• Nie Znaleziono Wyników

Synthesis, anticonvulsant activity and 5-HT

N/A
N/A
Protected

Academic year: 2022

Share "Synthesis, anticonvulsant activity and 5-HT"

Copied!
10
0
0

Pełen tekst

(1)

Synthesis, anticonvulsant activity and 5-HT 1A /5-HT 7 receptors affinity

of 1-[(4-arylpiperazin-1-yl)-propyl]-succinimides

Jolanta Obniska1, Iwona Chlebek1, Krzysztof Kamiñski1, Andrzej J. Bojarski2, Grzegorz Sata³a2

1Department of Medicinal Chemistry, Faculty of Pharmacy, Medical College, Jagiellonian University, Medyczna 9, PL 30-688 Kraków, Poland

2Department of Medicinal Chemistry, Institute of Pharmacology, Polish Academy of Sciences, Smêtna 12, PL 31-343 Kraków, Poland

Correspondence: Jolanta Obniska, e-mail: mfobnisk@cyf-kr.edu.pl

Abstract:

Background: Epilepsy is the most prevalent neurological disorder, affecting approximately 50 million people worldwide. Even though significant advances have been made in epilepsy research, convulsions in about 30% of epileptics are still inadequately con- trolled by standard drug therapy. For this reason, constant attempts are made to investigate new chemical agents and mechanisms through which epilepsy can be effectively controlled. Therefore, in the present studies, a series of sixteen new 1-[(4-arylpiperazin- 1-yl)-propyl]-3-methyl-3-phenyl- and 3-ethyl-3-methylpyrrolidine-2,5-dione derivatives as potential anticonvulsant agents was synthesized.

Methods: Anticonvulsant properties were evaluated in the maximal electroshock (MES), subcutaneous pentylenetetrazole (scPTZ) and psychomotor seizure (6-Hz) tests after intraperitoneal injection in mice. The acute neurological toxicity was determined in the motor impairment rotorod screen.

Results: The compounds showed activity at a dose of 30 mg/kg (4, 8, 16) or 100 mg/kg (6, 9, 10, 12, 17, 18) in the MES model in mice. Four or them (8, 10, 16, 17) were also evaluated after po administration in rats. In this series, the most active was 1-{3-[4-(3- chlorophenyl)-piperazin-1-yl]-propyl}-3-methyl-3-phenyl-pyrrolidine-2,5-dione (8) with the ED50 value of 28.2 mg/kg, TD50 value of 268.5 mg/kg and protective index of 9.52 after po administration in rats.

Conclusions: Taking into consideration the role of 5-HT1Aand 5-HT7receptor subtypes in relation to the control of seizures as well as the fact that all compounds obtained belong to the class of long-chain arylpiperazines, their serotonin 5-HT1Aand 5-HT7receptor affinity was determined. The most potent 5-HT1Areceptor ligands are 2-OCH3(11, 19) and 3-Cl (8, 16) derivatives with Ki= 72, 14 nM, and 109, 44 nM, respectively. With respect to the 5-HT7receptors, the best Kivalues were obtained for derivatives 8 and 11 (Ki= 76 nM and 63 nM, respectively).

Key words:

anticonvulsant activity, 5-HT1A/5-HT7receptor ligands, 3,3-disubstituted pyrrolidine-2,5-diones, arylpiperazines

(2)

Introduction

The currently available antiepileptic drugs are effec- tive in reducing the severity and number of seizures in less than 80% of patients [3]. Moreover, their usage is associated with undesirable side-effects from cos- metic (gingival hyperplasia) to life threatening (e.g., hepatotoxicity, megaloblastic anemia) [30]. For these reasons, constant attempts are made to investigate new agents and mechanisms through which epilepsy can be effectively inhibited.

It is well known that numerous derivatives with an- ticonvulsant activity contain 5- or 6-membered nitro- gen heterocycle rings, one or two carbonyl groups as well as an aromatic system [12]. Following these findings, in the course of the search of new anticon- vulsants our attention has been focused on a group of 3-substituted pyrrolidine-2,5-diones with different substituents at the imide nitrogen atom [9, 10]. It has been shown recently a high activity in the maximal electroshock (MES) test of great number of pyrro- lidine-2,5-diones with 4-arylpiperazines connected to the imide nitrogen atom by the alkylene spacer [22, 26]. Furthermore, the previous investigations proved also an essential role of aromatic moiety at position-3 of pyrrolidine-2,5-dione ring [21, 23, 25, 27]. There- fore, taking into consideration the above findings, in the present work we have synthesized a series of 1-[(4-arylpiperazin-1-yl)-propyl]-pyrrolidine-2,5-diones with two different substituents, namely methyl and phenyl (4–11) or only alkyl – ethyl and methyl groups (12–19) at the position-3 of pyrrolidine-2,5-dione. All the compounds obtained were tested for their anticon- vulsant activity within the Antiepileptic Drug Devel- opment (ADD) Program in Epilepsy Branch, Neuro- logical Disorders Program, National Institute of the Neurological and Communicative Disorders and Stroke (NIH/NINDS), Rockville, MD, USA.

The compounds synthesized may be regarded as long-chain arylpiperazines (LCAPs), that is molecules exhibiting a variety of pharmacological effects via in- teractions with several receptor types, however, the best known as ligands of different serotonin (5-HT) receptor subtypes. It is worthy of note that serotonin (5-hydroxytryptamine, 5-HT) is a major neurotrans- mitter involved in a large number of processes in cen- tral nervous system (CNS), including the regulation of feeding behavior, aggression, mood, perception, pain, anxiety and brain excitability. Furthermore, the wide distribution of the 5-HT receptors, both in the

CNS and in the peripheral tissues, is highly associated with their implications in psychiatric and neurological disorders such as depression, anxiety and epilepsy [1, 15]. It is well known that serotonin exerts its effects viaat least 14 different receptors subtypes, but the role of only a few of them, namely 5-HT1A, 5-HT2C and 5-HT7has been studied in relation to the control of sei- zures. This was the subject of several investigations and the obtained results are diversified (positive or negative role), depending on both the ligand type and the seizure model [19, 29]. Taking this into considera- tion, in the current studies the affinities for 5-HT1Aand 5-HT7receptor subtypes have been assessed.

Materials and Methods

Chemical syntheses

The synthesis of compounds 4–19 is shown in Figure 1.

All the chemicals and solvents were purchased from Sigma-Aldrich. Melting points (m.p.) were deter- mined on Büchi 353 melting point apparatus (Büchi Labortechnik, Flawil, Switzerland) and are uncor- rected. The chemical structures of the obtained com- pounds were confirmed by elemental and spectral analyses. The 1H NMR spectra were obtained using a Varian Mercury spectrometer (Varian Inc., Palo Alto, CA, USA), operating at 300 MHz. The chemical shifts were reported as parts per million (d, ppm) with (CH3)4Si (TMS) as an internal standard. Signal multi- plicities are represented by: s (singlet), brs (broad singlet), d (doublet), t (triplet), m (multiplet).

The purity of the compounds was checked by thin- layer chromatography (TLC) performed on Merck sil- ica gel GF254 aluminum sheets, using the developing system S1 consisted of chloroform : isopropanol : 25% ammonia (9:11:2, v/v/v). Spots were detected by their absorption under UV light. Elemental analyses for C, H, and N were carried out by a micro method using the elemental Vario EI III Elemental analyzer (Hanau, Germany). The elemental analyses for C, H, N were within ± 0.4% of the theoretical values.

The starting 2-methyl-2-phenylsuccinic acid (1), was obtained by the method described previously [11]. The appropriately substituted 1-(3-aminopro- pyl)-4-arylpiperazines were synthesized based on the described method [7].

Anticonvulsant activity and 5-HT1A/5-HT7affinity of certain succinimides

Joanna Obniska et al.

(3)

1-yl)-propyl]-3-methyl-3-phenyl-pyrrolidine-2,5-diones 4–11 which were isolated as hydrochloride salts.

Compounds 12–19 were synthesized using as a starting material commercially available 3-ethyl-3- methyl-succinimide (Sigma-Aldrich) (2), which was alkylated with 1-bromo-3-chloropropane yielding 1-(3-chloropropyl)-3-ethyl-3-methyl-pyrrolidine-2,5- dione (3). The intermediate 3 and given 4-arylpi- perazines were dissolved in acetone and then stirred in the presence of anhydrous K2CO3at 60°C for 24 h.

After this time, the crude products were purified by use of column chromatography and then converted into hydrochloride salts. Physicochemical and spec- tral data of compounds 5–19 are shown in experimen- tal section. The synthetic procedure and physico- chemical data of 4 were described elsewhere [26].

General procedure for the preparation of the 1-[(4-arylpiperazinyl-1-yl)-propyl]-3-methyl-3- phenylpyrrolidine-2,5-dione derivatives (4–11) A total of 0.01 mol of the appropriately substituted 1-(3-aminopropyl)-4-arylpiperazine was dissolved in 20 ml of water and 0.01 mol of 2-methyl-2-phenyl- succinic acid (1) was gradually added. The mixture was heated in termperature-regulated sand bath (ST 72 Roth, Karlsruhe, Germany) with simultaneous dis-

ucts were converted into hydrochloride salts in anhy- drous ethanol saturated with HCl gas. The obtained precipitates were crystallized from anhydrous ethanol.

General procedure for the preparation of the 1-(3-chloropropyl)-3-ethyl-3-methylpyrro- lidine- 2,5-dione (3)

A mixture of 3-ethyl-3-methyl-pyrrolidine-2,5-dione (Sigma-Aldrich) (2) (0.06 mol), 1-bromo-3-chlorop- ropane (0.06 mol) and anhydrous K2CO3(0.3 mol) in acetone was stirred at 60°C for 24 h. Then, the inor- ganic residue was filtered off and acetone was evapo- rated to dryness. The obtained 1-(3-chloropropyl)-3- ethyl-3-methylpyrrolidine-2,5-dione (3) was used to the next step of reaction without purification.

General procedure for the preparation of the 1-[(4-arylpiperazinyl-1-yl)-propyl]-3-ethyl-3- methylpyrrolidine-2,5-dione derivatives (12–19) A mixture of appropriately substituted 4-arylpipe- razine (0.0075 mol), 1-(3-chloropropyl)-3-ethyl-3- methylpyrrolidine-2,5-dione (3) (0.0075 mol) and an- hydrous K2CO3(0.0375 mol) in 50 ml of acetone was stirred at 60°C for 24 h. Then, the inorganic solid was filtered off and acetone was evaporated to dryness.

N N

O

C H3 C H3

O

N Cl N

O

C H3

C H3

O C

H3

NH

O O

C H3

N N

N O

O

C H3 OH

O OH O

C H3

a

1 4 - 11

b

c

2 3

12 -19

R R

Compound 4, 12 5, 13 6, 14 7, 15 8, 16 9, 17 10, 18 11, 19

R1 H 2-F 4-F 2-Cl 3-Cl 4-Cl 3-CF3 2-OCH3

Fig. 1. Synthetic pathways of compounds 4–19. Reagents and reaction condi- tions: (a) 1-(3-aminopropyl)-4-arylpi- perazine, 2 h, 180°C; (b) 1-bromo-3- chloropropane, K2CO3,acetone, 60°C;

24 h; (c) 4-arylpiperazine, acetone, K2CO3, 60°C, 24 h

(4)

The obtained oily residue was purified by column chromatography on Silica gel 60 (Merck, Darmstadt, Germany) using a chloroform-methanol (9:0.7, v/v) mixture as a solvent system. The crude products were converted into hydrochloride salts in anhydrous etha- nol saturated with HCl gas. The obtained precipitates were crystallized from anhydrous ethanol.

1-{3-[4-(2-Fluorophenyl)-piperazin-1-yl]-propyl}- 3-methyl-3-phenylpyrrolidine-2,5-dione mono- hydrochloride (5)

Yield: 30%. Analysis: for C24H28O2N3F × HCl (445.9):

C, H, N; m.p. 152–154°C; TLC Rf = 0.85 (S1), 1H NMR (DMSO, d, ppm): 1.65 (s, 3H, CH3), 2.24–2.34 (m, 2H, CH2-CH2-CH2), 2.89–3.00 (m, 5H, 4H, pipera- zine, 1H, imide), 3.18 (d, 1H, imide, J = 18.43 Hz), 3.41 (d, 2H, CH2-CH2-CH2, J = 10.22 Hz), 3.58 (t, 4H, piperazine, J = 6.95 Hz) 3.68–3.72 (m, 2H, CH2-CH2-CH2), 6.93–7.24 (m, 4H, ArH), 7.27–7.42 (m, 5H, ArH), 12.99 (brs, 1H, HCl).

1-{3-[4-(4-Fluorophenyl)-piperazin-1-yl]-propyl}- 3-methyl-3-phenylpyrrolidine-2,5-dione mono- hydrochloride (6)

Yield: 33%. Analysis: for C24H28O2N3F × HCl (445.9):

C, H, N; m.p. 203–206°C; TLC Rf = 0.84 (S1), 1H NMR (DMSO, d, ppm): 1.64 (s, 3H, CH3), 1.97–2.07 (m, 2H, CH2-CH2-CH2), 3.00–3.17 (m, 8H, 2H, CH2-CH2-CH2, 4H, piperazine, 2H, imide), 3.48–3.53 (t, 4H, piperazine, J = 6.92 Hz), 3.69 (d, 2H, CH2-CH2-CH2, J = 10 Hz), 6.98–7.12 (m, 4H, ArH), 7.24–7.41 (m, 5H, ArH), 11.27 (brs, 1H, HCl).

1-{3-[4-(2-Chlorophenyl)-piperazin-1-yl]-propyl}- 3-methyl-3-phenylpyrrolidine-2,5-dione mono- hydrochloride (7)

Yield: 48%. Analysis: for C24H28O2N3Cl × HCl (462.4):

C, H, N; m.p. 176–178°C; TLC Rf = 0.88 (S1), 1H NMR (DMSO, d, ppm): 1.65 (s, 3H, CH3), 2.04–2.09 (m, 2H, CH2-CH2-CH2), 2.94 (d, 1H, imide, J = 18.15 Hz), 3.00–3.09 (m, 4H, piperazine), 3.19 (d, 1H, imide, J = 18.43 Hz), 3.40 (d, 2H, piperazine, J = 12.65 Hz), 3.56 (d, 2H, piperazine, J = 11.83 Hz), 3.70–3.80 (m, 4H, CH2-CH2-CH2), 7.08–7.19 (m, 1H, ArH), 7.22–7.43 (m, 8H, ArH), 13.03 (brs, 1H, HCl).

1-{3-[4-(3-Chlorophenyl)-piperazin-1-yl]-propyl}- 3-methyl-3-phenylpyrrolidine-2,5-dione mono- hydrochloride (8)

Yield: 44%. Analysis: for C24H28O2N3Cl × HCl (462.4): C, H, N; m.p. 201–203°C; TLC Rf = 0.85 (S1), 1H NMR (DMSO, d, ppm): 1.75 (s, 3H, CH3), 2.18–2.37 (m, 2H, CH2-CH2-CH2), 2.84–2.97 (m, 5H, 4H, piperazine, 1H imide), 3.17 (d, 1H, imide, J = 18.17 Hz), 3.56–3.65 (t, 4H, piperazine, J = 12.31 Hz), 3.68–3.72 (m, 4H, CH2-CH2-CH2), 6.74–6.78 (m, 2H, ArH), 6.86–6.93 (m, 1H, ArH), 7.17–7.38 (m, 6H, ArH), 13.17 (brs, 1H, HCl).

1-{3-[4-(4-Chlorophenyl)-piperazin-1-yl]-propyl}- 3-methyl-3-phenylpyrrolidine-2,5-dione mono- hydrochloride (9)

Yield: 36%. Analysis: for C24H28O2N3Cl × HCl (462.4): C, H, N; m.p. 215–217°C; TLC Rf = 0.82 (S1), 1H NMR (DMSO, d, ppm): 1.60 (s, 3H, CH3), 2.26–2.32 (m, 2H, CH2-CH2-CH2), 2.85–2.97 (m, 5H, 1H, imide, 4H, piperazine), 3.17 (d, 1H, imide, J = 18.15 Hz), 3.48–3.61 (t, 4H, 4H piperazine, J = 18.29 Hz), 3.65–3.73 (m, 4H, CH2-CH2-CH2), 6.82–6.85 (m, 2H, ArH), 7.22–7.39 (m, 7H, ArH), 13.10 (brs, 1H, HCl).

1-{3-[4-(3-Trifluoromethylphenyl)-piperazin- 1-yl]-propyl}-3-methyl-3-phenylpyrrolidine-2,5- dione monohydrochloride (10)

Yield: 41%. Analysis: for C25H28O2N3F3 × HCl (495.9): C, H, N; m.p. 185–186°C; TLC Rf = 0.86 (S1), 1H NMR (DMSO, d, ppm): 1.76 (s, 3H, CH3), 2.28–2.33 (m, 2H, CH2-CH2-CH2), 2.92 (d, 1H, imide, J = 18.29 Hz), 2.97–3.03 (m, 4H, piperazine), 3.18 (d, 1H, imide, J = 18.29 Hz), 3.57–3.65 (t, 4H, piperazine, J = 12.43 Hz), 3.69–3.78 (m, 4H, CH2-CH2-CH2), 7.05–7.22 (m, 2H, ArH), 7.23–7.39 (m, 2H, ArH), 7.41–7.45 (m, 5H, ArH), 13.26 (brs, 1H, HCl).

1-{3-[4-(2-Methoxyphenyl)-piperazin-1-yl]-propyl}- 3-methyl-3-phenylpyrrolidine-2,5-dione mono- hydrochloride (11)

Yield: 44%. Analysis: for C25H31O3N3× HCl (458.0):

C, H, N; m.p. 212–214°C; TLC Rf= 0.79 (S1),1H NMR (DMSO, d, ppm): 1.64 (s, 3H, CH3), 1.98–2.03 (m, 2H, CH2-CH2-CH2), 2.97 (d, 1H, imide, J = 17.95 Hz), 3.04

Anticonvulsant activity and 5-HT1A/5-HT7affinity of certain succinimides

Joanna Obniska et al.

(5)

OCH3), 6.86–7.04 (m, 5H, ArH), 7.26–7.42 (m, 5H, ArH), 10.95 (brs, 1H, HCl).

1-[3-(4-Phenylpiperazin-1-yl)-propyl]-3-ethyl- -3methylpyrrolidine-2,5-dione monohydrochlo- ride (12)

Yield: 42%. Analysis: for C20H29O2N3× HCl (379.9):

C, H, N; m.p. 188–190°C; TLC Rf = 0.76 (S1), 1H NMR (DMSO, d, ppm): 0.78 (t, 3H, CH2-CH3, J = 7.44 Hz), 1,18 (s, 3H, CH3), 1.48–1.63 (m, 2H, CH2-CH3), 1.91–1.98 (m, 2H, CH2-CH2-CH2), 2.46 (d, 1H, imide, J = 8.21 Hz), 2.64 (d, 1H, imide, J = 18.21 Hz), 3.04–3.20 (m, 6H, 4H, piperazine, 2H, CH2-CH2-CH2), 3.40–3.51 (m, 4H, piperazine), 3.78 (d, 2H, CH2-CH2-CH2, J = 9.49 Hz), 6.84 (t, 1H, ArH, J = 7.31 Hz), 6.98 (d, 2H, ArH, J = 7.69 Hz), 7.22–7.27 (m, 2H, ArH), 11.04 (brs, 1H, HCl).

1-{3-[4-(2-Fluorophenyl)-piperazin-1-yl]-propyl}- 3-ethyl-3-methylpyrrolidine-2,5-dione monohy- drochloride (13)

Yield: 47%. Analysis: for C20H28O2N3F × HCl (397.9):

C, H, N; m.p. 195–197°C; TLC Rf = 0.80 (S1), 1H NMR (DMSO, d, ppm): 0.89 (t, 3H, -CH2-CH3, J = 7.44 Hz), 1,32 (s, 3H, CH3), 1.58–1.78 (m, 2H, CH2-CH3), 2.25 (brs, 2H, CH2-CH2-CH2), 2.47 (d, 1H, imide, J = 18.46 Hz), 2.65 (d, 1H, imide, J = 18.21 Hz), 3.13 (m, 2H, CH2-CH2-CH2), 3.58– 3.67 (m, 6H, 4H, piperazine, 2H, CH2-CH2-CH2), 3.80 (brs, 2H, piperazine), 4.34 (brs, 2H, piperazine), 7.16–7.24 (m, 3H, ArH), 7.62–7.70 (m, 1H, ArH), 12.04 (brs, 1H, HCl).

1-{3-[4-(4-Fluorophenyl)-piperazin-1-yl]-propyl}- 3-ethyl-3-methylpyrrolidine-2,5-dione monohy- drochloride (14)

Yield: 48%. Analysis: for C20H28O2N3F × HCl (397.9): C, H, N; m.p. 228–231°C; TLC Rf = 0.78 (S1), 1H NMR (DMSO, d, ppm): 0.78 (t, 3H, -CH2-CH3, J = 7.43 Hz), 1.19 (s, 3H, CH3), 1.47–1.63 (m, 2H, CH2-CH3), 1.94 (brs, 2H, CH2-CH2-CH2), 2.40 (d, 1H, imide, J = 18.46 Hz), 2.63 (d, 1H, imide, J= 18.21 Hz), 3.06 (brs, 6H, 2H, CH2-CH2-CH2, 4H, piperazine), 3.40–3.51 (m, 4H, piperazine) 3.69 (d,

1-{3-[4-(2-Chlorophenyl)-piperazin-1-yl]-propyl}- 3-ethyl-3-methylpyrrolidine-2,5-dione monohy- drochloride (15)

Yield: 41%. Analysis: for C20H28O2N3Cl × HCl (414.4): C, H, N; m.p. 167–170°C; TLC Rf = 0.82 (S1), 1H NMR (DMSO, d, ppm): 0.88 (t, 3H, -CH2-CH3, J = 7.43 Hz), 1.32 (s, 3H, CH3), 1.58–1.78 (m, 2H, CH2-CH3), 2.27 (brs, 2H, CH2-CH2-CH2), 2.47 (d, 1H, imide, J = 18.53 Hz), 2.65 (d, 1H, imide, J= 18.43 Hz), 3.05 (brs, 4H, piperazine), 3.39 (d, 2H, CH2-CH2-CH2, J = 12.10 Hz), 3.62–3.68 (m, 6H, 4H, piperazine, 2H, CH2-CH2-CH2), 7.03–7.10 (m, 1H, ArH), 7.22–7.25 (m, 2H, ArH), 7.27–7.39 (m, 1H, ArH), 13.00 (brs, 1H, HCl).

1-{3-[4-(3-Chlorophenyl)-piperazin-1-yl]-propyl}- 3-ethyl-3-methylpyrrolidine-2,5-dione monohy- drochloride (16)

Yield: 48%. Analysis: for C20H28O2N3Cl × HCl (414.4):

C, H, N; m.p. 192–195°C; TLC Rf = 0.81 (S1), 1H NMR (DMSO, d, ppm): 0.87 (t, 3H, -CH2-CH3, J = 7.44 Hz), 1.29 (s, 3H, CH3), 1.53–1.83 (m, 2H, CH2-CH3), 2.37–2.43 (m, 4H, 2H imide, 2H, CH2-CH2-CH2), 2.55–2.63 (m, 6H, 2H, CH2-CH2- CH2, 4H, piperazine), 3.19 (t, 4H, piperazine, J = 5.13 Hz), 3.57 (t, 2H, CH2-CH2-CH2, J = 7.18 Hz), 6.75–6.86 (m, 3H, ArH), 7.12–7.26 (m, 1H, ArH), 12.30 (brs, 1H, HCl).

1-{3-[4-(4-Chlorophenyl)-piperazin-1-yl]-propyl}- 3-ethyl-3-methylpyrrolidine-2,5-dione monohy- drochloride (17)

Yield: 52%. Analysis: for C20H28O2N3Cl × HCl (414.4): C, H, N; m.p. 155–157°C; TLC Rf = 0.80 (S1), 1H NMR (DMSO, d, ppm): 0.86 (t, 3H, -CH2-CH3, J = 7.44 Hz), 1.29 (s, 3H, CH3), 1.56–1.84 (m, 4H, CH2-CH3; CH2-CH2-CH2), 2.37–2.43 (m, 3H, CH2-CH2-CH2, 1H, imide), 2.56–2.68 (m, 5H, 4H, piperazine, 1H, imide), 3.14–3.23 (m, 4H, pipera- zine), 3.52–3.59 (m, 2H, CH2-CH2-CH2), 6.81–6.85 (m, 2H, ArH), 7.16–7.26 (m, 2H, ArH.), 11.50 (brs, 1H, HCl).

(6)

1-{3-[4-(3-Trifluoromethylphenyl)-piperazin-1- yl]-propyl}-3-ethyl-3-methylpyrrolidine-2,5- dione monohydrochloride (18)

Yield: 47%. Analysis: for C21H28O2N3F3 × HCl (447.9): C, H, N; m.p. 187–188°C; TLC Rf= 0.84 (S1),

1H NMR (DMSO, d, ppm): 0.76 (t, 3H, -CH2-CH3, J = 7.43 Hz), 1.31 (s, 3H, CH3), 1.55–1.79 (m, 2H, CH2-CH3), 2.21–2.31 (m, 2H, CH2-CH2-CH2), 2.46 (d, 1H, imide, J = 18.43 Hz), 2.64 (d, 1H, imide, J = 18.15 Hz), 2.94–3.10 (m, 4H, piperazine), 3.61–3.79 (m, 8H, 4H, CH2-CH2-CH2, 4H, piperazine), 7.05–

7.27 (m, 3H, ArH), 7.39 (t, 1H, ArH, J = 8.25 Hz), 13.20 (brs, 1H, HCl).

1-{3-[4-(2-Methoxyphenyl)-piperazin-1-yl]-propyl}- 3-ethyl-3-methylpyrrolidine-2,5-dione monohy- drochloride (19)

Yield: 35%. Analysis: for C21H31O3N3× HCl (409.9):

C, H, N; m.p. 197–200°C; TLC Rf = 0.75 (S1), 1H NMR (DMSO, d, ppm): 0.79 (t, 3H, CH2-CH3, J = 7.44 Hz), 1.20 (s, 3H, CH3), 1.48–1.64 (m, 2H, CH2-CH3), 1.93–1.98 (m, 2H, CH2-CH2-CH2), 2.49 (d, 1H, imide, J = 17.95 Hz), 2.64 (d, 1H, imide, J = 18.21 Hz), 3.03–3.18 (m, 6H, 4H, piperazine, 2H, CH2-CH2-CH2), 3.40–3.47 (m, 6H, 4H, piperazine, 2H, CH2-CH2-CH2), 3.77 (s, 3H, OCH3), 6.86–7.04 (m, 4H, ArH), 11.02 (brs, 1H, HCl).

PHARMACOLOGICAL PART

In vivoexperiments

Compounds 4–19 were pharmacologically pre- evaluated within the Antiepileptic Drug Development (ADD) Program, Epilepsy Branch, Neurological Dis- orders Program, National Institutes of the Neurologi- cal and Communicative Disorders and Stroke (NINCDS), Rockville using procedures described elsewhere [13, 14].

Male albino mice (CF-1 strain) and male albino rats (Sprague-Dawley) were used as experimental ani- mals. The animals were housed in metabolic cages and allowed free access to food and water. The com- pounds were suspended in 0.5% methylcellulose/wa- ter mixture. All the compounds were injected intrape- ritoneally into mice at the dose levels of 30, 100, and 300 mg/kg with anticonvulsant activity and motor im- pairment assessments at 0.5 and 4 h after administra- tion. The results are presented in Table 1.

Selected derivatives (8, 10, 16 and 17) were admin- istrated orally into rats using four animals at a fixed dose of 30 mg/kg (MES test) (Phase VIa). The motor impairment was studied in parallel. Rats were tested at five time periods ranging from one quarter to 4 h after substance administration.

In the maximal electroshock screen (MES), an electrical stimulus of 0.2 s in duration (50 mA in mice and 150 mA in rat at 60 Hz) is delivered via corneal electrodes primed with an electrolyte solution con- taining an anesthetic agent.

The motor impairment induced by compound was detected in mice or rats using standardized rotorod test [6]. Untreated control mice or rats when placed on the rod, can maintain their equilibrium for a pro- longed time period. The acute motor impairment can be demonstrated by the inability of animal to maintain equilibrium for a given time.

The quantitative determination of ED50and TD50val- ues for 8 and 17 was performed at previously estimated time of peak effect after oral administration into rats.

Groups of eight rats received various doses of the com- pound until at least three points were established in the range of 10–90% seizure protection or minimal motor impairment. From the plot of the data obtained, the re- spective ED50and TD50values, 95% confidence intervals, slope of the regression line, and standard error of the slope were calculated by means of a computer program written at NINDS/NIH. The results are shown in Table 2.

The 6-Hz model. This screen was carried out ac- cording to the protocol originally described by Brown et al. [4] and more recently by Barton et al. [2] and Kaminski et al. [8]. It is an alternative electroshock paradigm that uses low-frequency (6 Hz), long- duration (3 s) electrical stimulation. Corneal stimula- tion (0.2 ms-duration monopolar rectangular pulses at 6-Hz for 3 s) was delivered by a constant-current de- vice. During the stimulation, mice were manually re- strained and released into the observation cage imme- diately after the current application. The seizures manifest in “stunned” posture associated with rearing, forelimb, automatic movements and clonus, twitching of the vibrissae and Straub-tail. The duration of the seizure activity ranges from 60 to 120 s in untreated animals. At the end of the seizure, animals resume their normal exploratory behavior. The experimental end point is protection against the seizure. The animal is considered to be protected if it resumes its normal exploratory behavior within 10 s from the stimulation [8].

Anticonvulsant activity and 5-HT1A/5-HT7affinity of certain succinimides

Joanna Obniska et al.

(7)

5-HT1Aand 5-HT7receptors in vitro were assessed on the basis of their ability to displace [3H]-8-OH-DPAT (170 Ci/mmol, NEN Chemicals, USA) or [3H]-5-CT (93.0 Ci/mmol, Amersham), respectively. In both cases nonspecific binding was defined in the presence of 10 µM of 5-HT. Radioligand binding experiments for 5-HT1A receptors were carried out on rat brain (hippocampus) and for 5-HT7receptors on HEK 293 cells stably expressing human 5-HT7(b)receptor splice variant according to the previously published proce- dures [28]. Ki values were determined from at least three competitions binding experiments in which

analyzed using iterative curve fitting routines (Graph- Pad/Prism, Version 3.0, San Diego, CA, USA).

Results and Discussion

The preliminary anticonvulsant screening of the com- pounds investigated involved two mechanism- independent, animal seizure models: the maximal electroshock seizure (MES) test – to identify com-

Tab. 1. Chemical structures, anticonvulsant activity and the 5-HT1Aand 5-HT7receptor affinities of compounds 4–19

Cmpd R R1 MESa NTb Ki(nM)c

0.5 h 4 h 0.5h 4 h 5-HT1A 5-HT7

4* Phenyl H 30 100 100 313 263

5 Phenyl 2-F 3001 162 316

6 Phenyl 4-F 100 3001 1268 190

7 Phenyl 2-Cl 3001 157 112

8 Phenyl 3-Cl 300 30 300 109 76

9 Phenyl 4-Cl 300 100 1544 245

10 Phenyl 3-CF3 100 100 30014 107 187

11 Phenyl 2-OCH3 3001 72 63

12 Ethyl H 100 3001 180 269

13 Ethyl 2-F 3001 206 510

14 Ethyl 4-F 30023 3001 554 272

15 Ethyl 2-Cl 3001 44 138

16 Ethyl 3-Cl 30 44 186

17 Ethyl 4-Cl 100 454 312

18 Ethyl 3-CF3 100 100 3001 43 124

19 Ethyl 2-OCH3 100 14 187

PHTd 30 30 100 100

Doses of 30, 100 and 300 mg/kg were administrated intraperitoneally in mice. The figures indicate the minimum dose whereby anticonvulsant activity or neurotoxicity was demonstrated in 50% of the animals. A dash indicates the absence of anticonvulsant activity and neurotoxicity at the maximum dose administered (300 mg/kg). * The data from ref. [26].aMaximal electroshock test.bNeurotoxicity screening using rotorod test.cValues are the means of three experiments run in triplicate, SEM £ 18%.dPHT – phenytoin, reference drug, data from ref. [31]. Response comments:1death,14unable to grasp rotorod,23clonic seizures

(8)

pounds which prevent seizure spread, consistent with generalized tonic-clonic seizures in humans [14, 18], and the subcutaneous pentylenetetrazole seizure (scPTZ) test – to evaluate compound’s ability to raise the seizure threshold, a rodent model of absence epi- lepsy. The motor impairment of all the compounds was investigated by rotorod test (NT). The results are shown in Table 1.

The compounds studied were effective only in the MES test (4, 6, 8–10, 12 and 16–18) or were devoid of activity in both tests used – 5, 7, 11, 13–15, and 19.

Compounds 4, 8 and 16 were active at a dose of 30 mg/kg whereas 6, 9, 10, 12, 17 and 18 at a dose of 100 mg/kg. Five molecules: 4, 8, 9, 10 and 18 showed pro- tection in both time intervals – 0.5 h and 4 h. The other were active only 4 h after ip administration that indicates the slow onset and long duration of anticon- vulsant action. In the rotorod test (NT) compounds 9, 16 and 17 did not show motor impairment in the maximum dose administered (300 mg/kg) and emerged as anticonvulsants devoid of motor impair- ment properties. The other compounds exhibited mo- tor impairment at the dose of 100 mg/kg – 4, 19 or 300 mg/kg – 5–7, 10–15 and 18.

A valuable property of a candidate anticonvulsant is its ability to inhibit convulsions when given by the oral route. This screen discloses the time of onset, the approximate time of peak effect (TPE) and the dura- tion of anticonvulsant activity or neurotoxicity. On the basis of the ip screening data in mice, and according to Anticonvulsant Screening Project (ASP) disposi- tion, four compounds 8, 10, 16 and 17 were selected and examined in the MES test as well as rotorod screen after po administration into rats at a dose of 30 mg/kg.

The most active were compounds 16 and 17 that protected 100% of animals at 4 h (16, 17) and at 2 h (17). These molecules provided 50% protection at 2 h (16) or 1 h (17), and 25% at 1 h (16). The other com- pounds were less active, namely 8 showed the peak of 50% protection at 2 h and 4 h, as well as 25% at 1 h, whereas 10 was effective in 50% of animals only at 0.5 h. All derivatives tested did not cause motor im- pairment when given orally.

On the basis of the rats oral data, compound 8 and 17 were chosen for phase II evaluation for quantifica- tion of the pharmacological parameters (ED50 and TD50). The quantitative evaluation of the MES me- dian effective dose (ED50) and toxic dose (TD50) were performed after oral administration to rats at previ- ously estimated TPE. Results of the quantitative tests along with the data for the standard drug – phenytoin, are shown in Table 2. As can be seen from these data, compound 8 revealed an ED50of 28.20 mg/kg, TD50 of 268.50 mg/kg and the protection index (TD50/ED50) of 9.52, whereas 17 was less active and revealed ED50 120 mg/kg and TD50 > 500 mg/kg. The quantitative data showed that compound 8 have comparable ED50 to phenytoin which was used as reference drug for an- ticonvulsants active in the MES test.

According to the ASP dispositions compounds 6, 8, 9 and 16 were chosen for the evaluation of anticon- vulsant properties in the 6-Hz test. The selection was made randomly as a part of the search of molecules providing anti 6-Hz protection among chemically di- versified compounds pursued in the NIH/NINDS. The 6-Hz screen has been validated as a model of therapy-resistant epilepsy recently. Moreover, it is re- garded as capable for identifying anti-seizure agents

Anticonvulsant activity and 5-HT1A/5-HT7affinity of certain succinimides

Joanna Obniska et al.

Tab. 2. Quantification studies of 8 and 17 in the MES and rotorod tests in rats after po administration

Compound TPE (h)a ED50b

MES (mg/kg)

TD50c

(mg/kg)

Pid MES (TD50/ED50)

8 4 28.2

(5.4–59.1)e

268.5 (174.3–378.3)

9.5

17 4 > 120 > 500

PHTf 2 28.1

(20.7–35.2)

> 100 3.6

aTime of peak effect.bED50– median effective dose required to assure anticonvulsant protection in 50% animals.cTD50– median toxic dose eliciting minimal motor impairment in 50% animals.dPI protective index (TD50/ED50).e95% confidence limits given in parentheses.fReference drug, data for phenytoin ref. [32]

(9)

which administrated ip into mice at a dose of 100 mg/kg, protected 75% of animals at 0.25 h and 25% at 1 h. It’s 4-chloro analog 9 protected 50% of mice at 0.25 h. Compound 16 was less active and protected 25% of mice at 1 h and 4 h. Finally, derivative 8 was inactive in this test. The result of rotorod screen re- vealed that compounds 6, 8 and 16 caused the motor impairment at a dose of 100 mg/kg at 0.25 h (6, 8), whereas 16 at 0.5 h and 1 h. Compound 9 did not cause motor impairment in all time intervals.

In conclusion, the results of preliminary screening revealed that anticonvulsant activity in this series of compounds depended on the kind and the position of substituents at the 4-phenylpiperazine moiety as well as on the substitution mode at the position-3 of the succinimide. The most active were compounds with the phenyl moiety at position-3 of pyrrolidine-2,5- dione. The exchange of mentioned aromatic ring into aliphatic ethyl group decreased anticonvulsant activ- ity. Taking into consideration the 4-phenylpiperazine fragment it seems that the most favorable for activity is a lack of substituents or the presence of electron- withdrawing chloro atom or trifluoromethyl group at meta- or para-position.

In order to further characterize the compounds ob- tained and taking into account the presence of aryl- piperazine pharmacophore, their ability to displace [3H]-OH-DPAT and [3H]-5-CT binding to rat 5-HT1A and recombinant 5-HT7receptors was examined (Tab.

1). As had been anticipated, the novel arylpiperazines (4–19) exhibited diversified affinities for 5-HT1Are- ceptors, resulting from the substitution mode in the phenyl ring. It should be stressed here, that the influ- ence of various substituents in the aryl part on com- pound affinity at either receptor has been well docu- mented, and our results are in line with the general tendencies observed earlier [16, 17]. In short, 2-OCH3 (11, 19) and 3-Cl (8, 16) derivatives were the most po- tent 5-HT1A receptor ligands (Ki = 72, 14 nM, and 109, 44 nM, respectively), 2-Cl substituted com- pounds were usually only slightly less active or showed comparable activity, whereas introduction of halogen atoms in para- position of phenyl ring re- sulted in reduction of affinity. With respect to the 5-HT7receptors, the best Kivalues were obtained for derivatives 8 and 11 (Ki= 76 nM and 63 nM, respec- tively), other compounds demonstrated medium affin-

pharmacological studies, the results of radioligand ex- periments indicate the possibility of development this group as potential psychotropic agents, which activity is connected with modulation of investigated types of serotonin receptors. Furthermore, the moderate affini- ties to the 5-HT1A and 5-HT7 receptors, and on the other hand, the inhibition of electrically induced sei- zures may indicate rather the influence of compounds on voltage-depended sodium channels as the most probable mechanism of anticonvulsant action. Such interaction was documented for many succinimides with the arylpiperazine moieties that were active in the MES test [20].

Acknowledgments:

The authors wish to thank Dr. James Stables for providing pharmacological data through the Antiepileptic Drug Development (ADD) Program in Epilepsy Branch, Neurological Disorders Program, National Institute of the Neurological and Communicative Disorders and Stroke (NIH/NINDS, Rockville, MD, USA) and Prof.

Katarzyna Kieæ-Kononowicz (Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, Kraków, Poland) the coordinator of the ADD project from Polish site.

We are pleased to acknowledge the generous financial support of this work by the grant of the State Committee for Scientific Research, Poland (Grant No. N405 298536).

Radioligand binding experiments were financially supported by the grant PNRF-103-AI-1/07 from Norway through the Norwegian Financial Mechanism.

References:

1.Badarau E, Suzenet F, Bojarski A, Finaru A, Guillaumet G: Benzimidazolone-based serotonin 5-HT1Aor 5-HT7R ligands: Synthesis and biological evaluation. Bioorg Med Chem Lett, 2009, 19, 1600–1603.

2.Barton ME, Klein BD, Wolf HH, White HS: Pharma- cological characterization of the 6 Hz psychomotor sei- zure model of partial epilepsy. Epilepsy Res, 2001, 47, 217–227.

3.Brodie M J: Antiepileptic drug therapy the story so far.

Seizure, 2010, 19, 650–655.

4.Brown WC, Schiffman DO, Swinyard EA, Goodman LS:

Comparative assay of an antiepileptic drugs by psycho- motor seizure test and minimal threshold test. J Pharma- col Exp Ther, 1953, 107, 273–289.

5.Cheng Y, Prusoff WH: Relationship between the inhibi- tion constant (K1) and the concentration of inhibitor which causes 50 per cent inhibition (I50) of an enzymatic reaction. Biochem Pharmacol, 1973, 22, 3099–3108.

(10)

6.Dunham NW, Miya TA: A note on a simple apparatus for detecting neurological deficit in rats and mice. J Am Pharm Assoc Sci, 1957, 46, 208–209.

7.Glennon RA, Naiman NA, Lyon RA, Titeler M: Aryl- piperazine derivatives as high-affinity 5-HT1Aserotonin ligands. J Med Chem, 1988, 31, 1968–1971.

8.Kaminski RF, Livingood MR, Rogawski MA: Allopreg- nanolone analogues that positively modulate GABAAre- ceptors protect against partial seizures induced by 6-Hz electrical stimulation in mice. Epilepsia, 2004, 45, 864–873.

9.Kamiñski K, Obniska J: Design, synthesis and anticonvul- sant activity of N-phenylamino derivatives of 3.3-dialkyl- pirolidine-2,5-diones and hexahydro-isoindole-1,3-diones.

Bioorg Med Chem, 2008, 16, 4921–4931.

10.Kamiñski K, Obniska J: Synthesis and anticonvulsant properties of new N-pyridine-2-yl 3-substituted pirolidine-2,5-dione derivatives. Acta Pol Pharm Drug Res, 2008, 65, 457–465.

11.Katritzky AR, Satheesh KN, Witek RM, Hutchins SM:

Synthesis of 3,3-diarylpyrrolidines from diaryl ketones.

ARKIVOC, 2003, (v), 9–18.

12.Khan H N, Kulsoom S, Rashid H: Ligand based pharma- cophore model development for the identification of novel antiepileptic compound. Epilepsy Res, 2012, 98, 62–71.

13.Krall RL, Penry JK, White BG, Kupferberg HJ, Swinyar EA: Antiepileptic drug development II. Anticonvulsant drug screening. Epilepsia, 1978, 19, 400–428.

14.Kupferberg HJ: Antiepileptic drug development pro- gram: a cooperative effort of government and industry.

Epilepsia, 1989, 30, Suppl, 51–56.

15.Leopoldo M, Lacivita E, Berardi F, Perrone R, Hedlund PB: Serotonin 5-HT7 receptor agents: Structure-activity relationships and potential therapeutic applications in central system disorders. Pharmacol Ther, 2011, 129, 120–148.

16.Lewgowd W, Bojarski A, Szczesio M, Olczak A, Glowka M, Mordalski S, Stanczak A: Synthesis and structural investigation of some pyrimido[5,4-c]quinolin- 4(3H)-one derivatives with a long-chain arylpiperazine moiety as potent 5-HT1A/2Aand 5-HT7receptor ligands.

Eur J Med Chem, 2011, 4, 3348–3361.

17.López-Rodríguez M, Ayala D, Benhamú B, Morcillo M, Viso A: Arylpiperazine derivatives acting at 5-HT1Are- ceptors. Curr Med Chem, 2002, 9, 443–469.

18.Löscher W: Critical review of current animal models of seizures and epilepsy used in the discovery and develop- ment of new antiepileptic drugs. Seizure, 2011, 20, 359–368.

19.Mirski M, Ziai W, Chiang J, Hinich M, Sherman D: An- ticonvulsant serotonergic and deep brain stimulation in anterior thalamus. Seizure, 2009, 18, 64–70.

20.Obniska J, Byrtus H, Kamiñski K, Paw³owski M, Szczêsio M, Karolask-Wojciechowska J: Design, synthe- sis, and anticonvulsant activity of new N-Mannich bases

derived from spirosuccinimides. Bioorg Med Chem, 2010, 18, 6134–6142.

21.Obniska J, Chlebek I, Pichór J, Kopytko M, Kamiñski K:

Synthesis, physicochemical and anticonvulsant proper- ties of new N-[(4-arylpiperazin-1-yl)-alkyl]-3-phenyl- and 3-(3-methylphenyl)-pyrrolidine-2,5-diones. Acta Pol Pharm Drug Res, 2009, 66, 639–647.

22.Obniska J, Jurczyk S, Zejc A, Kamiñski K, Tatarczyñska, Stachowicz K: Anticonvulsant properties of N-(4-methyl- piperazin-1-yl) and N-[(4-methylpiperazin-1-yl)-propyl]

derivatives of 3-aryl and 3-spirocycloalkyl-pyrrolidine- 2,5-dione. Pharmacol Rep, 2005, 57, 170–175.

23.Obniska J, Kamiñski K, Skrzyñska D, Pichór J: Synthe- sis and anticonvulsant activity of new N-[(4-arylpiperazin- 1-yl)-alkyl]-derivatives of 3-phenylpyrrolidine-2,5-dione.

Eur J Med Chem, 2009, 44, 2224–2233.

24.Obniska J, Kamiñski K, Tatarczyñska E: Impact of aro- matic substitution on the anticonvulsant activity of new N-(4-arylpiperazin-1-yl)-alkyl-2-azaspiro[4.5]decane- 1,3-dione derivatives. Pharmacol Rep, 2006, 58, 207–214.

25.Obniska J, Kopytko M, Zagórska A, Chlebek I, Kamiñski K: Synthesis and anticonvulsant activity of new Mannich bases derived from 3-aryl-pyrrolidine-2,5-diones. Arch Pharm, 2010, 343, 333–341.

26.Obniska J, Kulig K, Zejc A: Synthesis and anticonvulsant properties of new 4-piperazinylalkyl imides of succinic acid. Acta Pol Pharm Drug Res, 1998, 55, 223–231.

27.Obniska J, Zagorska A: Synthesis and anticonvulsant proper- ties of new N-[(4-arylpiperazin-1-yl)-methyl] derivatives of 3-aryl pyrrolidine-2,5-dione and 2-aza-spiro[4.4]nonane-1,3- dione. Farmaco, 2003, 58, 1227–1234.

28.Paluchowska MH, Bugno R, Duszyñska B, Tatarczyñska E, Nikiforuk A, Lenda T, Chojnacka-Wójcik E: The in- fluence of modifications in imide fragment structure on 5-HT1Aand 5-HT7receptor affinity and in vivo pharma- cological properties of some new 1-(m-trifluoromethylphenyl- piperazines. Bioorg Med Chem, 2007, 15, 7116–7125.

29.Perièiæ D, Švob Štrac D: The role of 5-HT7receptors in the control of seizures. Brain Res, 2007, 1141, 48–55.

30.Perucca E, Tomson T: The pharmacological treatment of epilepsy in adults. Lancet Neurol, 2011, 10, 446–456.

31.Thirumurugan R, Sriram D, Saxena A, Stables J, Yogeeswari P: 2,4-Dimetoxyphenylsemicarbazones with anticonvulsant activity against tree animal models of seizures: Synthesis and pharmacological evaluation.

Bioorg Med Chem, 2006, 14, 3106–3112.

32.Yogeeswari P, Sriram D, Thirumurugan R, Raghaven- dran J, Sudhan K, Pavana P, Stables J: Discovery of N-(2,6-dimethylphenyl)-substituted semicarbazones as anticonvulsants: hybrid pharmacophore-based design.

J Med Chem, 2005, 48, 6202–6211.

Received: June 17, 2011; in the revised form: November 22, 2012;

accepted: December 5, 2011.

Anticonvulsant activity and 5-HT1A/5-HT7affinity of certain succinimides

Joanna Obniska et al.

Cytaty

Powiązane dokumenty

All the newly obtained compounds IIñX were tested for their anticonvulsant activity through the Anticonvulsant Screening Program (ASP) of the Na- tional Institute of

Evaluation of anticonvulsant activity and neu- rotoxicity was carried out according to the Anticonvulsant Screening Program (ASP) at National Institute of Neurological Disorders

The most potent in the maximum electroshock se- izure (MES) test were compounds with methyl [III] and chloro [XI] substituents at position -2 of the aromatic ring, whereas of all

These observations inspired us to: (i) search for triazole-based compounds endowed with anticonvulsant activity; and (ii) to check whether the obtained derivatives may act via

According to the Anticonvulsant Screening Project dis- positions, three compounds 13, 20 and 22 which showed protection in mice were selected and examined for their

the anticonvulsant activity without neurotoxic effects was shown in all the tested compounds; the most promising compound was VIII, which produced higher anticonvulsant protection

Comparing the activity and neurotoxicity between amides tested [I-V] and published previ- ously (12) and their amine analogues [VI-XVII], the achieved results of the

16 Given their localization and their ability to modulate several central neurotransmitter systems and as a continuation of our research, the H3R antagonist 2-18 with