1,186
Views
34
CrossRef citations to date
0
Altmetric
Research Paper

Inhibition of the α-carbonic anhydrase from Vibrio cholerae with amides and sulfonamides incorporating imidazole moieties

ORCID Icon, , ORCID Icon, , ORCID Icon, ORCID Icon, , , , , ORCID Icon & show all
Pages 798-804 | Received 30 Mar 2017, Accepted 28 Apr 2017, Published online: 01 Jun 2017

Abstract

We discovered novel and selective sulfonamides/amides acting as inhibitors of the α-carbonic anhydrase (CA, EC 4.2.1.1) from the pathogenic bacterium Vibrio cholerae (VchCA). This Gram-negative bacterium is the causative agent of cholera and colonises the upper small intestine where sodium bicarbonate is present at a high concentration. The secondary sulfonamides and amides investigated here were potent, low nanomolar VchCA inhibitors whereas their inhibition of the human cytosolic isoforms CA I and II was in the micromolar range or higher. The molecules represent an interesting lead for antibacterial agents with a possibly new mechanism of action, although their CA inhibition mechanism is unknown for the moment.

Introduction

Vibrio cholerae is a Gram-negative bacterium responsible of cholera, a severe and watery form of diarrhoea. Cholera is contracted by ingestion of food or water contaminated with the pathogen, and particularly with the V. cholerae serogroups (O1 and O139) associated with the expression of the cholera toxin. Following a short incubation period, symptoms begin and severe fluid loss can lead to severe dehydration, electrolyte imbalance and, ultimately, deathCitation1. Precise estimates of the global burden of cholera remain a challenge because the majority of cases are not declared due to limitations, in some countries, in health surveillance management systems and fear of negative impact on trade and tourism. The number of reported cholera cases remains high over the last decade; WHO gave, for 2015, an account of 172,454 cases and 1304 deaths notified by 42 countriesCitation2, though the global estimates range between 1.3 and 4 million cases and between 21,000 and 143,000 deaths per yearCitation3. The treatment of cholera infections, is mainly focused on the re-hydration by using saline–glucose solutions that can be combined, in case of severe dehydration, with antibiotics in order to stabilise highly dehydrated patients and reduce the duration of illnessCitation4. Tetracycline and quinolones have been widely used, but numerous multidrug-resistant strains of V. cholerae have been isolated from both clinical and environmental settings, and as a consequence, the use of antibiotics had to be restrictedCitation1 and alternative target need to be identified in order to develop more effective and safe drugs for cholera treatment.

V. cholerae survives and multiplies in the upper small intestine where sodium bicarbonate, described as a potential inducer of virulence gene expression, is present at a high concentrationCitation5. Moreover, V. cholerae can increase cytosolic bicarbonate levels by means of the carbonic anhydrase (CA), a metalloenzyme that catalyzes the hydration of CO2 to produce HCO3Citation6. The first class of CA from the bacterial pathogen V. cholerae was described by our group; it is an α-CA, denominated VchCA thatCitation7, similar to the other α-CA, has three His ligands, which coordinate the Zn(II) ion crucial for catalysis. An active site residue transfers a proton from the water coordinated to the Zn(II) ion to the environment, forming zinc hydroxide that represents the nucleophilic species of the enzymeCitation7. More in detail, the zinc hydroxide attacks the CO2, bound in a hydrophobic pocket near the metal ion, forming a labile intermediate where the bicarbonate is coordinated to the Zn(II) that readily reacts with an incoming water molecule, releasing the bicarbonate into solutionCitation8. On the basis of the role played by the bicarbonate ion as a virulence factor for V. cholerae, the VchCA has been suggested as a potential target for antibiotic development.

Experimental

Chemistry

Material and methods

All reagents, solvents, and deuterated were purchased from Sigma-Aldrich (Milan, Italy). Melting points were determined on Tottoli (Buchi) or Kofler apparatus and are uncorrected. Infrared spectra were recorded on a Spectrum One ATR Perkin Elmer FT-IR spectrometer, vibrational frequencies are given in ν, wave number (cm−1). Nuclear magnetic resonance (1H-NMR, 13C-NMR) spectra were acquired on AVANCE400 or AVANCE200 Bruker spectrometer, in DMSO, CD3OD or CDCl3 at 27 °C. Chemical shifts are reported in parts per million δ (ppm) relatively to TMS as internal reference, and the coupling constants (J) are expressed in Hertz (Hz). Splitting patterns are designated as follows: s, singlet; bs, broad singlet; d, doublet; t, triplet; q, quadruplet; and dd, double doublet. Mass spectra were recorded on a ThermoFinnigan LCQ Classic LC/MS/MS ion trap equipped with an ESI source and a syringe pump. Samples (10−4–10−5 M in MeOH/H2O 90:10) were infused in the electrospray system at a flow rate of 5–10 μl min−1. Elemental analyses for C, H, and N were obtained by a PE 2400 (Perkin-Elmer) analyzer, and the analytical results were within ±0.4% of the theoretical values for all compounds.

Synthesis of sulfonamides 1–4:

N-(3-(1H-imidazol-1-yl)propyl)methanesulfonamide 1

To a solution of 3-(1H-imidazol-1-yl)propan-1-amine (2.4 mmol) and TEA (3.6 mmol) in 3 ml of CH2Cl2, methanesulfonyl chloride (3.6 mmol) was added, and the mixture was kept overnight under stirring at room temperature. The solvent was evaporated under reduced pressure and the residue treated with a saturated solution of Na2CO3 (20 ml) and extracted in continuous for 24 h with CHCl3. After evaporation of the organic phase, 1 was purified by column chromatography on silica gel using CH2Cl2:MeOH (1:1) as mobile phase and obtained as a yellowish solid. M.p.: 89–91 °C (Tottoli-Buchi); 45% yield; 1H-NMR: δH (400 MHz, CDCl3) 7.52 (s, 1H), 7.09 (s, 1H), 6.97 (s,1H), 5.65 (bs, 1H), 4.12 (t, J = 6.5 Hz, 2H), 3.12–3.09 (m, 2H), 2.96 (s, 3H), 2.08 (m= 5, J = 6.5 Hz, 2H); 13C-NMR: δC (100 MHz, CDCl3): 137.4, 129.5, 118.8, 43.7, 39.97, 40.0, 31.5. MS-ESI+: m/z 203.93 [M + H]+.

N-(3-(1H-imidazol-1-yl)propyl)-4-fluorobenzenesulfonamide 2

To a solution of 3-(1H-imidazol-1-yl)propan-1-amine (0.8 mmol) and TEA (1.2 mmol) in 3 ml of CH2Cl2, 4-fluorobenzene-1-sulfonyl chloride (1.2 mmol) was added, and the mixture was kept overnight under stirring at room temperature. The reaction mixture was diluted with 5 ml of CH2Cl2 and washed with a saturated solution of NaHCO3 (5 ml). The aqueous phase was extracted with CH2Cl2 (2 × 5 ml), then the reunited organic layers were dried over Na2SO4, filtered and evaporated to give a residue that was purified by column chromatography on silica gel using as mobile phases firstly CH3CN and subsequently AcOEt:EtOH (1:1), 2 was obtained as a white solid. M.p.: 121–123 °C (Tottoli-Buchi); 40% yield; 1H-NMR: δH (400 MHz, DMSO-d6) 7.86–7.82 (m, 2H), 7.77 (bs, 1H), 7.55 (s, 1H), 7.47–7.43 (m, 2H), 7.09 (s, 1H), 6.86 (s, 1H), 3.96 (t, J = 7.0 Hz, 2H), 2.68 (t, J = 7.0 Hz, 2H), 2.51 (m = 5, J= 7.0 Hz, 2H); 13C-NMR: δC (100 MHz, DMSO-d6) 165.1 (J = 240.0 Hz), 137.2, 135.8 (J = 4.0 Hz), 129.8 (J = 9.0 Hz), 126.3, 119.5, 116.3 (22 Hz), 44.7, 39.5, 30.6; MS-ESI+: m/z 283.87 [M + H]+.

N-(3-(1H-imidazol-1-yl)propyl)-4-(trifluoromethyl)benzenesulfonamide 3

To a solution of 3-(1H-imidazol-1-yl)propan-1-amine (1.0 mmol) and TEA (1.5 mmol) in 5 ml of CH2Cl2, 4-(trifluoromethyl)benzene-1-sulfonyl chloride (1.5 mmol) was added, and the mixture was kept overnight under stirring at room temperature. The reaction mixture was washed with a saturated solution of Na2CO3 (3 × 5 ml). The organic layer, dried over Na2SO4, was evaporated and the residue was purified by column chromatography on silica gel using AcOEt:MeOH (8:2). The fractions with Rf = 0.21–0.40 were collected and purified using alumina and CH2Cl2:MeOH (9:1) to give a white residue that was left to solidify. M.p.: 127–128 °C (Kofler); 10% yield; 1H-NMR: δH (200 MHz, DMSO-d6) 8.06–7.98 (m, 5H), 7.67 (s, 1H), 7.14 (s, 1H), 6.92 (s, 1H), 3.96 (t, J = 6.4 Hz, 2H), 2.72 (t, J = 6.4 Hz, 2H), 2.08 (m = 5, J = 6.8 Hz, 2H); 13C-NMR: δC (100 MHz, DMSO-d6) 144.6, 137.6, 132.3 (J = 32.0 Hz), 128.3, 127.9, 127.0 (J = 3.7 Hz), 124.0 (J = 271.0 Hz), 119.9, 43.7, 40.07, 31.1; MS-ESI+: m/z 334.13 [M + H]+.

N-(3-(1H-imidazol-1-yl)propyl)-4-nitrobenzenesulfonamide 4

To a solution of 3-(1H-imidazol-1-yl)propan-1-amine (1.0 mmol) and TEA (1.5 mmol) in 5 ml of CH2Cl2, 4-nitrobenzene-1-sulfonyl chloride (1.5 mmol) was added, and the mixture was kept overnight under stirring at room temperature. The reaction mixture was washed with a saturated solution of Na2CO3 (3 × 5 ml). The organic layer, dried over Na2SO4, was evaporated and purified by column chromatography on silica gel using AcOEt:MeOH (8:2) as eluent, to give a white solid which was crystallised from AcOEt. M.p.: 140–141 °C (Kofler); 16% yield; 1H-NMR: δH (200 MHz, CD3OD) 8.41 (d, J = 9.1 Hz, 2H), 8.05 (d, J = 9.1 Hz, 2H), 7.62 (s, 1H), 7.10 (s, 1H), 6.95 (s, 1H), 4.10 (t, J = 6.8 Hz, 2H), 2.87 (t, J = 6.8 Hz, 2H),1.95 (m = 5, J = 6.8 Hz, 2H); 13C-NMR: δC (100 MHz, CD3OD) 150.1, 146.0, 137.1, 127.9, 127.8, 124.0, 119.2, 43.4, 39.3, 30.7; MS-ESI+: m/z 311.13 [M + H]+.

Synthesis of amides 5–9:

N-(3-(1H-imidazol-1-yl)propyl)acetamide 5

To a solution of 3-(1H-imidazol-1-yl)propan-1-amine (1.0 mmol) and TEA (1.5 mmol) in 5 ml of CH2Cl2, acetyl chloride (1.5 mmol) in 5 ml of CH2Cl2 was added dropwise. After 12 h at RT, the reaction mixture was evaporated under vacuum, the obtained residue was treated with a saturated solution of Na2CO3, and the aqueous solution was extracted in continuous with CHCl3 for 24 h. After solvent evaporation, the crude residue was purified by column chromatography on silica gel using CH2Cl2:MeOH (9:1) to give a colourless oil. IR ν: 1640 (C = O) cm−1; 65% yield; 1H-NMR: δH (400 MHz, CD3OD) 7.69 (s, 1H), 7.17 (s, 1H), 6.98 (s, 1H), 4.08 (t, J = 7.0 Hz, 2H), 3.18 (t, J = 7.0 Hz, 2H), 2.03–1.95 (m, 5H); 13C-NMR: δC (100 MHz, CDCl3) 170.8, 137.1, 128.9, 119.0, 44.8, 36.6, 31.0, 23.1; MS-ESI+: m/z 167.87 [M + H]+.

N-(3-(1H-imidazol-1-yl)propyl)-4-fluorobenzamide 6

To a solution of 3-(1H-imidazol-1-yl)propan-1-amine (1.0 mmol) and TEA (1.5 mmol) in 5 ml of CH2Cl2, 4-fluorobenzoyl chloride (1.5 mmol) was added. The reaction mixture was stirred for 12 h at RT and then washed with a saturated aqueous Na2CO3 (3 × 5 ml). The organic layer, dried over Na2SO4, was evaporated, and the obtained residue was purified by column chromatography on silica gel using CH2Cl2:MeOH (8:2) to give an oil which was solidified by treatment with petroleum ether M.p.: 70–72 °C (Tottoli-buchi); 60% yield. IR ν: 1650 (C = O) cm−1; 1H-NMR: δH (400 MHz, CD3OD) 7.90–7.86 (m, 2H), 7.72 (s, 1H), 7.23–7.18 (m, 3H), 6.99 (s, 1H), 4.14 (t, J = 7.0 Hz, 2H), 3.41 (t, J = 7.0 Hz, 2H), 2.12 (m = 5, J = 7.0 Hz, 2H); 13C-NMR: δC (100 MHz, CDCl3) 167.0, 164.7 (J = 250.0 Hz), 130.1, 130.4, 129.6 (J = 9.1 Hz), 128.6, 119.2, 115.4 (J = 21.0 Hz), 44.9, 37.1, 30.9; MS-ESI+: m/z 247.60 [M + H]+.

N-(3-(1H-imidazol-1-yl)propyl)-4-(trifluoromethyl)benzamide 7

To a solution of 3-(1H-imidazol-1-yl)propan-1-amine (0.8 mmol) and TEA (1.2 mmol) in 3 ml of CH2Cl2, 4-(trifluoromethyl)benzoyl chloride (1.2 mmol) was added, and the mixture was kept overnight under stirring at room temperature. The reaction mixture was diluted with 5 ml of CH2Cl2 and washed with a saturated aqueous NaHCO3 (5 ml). The aqueous phase was extracted with CH2Cl2 (2 × 5 ml), the combined organic layers were dried over Na2SO4, filtered and evaporated to give a residue that was purified by column chromatography on silica gel using CH2Cl2:MeOH (8:2). Compound 7 was obtained as a white solid. M.p.: 106–107 °C (Tottoli-Buchi); 80% yield; IR ν: 1657 (C = O) cm−1; 1H-NMR: δH (400 MHz, DMSO-d6) 8.75 (bs, 1H), 8.04 (d, J = 7.9 Hz, 2H), 7.86 (d, J = 7.9 Hz, 2H), 7.66 (s, 1H), 7.21 (s, 1H), 6.90 (s, 1H), 4.05–4.02 (m, 2H), 3.29–3.23 (m, 2H), 2.01–1.95 (m, 2H); 13C-NMR: δC (100 MHz, CD3OD) 167.6, 137.9, 137.1, 132.7 (q, J = 60.1 Hz), 127.8, 127.7, 125.1 (q, J = 10.2 Hz), 122.6, 119.2, 44.3, 36.9, 30.5; MS-ESI+: m/z 297.80 [M + H]+.

N-(3-(1H-imidazol-1-yl)propyl)-4-nitrobenzamide 8

To a solution of 3-(1H-imidazol-1-yl)propan-1-amine (0.8 mmol) and TEA (1.2 mmol) in 3 ml of dry CH2Cl2, 4-nitrobenzoyl chloride (1.2 mmol) was added, and the mixture was kept overnight under stirring at room temperature. The reaction mixture was then diluted with 5 ml of CH2Cl2 and washed with a saturated aqueous Na2CO3 (5 ml). The aqueous phase was extracted with CH2Cl2 (2 × 5 ml), and then the combined organic layers were dried over Na2SO4, filtered and evaporated under reduced pressure. The crude residue was purified by chromatography on silica gel using CH2Cl2:MeOH (9:1). The obtained yellowish solid was crystallised from CH3CN. M.p.: 152–154 °C (Tottoli-Buchi); 41% yield; IR ν: 1658 (C = O) cm−1; 1H-NMR: δH (400 MHz, DMSO-d6) 8.33 (d, J = 8.9 Hz, 2H), 8.02 (d, J = 8.9 Hz, 2H), 7.73 (s, 1H), 7.21 (s, 1H), 6.99 (s,1H), 4.16 (t, J = 6.9 Hz, 2H), 3.34 (t, J = 6.9 Hz, 2H), 2015 (m = 5, J = 6.9 Hz, 2H,); 13C-NMR: δC (100 MHz, CD3OD) 166.9, 149.6, 139.9, 137.2, 128.3, 127.8, 123.2, 119.2, 44.3, 37.0, 30.4; MS-ESI+: m/z 274.97 [M + H]+.

N-(3-(1H-imidazol-1-yl)propyl)-[1,1′-biphenyl]-4-carboxamide 9

To a solution of 3-(1H-imidazol-1-yl)propan-1-amine (1.0 mmol) and TEA (1.5 mmol) in 5 ml of dry CH2Cl2, biphenyl-4-carbonyl chloride (1.5 mmol) was added. The reaction mixture was kept under stirring at room temperature for 48 h, and then was washed with a saturated aqueous of Na2CO3 (3 × 5 ml). The organic layer, dried over Na2SO4, filtered and evaporated under reduced pressure. The crude residue was purified by column chromatography on silica gel using CH2Cl2:MeOH (9:1) to give a solid that was twice crystallised from AcOEt. M.p.: 135–136 °C; 26% yield. IR ν: 1639 (C = O) cm−1; 1H-NMR: δH (400 MHz, DMSO-d6) 8.57 (bs, 1H), 7.94 (d, J = 8.4 Hz, 2H), 7.78–7.72 (m, 4H), 7.67 (s, 1H), 7.51 (t, J = 7.2 Hz, 2H) 7.41 (t, J = 7.6 Hz, 1H), 7.22 (s, 1H), 6.90 (s, 1H), 4.03 (t, J = 6.8 Hz, 2H), 3.26 (q, J = 6.0 Hz, 2H), 1.98 (m =5, J = 6.8 Hz, 2H); 13C-NMR: δC (100 MHz, CD3OD) 168.7, 144.3, 139.8, 137.1, 132.8, 128.6, 127.8, 127.7, 127.5, 126.7, 126.6, 119.3, 44.4, 36.8, 30.7; MS-ESI+: m/z 306.07 [M + H]+.

Synthesis of 4-Nitro-N-(3-phenylpropyl)benzamide 10:

To a solution of 3-phenylpropan-1-amine (1.0 mmol) and TEA (1.5 mmol) in 5 ml of dry CH2Cl2, 4-nitrobenzoyl chloride (1.5 mmol) was added, and the mixture was kept overnight under stirring at room temperature. The reaction mixture was washed with a saturated aqueous of Na2CO3 (3 × 5 ml). The organic layer, was dried over Na2SO4, filtered and evaporated to give a white solid that was washed with petroleum ether and crystallised from toluene. M.p.: 95–97 °C (Tottoli-Buchi); 87% yield. IR ν: 1636 (C = O) cm−1; 1H-NMR δH (400 MHz, CD3OD) 8.33 (d, J = 7.9 Hz, 2H), 8.01 (d, J = 7.9 Hz, 2H), 7.30–7.15 (m, 5H), 3.45 (t, J = 6.9 Hz, 2H), 2.73 (t, J = 6.9 Hz, 2H), 1.98 (m = 5, J = 6.8 Hz, 2H); 13C-NMR: δC (100 MHz, CD3OD) 165.29, 149.51, 141.35, 140.16, 128.73, 128.41, 128.00, 126.26, 123.73, 40.31, 33.71, 30.80; MS-ESI+: m/z 283.07 [M + H]+.

Carbonic anhydrase assay

A stopped-flow methodCitation9 was used for assaying the CA catalysed CO2 hydration activity with phenol red as indicator, working at the absorbance maximum of 557 nm, following the initial rates of the CA-catalyzed CO2 hydration reaction for 10–100 s. For each inhibitor, at least six traces of the initial 5–10% of the reaction have been used for determining the initial velocity. The uncatalyzed rates were determined in the same manner and subtracted from the total observed rates. Stock solutions of inhibitor (0.01 mM) were prepared in distilled–deionised water with 5% DMSO and dilutions up to 0.1 nM were done thereafter with the assay buffer. The Inhibition constant (KI) was obtained by considering the classical Michaelis–Menten equation, which has been fitted by non-linear least squares by using PRISM 3. All CA isozymes used in the experiments were purified recombinant proteins obtained as reported earlier by our groupCitation10–18.

In vitro antibacterial assay

In order to evaluate the antibacterial activity of the synthesised compounds, an in vitro assay was performed using two V. cholerae O1 strains, the reference strain ATCC14103 (American Type Culture Collection; Manassas, VA) and ISS-Vc014, a clinical isolate from 1992 cholera outbreak in Luanda, Angola (courtesy of prof. Mauro Maria Colombo), previously characterised for a multiresistance profile (ampicillin, chloramphenicol, penicillin, streptomycin, spectinomycin, kanamycin, trimethoprim–sulfamethoxazole, tetracycline, and erythromycin)Citation19,Citation20. A procedure based on the method described in AndrewsCitation21 for broth dilution MICs was used for the tests: the studied compounds were dissolved in dimethyl sulfoxide (DMSO) and added to the liquid culture media (tryptic soy broth, Oxoid, Basingstoke, UK) in order to obtain final concentrations ranging from 0.1 μg ml−1 to 1 mg ml−1. The tests were performed in a microwell format (100 μl), and each strain was seeded at a concentration of 105 c.f.u. ml−1; the inoculated plates were incubated without shaking at 37 °C for 18–20 h. Ampicillin (Sigma–Aldrich; Saint Louis, MO), tested in the same dilutions of the compounds, was used in the experiments as a reference drug, and a reference strain for MIC tests (E. coli ATCC 25922) was included to validate the results. A minimum of two wells containing uninoculated medium, medium inoculated with the dilutions of the compounds only, and medium inoculated with the bacterial strains and DMSO in amounts equivalent to those used to dissolve the compounds were also included in each test to act respectively as sterility and growth controls. Compounds showing at least partial inhibition of the bacterial growth at 1 mg ml−1 were selected to be tested at higher concentrations, and tests were performed with the above described procedure for concentration ranging from 1 mg ml−1 to 5 mg ml−1, with 0.5 mg ml−1 increments.

Results and discussion

The rationale of this work was to design molecules, which may show enhanced affinity for CA from the bacterial pathogen V. cholerae, since in the last period many bacterial CA inhibitors were shown to possess interesting anti-infective actionCitation22–26. In addition, the imidazole ring is a group known for its ability to form coordination bonds, and there are many examples in the biological systems of complexes between the amino acid histidine and the zinc ion, although this type of binding has rarely been observed for CA inhibitorsCitation27–30. On these bases, we have assumed that the azole nitrogen could interact with the zinc ion or with the zinc ion coordinated water moleculeCitation31–36, blocking thus the enzyme activity, and therefore, a series of imidazole containing compounds was synthesised and evaluated as inhibitors of the bacterial enzyme VchCA; moreover, the ability to inhibit the human physiologically most important, CA I and II (hCA I and hCA II) was evaluated.

Two set of amides and sulfonamides were synthesised (Scheme 1), including compound 10, where the azole was replaced with a phenyl ring (Scheme 2). The latter compound was synthesised in order to demonstrate the possible involvement of the imidazole moiety in the inhibition of CA.

Scheme 1. General synthesis of 1–9. Reagents and conditions: (a): TEA, dry CH2Cl2, 12–48 h, RT.

Scheme 1. General synthesis of 1–9. Reagents and conditions: (a): TEA, dry CH2Cl2, 12–48 h, RT.

Scheme 2. Synthesis of 10. Reagents and conditions: (a): TEA, dry CH2Cl2, 16 h, RT.

Scheme 2. Synthesis of 10. Reagents and conditions: (a): TEA, dry CH2Cl2, 16 h, RT.

The synthesis of 1–9 was carried out by adding the appropriate sulfonylchloride or acyl chloride to a solution of 3-(1H-imidazol-1-yl)propan-1-amine and TEA in dry CH2Cl2 and stirring the reaction mixture at RT for 16–48 h. 10 was prepared from 3-phenylpropan-1-amine and 4-nitrobenzoyl chloride. All final compounds were purified by gravity column chromatography to afford products with high purity in yields between 10 and 87%; the analytical and spectroscopic data of the purified compounds are in agreement with the purposed structures. The detailed procedures for 1–10 are reported in the Supporting Information.

Carbonic anhydrase inhibition

We assessed the CA inhibitory activity of compounds 1–10, using the clinically acetazolamide (AAZ) as reference drug, for the inhibition of two human (h) isoforms, hCA I and II (cytosolic, widely distributed enzymes) as well as α-VchCA. The results are reported in .

Table 1. Inhibition activity of compounds 1–10 and AAZ against Vibrio cholerae α-carbonic anhydrase, obtained as described in the supplementary information.

Aromatic sulfonamide derivatives (2–4) exhibited inhibitory activity towards VchCA with inhibition constants ranging between 6.2 and 8.5 nM, comparable to that obtained by the simplest methyl derivative 1 and comparable or slightly higher than the AAZ (Ki = 5.0 nM). In the series of amides, the aromatic derivatives (6–9) generally showed a higher activity (Ki = 5.4–10.99 nM) than the acetoamide derivative 5 (Ki = 39.2 nM) with the most active compound being the biphenyl derivative 9, that exhibited a Ki value of 5.4 nM, comparable to AAZ.

However, all synthesised compounds are selective inhibitor for the bacterial isoform over the human ones, hCA I and hCA II. Indeed, unlike the standard drug that is not selective, sulfonamides and amides reported here were generally ineffective inhibitors against hCA I, showing inhibition constants up to values higher 50,000 nM, and hCA II, on which no activity has been reported.

Surprisingly, the compound 10, where a phenyl ring replaces the imidazole moiety of compound 8, maintain the inhibitory activity towards VchCA, thus suggesting that the imidazole ring does not represents an essential requirement for the VchCA inhibition activity of these compounds. It is thus quite challenging to understand the CA inhibition mechanism with these compounds, which probably act by the so-called inhibition mechanism 5Citation37, i.e. an unknown one. Several other classes of compounds, such as the secondary/tertiary sulfonamides and the protein tyrosine kinase inhibitors imatinib/nilotinib inhibit CAs by this mechanism of action which is not understood in details for the moment, since no X-ray crystal structures for adducts of the enzyme with such inhibitors could be obtained.

The antibacterial activity

Based on the high inhibition activity observed in the enzymatic tests, we decided to evaluate in vitro the antibacterial activity of the synthesised compounds. Compounds were tested on two V. cholerae O1 strains of, the ATCC14103 and strain ISS-Vc014, a clinical isolate previously characterised for antibiotic resistance. Ampicillin, used as reference drug, showed inhibition activity at concentration higher than 0.01 mg ml−1 against the ATCC14103 strain and at 1.0 mg ml−1 against the ISS-Vc014 strain.

Among the ten compounds tested in concentrations ranging from 0.1 μg ml−1 to 1 mg ml−1, a partial inhibition of the bacterial growth was observed for compounds 2, 7, and 8 at the concentration of 1 mg ml−1, while no growth inhibition was observed for the other compounds. Compounds 2, 7, and 8 were then selected to be tested at higher concentrations (1 mg ml−1 to 5 mg ml−1). Compound 7 resulted not active until the concentration of 3 mg ml−1 (the limit of solubility), while compound 8 resulted partially active at the concentration of 3 mg ml−1 (the limit of solubility). Finally, compound 2 inhibited the bacterial growth of both V. cholerae ATCC14103 and ISS-Vc014 strain at a concentration of 2 mg ml−1. Compared to ampicillin, this compound resulted less active against the ATCC14103 strain, but possessed a comparable activity in the test on the strain ISS-Vc014.

Despite the compounds 2, 7, and 8 possess a high power on isolated VchCA, they resulted not particularly active in the cellular assay. This fact could be related to limited permeability of the cell wall to such compounds, leading to a reduced uptake into the cell, or to an efficient active efflux from the cell itself, a mechanism largely exploited by V. cholerae for antibiotic resistanceCitation1.

Conclusions

In conclusion, on the basis of the role played by the bicarbonate ion as a virulence factor of V. cholerae, the VchCA could represent an interesting molecular target for the development of antibacterial drugs. For this purpose, we have synthesised a series of imidazole derivatives which were able to inhibit VchCA at nM concentrations; these compounds were also highly selective because they were inactive toward the human CA isoforms hCA I and II.

However, much work remains to be done, on one side to clarify the importance of the imidazole moiety in the enzyme inhibition processes, and on the other one to make these compounds more active both in the enzymatic and cellular assays, in order to identify novel antiinfective leads.

Disclosure statement

CTS declares conflict of interest, being author of several patents in the field of CA inhibitors/activators. This research was financed in part, by several EU projects (Euroxy, Metoxia, DeZnIt and Dynano, to CTS). The other authors do not declare conflict of interest.

Additional information

Funding

This research was financed in part, by several EU projects (Euroxy, Metoxia, DeZnIt and Dynano, to CTS).

References

  • Kitaoka M, Miyata ST, Unterweger D, Pukatzki S. Antibiotic resistance mechanisms of Vibrio cholerae. J Med Microbiol 2011;60:397–407.
  • World Health Organization (WHO). Cholera Annual Report 2015, Weekly epidemiological record, 23 September 2016, 91, 38:433–440. Available from: http://www.who.int/wer/2016/wer9138/en/
  • Ali M, Nelson AR, Lopez AL, Sack DA. Updated global burden of cholera in endemic countries. PLoS Negl Trop Dis 2015;9:e0003832. doi:10.1371/journal.pntd.0003832
  • WHO. Cholera outbreak: assessing the outbreak response and improving preparedness; 2010. Available from: http://apps.who.int/iris/bitstream/10665/43017/1/WHO_CDS_CPE_ZFk_2004.4_eng.pdf
  • Abuaita BH, Withey JH. Bicarbonate induces Vibrio cholerae virulence gene expression by enhancing ToxT activity. Infect Immun 2009;77:4111–20.
  • Alafeefy AM, Ceruso M, Al-Tamimi A-MS, et al. Quinazoline–sulfonamides with potent inhibitory activity against the α-carbonic anhydrase from Vibrio cholera. Bioorg Med Chem 2014;22:5133–40.
  • Del Prete S, De Luca V, Scozzafava A, et al. Biochemical properties of a new α-carbonic anhydrase from the human pathogenic bacterium, Vibrio cholerae. J Enzyme Inhib Med Chem 2014;29:23–7.
  • Supuran CT. Structure and function of carbonic anhydrases. Biochem J 2016;473:2023–32.
  • Khalifah RG. The carbon dioxide hydration activity of carbonic anhydrase. I. Stop-flow kinetic studies on the native human isoenzymes B and C. J Biol Chem 1971;246:2561–73.
  • Diaz JR, Fernández Baldo M, Echeverría G, et al. A substituted sulfonamide and its Co (II), Cu (II), and Zn (II) complexes as potential antifungal agents. J Enzyme Inhib Med Chem 2016;31:51–62.
  • Chohan ZH, Scozzafava A, Supuran CT. Unsymmetrical 1,1′-disubstituted ferrocenes: synthesis of Co(ii), Cu(ii), Ni(ii) and Zn(ii) chelates of ferrocenyl-1-thiadiazolo-1′-tetrazole, -1-thiadiazolo-1′-triazole and -1-tetrazolo-1′-triazole with antimicrobial properties. J Enzyme Inhib Med Chem 2002;17:261–6.
  • Del Prete S, Vullo D, Fisher GM, et al. Discovery of a new family of carbonic anhydrases in the malaria pathogen Plasmodium falciparum – the η-carbonic anhydrases. Bioorg Med Chem Lett 2014;24:4389–96.
  • Supuran CT, Scozzafava A, Mastrolorenzo A. Bacterial proteases: current therapeutic use and future prospects for the development of new antibiotics. Expert Opin Ther Pat 2001;11:221–59.
  • Supuran CT. Carbonic anhydrases: novel therapeutic applications for inhibitors and activators. Nat Rev Drug Discov 2008;7:168–81.
  • Neri D, Supuran CT. Interfering with pH regulation in tumours as a therapeutic strategy. Nat Rev Drug Discov 2011;10:767–77.
  • Capasso C, Supuran CT. An overview of the alpha-, beta- and gamma-carbonic anhydrases from bacteria: can bacterial carbonic anhydrases shed new light on evolution of bacteria? J Enzyme Inhib Med Chem 2015;30:325–32.
  • Supuran CT. Structure-based drug discovery of carbonic anhydrase inhibitors. J Enzyme Inhib Med Chem 2012;27:759–72.
  • Supuran CT. Carbonic anhydrase inhibitors. Bioorg Med Chem Lett 2010;20:3467–74.
  • Colombo MM, Mastrandrea S, Leite F, et al. Tracking of clinical and environmental Vibrio cholerae O1 strains by combined analysis of the presence of toxin cassette, plasmid content and ERIC PCR. FEMS Immunol Med Microbiol 1997;19:33–45.
  • Ceccarelli D, Salvia AM, Sami J, et al. New cluster of plasmid-located class 1 integrons in Vibrio cholerae O1 and a dfrA15 cassette-containing integron in Vibrio parahaemolyticus isolated in Angola. Antimicrob Agents Chemother 2006;50:2493–9.
  • Andrews JM. Determination of minimum inhibitory concentrations. J Antimicrob Chemother 2001;48:5–16.
  • Supuran CT. Carbonic anhydrases: from biomedical applications of the inhibitors and activators to biotechnological use for CO(2) capture. J Enzyme Inhib Med Chem 2013;28:229–30.
  • Capasso C, Supuran CT. Anti-infective carbonic anhydrase inhibitors: a patent and literature review. Expert Opin Ther Pat 2013;23:693–704.
  • Capasso C, Supuran CT. Sulfa and trimethoprim-like drugs - antimetabolites acting as carbonic anhydrase, dihydropteroate synthase and dihydrofolate reductase inhibitors. J Enzyme Inhib Med Chem 2014;29:379–87.
  • Supuran CT. Bacterial carbonic anhydrases as drug targets: toward novel antibiotics? Front Pharmacol 2011;2:34.
  • Del Prete S, Vullo D, De Luca V, et al. Biochemical characterization of recombinant beta-carbonic anhydrase (PgiCAb) identified in the genome of the oral pathogenic bacterium Porphyromonas gingivalis. J Enzyme Inhib Med Chem 2015;30:366–70.
  • 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.
  • Carta F, Aggarwal M, Maresca A, et al. Dithiocarbamates strongly inhibit carbonic anhydrases and show antiglaucoma action in vivo. J Med Chem 2012;55:1721–30.
  • Scozzafava A, Menabuoni L, Mincione F, Supuran CT. Carbonic anhydrase inhibitors. A general approach for the preparation of water soluble sulfonamides incorporating polyamino-polycarboxylate tails and of their metal complexes possessing long lasting, topical intraocular pressure lowering properties. J Med Chem 2002;45:1466–76.
  • Fabrizi F, Mincione F, Somma T, et al. A new approach to antiglaucoma drugs: carbonic anhydrase inhibitors with or without NO donating moieties. Mechanism of action and preliminary pharmacology. J Enzyme Inhib Med Chem 2012;27:138–47.
  • Winum JY, Scozzafava A, Montero JL, Supuran CT. Therapeutic potential of sulfamides as enzyme inhibitors. Med Res Rev 2006;26:767–92.
  • Pacchiano F, Aggarwal M, Avvaru BS, et al. Selective hydrophobic pocket binding observed within the carbonic anhydrase II active site accommodate different 4-substituted-ureido-benzenesulfonamides and correlate to inhibitor potency. Chem. Commun. (Camb.) 2010;46:8371–3.
  • Carta F, Garaj V, Maresca A, et al. Sulfonamides incorporating 1,3,5-triazine moieties selectively and potently inhibit carbonic anhydrase transmembrane isoforms IX, XII and XIV over cytosolic isoforms I and II: solution and X-ray crystallographic studies. Bioorg Med Chem 2011;19:3105–19.
  • Garaj V, Puccetti L, Fasolis G, et al. Carbonic anhydrase inhibitors: Synthesis and inhibition of cytosolic/tumor-associated carbonic anhydrase isozymes I, II and IX with sulfonamides incorporating 1,2,4-triazine moieties. Bioorg Med Chem Lett 2004;14:5427–33.
  • Garaj V, Puccetti L, Fasolis G, et al. Carbonic anhydrase inhibitors. Novel sulfonamides incorporating 1,3,5-triazine moieties as inhibitors of the cytosolic and tumor-associated carbonic anhydrase isozymes I, II and IX. Bioorg Med Chem Lett 2005;15:3102–8.
  • Supuran CT. Carbonic anhydrase inhibitors. In: Puscas I, ed. Carbonic anhydrase and modulation of physiologic and pathologic processes in the organism. Helicon Press: Helicon, Timisoara; 1994:29–111.
  • Supuran CT. How many carbonic anhydrase inhibition mechanisms exist? J Enzyme Inhib Med Chem 2016;31:345–60.