1,003
Views
51
CrossRef citations to date
0
Altmetric
Research Article

Synthesis and carbonic anhydrase isoenzymes I and II inhibitory effects of novel benzylamine derivatives

, , &
Pages 168-174 | Received 30 Nov 2012, Accepted 30 Dec 2012, Published online: 07 Feb 2013

Abstract

Synthesis and carbonic anhydrase inhibitory properties of novel diarylmethylamines 2225 and sulfonamide derivatives 2628 were investigated. Acylation of methoxy-substituted benzenes with benzene carboxylic acids, reduction of ketones with NaBH4, conversion of alcohols to azides, Pd-C catalyzed hydrogenation of azides afforded title compounds 2225. Compounds 22, 24 and 25 were converted to sulfonamide derivatives 2628 with MeSO2Cl. The inhibitory effects of novel benzylamine derivatives 2228 were tested on human carbonic anhydrase (hCA, EC 4.2.1.1) isozymes hCA I and II. The results demonstrated that compound 28 was found to be the best inhibitor against both hCA I (Ki: 3.68 µM) and hCA II (Ki: 9.23 µM).

Introduction

Sulfonamides are important biologically active compounds. There are many drugs containing this functional group in their structuresCitation1. Anti-glaucoma agent dorzolamide (1), commercially known as trusopt, is a carbonic anhydrase (CA) inhibitorCitation2. Brinzolamide (2) is a CA inhibitor used to lower intraocular pressure in patients with open-angle glaucoma or ocular hypertensionCitation3. CA inhibitory properties of dichlorophenamide (3) have been reported by Alterio and co-workersCitation4. A CA inhibitor drug acetazolamide (AZA) (4) is used to treat glaucomaCitation5 and idiopathic intracranial hypertensionCitation6 ().

Figure 1.  Structures of some biologically active sulfonamides derivatives.

Figure 1.  Structures of some biologically active sulfonamides derivatives.

CA (EC 4.2.1.1.) is a pH regulatory/metabolic enzyme in all life kingdoms, found in organisms all over the phylogenetic treeCitation3,Citation7,Citation8 catalyzing the hydration of carbon dioxide (CO2) to bicarbonate () and the corresponding dehydration of in acidic medium with regeneration of CO2 following a three-step catalytic mechanismCitation3.

In the hydration direction, the first step is the nucleophilic attack of a Zn2+-bound hydroxide ion on CO2 (Equation (1)) with consequent formation of (Equation (3)), which is then displaced from the active site by a water molecule (Equation (2)). The third step, which is rate limiting, regenerates the catalytically active Zn2+-bound hydroxide ion through a proton transfer reaction from the Zn2+-bound water molecule to an exogenous proton acceptor or to an active site residue, generically represented by B (Equation (3)).

Recently, the structure of the human carbonic anhydrase (hCA) II/ complex was also providedCitation9. The lies in the same plane defined by the CO2 molecule and the Zn2+-bound OH and is tetrahedrally coordinated to the catalytic metal ion through one of its oxygen atoms. The availability of both the hCA II/CO2 and hCA II/ complex structures made it possible to obtain for the first time a detailed description of the first step of the enzymatic reaction providing new perspectives in the structural studies of this extremely interesting enzyme familyCitation3.

This metalloenzyme family is ubiquitous and present in majority of living organisms and divided to five unrelated gene families: the α-, β-, γ-, δ- and ζ-carbonic anhydrases (CAs). The α-, β- and δ-CAs contain a Zn2+ at the active site, the γ-CAs are almost certainly Fe2+ enzymes but they are active also with bound Zn2+ or Co2+, while the metal ion is usually replaced by cadmium in the ζ-CAsCitation2,Citation10. All hCAs belong to the α-class; up to this time, 16 isozymes have been identified, which differ by molecular features, oligomeric arrangement, cellular localization, distribution in organs and tissues, expression levels, kinetic properties and response to different classes of inhibitorsCitation3,Citation11. Some of these are cytosolic (CA I, CA II, CA III, CA VII and CA XIII), others are membrane bound (CA IV, CA IX, CA XII and CA XIV), two are mitochondrial (CA VA and CA VB) and one is secreted in saliva (CA VI). It has been reported that CA XV isoform is not expressed in humans or in other primatesCitation12,Citation13. Twelve isoforms (CAs I–IV, VA–VB, VI–VII, IX, and XII–XIV) show a variable degree of enzymatic activity, whereas three isoforms (VIII, X and XI), the so-called CA-related proteins, are devoid of any catalytic activityCitation3,Citation14. Several studies demonstrated the important roles of CAs in a variety of physiological processes, such as acid–base balance, respiration, carbon dioxide and ion transport, bone resorption, ureagenesis, gluconeogenesis, lipogenesis and body fluid generation. In addition, CAs demonstrated that the abnormal levels or activities of these enzymes have been frequently associated with different human diseasesCitation7,Citation8,Citation15.

It was reported that none of the current clinically used CA inhibitors displayed selectivity for a specific isoenzymeCitation3,Citation16. Therefore, developing isozyme-specific CA inhibitors is extremely important in order to obtain novel classes of drugs devoid of various undesired side effects. Consequently, in recent years CA isozymes have become an interesting target for the design of inhibitors or activators with biomedical applicationsCitation3,Citation17–19. Recently, numerous structural studies have provided a scientific basis for the rational drug design of more selective enzyme inhibitorsCitation20. Nevertheless, although X-ray crystal structures are available for the majority of the 12 catalytically active members of the hCA familyCitation21–23, most of the reported complexes with inhibitors regards just isozyme II, which is the most thoroughly characterized CA isoformCitation2.

In the current research, the first synthesis of a number of novel diarylmethylamine hydrochloride salts 22–25 and their sulfonamide derivatives 26–28 was studied. CA inhibitors are valuable molecules for therapeutic and pharmacological applications.

Experimental section

General information

All chemicals and solvents are commercially available. All solvents were distilled and dried according to standard procedures. Silica gel (SiO2, 60 mesh; Merck, Darmstadt, Germany) was used for column chromatography. About 1 mm of SiO2 60 PF (Merck) on glass plates was used for preparative thick layer chromatography. Melting point (m.p.) of all compounds was determined with cap melting-point apparatus (BUCHI 530, Flawil, Switzerland) and uncorrected. Infrared (IR) spectra were recorded as solutions in 0.1 mm cells with a Mattson 1000 FT-IR spectrophotometer (Cambridge, England). 1H- and 13C-NMR spectra were recorded on 400 (100)-MHz Varian spectrometer (Danbury, CT) in deuterated solvents (CDCl3 and D2O) with tetramethylsilane (SiMe4) as an internal standard for protons and solvent signals, as internal standard for carbon spectra. Chemical shift values were mentioned δ in ppm. Elemental analyses were recorded on Leco CHNS-932 apparatus (Saint Joseph, MI). CA inhibitory properties of samples were determined on a spectrophotometer (UV-1208, Shimadzu, Kyoto, Japan).

Syntheses of compounds (10Citation24, 11Citation25, 12Citation26, 13Citation27, 14Citation28, 15Citation28, 16Citation26, 17Citation29 and 22Citation30) have been reported previously.

General procedure for synthesis of azides 18–21

4,4′-(Azidomethylene)bis(1,2-dimethoxybenzene) (18)

To a stirred solution of alcohol 14 (3.12 g, 10.25 mmol) in CH2Cl2 (50 mL) were added NEt3 (3.11 g, 30.76 mmol) and MeSO2Cl (2.94 g, 25,63 mmol) at room temperature (r.t.) for 4 h. After the solvent was evaporated, the residue was dissolved in DMF (60 mL). NaN3 (2 g, 30.76 mmol) was added to this solution and the mixture was refluxed at 60–70 °C for 18 h. H2O (100 mL) and EtOAc (80 mL) were added to the cooled mixture. The organic layer was separated and washed with H2O (3 × 60 mL). After drying the organic layer over Na2SO4 and evaporation of the solvent, chromatography of the liquid residue on silica gel (SiO2, 50 g) with ethyl acetate/hexane (1:4) afforded oily azide 18 (2.38 g, 71%). 1H-NMR (400 MHz, CDCl3) δ 6.86–6.78 (m, 6H), 5.61 (s, 1H), 3.86 (s, 6H, 2OCH3), 3.83 (s, 6H, 2OCH3). 13C-NMR (100 MHz, CDCl3) δ 149.4 (2C), 149.1 (2C), 132.5 (2C), 120.1 (2CH), 111.2 (2CH), 110.7 (2CH), 68.4 (CHN3), 56.14 (2OCH3), 56.13 (2OCH3). IR (CH2Cl2, cm−1) 2997, 2936, 2836, 2098, 1600, 1592, 1515, 1464, 1418, 1252, 1234, 1158, 1026 and 855. Anal. Calcd for C17H19N3O4: C 62.00, H 5.81 and N 12.76; Found: C 61.96, H 5.80 and N 12.74.

4-[Azido(2,4-dimethoxyphenyl)methyl]-1,2-dimethoxybenzene (19)

The general procedure for the synthesis of 18 was applied to 15 (0.5 g, 1.64 mmol) to give an oily azide 19 (332.0 mg, 61%). 1H-NMR (400 MHz, CDCl3) δ 7.09 (d, 1H, J = 8.1 Hz), 6.88–6.81 (m, 3H), 6.48–6.45 (m, 2H), 6.05 (s, 1H), 3.85 (s, 3H, OCH3), 3.84 (s, 3H, OCH3), 3.80 (s, 3H, OCH3), 3.78 (s, 3H, OCH3). 13C-NMR (100 MHz, CDCl3) δ 160.9 (C), 158.0 (C), 149.1 (C), 148.6 (C), 132.5 (C), 129.2 (CH), 121.0 (C), 119.8 (CH), 111.1 (CH), 110.7 (CH), 104.6 (CH), 98.7 (CH), 62.1 (CHN3), 56.1 (2OCH3), 55.7 (OCH3), 55.6 (OCH3). IR (CH2Cl2, cm−1) 2998, 2936, 2837, 2098, 1681, 1611, 1588, 1515, 1464, 1419, 1256, 1209, 1182, 1158, 1140, 1115, 1030 and 926. Anal. Calcd for C17H19N3O4: C 62.00, H 5.81 and N 12.76; found: C 61.95, H 5.86 and N 12.82.

1-[Azido(3,4-dimethoxyphenyl)methyl]-4,5-dimethoxy-2-methylbenzene (20)

The general procedure for the synthesis of 18 was applied to 16 (3.25 g, 10.21 mmol). After crystallization from MeOH, azide 20 (2.48 g, 71%) was obtained as colorless block crystals. M.p. 102–104 °C. 1H-NMR (400 MHz, CDCl3) δ 69.92 (s, 1H), 6.82 (d, 1H, J = 8.6 Hz), 6.78–6.76 (m, 2H), 6.68 (s, 1H), 5.78 (s, 1H), 3.87 (s, 3H, OCH3), 3.86 (s, 3H, OCH3), 3.85 (s, 3H, OCH3), 3.83 (s, 3H, OCH3), 2.18 (s, 3H, CH3). 13C-NMR (100 MHz, CDCl3) δ 149.3 (C), 148.9 (C), 148.5 (C), 147.3 (C), 131.8 (C), 129.5 (C), 128.2 (C), 120.2 (CH), 114.0 (CH), 111.1 (CH), 111.0 (CH), 110.7 (CH), 65.6 (CHN3), 56.3 (OCH3), 56.1 (3OCH3), 19.2 (CH3). IR (CH2Cl2, cm−1) 3429, 3003, 2958, 2835, 2098, 1646, 1515, 1464, 1417, 1399, 1342, 1261, 1234, 1212, 1183, 1140, 1098, 1027, 1002 and 848. Anal. Calcd for C18H21N3O4: C 62.96, H 6.16 and N 12.24; Found: C 63.01, H 6.12 and N 12.26.

3,5′-(Azidomethylene)bis(1,2-dimethoxybenzene) (21)

Compound 21 was synthesized according to the general procedure given for 18. From 17 (1.22 g, 4.01 mmol), azide 21 (0.986 g, 75%) was obtained, after crystallized from CH2Cl2. Colorless block crystals. M.p. 78–80 °C. 1H-NMR (400 MHz, CDCl3) δ 7.08 (t, 1H, J = 8.1 Hz), 6.96 (d, 1H, J = 7.7 Hz), 6.89–6.80 (m, 4H), 6.10 (s, 1H), 3.86 (s, 3H, OCH3), 3.85 (s, 3H, OCH3), 3.84 (s, 3H, OCH3), 3.70 (s, 3H, OCH3). 13C-NMR (100 MHz, CDCl3) δ 152.8 (C), 149.2 (C), 148.8 (C), 146.6 (C), 133.9 (C), 132.5 (C), 124.4 (CH), 120.0 (CH), 119.7 (CH), 112.3 (CH), 111.1 (CH), 110.7 (CH), 62.6 (CHN3), 61.0 (OCH3), 56.12 (OCH3), 56.10 (OCH3), 56.0 (OCH3). IR (CH2Cl2, cm−1) 3423, 2829, 2098, 1645, 1516, 1480, 1415, 1271, 1234, 1140, 1072, 1027 and 1001. Anal. Calcd for C17H19N3O4: C 62.00, H 5.81 and N 12.76; Found: C 61.90, H 5.83 and N 12.77.

General procedure for synthesis amine hydrochloride salts 23–25

(2,4-Dimethoxyphenyl)(3,4-dimethoxyphenyl)methanamine hydrochloride (23)

Pd–C (100 mg) and 4-(azido(2,4-dimethoxyphenyl)methyl)-1,2-dimethoxybenzene (19) (0.42 g, 1.28 mmol) in MeOH (40 mL) and CHCl3 (6 mL) were placed in a 100 mL two-necked flask. A balloon filled with H2 gas (3 L) was fitted to the flask. The mixture was deoxygenated by flushing with H2 and then hydrogenated at r.t. for one day. The catalyst was removed by filtration. Recrystallization of the residue from MeOH–Et2O yielded (2,4-dimethoxyphenyl)(3,4-dimethoxyphenyl)methanamine hydrochloride (23) (0.378 g, 87%) as a white solid. M.p. 230–232 °C. 1H-NMR (400 MHz, D2O) δ 6.93 (d, 1H, J = 8.8 Hz), 6.88 (d, 1H, 8.1 Hz), 6.84–6.81 (m, 2H), 6.52 (s, 1H), 6.42 (qd, 1H, J = 8.4), 5.51 (s, 1H), 4.64 (bs, OH of D2O and NH3), 3.68 (s, 6H, 2OCH3), 3.66 (s, 3H, OCH3), 3.64 (s, 3H, OCH3). 13C-NMR (100 MHz, CDCl3) δ 161.2 (C), 157.9 (C), 148.4 (C), 129.8 (CH), 129.2 (2C), 120.1 (CH), 117.2 (C), 111.9 (CH), 110.6 (CH), 105.4 (CH), 99.1 (CH), 55.8 (CH–NH3Cl), 55.75 (OCH3), 55.6 (2OCH3), 54.4 (OCH3). Anal. Calcd for C17H22ClNO4: C 60.09, H 6.53 and N 4.12; Found: C 60.02, H 6.59 and N 4.13.

(4,5-Dimethoxy-2-methylphenyl) (3,4-dimethoxyphenyl) methanamine hydrochloride (24)

The general procedure was carried out with 1-(azido(3,4-dimethoxyphenyl)methyl)-4,5-dimethoxy-2-methylbenzene (20) (1.34 g, 3.90 mmol). Recrystallization of the residue from MeOH–Et2O afforded (4,5-dimethoxy-2-methylphenyl)(3,4-dimethoxyphenyl)methanamine hydrochloride (24) (1.28 g, 93%) as a white solid. M.p. 235–237 °C. 1H-NMR (400 MHz, CDCl3) δ 6.93 (s, 1H), 6.87–6.77 (m, 4H), 5.61 (s, 1H), 4.64 (bs, OH of D2O and NH3), 3.72 (s, 3H, OCH3), 3.71 (s, 3H, OCH3), 3.69 (s, 3H, OCH3), 3.66 (s, 3H, OCH3), 2.04 (s, 3H, CH3). Citation13C-NMR (100 MHz, CDCl3) δ 148.9 (C), 148.8 (C), 148.3 (C), 146.9 (C), 129.8 (C), 129.4 (C), 127.3 (C), 120.8 (CH), 115.0 (CH), 112.3 (CH), 111.4 (CH), 109.7 (CH), 56.4 (CH–NH3Cl), 56.1 (2OCH3), 56.0 (OCH3), 54.6 (OCH3), 18.0 (CH3). Anal. Calcd for C18H24ClNO4: C 61.10, H 6.84 and N 3.96; Found: C 61.07, H 6.86 and N 3.94.

(2,3-Dimethoxyphenyl)(3,4-dimethoxyphenyl)methanamine hydrochloride (25)

The general procedure was carried out with 3,5′-(azidomethylene)bis(1,2-dimethoxybenzene) (21) (2.14 g, 6.50 mmol). Recrystallization of the residue from MeOH–Et2O yielded (2,3-dimethoxyphenyl)(3,4-dimethoxyphenyl)methanamine hydrochloride (25) (2.02 g, 92%) as a white solid. M.p. 221–223 °C. 1H-NMR (400 MHz, D2O) δ 7.08–7.06 (m, 1H), 6.98–6.96 (m, 1H), 6.90–6.82 (m, 4H), 5.66 (s, 1H), 4.64 (bs, OH of D2O and NH3), 3.69 (s, 3H, OCH3), 3.68 (s, 3H, OCH3), 3.65 (s, 6H, 2OCH3). Citation13C-NMR (100 MHz, CDCl3) δ 152.7 (C), 148.7 (C), 148.5 (C), 145.6 (C), 130.4 (C), 129.4 (C), 125.4 (CH), 120.5 (CH), 118.7 (CH), 114.1 (CH), 112.0 (CH), 110.9 (CH), 60.8 (CH–NH3Cl), 55.9 (OCH3), 55.8 (2OCH3), 53.6 (OCH3). Anal. Calcd for C17H22ClNO4: C 60.09, H 6.53 and N 4.12; Found: C 60.15, H 6.57 and N 4.09.

General procedure for the synthesis of sulfonamides 26–28

N-(bis(3,4-dimethoxyphenyl)methyl)methanesulfonamide (26)

To a solution of bis(3,4-dimethoxyphenyl)methanamine hydrochloride (22) (0.197 g, 0.580 mmol) in MeOH (15 mL) was added 6 M NaOH (40 mL). The mixture was stirred at r.t. for 3 h. After most of the MeOH was evaporated, H2O (20 mL) and CH2Cl2 (50 mL) were added to the residue. Organic phase was separated and H2O phase was extracted with CH2Cl2 (3 × 30 mL). Combined organic layers were dried over Na2SO4 and the solvent was evaporated. After the residue was dissolved in CH2Cl2 (30 mL), NEt3 (70 mg, 0.696 mmol) and MeSO2Cl (86 mg, 0.754 mmol) were added to this solution at r.t. for 20 h. The solvent was evaporated and to the residue were added H2O (20 mL) and EtOAc (30 mL). The organic layer was separated and H2O layer was extracted with EtOAc (2 × 30 mL). The combined organic layers were dried over Na2SO4 and the solvent was evaporated. The chromatography of the residue on silica gel (SiO2, 60 g) with ethyl acetate/hexane (1:9) gave a yellowish solid N-(bis(3,4-dimethoxyphenyl)methyl)methanesulfonamide (26) (0.202 g, 91%). Solidified; m.p. 123–125 °C. 1H-NMR (400 MHz, CDCl3) δ 6.83–6.78 (m, 6H), 5.60 (d, 1H, CH, J = 8.1 Hz), 5.57 (d, 1H, NH, J = 8.1 Hz), 3.83 (s, 6H, 2OCH3), 3.80 (s, 6H, OCH3), 2.60 (s, 3H, CH3). Citation13C-NMR (100 MHz, CDCl3) δ 149.5 (2C), 148.9 (2C), 133.5 (2C), 119.8 (2CH), 111.2 (2CH), 110,7 (2CH), 61.0 (CH-NH), 56.2 (2OCH3), 56.1 (2OCH3), 42.0 (CH3). IR (CH2Cl2, cm−1) 3565, 3261, 2829, 2090, 1645, 1515, 1464, 1418, 1318, 1252, 1189, 1140, 1063, 1025, 1063, 1025 and 979. Anal. Calcd for C18H23NO6S: C 56.68, H 6.08, N 3.67 and S 8.41; Found: C 56.73, H 6.02, N 3.62 and S 8.40.

N-((4,5-dimethoxy-2-methylphenyl) (3,4-dimethoxyphenyl)methyl) methane sulfonamide (27)

The general procedure for 26 was applied to (4,5-dimethoxy-2-methylphenyl)(3,4-dimethoxyphenyl)methanamine hydrochloride (24) (0.250 g, 0.707 mmol) to give yellowish solid 27 (0.260 g, 93%). M.p. 104–106 °C. 1H-NMR (400 MHz, CDCl3) δ 6.84 (bs, 1H), 6.83 (s, 1H), 6.78 (d, 1H, J = 8.3 Hz), 6.75 (qd, 1H, J = 8.3 Hz), 6.68 (s, 1H), 5.83 (d, 1H, CH, J = 7.2 Hz), 5.24 (d, 1H, NH, J = 7.2), 3.86 (s, 3H, OCH3), 3.84 (s, 3H, OCH3), 3.81 (s, 6H, 2OCH3), 2.63 (s, 3H, CH3), 2.28 (s, 3H, CH3). Citation13C-NMR (100 MHz, CDCl3) δ 149.5 (C), 148.9 (C), 148.5 (C), 147.6 (C), 132.9 (C), 130.5 (C), 128.5 (C), 119.9 (CH), 114.2 (CH), 111.3 (CH), 111.0 (CH), 110.7 (CH), 57.7 (CH-NH), 56.4 (OCH3), 56.2 (OCH3), 56.11 (OCH3), 56.10 (OCH3), 42.2 (CH3), 19.1 (CH3). IR (CH2Cl2, cm−1) 3618, 3161, 2061, 1635, 1514, 1457, 1320, 1242, 1208, 1149, 1102, 1027 and 979. Anal. Calcd for C19H25NO6S: C 57.70, H 6.37, N 3.54 and S 8.11; Found: C 57.75 H 6.32, N 3.51 and S 8.09.

N-((2,3-dimethoxyphenyl)(3,4-dimethoxyphenyl)methyl)methanesulfonamide (28)

The general procedure described above was applied to 25 (0.300 g, 0.883 mmol) to yield an oily N-((2,3-dimethoxyphenyl)(3,4-dimethoxyphenyl)methyl)methanesulfonamide (28) (0.328 g, 97%). 1H-NMR (400 MHz, CDCl3) δ 7.08 (t, 1H, J = 7.9 Hz), 6.93–6.90 (m, 3H), 6.76 (bs, 2H), 5.77 (d, 1H, CH, J = 9.1 Hz), 5.71 (d, 1H, NH, J = 9.1 Hz), 3.85 (OCH3), 3.82 (OCH3), 3.51 (OCH3), 2.68 (CH3). Citation13C-NMR (100 MHz, CDCl3) δ 153.4 (C), 149.2 (C), 148.5 (C), 146.6 (C), 135.0 (C), 134.0 (C), 124.5 (CH), 120.8 (CH), 119.0 (CH), 112.8 (CH), 111.0 (CH), 110.4 (CH), 60.7 (CH-NH), 58.0 (OCH3), 56.2 (OCH3), 56.1 (OCH3), 56.0 (OCH3), 41.7 (CH3). IR (CH2Cl2, cm−1) 3713, 3279, 3003, 2938, 2837, 1589, 1516, 1481, 1463, 1323, 1272, 1235, 1147, 1086, 1046, 1027, 1003, 982. Anal. Calcd for C18H23NO6S: C 56.68, H 6.08, N 3.67 and S 8.41; Found: C 56.75, H 6.04, N 3.65 and S 8.42.

Purification of CA isoenzymes from human erythrocytes by affinity chromatography

CA isoenzymes were purified via a simple single-step method using sepharose-4B-L tyrosine-sulfanilamide affinity gel chromatography, described previouslyCitation31,Citation32. The serum was obtained from fresh human blood acquired from the Blood Centre of the Research Hospital at Atatürk University. The blood samples were centrifuged at 5000 rpm for 15 min and the precipitant was removed and the serum was isolated. The pH was adjusted to 8.7 with solid Tris. sepharose-4B-tirozyne-sulfanylamide affinity column equilibrated with 25 mM Tris–HCl/0.1 M Na2SO4 (pH 8.7). The affinity gel was washed with 25 mM Tris–HCl/22 mM Na2SO4 (pH 8.7). The hCA VI isozyme was eluted with 0.25 M H2NSO3H/25 mM Na2HPO4 (pH 6.7). All procedures were performed at 4 °CCitation33,Citation34.

Hydratase activity assay

The hydratase activities of hCA I and II were assayed by following the hydration of CO2 according to the method described by Wilbur and AndersonCitation35. The activity of CO2-hydratase in enzyme units (EU) was calculated by using the equation (to – tc/tc) where to and tc are the times for of pH change of the non-enzymatic and the enzymatic reactions, respectivelyCitation36.

Esterase activity assay

CA activity was assayed by following the change in absorbance at 348 nm of 4-nitrophenylacetate (NPA) to 4-nitrophenylate ion over a period of 3 min at 25°C using a spectrophotometer (Shimadzu, UVmini-1240 UV-VIS spectrophotometer) according to the method described by Verpoorte et al.Citation37 The enzymatic reaction contained 1.4 mL 0.05 M Tris–SO4 buffer (pH 7.4), 1 mL 3 mM 4-NPA, 0.5 mL H2O and 0.1 mL enzyme solution (total volume, 3.0 mL). A reference measurement was obtained by preparing the mixture without the enzyme solution. All measurements were recorded in triplicate. The Ki values were determined from a series of experiments using three different bromophenols concentrations and 4-NPA as the substrate at five different concentrations to construct Lineweaver–Burk curvesCitation38.

Protein determination

The yield of protein during the purification steps was determined spectrophotometrically at 595 nm according to the Bradford methodCitation39, using bovine serum albumin as the standardCitation40.

SDS polyacrylamide gel electrophoresis

The purity of the enzymes was confirmed using SDS polyacrylamide gel electrophoresis. The running and stacking gels contained 10% and 3% acrylamide, respectively, and 0.1% SDS, according to the Laemmli procedureCitation41 described previouslyCitation12. A 20 mg sample was applied to the electrophoresis medium. Gels were stained for 1.5 h in 0.1% Coomassie Brilliant Blue R-250 in 50% methanol and 10% acetic acid, then distained with several changes of the same solvent without the dye.

Results and discussion

Synthesis

Polyphosphoric acid (PPA) prepared from phosphoric acid and P2O5 catalyzed acylation or alkylation of aromatic compounds has been used in the literatureCitation42–45. Following this procedure, acylation of electron rich aromatic compounds 5–7 with carboxylic acid 8 gave diarylmethanone derivatives 10Citation46, 11Citation25 and 12Citation26. By a similar approach, the reaction of 5 with acid 9 afforded 13Citation46. Reduction of diarylmethanones with NaBH4 in MeOH at 0–25 °C by the procedure described by Lantano et al.Citation47 furnished diarylmethanols 14, 15Citation28, 16Citation26 and 17Citation29 in excellent yields. Alcohols can be converted to their azide derivatives by Mitsunobu reactionCitation48 or via the substitution of the corresponding mesylate esters with NaN3Citation49. Here, the later procedure was chosen. For this purpose, esterification of alcohols 14–17 with MeSO2Cl in CH2Cl2 in the presence of Et3N at 25 °C and then substitution of the corresponding mesylate esters with NaN3 in DMF at 60–70 °C afforded four novel diarylmethylazides 18–21 in good yields. The most critical step of our synthesis was the reduction of azides to amine hydrochloride salts. Due to the dibenzylic position of the azide groups, the Pd--C catalyzed hydrogenation could have possibly failed by leading to a deamination reaction. Fortunately, the reduction of azide groups to the corresponding amine derivatives was carried out without encountering any problems. The reduction of azides 18–21 with Pd-C catalyzed hydrogenation in the presence of CHCl3 (for in situ production of HCl)Citation49 in MeOH yielded amine hydrochlorides 22–25 from which only 22 has previously been reportedCitation30. Amine hydrochloride salts 22, 24 and 25 were converted to their amines with 6 M NaOH. Then the reaction of these amines with MeSO2Cl in the presence of Et3N gave new sulfonamide derivatives 26–28 in high yields (). The structures of all synthesized compounds were characterized by 1H-, 13C-NMR and azide functional groups were characterized by IR. Especially, in the Citation13C-NMR spectra, carbonyl carbons of diarylmethanones 10–13 resonating at 194.5–196.8 which displayed positive evidence for the formation of the ketone group. In this context, diarylcarbinol carbons of compounds 14–17 resonating at 71.8–76.0 ppm are in good agreement with the structure. On the other hand; benzylic carbons of the azides resonated at upper field (62.1–68.4 ppm) than the corresponding alcohols as expected. Similarly, characteristic IR bands of N3 groups at 2098 cm−1 were good evidence for the presence of azide functional groups. Benzylic carbons of amine hydrochlorides resonating at 55.8–60.8 ppm were in agreement with the structures.

Scheme 1. Reagents and conditions: (i) PPA, 80 °C, 1 h; (ii) NaBH4, THF/MeOH, 30 min 0 °C, then 4 h, r.t.; (iii) CH2Cl2, NEt3, MeSO2Cl, r.t., 4 h, then DMF, NaN3, 60–70 °C, 18 h; (iv) Pd–C, MeOH/CHCl3, H2, r.t., 1 d and (v) MeOH, 6 M NaOH r.t., 3 h then CH2Cl2, NEt3, MeSO2Cl r.t., 20 h.

Scheme 1. Reagents and conditions: (i) PPA, 80 °C, 1 h; (ii) NaBH4, THF/MeOH, 30 min 0 °C, then 4 h, r.t.; (iii) CH2Cl2, NEt3, MeSO2Cl, r.t., 4 h, then DMF, NaN3, 60–70 °C, 18 h; (iv) Pd–C, MeOH/CHCl3, H2, r.t., 1 d and (v) MeOH, 6 M NaOH r.t., 3 h then CH2Cl2, NEt3, MeSO2Cl r.t., 20 h.

CA purification and activity assay

In this study, CA isoenzymes I and II (hCA I and hCA II) were purified from human erythrocytes (). The purification of both CA isozymes was performed using a simple one-step method with a sepharose-4B-thyrosine-sulfanilamide affinity column chromatography. Human erythrocyte CA I isoenzyme was purified, 312.2-fold with a specific activity of 626 EU/mg and overall yield of 60.7%; CA II isoenzyme was purified, 296-fold with a specific activity of 594 EU/mg and an overall yield of 44.1%. We report here the first study on the inhibitory effects of novel benzylamine derivatives (22–28) on the esterase activity of hCA I and II. The inhibitor concentrations that caused 50% inhibition (IC50) were determined from activity versus (%)-[benzylamines] plots and the Ki values were calculated from the Lineweaver–Burk plots (). The data of show the following inhibition of hCA I and II with novel benzylamine derivatives (22–28), by an esterase assay, with 4-NPA as substrate:

Table 1. Summary of purification procedure for hCA I and hCA II isoenzymes by a sepharose-4B-thyrosine-sulfanilamide affinity column chromatography.

Table 2. hCA I and II isoenzymes inhibition data with some synthesized benzylamine derivatives (2228) by an esterase assay with 4-NPA as substrate.

CA isoenzymes inhibition effects

CA isoenzymes (hCA I and hCA II) inhibitory effects of the novel benzylamine derivatives (22–28) were tested under in vitro conditions and Ki values were calculated and given in .

The data in show the following regarding the inhibition of hCA I and II by novel benzylamine derivatives 22–28. The strongest inhibitory activity has been observed with compound 28, investigated here for the inhibition of the rapid cytosolic isozymes hCA I and II (). According to these activities, the novel benzylamine derivative of 28 showed the strongest hCA I inhibitory activity with Ki of 3.68 ± 1.28 µM and hCA II inhibitory activity with Ki of 9.23 ± 2.73 µM (). Also, the remaining other benzylamine derivatives were quite effective cytosolic isozyme hCA I inhibitors with Ki-s within the range of 3.68–66.89 µM and hCA II inhibitors with Ki-s in the range of 9.23–73.31 µM (). It is widely known that amine compounds are CA isoenzymeCitation50.

In our study, all of the novel benzylamine derivatives 22–28 had four methoxy moieties in both the aromatic rings. In addition, two of the novel benzylamine derivatives 24 and 27 have a methyl group in the aromatic ring. We suppose that these groups make aromatic rings electronically richer. Thus, these molecules easily connect to zinc ions in active sides of CA isoenzymes.

The synthesized novel benzylamine derivatives 22–28 have two aromatic rings. These new synthesized compounds have been investigated as CA izoenzymes inhibitors in this study. The rationale of investigating these compounds as CA inhibitors exists in the fact that the compounds with aromatic rings have been shown to be the only competitive inhibitor with CO2 as the substrate for the main isoform of CA, i.e. hCA I and II. All new synthesized compounds 22, 23, 26 and 28 were much stronger CA I inhibition effects than the clinically used AZA included in the assays as a standard inhibitor. On the other hand, CA II inhibition effects of all new synthesized compounds 22–28 are weaker than that of AZA ().

Conclusion

In summary, diarylmethylamine hydrochloride salts 22–25 were synthesized starting from methoxy-substituted benzenes in five steps with high yields. Compounds 22, 24 and 25 were converted to their sulfonamide derivatives 26–28. All synthesized compounds can be important synthons for further synthetic and biological purposes. In addition, CA activities of the synthesized benzylamines 22–25 and their sulfonamide derivatives 26–28 were also investigated. All of the new synthesized compounds were found as effective CA I and CA II isoenzymes inhibitor. Further, these findings point out that these compounds 22–28 may potentially be used as leads for generating a useful CA isoenzymes inhibitor.

Declaration of interest

The authors report no conflicts of interest. The authors alone are responsible for the content and writing of this article.

We thank Atatürk University for the financial supports of this work (BAP, Project no: 2009/84 for YÇ and BAP, Project no: 2009/251 for IG) and the Scientific and Technological Research Council of Turkey (TUBITAK, Project no: 109T241 for SG) and Prof Dr Hasan Seçen for his contribution.

References

  • Supuran CT, Scozzafava A. Carbonic anhydrase inhibitors and their therapeutic potential. Expert Opin Ther Pat 2000;10:575–601
  • Alterio V, Di Fiore A, D’Ambrosio K, et al. Multiple binding modes of inhibitors to carbonic anhydrases: how to design specific drugs targeting 15 different isoforms. Chem Rev 2012;112:4421–68
  • Supuran CT. Carbonic anhydrases: novel therapeutic applications for inhibitors and activators. Nat Rev Drug Discov 2008;7:168–81
  • Alterio V, De Simone G, Monti SM, et al. Carbonic anhydrase inhibitors: inhibition of human, bacterial, and archaeal isozymes with benzene-1,3-disulfonamides – solution and crystallographic studies. Bioorg Med Chem Lett 2007;17:4201–7
  • Kaur IP, Smitha R, Aggarwal D, Kapil M. Acetazolamide: future perspective in topical glaucoma therapeutics. Int J Pharm 2002;248:1–14
  • Celebisoy N, Gökçay F, Sirin H, Akyürekli O. Treatment of idiopathic intracranial hypertension: topiramate vs acetazolamide, an open-label study. Acta Neurol Scand 2007;116:322–7
  • Supuran CT. Carbonic anhydrases-an overview. Curr Pharm Des 2008;14:603–14
  • Supuran CT. Diuretics: from classical carbonic anhydrase inhibitors to novel applications of the sulphonamides. Curr Pharm Des 2008;14:641–8
  • Sjoüblom B, Polentarutti M, Djinovic-Carugo K. Structural study of X-ray induced activation of carbonic anhydrase. Proc Natl Acad Sci USA 2009;106:10609–13
  • Şentürk M, Gülçin İ, Daştan A, et al. Carbonic anhydrase inhibitors. Inhibition of human erythrocyte isozymes I and II with a series of antioxidant phenols. Bioorg Med Chem 2009;17:3207–11
  • Clare BW, Supuran CT. A perspective on quantitative structure-activity relationships and carbonic anhydrase inhibitors. Expert Opin Drug Metab Toxicol 2006;2:113–37
  • Çoban TA, Beydemir Ş, Gülçin İ, Ekinci D. Morphine inhibits erythrocyte carbonic anhydrase in vitro and in vivo. Biol Pharm Bull 2007;30:2257–61
  • Innocenti A, Hilvo M, Scozzafava A, et al. Carbonic anhydrase inhibitors: inhibition of the new membrane-associated isoform XV with phenols. Bioorg Med Chem Lett 2008;18:3593–6
  • Lindskog S. Structure and mechanism of carbonic anhydrase. Pharmacol Ther 1997;74:1–20
  • Öztürk Sarıkaya SB, Topal F, Şentürk M, et al. In vitro inhibition of α-carbonic anhydrase isozymes by some phenolic compounds. Bioorg Med Chem Lett 2011;21:4259–62
  • Supuran CT, Scozzafava A, Conway J. Carbonic anhydrase: its inhibitors and activators. Boca Raton (FL): CRC Press; 2004:243–54
  • Supuran CT. Carbonic anhydrases as drug targets-an overview. Curr Top Med Chem 2007;7:825–33
  • Supuran CT, Scozzafava A. Carbonic anhydrases as targets for medicinal chemistry. Bioorg Med Chem 2007;15:4336–50
  • Innocenti A, Gülçin İ, Scozzafava A, Supuran CT. Carbonic anhydrase inhibitors. Antioxidant polyphenol natural products effectively inhibit mammalian isoforms I-XV. Bioorg Med Chem Lett 2010;20:5050–3
  • Alterio V, Di Fiore A, D’Ambrosio K, et al. X-ray crystallography of CA inhibitors and its importance in drug design. In: Supuran CT, Winum JY, eds. In drug design of zinc-enzyme inhibitors: functional, structural, and disease applications. Hoboken (NJ): Wiley; 2009:73–138
  • Duda DM, Tu C, Fisher SZ, et al. Human carbonic anhydrase III: structural and kinetic study of catalysis and proton transfer. Biochemistry 2005;44:10046–53
  • Di Fiore A, Monti M, Hilvo MS, et al. Crystal structure of human carbonic anhydrase XIII and its complex with the inhibitor acetazolamide. Proteins 2008;74:164–75
  • Alterio V, Hilvo M, Di Fiore A, et al. Crystal structure of the catalytic domain of the tumor-associated human carbonic anhydrase IX. Proc Natl Acad Sci USA 2009;106:16233–8
  • Harig M, Neumann B, Stammler HG, Kuck D. 2,3,6,7,10,11-Hexamethoxytribenzo-triquinacene: synthesis, solid-state structure and functionalization of a rigid analogue of cyclotriveratrylene. Eur J Org Chem 2004;11:2381–97
  • Mitter PC. Some derivatives of 4-phenylchroman. J Ind Chem Soc 1931;8:271–6
  • Garofano T, Oliverio A. The influence of o-substituents on the reactivity of aryl ketones. I. Influence of o-substituents on the reactivity of benzophenone. Annal di Chim 1957;47:260–84
  • Gazave JM, Rancurel A, Grenier G. Pharmaceutical composition containing at least one diphenyl-substituted compound. Ger Offen Patent 1975, DE 2501443. Chem Abstr 1975;83:188522
  • Konig B, Kostanecki Sv. Leuco-compounds of hydroxyketones. Ber Deutsch Chem Gesellschaft 1907;39:4027–31
  • Fukazawa N, Odate M, Suzuki T, et al. Preparation of heterocyclic compounds as anticancer drug potentiators. Eur Patent Appl 1990, Chem Abstr 1990;113:152275
  • Valette. Mono- and polyalkyl- or halogen-substituted derivatives of benzohydrylamine and symmetrical diphenylethylamine. Bull Soc Chim Fr 1930;47:289–300
  • Öztürk Sarıkaya SB, Gülçin İ, Supuran CT. Carbonic anhydrase inhibitors. Inhibition of human erythrocyte isozymes I and II with a series of phenolic acids. Chem Biol Drug Design 2010;75:515–20
  • Şentürk M, Gülçin İ, Beydemir Ş, et al. In vitro inhibition of human carbonic anhydrase I and II isozymes with natural phenolic compounds. Chem Biol Drug Design 2011;77:494–9
  • Gülçin İ, Beydemir Ş, Büyükokuroğlu ME. In vitro and in vivo effects of dantrolene on carbonic anhydrase enzyme activities. Biol Pharm Bull 2004;27:613–16
  • Beydemir Ş, Gülçin İ. Effect of melatonin on carbonic anhydrase from human erythrocyte in vitro and from rat erythrocyte in vivo. J Enzyme Inhib Med Chem 2004;19:193–7
  • Wilbur KM, Anderson NG. Electrometric and colorimetric determination of carbonic anhydrase. J Biol Chem 1976;176:147–54
  • Aras Hisar Ş, Hisar O, Beydemir Ş, et al. Effect of vitamin E on carbonic anhydrase enzyme activity in rainbow trout (Oncorhynchus mykiss) erythrocytes in vitro and in vivo. Acta Vet Hung 2004;52:413–22
  • Verpoorte JA, Metha S, Edsall JT. Esterase activities of human carbonic anhydrase B and C. J Biol Chem 1967;242:4221–9
  • Lineweaver H, Burk D. The determination of enzyme dissociation constants. J Am Chem Soc 1934;57:685–93
  • Bradford M. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 1976;72:248–54
  • Gülçin İ, Küfrevioğlu Öİ, Oktay M. Purification and characterization of polyphenol oxidase from nettle (Urtica dioica L.) and inhibition effects of some chemicals on the enzyme activity. J Enzyme Inhib Med Chem 2005;20:297–302
  • Laemmli DK. Cleavage of structural proteins during the assembly of the head of bacteriophage T4. Nature 1970;227:680–5
  • Çetinkaya Y, Menzek A, Sahin E, Balaydın HT. Selective O-demethylation during bromination of (3,4-dimethoxyphenyl)(2,3,4-trimethoxyphenyl)methanone. Tetrahedron 2011;67:3483–9
  • Çetinkaya Y, Göçer H, Menzek A, Gülçin İ. Synthesis and antioxidant properties of (3,4-dihydroxyphenyl)(2,3,4-trihydroxyphenyl)methanone and its derivatives. Arch Pharm Chem Life Sci 2011;345:323–34
  • Nar M, Çetinkaya Y, Gülçin İ, Menzek A. (3,4-Dihydroxyphenyl)(2,3,4-trihydroxyphenyl)methanone and its derivatives as carbonic anhydrase isoenzymes inhibitors. J Enzyme Inhib Med Chem 2012. [Epub ahead of print]. doi: 10.3109/14756366.2012.670807
  • Balaydın HT, Akbaba Y, Menzek A, et al. First and short syntheses of biologically active, naturally occurring brominated mono- and dibenzyl phenols. Arkivoc 2009;14:75–87
  • Hosangadi BD, Kasbekar AB, Nabar MJ, Desai RC. Novel reactions with polyphosphoric acid II. Decarboxylative acetylation, trans-carbonylation, and other reactions of substituted aromatic carboxylic acids. Ind J Chem 1973;11:711–13
  • Lantano B, Aguirre JM, Ugliarolo EA, et al. Scope of the formal [3 + 2] cycloaddition for the synthesis of five-membered ring of functionalized indanes. Tetrahedron 2012;68:913–21
  • Göksu S, Seçen H, Sütbeyaz Y. One-pot and stereospecific synthesis of cis-1,2-diazides via Mitsunobu reaction of epoxides. Synthesis-Stuttgart 2002;16:2373–8
  • Göksu S, Seçen H. Concise syntheses of 2-aminoindans via indan-2-ol. Tetrahedron 2005;61:6801–7
  • Pastorekova S, Vullo D, Nishimori I, et al. Carbonic anhydrase activators: activation of the human tumor-associated isozymes IX and XII with amino acids and amines. Bioorg Med Chem 2008;16:3530–6

Reprints and Corporate Permissions

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

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

Academic Permissions

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

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

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