2,577
Views
22
CrossRef citations to date
0
Altmetric
Short Communication

Design, synthesis and biological activity of pyrazinamide derivatives for anti-Mycobacterium tuberculosis

, &
Pages 1183-1186 | Received 03 Jun 2017, Accepted 01 Aug 2017, Published online: 04 Sep 2017

Abstract

A total of 11 pyrazinamide derivatives were designed and synthesised using pyrazinamide as the lead compound, which was optimised by structural modification with alkyl chains, six-membered rings, and bioisosterism, respectively. The target compounds were synthesised using pyrazinecarboxylic acid as the starting material by acylation, amidation, and alkylation, respectively. Their structures were confirmed by 1H NMR, 13C NMR, HRESIMS, and elemental analysis, respectively. The bioactivities of derivatives were assayed using bacteriostatic experiment and minimum inhibitory concentration experiment. It was showed that the derivatives had good inhibitory effect on Mycobacterium tuberculosis. The biological activity of derivative 1f was the best among all compounds, its antibacterial activity was 99.6%, and the minimum inhibitory concentration was 8.0 µg/mL.

Introduction

Tuberculosis (TB) is a common and fatal chronic infectious disease, and it is easy to occur in young people. Its clinical manifestations include low fever, night sweats, weakness or emaciation, and respiratory symptoms, such as cough and hemoptysis. The disease is mainly caused by Mycobacterium tuberculosis infection, also from M. bovis, M. africanum, M. canetti, M. microti, and other species of M. tuberculosis infection, but the mycobacteria do not usually infect healthy adultsCitation1–3. TB usually infects the lungs, brain, lymph system, central nervous system, urinary system, circulatory system, bones, joints, and even some skin can also be infected. At present, there are about 600 million people worldwide to have the Bacillus M. tuberculosis infection, and most infected people are in the developing world. The infection rate is the highest in Africa’s Per Capita, and most infected people have no corresponding symptoms for latent tuberculosis infectionCitation4–6. With the impact of environmental pollution and the spread of AIDS, the incidence of TB is increasing. The WHO announced in 1993 that TB was a global health emergency, while the termination of TB partners proposed the “global TB prevention program”. In 1882, scientists found that the pathogenic bacterium of tuberculosis for M. tuberculosis. Until the 50s of the last century, the researchers found drugs which could inhibit M. tuberculosis, and the TB infection control was effective. At present, the drugs commonly used in clinical trials for anti-tuberculosis include isoniazid, rifampicin, pyrazinamide, and ethambutolCitation7. Latent TB was usually used as a single drug, while active TB was most appropriate to take several drugs at the same time for reducing the risk of drug-resistant bacteriaCitation8,Citation9. In view of the great harm of TB, it is of great significance to study new anti-disease drugs. Pyrazinamide has good inhibitory effect on M. tuberculosis at pH 5–5.5, and the inhibition of bacteria is the strongest. The minimum inhibitory concentration of pyrazinamide in the human body is 12.5 µg/mL, and it can directly kill M. tuberculosis in vivo when the concentration reaches 50 µg/mL. The metabolite of pyrazinamide during antibacterial mechanism is pyrazinoic acid. When pyrazinamide gets into M. tuberculosis bacteria in vivo, the pyrazinoic acid metabolite achieves the bacteriostatic effect. In addition, it can also replace the nicotinamide to prevent dehydrogenase interference and dehydrogenation, interfere with M. tuberculosis on oxygen utilisation, affect the normal metabolism, and result in the death of bacteria M. tuberculosis. The pyrazinamide has good antibacterial effect, but there are side effects, and the main side effect is to cause liver damage. In addition, the water/fat-solubility of pyrazinamide is not good. Our research group designed and synthesised a new class of pyrazinamide compounds that achieved better bacteriostatic effect. The structure of pyrazinamide was optimised by structure modification with alkyl chains/six membered rings and bioisosterism. The design () of pyrazinamide derivatives was as follows.

Figure 1. Design of pyrazinamide derivatives.

Figure 1. Design of pyrazinamide derivatives.

Experimental

Synthetic experiments

Synthesis of pyrazine-2-carboxylic acid chloride (3)

The pyrazine carboxylic acid (12.41 g, 0.10 mol) was put into 250 mL round bottom flask, then 100 mL methylene chloride as solvent and 5 drops of N,N-dimethyl formamide (DMF) used as catalyst for the reaction were added. The flask was placed in the ice water bath, and a magnetic mixer was used to stir until the reaction liquid became clarified. The thionyl chloride (29 mL, 0.40 mol) was constantly dropped into the flask under stirring, and the drop speed rate and reaction temperature were controlled. After the reactants were added, the reaction lasted 8 h under refluxing. The methylene chloride and excessive thionyl chloride were removed under vacuum. The mixture was dried to get the crude product of pyrazine formyl chloride. The crude product was recrystallised with toluene, filtered, and dried, respectively. The pure pyrazine-2-carboxylic acid chloride (3) was obtained as the white crystalCitation10.

A general method for synthesis of compounds 4a to 4c

The pyrazine formyl chloride (14.25 g, 0.10 mol) was put into 250 mL round bottom flask, and 16 mL (0.20 mol) bromide methylamine and 100 mL toluene solvent were added. The reaction liquid was magnetically stirred, and the reaction lasted 8 h under refluxing. The reaction mixture was thermally filtrated and cooled. The crude product was recrystallised with toluene, filtered, and dried, respectively. The pure N-(1-bromine methyl) pyrazine formamide (4a) as white crystal was obtained. The general method was used to synthesise compounds 4b4c.

N-(bromomethyl)pyrazine-2-carboxamide (4a): white crystal; 1H NMR (CDCl3, 300 MHz): δ = 9.96 (1H, s, pyrazine-H), 9.38 (1H, s, –NH–), 9.10 (1H, dd, J = 8.0 Hz, pyrazine-H), 8.89 (1H, dd, J = 8.0 Hz, pyrazine-H), 4.97 (2H, s, J = 6.4 Hz, –CH2–); 13C NMR (CDCl3, 75 MHz): δ = 161.0 (C=O), 146.0, 145.0, 144.7, 144.6 (pyrazine-C), 45.4 (CH2); HRESIMS m/z (pos): 216.0376 C6H6N3OBr (calcd. 215.7230); Anal. Calcd. for C6H6N3OBr: C, 33.36; H, 2.80; N, 19.45; Br, 36.99. Found: C, 33.37; H, 2.81; N, 19.43; Br, 36.98%.

N-(2-bromoethyl)pyrazine-2-carboxamide (4b): white crystal; 1H NMR (CDCl3, 300 MHz): δ = 9.96 (1H, s, pyrazine-H), 9.10 (1H, dd, J =7.9 Hz, pyrazine-H), 8.98 (1H, s, –NH–), 8.89 (1H, dd, J = 7.9 Hz, pyrazine-H), 3.74 (4H, s, J = 6.3 Hz, –CH2–); 13 C NMR (CDCl3, 75 MHz): δ = 160.7 (C=O), 146.0, 145.0, 144.7, 144.6 (pyrazine-C), 46.6, 30.9 (CH2CH2); HRESIMS m/z (pos): 230.0653 C7H8N3OBr (calcd. 229.3426); Anal. Calcd. for C7H8N3OBr: C, 36.54; H, 3.51; N, 18.26; Br, 34.73. Found: C, 36.55; H, 3.52; N, 18.26; Br, 34.72%.

N-(3-bromopropyl)pyrazine-2-carboxamide (4c): white crystal; 1H NMR (CDCl3, 300 MHz): δ = 9.96 (1H, s, pyrazine-H), 9.10 (1H, dd, J =7.8 Hz, pyrazine-H), 8.98 (1H, s, –NH–), 8.89 (1H, dd, J =7.8 Hz, pyrazine-H), 3.51 (2H, m, J = 6.0 Hz, –CH2–), 3.18 (2H, m, J = 6.0 Hz, –CH2–), 2.05 (2H, m, J = 6.0 Hz, –CH2–); 13C NMR (CDCl3, 75 MHz): δ = 160.7 (C=O, 146.0, 145.0, 144.7, 144.6 (pyrazine-C), 38.4, 32.6, 30.8 (CH2CH2CH2); HR-ESI-MS m/z (pos): 244.0921 C8H10N3OBr (calcd. 243.5438); Anal. Calcd. for C8H10N3OBr: C, 39.37; H, 4.13; N, 17.22; Br, 32.74. Found: C, 39.38; H, 4.14; N, 17.21; Br, 32.73%.

A general method for all titled derivatives 1a–1k

N-(1-bromine methyl) pyrazine formamide (21.60 g, 0.10 mol) was put into 250 mL round-bottomed flask, and 21.5 mL (0.20 mol) six-membered ring compound and 100 mL toluene were added. The reaction liquid was magnetically stirred, and the reaction lasted 12 h under refluxing. The reaction mixture was filtrated and cooled. The crude product was recrystallised with toluene, filtered, and dried, respectively. The target (1) as white crystal was obtained.

N-(piperidin-1-ylmethyl)pyrazine-2-carboxamide (1a): white crystal; yield 86.2%; m.p. 183–185 °C; 1H NMR (CDCl3, 300 MHz): δ = 9.96 (1H, s, pyrazine-H), 9.38 (1H, s, –NH–), 9.10 (1H, dd, J = 7.8 Hz, pyrazine-H), 8.89 (1H, dd, J = 7.8 Hz, pyrazine-H), 4.03 (2H, s, J = 6.0 Hz, N–CH2–), 2.39 (4H, m, –CH2–), 1.43 (4H, m, –CH2–), 1.30 (2H, m, –CH2–); 13 C NMR (CDCl3,75 MHz): δ = 161.0 (C=O), 146.6, 145.0, 144.7, 144.6 (pyrazine-C), 66.1 (CH2, N–CH2), 54.2, 25.6, 24.5 (CH2, CH2–CH2); HRESIMS m/z (pos): 220.2760 C11H16N4O (calcd. 220.1324); Anal. Calcd. for C11H16N4O: C, 59.98; H, 7.32; N, 25.44. Found: C, 59.99; H, 7.31; N, 25.43.

N-(piperazin-1-ylmethyl)pyrazine-2-carboxamide (1b): white crystal; yield 88.2%; m.p. 190–192 °C; 1H NMR (CDCl3, 300 MHz): δ = 9.96 (1H, s, pyrazine-H), 9.38 (1H, s, –NH–), 9.10 (1H, dd, J = 7.6 Hz, pyrazine-H), 8.89 (1H, dd, J = 7.6 Hz, pyrazine-H), 4.03 (2H, s, J = 5.9 Hz, N–CH2–), 2.65 (4H, m, –CH2–), 2.34 (4H, m, –CH2–), 1.07 (1H, s, –NH–); 13C NMR(CDCl3, 75 MHz): δ = 161.0 (C=O), 146.6, 145.0, 144.7, 144.6 (pyrazine-C), 66.1 (CH2, N–CH2), 54.7, 45.9 (CH2, CH2–CH2); HRESIMS m/z (pos): 221.2640 C10H15N5O (calcd. 221.1277); Anal. Calcd. for C10H15N5O: C, 54.28; H, 6.83; N, 31.65. Found: C, 54.29; H, 6.83; N, 31.64.

N-(thiomorpholinomethyl)pyrazine-2-carboxamide (1c): white crystal; yield 86.3%; m.p. 201–203 °C; 1H NMR (CDCl3, 300 MHz): δ = 9.96 (1H, s, pyrazine-H), 9.38 (1H, s, –NH–), 9.10 (1H, dd, J = 7.7 Hz, pyrazine-H), 8.89 (1H, dd, J = 7.7 Hz, pyrazine-H), 4.03(2H, s, J = 6.0 Hz, N–CH2–), 2.73 (4H, m, –CH2–), 2.54 (4H, m, –CH2–); 13 C NMR (CDCl3, 75 MHz): δ = 161.0 (C=O), 146.6, 145.0, 144.7, 144.6 (pyrazine-C), 65.4 (CH2, N–CH2), 57.7, 28.0 (CH2, CH2–CH2); HRESIMS m/z (pos): 238.3090 C10H14N4OS (calcd. 238.0888); Anal. Calcd. for C10H14N4OS: C, 50.40; H, 5.92; N, 23.51; S, 13.45. Found: C, 50.41; H, 5.91; N, 23.50; S, 13.46%.

N-(2-(piperidin-1-yl)ethyl)pyrazine-2-carboxamide (1d): white crystal; yield 84.0%; m.p. 185–187 °C; 1H NMR (CDCl3, 300 MHz): δ = 9.96 (1H, s, pyrazine-H), 9.10 (1H, dd, J = 7.6 Hz, pyrazine-H), 8.89 (1H, dd, J = 7.6 Hz, pyrazine-H), 8.81 (1H, s, –NH–), 3.36 (2H, s, J = 5.8 Hz, N–CH2–), 2.46 (2H, s, –CH2–), 2.42 (4H, m, –CH2–), 1.49 (4H, m, –CH2–), 1.37 (2H, m, –CH2–); 13C NMR (CDCl3, 75 MHz): δ = 160.7 (C=O), 146.6, 145.0, 144.7, 144.6 (pyrazine-C), 57.2 (CH2, N–CH2), 56.8, 54.0, 25.9, 24.5 (CH2, CH2–CH2); HRESIMS m/z (pos): 234.1481 C12H18N4O (calcd. 234.3030); Anal. Calcd. for C12H18N4O: C, 61.52; H, 7.74; N, 23.91. Found: C, 61.53; H, 7.75; N, 23.90.

N-(2-(piperazin-1-yl)ethyl)pyrazine-2-carboxamide (1e): white crystal; yield 90.4%; m.p. 192–193 °C; 1H NMR (CDCl3, 300 MHz): δ = 9.96 (1H, s, pyrazine-H), 9.10 (1H, dd, J = 7.7 Hz, pyrazine-H), 8.89 (1H, dd, J = 7.7 Hz, pyrazine-H), 8.81 (1H, s, –NH–), 3.36 (2H, s, J = 5.9 Hz, N–CH2–), 2.65 (4H, m, –CH2–), 2.46 (2H, s, –CH2–), 2.34 (4H, m, –CH2–), 1.07 (1H, s, –NH2–); 13C NMR (CDCl3, 75 MHz): δ = 160.7 (C=O), 146.6, 145.0, 144.7, 144.6 (pyrazine-C), 57.3 (CH2, N–CH2), 57.2, 56.8, 53.7, 46.2 (CH2, CH2–CH2); HRESIMS m/z (pos): 235.1433 C11H17N5O (calcd. 235.2910); Anal. Calcd. for C11H17N5O: C, 56.15; H, 7.28; N, 29.77. Found: C, 56.16; H,7.27; N, 29.78.

N-(2-morpholinoethyl)pyrazine-2-carboxamide (1f): white crystal; yield 91.2%; m.p. 202–204 °C; 1H NMR (CDCl3, 300 MHz): δ = 9.96 (1H, s, pyrazine-H), 9.10 (1H, dd, J = 7.8 Hz, pyrazine-H), 8.89 (1H, dd, J = 7.8 Hz, pyrazine-H), 8.81 (1H, s, –NH–), 3.50 (4H, m, –CH2–), 3.36 (2H, s, J = 6.0 Hz, N–CH2–), 2.46 (2H, s, –CH2–), 2.40 (4H, m, –CH2–); 13C NMR (CDCl3, 75 MHz): δ = 160.7 (C=O), 146.6, 145.0, 144.7, 144.6 (pyrazine-C), 66.7 (CH2, N–CH2), 57.2, 55.8, 54.0 (CH2, CH2–CH2); HRESIMS m/z (pos): 236.2750 C11H16N4O2 (calcd. 236.1273); Anal. calcd for C11H16N4O2: C, 55.92; H, 6.83; N, 23.71. Found: C, 55.91; H, 6.84; N, 23.70.

N-(2-thiomorpholinoethyl)pyrazine-2-carboxamide (1g): white crystal; yield 90.4%; m.p. 192–193 °C; 1H NMR (CDCl3, 300 MHz): δ = 9.96 (1H, s, pyrazine-H), 9.10 (1H, dd, J = 7.7 Hz, pyrazine-H), 8.89 (1H, dd, J = 7.7 Hz, pyrazine-H), 8.81 (1H, s, –NH–), 3.36 (2H, s, J = 5.9 Hz, N–CH2–), 2.73 (4H, m, –CH2–), 2.54 (4H, m, –CH2–), 2.46 (2H, s, –CH2–); 13C NMR (CDCl3, 75 MHz): δ = 160.7 (C=O), 146.6, 145.0, 144.7, 144.6 (pyrazine-C), 60.3 (CH2, N–CH2), 57.2, 53.3, 28.3 (CH2, CH2–CH2); HRESIMS m/z (pos): 252.3360 C11H16N4OS (calcd. 252.1045); Anal. Calcd. for C11H16N4OS: C, 52.36; H, 6.39; N, 22.20; S, 12.71. Found: C, 52.35; H, 6.40; N, 22.18; S, 12.72%.

N-(3-(piperidin-1-yl)propyl)pyrazine-2-carboxamide (1h): white crystal; yield 81.0%; m.p. 204–205 °C; 1H NMR (CDCl3, 300 MHz): δ = 9.96 (1H, s, pyrazine-H), 9.10 (1H, dd, J = 7.5 Hz, pyrazine-H), 8.98 (1H, s, –NH–), 8.89 (1H, dd, J = 7.5 Hz, pyrazine-H), 3.18 (2H, s, J = 5.8 Hz, N–CH2–), 2.42 (4H, m, –CH2–), 2.36 (2H, s, –CH2–), 1.73 (2H, s, –CH2–), 1.49 (4H, m, –CH2–), 1.37 (2H, m, –CH2–); 13C NMR(CDCl3, 75 MHz): δ = 160.7 (C=O), 146.6, 145.0, 144.7, 144.6 (pyrazine-C), 57.1 (CH2, N–CH2), 40.4, 39.7, 27.0, 25.9, 24.5 (CH2, CH2–CH2); HRESIMS m/z (pos): 248.3300 C13H20N4O (calcd. 248.1637); Anal. Calcd. for C13H20N4O: C, 62.88; H, 8.12; N, 22.56. Found: C, 62.89; H, 8.13; N, 22.55.

N-(3-(piperazin-1-yl)propyl)pyrazine-2-carboxamide (1i): white crystal; yield 84.6%; m.p. 210–211 °C; 1H NMR (CDCl3, 300 MHz): δ = 9.96 (1H, s, pyrazine-H), 9.10 (1H, dd, J = 7.6 Hz, pyrazine-H), 8.98 (1H, s, –NH–), 8.89 (1H, dd, J = 7.6 Hz, pyrazine-H), 3.18 (2H, s, J = 5.7 Hz, N–CH2–), 2.65 (4H, m, –CH2–), 2.36 (2H, s, –CH2–), 2.34 (4H, m, –CH2–), 1.73 (2H, s, –CH2–), 1.07 (2H, s, –CH2–); 13C NMR (CDCl3, 75 MHz): δ = 160.7 (C=O), 146.6, 145.0, 144.7, 144.6 (pyrazine-C), 57.6 (CH2, N–CH2), 51.6, 46.2, 39.7, 27.0 (CH2, CH2–CH2); HRESIMS m/z (pos): 249.3180 C12H19N5O (calcd. 249.1590); Anal. Calcd. for C12H19N5O: C,57.81; H, 7.68; N, 28.09. Found: C, 57.80; H, 7.69; N, 28.10.

N-(3-morpholinopropyl)pyrazine-2-carboxamide (1j): white crystal; yield 87.3%; m.p. 221–223 °C; 1H NMR (CDCl3, 300 MHz): δ = 9.96 (1H, s, pyrazine-H), 9.10 (1H, dd, J = 7.7 Hz, pyrazine-H), 8.98 (1H, s, –NH–), 8.89 (1H, dd, J = 7.7 Hz, pyrazine-H), 3.55 (4H, m, –CH2–), 3.18 (2H, s, J = 5.7 Hz, N–CH2–), 2.37 (2H, s, –CH2–), 2.33 (4H, m, –CH2–), 1.73 (2H, s, –CH2–); 13C NMR (CDCl3, 75 MHz): δ = 160.7 (C=O), 146.6, 145.0, 144.7, 144.6 (pyrazine-C), 62.9 (CH2, N–CH2), 59.4, 39.7, 27.0 (CH2, CH2–CH2); HRESIMS m/z (pos): 250.3020 C12H18N4O2 (calcd. 250.1430); Anal. Calcd. for C12H18N4O2: C, 57.58; H, 7.25; N, 22.38. Found: C, 57.59; H, 7.24; N, 22.37.

N-(3-thiomorpholinopropyl)pyrazine-2-carboxamide (1k): white crystal; yield 84.3%; m.p. 230–231 °C; 1H NMR (CDCl3, 300 MHz): δ = 9.96 (1H, s, pyrazine-H), 9.10 (1H, dd, J = 7.6 Hz, pyrazine-H), 8.98 (1H, s, –NH–), 8.89 (1H, dd, J = 7.6 Hz, pyrazine-H), 3.18 (2H, s, J = 5.6 Hz, N–CH2–), 2.73 (4H, m, –CH2–), 2.54 (4H, m, –CH2–), 2.36 (2H, s, –CH2–), 1.73 (2H, s, –CH2–); 13C NMR (CDCl3, 75 MHz): δ = 160.7 (C=O), 146.6, 145.0, 144.7, 144.6 (pyrazine-C), 60.6 (CH2, N–CH2), 51.2, 39.7, 28.3, 27.0 (CH2, CH2–CH2); HRESIMS m/z (pos): 266.3630 C12H18N4OS (calcd. 266.1201); Anal. Calcd. for C12H18N4OS: C, 54.11; H, 6.81; N, 21.03; S, 12.04. Found: C, 54.10; H, 6.82; N, 21.04; S, 12.04%.

Biological activity experiments

TB bacillus strain was the standard strain H37Rv. The modified roche medium was used for bacteriostatic experiment, and the MH agar culture medium was used for the experiment of minimal inhibitory concentration (MIC). The standard strains were dissolved in sterile distilled water, and the bacteria liquid concentration was adjusted to 103 CFU/mL. The 0.1 mL bacteria were accurately inoculated by pipetting gun on modified roche medium, then pyrazinamide derivative (0.1 mL, 25 µg/mL) was put into culture medium. Each compound was inoculated in five culture tubes, respectively. The culture tubes were put into an incubator with 5% CO2 at 36 °C for 20 days, and the wedding difference of coli growth was observed. Blank control experiment did not need to take drug, and directly inoculated with the microorganisms. For minimum bacteriostatic concentration experiment, various derivatives were mixed into different concentration solutions firstly, then they were moved into MH agar medium, respectively. After sterilisation, 0.1 mL standard H37Rv bacteria liquid with multi-point inoculator was inoculated in the agar plate surface. Vaccination was added into the incubator with 5% CO2 at 36 °C for 24 h, and the growth differences of n/med Tuberculosis bacili were observed.

Results and discussion

The design and synthesis of pyrazinamide derivatives

The pyrazinamide derivatives for anti-Mycobacterium tuberculosis bacilli were designed using the pyrazinamide as lead compound, which was modified with the alkyl chains/six-member rings and bioisosterism. In the structure transformation with alkyl chains, the number of carbon atom was 1, 2, and 3, respectively. In the structure transformation with six-member rings, the X1 = N; X2 = CH2, NH, O, S were selected, respectively. Through the optimisation of lead compound structure, 20 compounds were designed. Using pyrazinecarboxylic acid as the starting material, the pyrazinamide derivatives were synthesised by acylation, amidation, and alkylation, respectively. This synthetic route () was simple, and the yield was higher (79.6%–91.2%). It indicated that the length of alkyl chain did not obviously affect the reaction yield, but the six-member ring type more obviously influenced the yield. In general, the production rate with six-member ring (X1 = N and X2= O) was obviously higher than other combinations. In addition, it was showed that the chemical shift of –NH(CH2)n– changed from compound 4 to target product 1. So, the synthesis was successful.

Figure 2. Synthetic route of pyrazinamide derivatives.

Figure 2. Synthetic route of pyrazinamide derivatives.

Biological activities

Bioactive experiment process was based on standard H37Rv tuberculosis bacillus strains differences, the bacteriostatic experiment and minimum bacteriostatic concentration experiment were carried out, respectively. The modified roche medium was used for the bacteriostatic experiment, and the MH agar was used for the minimum bacteriostatic concentration experiment. Pure culture method and agar dilution method were used, respectively. The results of biological activity experiment are shown in . It showed that the length of alkyl chain and six-member ring kind had certain effect on the biological activity. For compound 1f with the alkyl chain of carbon atom (n = 2) and X1 =N, X2 =O, its bacteriostatic effect was 99.2%, and the minimum bacteriostatic concentration was the lowest (8.0 µg/mL) among all compounds. Other bioactive compounds were ideal, the bacteriostatic rate was from 79.2% to 93.6%, and the minimum bacteriostatic concentration ranged from 10.2 µg/mL to 32.2 µg/mL. For the structure-activity, the alkyl chain and the six-member ring kind influenced the activity of pyrazinamide derivatives. When the carbon atom number of alkyl chain was 2, the molecular space structure was the best. In addition, the derivative with the six-member ring (X1=N, X2=O) had the biggest combining ability, so its biological activity was the best too. In the experiment, the pyrazinamide was used as a positive control.

Table 1. Biological activities of pyrazinamide derivatives (0.1 mL, 25 μg/mL).

Conclusion

A total of 11 pyrazinamide derivatives were designed and synthesised using pyrazinamide as the lead compound, which was modified with the alkyl chains/six-member rings and bioisosterism, respectively. The chemical structures of these derivatives were characterised by 1H NMR, 13 C NMR, MS, and elemental analysis, respectively. These derivatives had been carried out the assay of biological activity. It showed that some derivatives had good function of resisting M. tuberculosis, high bacteriostatic rates, and low minimum bacteriostatic concentrations. In addition, the derivative 1f had the best biological activity among all compounds, its antibacterial rate was 99.6%, and the minimum concentration of bacteriostasis was 8.0 µg/mL.

Disclosure statement

No potential conflict of interest was reported by the authors.

Additional information

Funding

The project was sponsored by the Scientific Research Foundation for the Returned Overseas Chinese Scholars, State Education Ministry. The work was also supported by Chongqing Scientific & Technological Innovation Special Project of Social Undertakings and People's Livelihood Guarantee [No. cstc2015shmszx80060], Chongqing Key Research Project of Basic Science & Frontier Technology [No. cstc2017jcyjBX0012], Scientific and Technological Research Program of Chongqing Municipal Education Commission [Grant No. KJ1400523], Foundation Project of Chongqing Normal University [No. 14XYY020], Chongqing General Research Project of Basic Science & Frontier Technology [No. cstc2015jcyjA10054], Chongqing Normal University Postgraduate’s Research and Innovation Project [No. YKC17004], Open Foundation Project of University Engineering Research Center for Bioactive Substances in Chongqing [No. AS201614], Chongqing University Students’ Training Project of Innovation & Undertaking [No. 201610637076], China.

References

  • Gangjee A, Zaware N, Raghavan S. Synthesis and biological activity of 5-chloro-N4-substituted phenyl-9H-pyrimido [4, 5-b] indole-2, 4-diamines as vascular endothelial growth factor receptor-2 inhibitors and antiangiogenic agents. Bioorg Med Chem 2013;21:1857–64.
  • Liu Q, Sabnis Y, Zhao Z. Developing irreversible inhibitors of the protein kinase cysteinome. Chem Biol 2013;20:146–59.
  • Wissner A, Fraser HL, Ingalls CL. Dual irreversible kinase inhibitors: quinazoline-based inhibitors incorporating two independent reactive centers with each targeting different cysteine residues in the kinase domains of EGFR and VEGFR-2. Bioorg Med Chem 2007;15:3635–48.
  • Kumar S, Tiwari M. Topomer-CoMFA-based predictive modelling on 2,3-diaryl substituted-1,3-thiazolidin-4-ones as non-nucleoside reverse transcriptase inhibitors. Med Chem Res 2015;24:245–57.
  • Sean M, Bruce H. Chemoselective reductions of nitroaromatics in water at room temperature. Org Lett 2014;16:98–101.
  • Chachignon H, Scalacci N, Petricci E. Synthesis of 1,2,3-substituted pyrroles from propargylamines via a one-pot tandem enyne cross metathesis-cyclization reaction. Org Chem 2015;80:5287–95.
  • Dickinson JM, Aber VR, Mitchison DA. Bactericidal activity of streptomycin, isoniazid, rifampin, ethambutol, and pyrazinamide alone and in combination against Mycobacterium tuberculosis. Am Rev Resp Dis 1977;116:627–35.
  • Palmer R, Weiss R, Zusman R. Effects of nabumetone, celecoxib, and ibuprofen on blood pressure control in hypertensive patients on angiotensin converting enzyme inhibitors. Am J Hypertens 2003;16:135.
  • Maria J, Avelino C, Sara I. MgO nanoparticle-based multifunctional catalysts in the cascade reaction allows the green synthesis of anti-inflammatory agents. J Cata 2007;247:223–30.
  • Dolezal M, Miletin M, Kunes J, Kralova K. Substituted amides of pyrazine-2-carboxylic acids: synthesis and biological activity. Molecules 2002;7:363–73.