of reference in this assay, showed a high antinocicep-

tive potency in this assay (ED50 = 3.4 mg/kg).

Writhing test

In the ethanoic acid-induced writhing test, compound

18 diminished the number of abdominal constrictions

in a dose-dependent manner (Tab. 5). The mean number

of writhing responses in the vehicle-treated mice was

51.7. Compound 18 (30 mg/kg) and indomethacin

(10 mg/kg) diminished the number of nociceptive reac-

tions by 87 and 95%, respectively (significant at p <

0.001), as compared to the methylcellulose-pretreated

animals. The ED50 value established for compound 18 in

this assay was 15.3 mg/kg.

Formalin test

Compound 18 was highly effective in the formalin

test reducing the duration of nociceptive responses

in both phases of this assay. The results were statisti-

cally significant at p < 0.01. The ED50 value obtained

for compound 18 in the first phase of the test was

828 Pharmacological Reports, 2012, 64, 817�833

Tab. 2. The influence of compound 18 on the locomotor activity

Dose(mg/kg)

Number of crossings(the mean ± SEM)

Locomotor activityincrease (%)

Vehicle (0.5% MC) 447 ± 30 –

3.75 508 ± 35 13.7

7.50 619 ± 44* 38.5

30.00 552 ± 45 23.0

Locomotor activity was recorded in photoresistor actimeters con-nected to a counter for the recording of light-beam interruptions. Thenumber of light-beam crossings was counted during a 30-min ses-sion. The compound and the vehicle were administered ip 30 min be-fore the assay. Each value represents the mean ± SEM obtained from8 mice. MC = methylcellulose (vehicle). Statistical analysis: one-wayANOVA, followed by Dunnett’s test. F (3, 23) = 3.425. * indicates a sig-nificant difference compared to the vehicle-treated group: * p < 0.05

Tab. 3. Anxiolytic-like activity of compound 18 and diazepam in the four-plate test

Compound Dose(mg/kg)

Number of punishedpassages

Effect(%)

ED50

(mg/kg)

Vehicle (0.5% MC) – 3.8 ± 0.4 – –

18

3.75 4.2 ± 0.4 10.5

9.3 (3.9–22.5)7.50 5.8 ± 0.6 52.6

15.00 5.6 ± 0.9 47.4

30.00 6.8 ± 0.9* 78.9

Diazepam 1.00 6.3 ± 0.7* 65.8 –

Each value represents the mean ± SEM obtained from 10 mice. The compound and the vehicle were administered ip 30 min before the assay.MC = methylcellulose (vehicle). Statistical analysis: one-way ANOVA, followed by Dunnett’s test. F (5, 50) = 3.01. Significant difference com-pared to MC: * p < 0.05

Page 13: Synthesis and pharmacological properties of new GABA ...if-pan.krakow.pl/pjp/pdf/2012/4_817.pdf · Synthesis and pharmacological properties of new GABA uptake inhibitors ... a selective

5.3 mg/kg. This analgesic potency was similar to mor-

phine for which the ED50 value was 3.0 mg/kg (Tab. 6).

Chimney test

Compound 18 (30 mg/kg, ip) had no adverse effect on

the animals’ motor performance in the chimney test.

The motor coordination in the vehicle-treated group

of mice and in mice treated with compound 18 was

impaired to similar degree – in both groups 8% of

mice were unable to perform the test within 1 min.

Also the mean time necessary to leave the cylinder

was similar in drug-treated and vehicle-treated ani-

mals (10.2 s and 9.1 s, respectively) (Tab. 7).

Discussion

In this paper, the synthesis of new GABA uptake in-

hibitors and their influence on [3H]GABA uptake and

NO 711 MS-binding in the assays based on stably

transfected HEK cells are described. Since compound

18 demonstrated the highest affinity for all four

murine GAT (mGAT1–4) in vitro, it was chosen for

further extended pharmacological research in behav-

ioral tests. The IC50 values obtained for compound 18

in the in vitro assays (38 µM for mGAT1; 28 µM for

mGAT2; 8.5 µM for mGAT3 and 18 µM for mGAT4)

suggest that this activity may explain many but not all

Pharmacological Reports, 2012, 64, 817�833 829

Pharmacological activity of new GABA uptake inhibitorsKinga Sa³at et al.

Tab. 4. Antinociceptive activity of compound 18 and morphine in the hot plate test

Compound Dose(mg/kg)

Time latency(mean ± SEM)

(s)

Effect(%)

ED50

(mg/kg)

Vehicle (0.5% MC) – 14.5 ± 1.6 – –

187.5 15.6 ± 2.1 7.6 –

30.0 22.1 ± 3.7 52.4

Vehicle (0.5% MC) – 18.1 ± 0.5 – –

Morphine 1 19.0 ± 0.5 5.0

3.4 (2.2–5.1)3 29.2 ± 0.4**** 61.3

6 30.0 ± 0.0**** 65.7

Each value represents the mean ± SEM obtained from 8 mice. The compound and the vehicle were administered ip 30 min before the assay.MC = methylcellulose (vehicle). Statistical analysis: one-way ANOVA, followed by Dunnett’s test. F (2, 26) = 2.816 (compound 18); F (3, 30) =226.6 (morphine). Significant difference compared to MC: **** p < 0.001

Tab. 5. Antinociceptive activity of compound 18 and indomethacin in the ethanoic acid-induced writhing test

Compound Dose(mg/kg)

Number of writhes(mean ± SEM)

Effect(%)

ED50

(mg/kg)

Vehicle (0.5% MC) – 51.7 ± 7.5 – –

18

7.5 44.7 ± 2.4 13.515.3

(10.2–23.0)15.0 28.2 ± 7.4*** 45.5

30.0 6.8 ± 1.6**** 86.8

Indomethacin 10.0 2.8 ± 1.3 **** 94.6 -

Each value represents the mean ± SEM obtained from 8 mice. The compound and the vehicle were administered ip 30 min before the assay.MC = methylcellulose (vehicle). Statistical analysis: one-way ANOVA, followed by Dunnett’s test. F (4, 29) = 23.05. Significant difference com-pared to MC: *** p < 0.01, **** p < 0.001

Page 14: Synthesis and pharmacological properties of new GABA ...if-pan.krakow.pl/pjp/pdf/2012/4_817.pdf · Synthesis and pharmacological properties of new GABA uptake inhibitors ... a selective

effects observed in animal models. For instance, the

inhibition of GABA transport would be expected to

diminish or completely abolish the locomotor activity

of experimental animals [6]. Surprisingly, in our stud-

ies, compound 18 did not decrease the locomotor ac-

tivity. Although this observation requires further stud-

ies, it can be explained as follows: the GABA uptake

inhibition evoked by the investigated compound 18

may lead to the increase of GABA within the synaptic

cleft and indirectly activate the excitatory pathways in

the CNS via the disinhibition of the inhibitory sys-

tems. It is also possible that the increased locomotor

activity is due to some additional and not defined yet,

unknown properties of compound 18 which are not

directly related to GABA enhancement.

In the four-plate test, compound 18 exerted signifi-

cant anxiolytic-like properties increasing the mean

number of punished crossings as compared to the

vehicle-treated mice. This action is likely to result

from its affinity for mGAT1. Recent evidence sug-

gests that mGAT1 (SLC6A1) plays a role in the pa-

thophysiology and treatment of anxiety disorders [52]

and tiagabine, a selective mGAT1 inhibitor, has

anxiolytic-like properties in humans [1, 43]. It is well

known that the inhibition of GABA uptake from the

synaptic cleft by potent and selective inhibitors of the

GABA transporter GAT1 is used therapeutically in the

treatment of epilepsy or some psychiatric disorders,

including anxiety [29, 43–45]. Moreover, the results

from animal studies indicate that acute or chronic ad-

ministration of tiagabine decreases anxiety-related be-

havior in rodents [53].

In the present work, we also evaluated the antino-

ciceptive effects of compound 18 in acute (hot plate,

830 Pharmacological Reports, 2012, 64, 817�833

Tab. 6. Antinociceptive activity of compound 18 and morphine in the formalin test

Compound Dose(mg/kg)

Phase I latency(mean ± SEM)

(s)

Effect(%)

ED50

(mg/kg)Phase II latency

(the mean ± SEM)(s)

Effect(%)

Vehicle (0.5% MC) – 54.8 ±10.8 – – 152.6 ± 18.3 –

18

3.75 34.6 ± 5.6 36.4

5.3(2.6–10.9)

52.6 ± 14.7**** 65.5

7.5 17.9 ± 4.1**** 67.3 57.3 ± 13.4**** 62.5

15.0 15.0 ± 4.2**** 72.7 57.1 ± 13.8**** 62.6

30.0 15.4 ± 5.2**** 72.7 43.3 ± 10.6**** 71.9

Vehicle (0.5% MC) – 91.2 ± 11.1 – – 133.8± 22.6 –

Morphine

2.1 64.7 ± 12.4 29.063.0

(1.9–4.7)

3.4 ± 1.7**** 97.5

3.0 41.2 ± 5.7*** 54.8 18.6 ± 6.6**** 86.2

5.0 25.1 ± 3.9**** 72.5 17.9 ± 8.0**** 86.6

Each value represents the mean ± SEM obtained from 8 mice. The compound and the vehicle administered ip 30 min before the assay.MC = methylcellulose (vehicle). Statistical analysis of the results performed by one-way ANOVA followed by Newman-Keuls test. Compound18: F (4, 38) = 7.656 (phase I); F (4, 38) = 6.778 (phase II); morphine: F (3, 31) = 8.743 (phase I); F (3, 31) = 26. (phase II). Significant differencecompared to MC: *** p < 0.01; **** p < 0.001

Tab. 7. Effect of compound 18 on the motor coordination in the chimney test

Compound Dose(mg/kg)

Mean time to leave the cylindera

(mean ± SEM)(s)

Effect(%)

% of mice showingmotor impairmentb

Vehicle (0.5% MC) – 9.14 ± 1.83 – 8

18 30.0 10.24 ± 0.9 12 8

Values shown as the mean ± SEM (time to leave the cylinder) or as percent of animals with motor impairment (animals per group were n = 12).The compound and the vehicle were administered ip 30 min before the assay. MC = methylcellulose (vehicle). a, b Statistical analysis of dataperformed by using a Student’s t-test and b Fisher’s exact probability test: the results were not found statistically significant

Page 15: Synthesis and pharmacological properties of new GABA ...if-pan.krakow.pl/pjp/pdf/2012/4_817.pdf · Synthesis and pharmacological properties of new GABA uptake inhibitors ... a selective

writhing tests) and tonic (formalin) pain models. Ac-

cumulating evidence indicates that the GABAergic

transmission plays a pivotal role in the inhibitory

regulation of the nociceptive process, especially

within the dorsal horn of the spinal cord [14, 51, 59]

and many GABAergic drugs, including tiagabine and

vigabatrin, are analgesic in rodent models of acute

(hot plate, tail immersion, grid-shock) and chronic

(formalin test, dynorphin-induced allodynia) pain [14,

22, 27, 32–35, 51]. The inhibition of GAT1 was re-

ported to attenuate the excitatory amino acids neuro-

transmission, which results in the pain-relieving effect

at the spinal cord level [4]. This activity may be due

to the enhanced GABAergic transmission and subse-

quent GABAB receptor activation [22, 51]. The in-

volvement of both GABA receptors (A and B) in

tiagabine-induced analgesia in the formalin test was

also reported [27].

Compound 18 demonstrated a weak analgesic ac-

tivity in the hot plate test, despite its affinity for GAT

in vitro. The hot plate assay is used to reveal com-

pounds with central antinociceptive properties,

whereas peripherally acting analgesics are generally

not active in it. The analgesic effect in the hot plate

test measured as the prolongation of the latency time

to nocifensive reaction is a consequence of supraspi-

nal attenuation of ascending nociceptive input [40].

The lack of analgesic properties of compound 18 in

the hot plate test may account for other peripheral

mechanisms underlying its antinociceptive activity.

This was also confirmed in another pain test, namely

the writhing test which is a chemogenic model [50,

58] used for testing peripherally acting analgesic

drugs and additionally in the formalin test. In both as-

says, compound 18 was able to reduce nociceptive re-

actions. Comparing the same dose ranges effective in

the writhing test and in the second (inflammatory)

phase of the formalin test, it seems that the investi-

gated compound was more efficacious in the formalin

model. This may be due to the difference in the inten-

sity of pain generated either by formalin or ethanoic

acid administration.

In the formalin test, compound 18 demonstrated

a very high and statistically significant antinocicep-

tive activity. The ED50 value obtained for the first

(neurogenic) phase of the test was similar to morphine

(5.3 mg/kg and 3.0 mg/kg, respectively). It was also

active in the second (late) phase which appears to be

dependent on the combination of inflammatory reac-

tions in the peripheral tissues and functional changes

in the dorsal horn of the spinal cord [21, 56].

Noteworthy, the analgesic effect exerted by com-

pound 18 was observed at doses that did not impair

the motor coordination in the chimney test. At the

dose of 30 mg/kg, it did neither significantly influ-

ence the mean time to get off the cylinder nor the

number of animals that demonstrated motor deficits.

Several lines of evidence suggest that sedation in men

[7, 38], hypolocomotion and impaired motor coordi-

nation in rodents are typical for compounds acting at

mGAT1 (e.g., tiagabine) [5, 7, 22]. The results ob-

tained for compound 18 suggest that the effects ob-

served both in the locomotor activity test and in the

chimney test are independent of its affinity for

mGAT1.

Concluding, although different compounds were

shown to have affinity for GABA transporters, only ti-

agabine is used in therapy as an anticonvulsant agent

for the treatment of partial seizures in men [7, 8,

11–13, 35, 41]. The physiological role of compounds

targeting at GAT other than GAT1 is up to date poorly

established. Several reports suggest that non-GAT1

inhibitors of GABA transport, especially agents acting

at mGAT2 and mGAT4, are very interesting as poten-

tial prospects for the future epilepsy treatment [36].

For tiagabine, new medical indications are being

evaluated in clinic, as well [43–45, 48, 49].

In the present paper, the anxiolytic-like and analge-

sic activities of a new non-selective GABA uptake in-

hibitor, namely compound 18, have been described.

The results obtained in this study are promising, how-

ever, the therapeutic potential of compound 18 should

be investigated in greater detail. Therefore, in the

search for future analgesic and anxiolytic-like drugs,

we suggest further research of the derivatives of 2-

substituted-4-(1,3-dioxoisoindolin-2-yl)-butanamides

and 2-substituted-4-aminobutanoic acids.

Acknowledgments:

Financial support of this work by the Jagiellonian University

Medical College grants K/ZBW/000603 and K/ZDS/001919

is gratefully acknowledged.

References:

1. Baldwin D, Woods R, Lawson R, Taylor D: Efficacy of

drug treatments for generalised anxiety disorder: system-

Pharmacological Reports, 2012, 64, 817�833 831

Pharmacological activity of new GABA uptake inhibitorsKinga Sa³at et al.

Page 16: Synthesis and pharmacological properties of new GABA ...if-pan.krakow.pl/pjp/pdf/2012/4_817.pdf · Synthesis and pharmacological properties of new GABA uptake inhibitors ... a selective

atic review and meta-analysis. BMJ, 2011,

doi: 10.1136/bmj.d1199.

2. Boissier JR, Tardy J, Divierres JC: A new simple method

to explore the tranquilizing action: the chimney test

(French). Med Exp, 1960, 3, 81–84.

3. Bourin M, Masse F, Dailly E, Hascoët M: Anxiolytic-like

effect of milnacipran in the four-plate test in mice: mecha-

nism of action. Pharmacol Biochem Behav, 2005, 81,

645–656.

4. Bowery NG, Smart TG: GABA and glycine as neuro-

transmitters: a brief history. Br J Pharmacol, 2006, 147,

S109–S119.

5. Castelli MP: Multi-faceted aspects of gamma-hydroxybutyric

acid: a neurotransmitter, therapeutic agent and drug of

abuse. Mini Rev Med Chem, 2008, 8, 1188–1202.

6. Cazalets JR, Bertrand S, Sqalli-Houssaini Y, Clarac F:

GABAergic control of spinal locomotor networks in the

neonatal rat. Ann NY Acad Sci, 1998, 860, 168–180.

7. Chiu CS, Brickley S, Jensen K, Southwell A, McKinney

S, Cull-Candy S, Mody I, Lester HA: GABA transporter

deficiency causes tremor, ataxia, nervousness, and in-

creased GABA-induced tonic conductance in cerebel-

lum. J Neurosci, 2005, 25, 3234–3245.

8. Chroœciñska-Krawczyk M, Ratnaraj N, Patsalos PN,

Czuczwar SJ: Effect of caffeine on anticonvulsant effects

of oxcarbazepine, lamotrigine and tiagabine in a mouse

model of generalized tonic-clonic seizures. Pharmacol

Rep, 2009, 61, 819–826.

9. Clausen RP, Frølund B, Larsson OM, Schousboe A,

Krogsgaard-Larsen P, White HS: A novel selective g-am-

inobutyric acid transport inhibitor demonstrated a func-

tional role for GABA transporter subtype GAT2/BGT-1

in the CNS. Neurochem Int, 2006, 48, 637–642.

10. Cope DW, Di Giovanni G, Fyson SJ, Orban G, Errington

AC, Lorincz ML, Gould TM et al.: Enhanced tonic

GABAA inhibition in typical absence epilepsy. Nat Med,

2009, 15, 1392–1398.

11. Czapinski P, Blaszczyk B, Czuczwar SJ: Mechanisms of action

of antiepileptic drugs. Curr Top Med Chem, 2005, 5, 3–14.

12. Dalby NO, Thomsen Ch, Fink-Jensen A, Lundbeck J,

Sokilde B, Man CM, Sorensen PO, Meldrum B: Anti-

convulsant properties of two GABA uptake inhibitors

NNC 05-2045 and NNC 05-2090, not acting preferen-

tially on GAT-1. Epilepsy Res, 1997, 28, 51–61.

13. Dalby NO: Inhibition of g-aminobutyric acid uptake:

anatomy, physiology and effects against epileptic sei-

zures. Eur J Pharmacol, 2003, 479, 127–137.

14. Di Lio A, Benke D, Besson M, Desmeules J, Daali Y,

Wang ZJ, Edwankar R et al.: HZ166, a novel GABAA re-

ceptor subtype-selective benzodiazepine site ligand, is an-

tihyperalgesic in mouse models of inflammatory and neu-

ropathic pain. Neuropharmacology, 2011, 60, 626–632.

15. Eddy N, Leimbach D: Synthetic analgesics. II. Dithien-

ylbutenyl - and dithienylbutylamines. J Pharmacol Exp

Ther, 1953, 107, 385–393.

16. Ettinger AB, Argoff CE: Use of antiepileptic drugs for

non-epileptic conditions: psychiatric disorders and

chronic pain. Neurotherapeutics, 2007, 4, 75–83.

17. Gajcy K, Lochynski S, Librowski T: A role of GABA

analogues in the treatment of neurological diseases.

Curr Med Chem, 2010, 17, 2338–2347.

18. Giardina WJ, Decker MW, Porsolt RD, Roux S, Collins

SD, Kim DJB, Bannon AW: An evaluation of the GABA

uptake blocker tiagabine in animal models of neuropathic

and nociceptive pain. Drug Dev Res, 1998, 44, 106–113.

19. Gowin JL, Green CE, Alcorn JL, Swann AC, Moeller

FG, Lane SD: Chronic tiagabine administration and ag-

gressive responding in individuals with a history of sub-

stance abuse and antisocial behavior. J Psychopharma-

col, 2012, 26, 982–993.

20. Höfner G, Wanner KT: Using short columns to speed up

LC-MS quantification in MS binding assays. J Chromatogr

B Analyt Technol Biomed Life Sci, 2010, 878, 1356–1364.

21. Hunskaar S, Hole K: The formalin test in mice: dissocia-

tion between inflammatory and non-inflammatory pain.

Pain, 1987, 30, 103–114.

22. Ipponi A, Lamberti C, Medica A, Bartolini A,

Malmberg-Aiello P: Tiagabine antinociception in rodents

depends on GABAB receptor activation: parallel antino-

ciception testing and medial thalamus GABA micro-

dialysis. Eur J Pharmacol, 1999, 368, 205–211.

23. Iversen L: Neurotransmitter transporters and their impact

on the development of psychopharmacology. Br J Phar-

macol, 2006, 147; S82–S88.

24. Johannessen LC: Antiepileptic drugs in non-epilepsy dis-

orders: relations between mechanisms of action and

clinical efficacy. CNS Drugs, 2008, 22, 27–47.

25. Kragler A, Höfner G, Wanner KT: Synthesis and biologi-

cal evaluation of aminomethylphenol derivatives as in-

hibitors of the murine GABA transporters mGAT1-

mGAT4. Eur J Med Chem, 2008, 43, 2404–2411.

26. Kulig K, Wiêckowski K, Wiêckowska A, Gajda J,

Pochwat B, Höfner GC, Wanner KT, Malawska B:

Synthesis and biological evaluation of the new amides

of 2-substituted 4-hydroxybutyric acid derivatives as

GABA uptake inhibitors. Eur J Med Chem, 2011, 46,

183–190.

27. Laughlin TM, Tram KV, Wilcox GL, Birnbaum AK:

Comparison of antiepileptic drugs tiagabine, lamotrigine,

and gabapentin in mouse models of acute, prolonged and

chronic nociception. J Pharmacol Exp Ther, 2002, 302,

1168–1175.

28. Lewin L, Mattsson MO, Sellström A: Inhibition of trans-

porter mediated gamma-aminobutyric acid (GABA) re-

lease by SKF 89976-A, a GABA uptake inhibitor, stud-

ied in a primary neuronal culture from chicken. Neuro-

chem Res, 1992, 17, 577–584.

29. Lieving LM, Cherek DR, Lane SD, Tcheremissine OV,

Nouvion SO: Effects of acute tiagabine administration

on aggressive responses of adult male parolees. J Psy-

chopharmacol, 2008, 22, 144–152.

30. Litchfield JT, Wilcoxon E: A simplified method of evalu-

ating dose-effect experiments. J Pharmacol Exp Ther,

1949, 96, 99–113.

31. Liu GX, Cai GQ, Cai YQ, Sheng ZJ, Jiang J, Mei Z,

Wang ZG et al.: Reduced anxiety and depression-like be-

haviors in mice lacking GABA transporter subtype I.

Neuropsychopharmacology, 2007, 32, 1531–1539.

32. £uszczki JJ, Czuczwar SJ: Dose-response relationship

analysis of vigabatrin doses and their antinociceptive ef-

fects in the hot-plate test in mice. Pharmacol Rep, 2008,

60, 409–414.

832 Pharmacological Reports, 2012, 64, 817�833

Page 17: Synthesis and pharmacological properties of new GABA ...if-pan.krakow.pl/pjp/pdf/2012/4_817.pdf · Synthesis and pharmacological properties of new GABA uptake inhibitors ... a selective

33. Luszczki JJ, Czuczwar SJ: Isobolographic characteriza-

tion of interactions between vigabatrin and tiagabine in

two experimental models of epilepsy. Prog Neuropsy-

chopharmacol Biol Psychiatry, 2007, 31, 529–538.

34. £uszczki JJ, Ko³acz A, Wojda E, Czuczwar M, Przesmycki

K, Czuczwar SJ: Synergistic interaction of gabapentin with

tiagabine in the hot-plate test in mice: an isobolographic

analysis. Pharmacol Rep, 2009,61, 459–467.

35. Luszczki JJ, Swiader M, Parada-Turska J, Czuczwar SJ:

Tiagabine synergistically interacts with gabapentin in the

electroconvulsive threshold test in mice. Neuropsycho-

pharmacology, 2003, 28, 1817–1830.

36. Madsen K, White HS, Clausen RP, Frølund B, Larsson

OM, Krogsgaard-Larsen P, Schousboe A: Functional and

pharmacological aspects of GABA transporters. Hand-

book of Neurochemistry and Molecular Neurobiology.

Neural Membranes and Transport. Springer Science +

Business Media, LLC, New York, 2007.

37. Madsen KK, Clausen RP, Larsson OM, Krogsgaard-

Larsen P, Schousboe A, White HS: Synaptic and extra-

synaptic GABA transporters as targets for anti-epileptic

drugs. J Neurochem, 2009, 109, 139–144.

38. Madsen KK, White HS, Schousboe A: Neuronal and

non-neuronal GABA transporters as targets for antiepi-

leptic drugs. Pharmacol Ther, 2010, 125, 394–401.

39. Malawska B, Zejc A: Search for new anticonvulsant

compounds. Part 1: Synthesis, physicochemical and anti-

convulsant properties of new derivatives of alpha-

amino-gamma-phthalimidobutyric acid. Pharmazie,

1995, 50, 722–725.

40. Manzanares J, Julian MD, Carrascosa A: Role of the

cannabinoid system in pain control and therapeutic im-

plications for the management of acute and chronic pain

episodes. Curr Neuropharmacol, 2006, 4, 239–257.

41. Meldrum BS, Rogawski MA: Molecular targets for an-

tiepileptic drug development. Neurotherapeutics, 2007,

4, 18–61.

42. Narita M, Niikura K, Nanjo-Nikura K, Narita M, Furuya

M, Yamashita A, Saeki M et al.: Sleep disturbances in

a neuropathic pain-like condition in the mouse are asso-

ciated with altered GABAergic transmission in the cin-

gulate cortex. Pain, 2011, 152, 1358–1372.

43. Pollack MH, Roy-Byrne PP, Van Ameringen M, Snyder

H, Brown C, Ondrasik J, Rickels K: The selective

GABA reuptake inhibitor tiagabine for the treatment of

generalized anxiety disorder: results of a placebo-

controlled study. J Clin Psychiatry, 2005, 66, 1401–1408.

44. Rogawski MA, Loescher W: The neurobiology of antie-

pileptic drugs for the treatment of non-epileptic condi-

tions. Nat Med, 2004, 10, 685–692.

45. Rosenthal M: Tiagabine for the treatment of generalized

anxiety disorder: a randomized, open-label, clinical trial

with paroxetine as a positive control. J Clin Psychiatry,

2003, 64, 1245–1249.

46. Salat K, Kulig K: GABA transporters as targets for new

drugs. Fut Med Chem, 2011, 3, 211–222.

47. Sa³at K, Kulig K, Sa³at R, Filipek B, Malawska B: Anal-

gesic and anticonvulsant activity of new derivatives of

2-substituted 4-hydroxybutanamides in mice. Pharmacol

Rep, 2012, 64, 102–112.

48. Schwartz TL, Nihalani N: Tiagabine in anxiety disorders.

Expert Opin Pharmacother, 2006, 7, 1977–1987.

49. Spina E, Perugi G: Antiepileptic drugs: indications other

than epilepsy. Epileptic Disord, 2004, 6, 57–75.

50. Shin YH, Jung OM, Nah JJ, Nam KY, Kim CY, Nah SY:

Ginsenosides that produce differentia antinociception in

mice. Gen Pharmacol, 1999, 32, 653–659.

51. Smith CGS, Bowery NG, Whitehead KJ: GABA trans-

porter type 1 (GAT-1) uptake inhibition reduces stimu-

lated aspartate and glutamate release in the dorsal spinal

cord in vivo via different GABAergic mechanisms. Neu-

ropharmacology, 2007, 53, 975–981.

52. Soudijn W, van Wijngaarden I: The GABA transporter

and its inhibitors. Curr Med Chem, 2000, 7, 1063–1079.

53. Thoeringer CK, Erhardt A, Sillaber I, Mueller MB, Ohl

F, Holsboer F, Keck ME: Long-term anxiolytic and

antidepressant-like behavioural effects of tiagabine, a se-

lective GABA transporter-1 (GAT-1) inhibitor, coincide

with a decrease in HPA system activity in C57BL/6

mice. J Pharmacol, 2010, 24, 733–743.

54. Thoeringer CK, Ripke S, Unschuld PG, Lucae S, Ising

M, Bettecken T, Uhr M et al.: The GABA transporter 1

(SLC6A1): a novel candidate gene for anxiety disorders.

J Neural Transm, 2009, 116, 649–657.

55. Thomsen C, Sorensen PO, Egebjerg J: 1-(3-(9H-

carbazol-9-yl)-1-propyl)-4-(2-methoxyphenyl)-4-

piperidinol, a novel subtype selective inhibitor of the

mouse type II GABA-transporter. Br J Pharmacol, 1997,

120, 983–985.

56. Tjølsen A, Berge OG, Hunskaar S, Rosland JH, Hole K:

The formalin test: an evaluation of the method. Pain,

1992, 51, 5–17.

57. White HS, Watson WP, Hansen SL, Slough S, Perregaard

J, Sarup A, Bolvig T et al.: First demonstration of a func-

tional role for central nervous system betaine/g-aminobu-

tyric acid transporter (mGAT2) based on synergistic anti-

convulsant action among inhibitors of mGAT1 and

mGAT2. J Pharmacol Exp Ther, 2005, 312, 866–874.

58. Wilson SG, Bryant CD, Lariviere WR, Olsen MS, Giles

BE, Chesler EJ, Mogil JS: The heritability of antino-

ciception II: pharmacogenetic mediation of three over-

the-counter analgesics in mice. J Pharmacol Exp Ther,

2003, 305, 755–764.

59. Zeilhofer HU, Möhler H, Di Lio A: GABAergic analge-

sia: new insights from mutant mice and subtype-selective

agonists. Trends Pharmacol Sci, 2009, 30, 397–402.

60. Zepperitz C, Höfner G, Wanner KT: MS-binding assays: ki-

netic, saturation, and competitive experiments based on

quantitation of bound marker as exemplified by the GABA

transporter mGAT1. Chem Med Chem, 2006, 1, 208–217.

Received: October 26, 2011; in the revised form: February 15, 2012;

accepted: April 5, 2012.

Pharmacological Reports, 2012, 64, 817�833 833

Pharmacological activity of new GABA uptake inhibitorsKinga Sa³at et al.