Next Article in Journal
The Microfluidic Toolbox for Analyzing Exosome Biomarkers of Aging
Next Article in Special Issue
Antibacterial Activity of a Promising Antibacterial Agent: 22-(4-(2-(4-Nitrophenyl-piperazin-1-yl)-acetyl)-piperazin-1-yl)-22-deoxypleuromutilin
Previous Article in Journal
Experimental and Computational Study of a Liquid Crystalline Dimesogen Exhibiting Nematic, Twist-Bend Nematic, Intercalated Smectic, and Soft Crystalline Mesophases
Previous Article in Special Issue
New Heteroleptic 3D Metal Complexes: Synthesis, Antimicrobial and Solubilization Parameters
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Microwave-Assisted Synthesis, Structural Characterization and Assessment of the Antibacterial Activity of Some New Aminopyridine, Pyrrolidine, Piperidine and Morpholine Acetamides

by
Abdulmajeed S. H. Alsamarrai
* and
Saba S. Abdulghani
Department of Chemistry, College of Applied Sciences, University of Samarra, Salahaldin 13/1333, Iraq
*
Author to whom correspondence should be addressed.
Submission received: 28 September 2020 / Revised: 9 November 2020 / Accepted: 11 November 2020 / Published: 20 January 2021

Abstract

:
A series of new acetamide derivatives 2228 of primary and secondary amines and para-toluene sulphinate sodium salt have been synthesized under microwave irradiation and assessed in vitro for their antibacterial activity against one Gram-positive and two Gram-negative bacterial species such as S. pyogenes, E. coli, and P. mirabilis using the Mueller-Hinton Agar diffusion (well diffusion) method. The synthesized compounds with significant differences in inhibition diameters and MICs were compared with those of amoxicillin, ampicillin, cephalothin, azithromycin and doxycycline. All of the evaluated acetamide derivatives were used with varying inhibition concentrations of 6.25, 12.5, 37.5, 62.5, 87.5, 112.5 and 125 µg/mL. The results show that the most important antibacterial properties were displayed by the synthetic compounds 22 and 24, both of bear a para-chlorophenyl moiety incorporated into the 2-position moiety of acetamide 1. The molecular structures of the new compounds were determined using the FT-IR and 1H-NMR techniques.

1. Introduction

Several acetamide derivatives have been reported to act as antimicrobial agents which have been shown to be highly effective against Gram-positive and Gram-negative species [1,2,3,4,5]. There are many reports on the use of α-chloroacetamides as useful building blocks for the synthesis of complex heterocyclic compounds. Further possible biological effects have also been shown by derivatives of the acetamide moiety 1 (Figure 1), notably as antimalarial [6], anticancer [7], anti-diabetic [8], anti-tuberculosis [9] and anti-inflammatory agents [10], as well as in industrial applications such as stabilizer synthesis [11], plastic releasing agents [12], film [13], surfactants and soldering flux [14], organic fibers [15] and dyes [16].
The compounds atorvastatin (2) [17], lidocaine (3) [18], paracetamol (4) [19], amoxicillin (5) [20], levobupivacaine (6) [21] are the most widely used drugs in medicine that contain the acetamide moiety 1 (Figure 1). Pyrrolidine, piperidine, morpholine, and piperazine are secondary amines that are very valuable saturated heterocyclic compounds because of their wide and diverse range of promising biological activities, such as antibacterial and anti-inflammatory activity [22,23,24,25,26]. In addition, many derivatives of 1 containing secondary cyclic amines linked to the 1 or 2-positions were reported to be anticonvulsant agents. For instance, anticonvulsant compounds such as 7 and 8 are used to treat epilepsy [27], while the combination of compounds 9 and 10 is used to inhibit kinase enzyme and antihistamines [28].
Pyridine heterocycles are present in certain biologically active molecules (Figure 2). One of the pyridine-containing medications is Lunesta with the active ingredient 11 that is used to treat insomnia, and pioglitazone 12 which is effective in diabetes care.
In addition, previous studies conducted by Nakamoto et al. [29] and Tang et al. [30] evaluated the compounds 13 and 14 as an antifungal and antituberculous agent, respectively.
The synthesis of compounds with broad antibacterial activity is required because of the extensive use of antibiotics in medicine and the prevailing resistance of microorganisms. Conventional organic preparation methods are too sluggish to meet the need for the rapid synthesis of these compounds. Nowadays microwave irradiation represents an interesting technique to be applied in organic synthesis [31]. Herein, we report the use of microwaves in the synthesis of seven new acetamide derivatives 2228 by reaction of chloroacetyl chloride 15 with primary, secondary amines and para-toluene sulphinate sodium salts. In addition, the aim of this study was to assess the susceptibility profile of bacterial isolates to the synthesized compounds 2228 and reference antibiotics, and to determine their minimum inhibition concentrations (MICs). The structures of compounds 2228 were determined by 1H-NMR, and FT-IR and elemental analysis. Table 1 shows the approach followed.

2. Results and Discussion

2.1. Chemistry

We are engaged in intensive efforts to synthesize nitrogen-heterocyclic compounds in high yields. Following a previously described study [32], the α-chloroacetamide intermediates 1621 and the new targeted derivatives 2228 were designed and synthesized by introducing para-toluene sulphinate, pyrrolidinyl, morpholinyl, and piperidinyl moieties into the acetamide scaffold as can be seen in Scheme 1.
Over the past thirty years, microwave irradiation has been used to enhance reaction rates [33,34]. We have thus exploited the advance of microwave technology to accelerate the creation of C-N bonds. For the synthesis of the precursors 1621, the process was performed under mild conditions involving the addition of chloroacetyl chloride 15 to a mixture of amines and para-toluene sulphinate sodium salt in dry CH2Cl2 at 0 °C and the isolated yields of these compounds ranged from 60 to 90% (see Table 1). The synthesis of targeted compounds 2228 was achieved through two routes: conventional heating and microwave irradiation. The heating process involved precursors 1621, amine, and sodium para-toluene sulphinate reactions in a CH3CN solution containing Et3N as a catalyst giving the compounds 2228 in moderate yields reaching 60% at 70 °C (see Scheme 1 and Table 1), On the other hand, treatment of the precursors 1621 with amines as well as sodium para-toluene sulphinate in dry CH3CN provided the desired compounds 2228 at 65–70 °C in good yields under microwave irradiation. The reactions took 5–10 min to complete and one of the advantages of this technique is allowed us to isolate clean products in good yields and without side products (see Table 1 and Table 2 for the corresponding 1H-NMR data).

2.2. In Vitro Antibacterial Activity Testing

Instructive bacterial infections such as tuberculosis, urinary tract infection, pneumonia, brain abscess, phyaryngitis, and tonsillitis are a series of life-threatening diseases commonly recognized in immunocompromised patients. Such infectious diseases are becoming more and more resistant to antibiotics. Resistance to microbial drugs is an inevitable consequence of the overuse of antibacterial drugs. Susceptibility to antibiotics by pathogens has now become a crucial factor in the effectiveness of chosen antibacterial drugs. Amoxicillin, ampicillin, doxycycline, azithromycin and cephalothin, the five reference antibiotics used in this research, are commonly used for the treatment of infections. Researchers continue to seek new antimicrobial agents. This research was carried out to determine the susceptibility of certain bacterial organisms toward seven newly synthesized compounds. The synthesized compounds 2228 have been evaluated for antibacterial activity in comparison with the abovementioned reference antibiotics.
Acetamide derivatives are well known to have a broad range of antibacterial actions [35]. With regards to our recent research [32] on the synthesis of acetamide derivatives and the assessment of their biological role, in this in vitro study, three species of microorganisms were included, and all isolates of the three species were susceptible to the synthesized compounds 2228 at MICs greater than 6.25 μg/mL.
Tested compounds 22 and 23, exhibited inhibition zones ranging from 6.0 to 8.4 mm at MIC of 12.5 μg/mL against the Gram-negative species E. coli and P. mirabilis that cause urinary tract infections (UTI) in patients, while isolates of these bacterial species were not susceptible to compounds 26 and 28 even at an MIC of 37.5 μg/mL. Compounds 24, 25, and 27 showed inhibition zones ranging from 6 to 14 mm at a MIC of 37.5 μg/mL (Figure 3 and Figure 4, and Table S1). These intermediate results are in agreement with other authors who have reported inhibition zones ranging from between 11 to 12 mm using amoxicillin (20 μg) and ampicillin (20 μg) [36,37,38] antimicrobial disks (Tables S1 and S2).
The Gram-positive species S. pyogenes are one of the main causes of urinary tract infections and poses a significant health concern causing a number of human diseases [39,40]. S. pyogenes is also considered to be the most common and significant cause of bacterial tonsillitis in children and adults. Isolates of this bacterial species obtained from the Tikrit Teaching Hospital (Tikrit, Iraq) were all susceptible to the synthesized compounds 2228 at MICs of 12.5 μg/mL, and compounds 27 and 28 did not display any inhibition zones in this test, even at a MIC of 37.5 μg/mL (see Figure 5 and Table S3).
For the sake of comparison, we compared the antibacterial susceptibility of all the isolates to the five antibacterial agents amoxicillin (20 μg), ampicillin (25 μg), cephalothin (30 μg), azithromycin (15 μg) and doxycycline (30 μg) by the disc diffusion process. Isolates of the three species were susceptible to the five antibiotics and displayed inhibition zones ranging from 5.8–19 mm. S. pyogenes isolates showed intermediate zones ranging from 5.8–7.6 mm compared to other Gram-negative species isolates (see Figure 5, Table S3).
All species included in this research, however, were more susceptible to the synthesized compounds 2228 at higher MICs ranging from 37.5 to 125 μg/mL compared to the control antibiotics. This low activity may, potentially, be caused by resistance-acquiring bacteria due to the arbitrary use of antibiotics by patients [41,42,43].
The aim of this study was also to observe any relation between the structure of the synthesized compounds 2228 and the susceptibility of the bacterial isolates towards them. Thus, as described earlier, the antibacterial susceptibility of compounds 2228 and antibiotics given as MIC values in μg/mL were used to achieve this goal. For compounds 2228, Figure 3, Figure 4 and Figure 5 clearly indicate that E. coli is more susceptible to compounds 22, 23, and 12, while P. mirabilis is more susceptible to compounds 22 and 24 as is S. pyogenes (see Figure 5). The presence of a para-chlorophenyl moiety at the 2-position of the 1 unit of these compounds appears to be responsible for this effect. The presence of a chlorine atom presumably makes such compounds more active than other compounds against the isolates of the three bacterial species. In case of antibiotics, E. coli tends to be more susceptible to these five antibiotics, and there were few variations among the isolates’ responses to the antibiotics (see Figure 3, Figure 4 and Figure 5).
As previously mentioned, several pyridine compounds have been shown to possess antibacterial activity [29,30]. In the present study, it appears that the use of aminopyridines in an attempt to enhance the antibacterial activity of the targeted compounds 2228 is not encouraging. Recently, there is a growing interest in the use of drugs of choice as quaternary pyridinium salts to improve their solubility and thus to boost the antibacterial activity due to the presence of quaternary ammonium salts [44].
In order to equate the susceptibility of the more active synthesized compounds, which in this case were compounds 22 and 24 with antibiotics against the bacterial species used in the study, the antibiotics exhibited susceptibility against E. coli two-fold greater than compounds 22 and 24, while antibiotics exhibited susceptibility against S. pyogenes two-fold lower than compounds 22 and 24. Antibiotics demonstrated the same activity against P. mirabilis as compounds 22 and 24. The comparison was made using a concentration of 37.5 µg/mL for compounds 22 and 24 close to the contents of antibiotic discs (see Table 3, Figure 6).

3. Materials and Methods

3.1. General Information

Melting points were measured using an open capillary on a Buchi melting point apparatus Buchi labortechnik AG, Essen, Germany, and are uncorrected. All the required chemicals used were purchased from Aldrich (Hamburg, Germany). Thin layer chromatography (TLC) was carried out on 5 × 5 plates coated with silica 0.25 cm N-HR/UV254 obtained from Merck (Dramstadt, Germany). IR spectra were recorded over a frequency range of 4000–400 cm−1 on a FT-IR 8400S spectrophotometer (Shimadzu, Tokyo, Japan). 1H-NMR spectra were recorded on a 300 MHz spectrometer (Tehran, Islamic Republic of Iran) using different solvents and TMS as internal reference Chemical shifts are expressed relative to the internal standard on the δ scale in ppm. Elemental analyses (CHN) were determined on an Eur.Vector EA 3000A system (Rome, Italy). Microwave experiments were conducted using a Microwave Synthesis WorkStation (MAS-II, Microwave Chemistry Technology, Shanghai, China)

3.2. Chemistry

3.2.1. General procedure for the preparation of derivatives 16–21

Following a previous report [32], an equivalent quantity of 2-chloroacetyl chloride (2 g, 0.0176 mol) was added to a stirred and cooled solution of the appropriate secondary amine pyrrolidine, morpholine, piperidine, 2,4-dimethylaminopyridine, 2-aminopyridine, 3-aminopyridine and 4-aminopyridine (0.0176 mol) in dry dichloromethane (12 mL), containing equivalent quantities of triethylamine (0.0178 mol) as a base. The reaction mixtures were then stirred at room temperature for 1–2 h. Reaction progress was tracked via TLC. Once the reactions were complete, aqueous sodium carbonate was added with shaking until the medium became neutral. The organic layers were washed with water separately and dried over anhydrous magnesium sulfate. The solvents were removed under vacuum and the residual materials treated with ether to give crystalline samples of of 2-chloro (pyrrolidinyl, morpholinyl, piperazinyl, pyperidinyl, pyridin-2-yl, pyridin-3-yl, pyridin-4-yl, 4,6-dimethyl pyridine-2-yl) acetamides 1621, and recrystallization from different solvents gave pure derivatives 1621.
2-Chloro-1-(pyrrolidine-1-yl)ethan-1-one (16). Grey crystals, m.p., 143–145 °C; (Yield: 2.8 g, 78%); υmax [KBr]: 2986 cm−1 (C-H aliphatic); 1635 cm−1 (C=O amide); 1452 cm−1 (C-N); 707 cm−1 (C-Cl).
2-Chloro-1-(piperidin-1-yl)ethan-1-one (17). White crystals, m.p., 202–203 °C; (Yield: 2.5 g, 85%); υmax [KBr]: 2945 cm−1 (C-H aliphatic); 1708 cm−1 (C=O amide); 1506 cm−1 (C-N); 746 cm−1 (C-Cl).
2-Chloro-1-morpholinoethan-1-one (18). Brown crystals, m.p., 198–200 °C; (Yield: 2.7 g, 60%); υmax [KBr]: 2975 cm−1 (C-H aliphatic); 1650 cm−1 (C=O amide); 1437 cm−1 (C-N); 732 cm−1 (C-Cl).
2-Chloro-N-(pyridin-3-yl)acetamide (19). Brown crystals, m.p., 196–198 °C; (Yield: 4.1 g, 83%); υmax [KBr]: 3500 cm−1 (N-H amide); 3040 cm−1 (C-H aromatic); 2981 cm−1 (C-H aliphatic); 1654 cm−1 (C=O amide); 1575 cm−1 (C=C); 1477 cm−1 (C-N); 740 cm−1 (C-Cl).
2-Chloro-N-(pyridin-4-yl)acetamide (20). White crystals, m.p., 251–253 °C; (Yield:3.9 g, 80%); υmax [KBr]: 3346 cm−1 (N-H amide); 3051 cm−1 (C-H aromatic); 2941 cm−1 (C-H aliphatic); 1677 cm−1 (C=O amide); 1550 cm−1 (C=C); 1460 cm−1 (C-N); 578 cm−1 (C-Cl).
2-Chloro-N-(4,6-dimethylpyridin-2-yl)acetamide (21). Brown crystals, m.p., 267–278 °C; (Yield: 2.6 g, 90%); υmax [KBr]: 3307 cm−1 (N-H amide); 3074 cm−1 (C-H aromatic); 2947 cm−1 (C-H aliphatic); 1656 cm−1 (C=O amide); 1575 cm−1 (C=C); 1545 cm−1 (C-N); 743 cm−1 (C-Cl).

3.2.2. General Procedure for the Preparation of Derivatives 22–28

(A) Conventional Method
The compounds 2228 synthesized as previously reported [32] by combining equivalent amounts of the acetamide 16 (0.2 g, 0.0013 mol) with substituted para-chloro aniline (0.17 g, 0.0013 mol) in dry acetonitrile (7 mL) containing an equivalent amount of triethylamine as a catalyst. The mixture is heated for 2–3 h at 70 °C, and the progress of reactions tracked by TLC until the starting materials disappeared. Upon reaction completion, the reaction mixture was cooled to room temperature, the solvent was removed under vacuum and the residue taken up in CH2Cl2 (10 mL), treated with aqueous K2CO3 until neutralization, the organic layer washed with water, isolated and dried over anhydrous magnesium sulphate. The solvent was removed in vacuo, and the resulting gummy materials treated with ether, resulting in crystalline derivatives of 22 (see Table 1). Compounds 2328 were synthesized similarly by using equivalent amounts of starting materials.
((4-Chlorophenyl)amino)-1-(3-rrolidine-1-yl)ethan-1-one (22). Grey crystals, m.p., 183–185 °C; (Yield: 0.17 g, 50%); calculated. C, 60.31; H, 6.28; N, 11.76. C12H15N2OCl Founded. C, 60.01; H, 6.02; N, 11.45; υmax [KBr]: 3132 cm−1 (N-H); 3078 cm−1 (C-H aromatic); 2989 cm−1 (C-H aliphatic); 1679 cm−1 (C=O amide); 1583 cm−1 (C=C); 1541 cm−1 (C-N); 690 cm−1 (C-Cl). 1H-NMR (DMSO-d6): δ 7.20–6.75 (4H, dd, J2,3 5.25, J2,6 1.30 Hz, aromatic); 6.32 (1H, s, N-H); 3.36 and 1.60 (8H, 2m, pyrrolidinyl protons); and ppm 3.30 (2H, s, -CH2CO-).
1-(Pyrrolidin-1-yl)-2-tosylethan-1-one (23). Yellow crystals, m.p., 188–189 °C; (Yield: 0.29 g, 85%); calculated. C, 58.42; H, 6.23; N, 5.24. C13H17NO3S Founded. C, 58.10; H, 6.02; N, 5.11; υmax [KBr]: 3068 cm−1 (C-H aromatic); 2931 cm−1 (C-H aliphatic); 1676 cm−1 (C=O amide); 1562 cm−1 (C=C); 1515 cm−1 (C-N); 1321 cm−1; 1153 cm−1; 611 cm−1 (S=O); 1H-NMR (DMSO-d6): δ 7.75–7.42 (4H, dd, J2,3 5.35, J2,6 1.30 Hz, aromatic, tolyl protons); 3.20 (2H, s, -CH2CO-); 2.90 and 1.45 (8H, 2m, pyrrolidinyl protons), 2.35 ppm (3H, s, CH3).
1-(Piperidin-1-yl)-2-tosylethan-1-one (24). White crystals, m.p., 204–207 °C; (Yield: 0.27 g, 78%); calculated. C, 61.17; H, 6.73; N, 11.08. C13H17N2OCl Founded. C, 60.85; H, 6.90; N, 10.30; υmax [KBr]: 3298 cm−1 (N-H); 3031 cm−1 (C-H aromatic); 2977 cm−1 (C-H aliphatic); 1647 cm−1 (C=O amide); 1515 cm−1 (C=C); 1460 cm−1 (C-N); 1357 cm−1; 1174 cm−1; 580 cm−1 (S=O); 1H-NMR (DMSO-d6): δ 7.20–6.70 (4H, dd, J2,3 5.30, J2,6 1.45 Hz, aromatic); 6.35 (1H, s, N-H); 3.20 (2H, s, -CH2CO-); 2.90 and 1.45 ppm (10H, 2m, piperidinyl protons).
1-Morpholino-2-tosylethan-1-one (25). White crystals, m.p., 170–172 °C; (Yield: 0.31 g, 90%); calculated. C, 55.12; H, 6.0; N, 4.94. C13H17NO4S Founded. C, 54.90; H, 6.10; N, 5.08; υmax [KBr]: 3099 cm−1 (C-H aromatic); 2974 cm−1 (C-H aliphatic); 1679 cm−1 (C=O amide); 1581 cm−1 (C=C); 1539 cm−1 (C-N); 1247 cm−1; 1172 cm−1; 584 cm−1 (S=O); 1H-NMR (DMSO-d6): δ 7.75–7.42 (4H, dd, J2,3 5.25 Hz, J2,6 1.4 Hz, tolyl protons), 4.15 (2H, 2, -CH2CO-); 3.70 and 2.50 (8H, mm, morpholinyl protons), 2.40 ppm (3H, s, CH3).
N-(Pyridin-3-yl)-2-tosylacetamide (26). White crystals, m.p., 204–207 °C; (Yield: 0.27 g, 78%); calculated. C, 57.93; H, 4.82; N, 9.65. C14H14N2O3S Founded. C, 58.20; H, 5.02; N, 9.50; υmax [KBr]: 3298 cm−1 (N-H); 3031 cm−1 (C-H aromatic); 2977 cm−1 (C-H aliphatic); 1647 cm−1 (C=O amide); 1515 cm−1 (C=C); 1460 cm−1 (C-N); 1357 cm−1; 1174 cm−1; 580 cm−1 (S=O); 1H-NMR (DMSO-d6): δ 10.61 ppm (N-H, s); δ 10.65 (1H, s, N-H); 9.0 (1H, s, pyridinyl proton); 8.41–7.10 (3H, dd, J4,5 5.31, J5,6 5.25 Hz, J2,6 1.5 Hz, pyridinyl proton); 7.70–7.35 (4H, dd, J2,3 5.25 Hz, tolyl protons); 4.20 (2H, s, -CH2CO-); and 2.35 ppm (3H, s, CH3).
N-(Pyridin-4-yl)-2-tosylacetamide (27).White crystals, m.p., 251–253 °C; (Yield: 0.26 g, 76%); calculated. C, 57.93; H, 4.82; N, 9.65. C14H14N2O3S Founded. C, 57.60; H, 5.0; N, 9.61; υmax [KBr]: 3344 cm−1 (N-H); 3080 cm−1 (C-H aromatic); 2977 cm−1 (C-H aliphatic); 1639 cm−1 (C=O amide); 1531 cm−1 (C=C); 1461 cm−1 (C-N); 1365 cm−1; 1103 cm−1; 557 cm−1 (S=O); 1H-NMR (DMSO-d6): δ 9.90 (1H, s, N-H); δ 8.41–7.91(4H, dd, J2,3 5.31 Hz, J2,6 1.5 Hz, pyridinyl protons); δ 7.65–6.40 (4H, dd, J2,3 5.20 Hz, J2,6 1.5 Hz, tolyl protons); δ 4.25 (2H, s, -CH2CO-); and 2.32 ppm (3H, s, CH3).
N-(4,6-Dimethylpyridin-2-yl)-2-tosylacetamide (28). Brown crystals, m.p., 212–213 °C; (Yield: 0.21 g, 62%); calculated. C, 60.37; H, 5.66; N, 8.80. C16H18N2O3S Founded. C, 60.57; H, 5.42; N, 8.91; υmax [KBr]: 3463 cm−1 (N-H); 3085 cm−1 (C-H aromatic) ; 2945 cm−1 (C-H aliphatic); 1677 cm−1 (C=O amide); 1596 cm−1 (C=C); 1544 cm−1 (C-N); 1336 cm−1; 1150 cm−1; 524 cm−1 (S=O); 1H-NMR (DMSO-d6): δ 10.40 (1H, s, N-H); 8.30–7.40 (2H, 2s, pyridinyl protons); 7.60–7.45 (4H, dd, J2,3 5.25 Hz, J2,6 1.55 Hz, tolyl protons); 4.25 (2H, s, -CH2CO-); and 2.45–2.35 ppm (9H, s, 3CH3).
(B) Microwave Method
The compounds 2228 synthesized as previously stated [32] by combining the equivalent quantities mentioned in the above experiments, substituted aniline and para-toluene sulfonate sodium salt with α-chloroacetamides 1621 in dry acetonitrile (7 mL) containing equivalent quantities of triethylamine as a catalyst. The mixture was heated at 65–70 °C under 400 Watt microwave irradiation for 5–10 min. The reaction progress was monitored with TLC until the starting materials had disappeared. After the reaction was completed, the mixture was cooled to room temperature, the solvent removed under vacuum, and the residue taken in CH2Cl2 (10 mL) and treated with aqueous K2CO3 until neutralization, then the organic layer was washed with water, separated and dried over anhydrous magnesium sulfate. The solvent was removed under vacuum, and the resulting gummy products 2228 treated with ether to give crystalline products 2228 (See Table 1).

3.3. Antibiotics

Ready-impregnated antibiotic disks of amoxicillin (20 mcg), ampicillin (25 mcg), cephalothin (30 mcg), azithromycin (15 mcg) and doxycycline (30 mcg) were obtained from Samarra Drug Industries, (Samarra, Iraq) and were used as standard broad-spectrum antibacterial agents in the disk diffusion method.

3.4. Bacterial Species

Isolates of the three different bacterial species, namely, two Gram-negative bacteria (E. coli plus P. mirabilis) and one Gram-positive bacterium (S. pyogenes) were collected from the Tikrit Teaching Hospital (Tikrit, Iraq). All the isolates of E. coli and P. mirabilis were collected from urine samples of urinary tract infection (UTI) patients and the isolates of S. pyogenes were collected from tonsillitis patients.

3.5. Antibacterial Susceptibility Testing

Antibacterial resistance to seven new synthesized compounds 2228 of the acetamide class and five antibiotics, including amoxicillin, ampicillin, cephalothin, azithromycin and doxycycline, was evaluated using the standard disk diffusion method [45] and the diffusion method (well diffusion) for synthesized compounds 2228. Both studied bacterial species were pre-cultivated on 24 h incubated nutrient agar plates and bacterial suspensions of each pure isolate were prepared for in vitro antibacterial treatment in 0.6 McFarland turbidity nutrient broth tubes. Mueller Hinton agar plates (Oxoid Ltd., Hampshire, UK), 12 cm in diameter, were prepared as directed by the manufacturer and incubated at 37 °C for 24 h. So, a sterile borer was used to make equidistant wells with a diameter of 6 mm. 50 mg of each of the synthetic compounds 2228 was dissolved in DMSO (1 mL), followed by dilution of 2.5, 5, 15, 25, 35, 45 and 50 μL of each to 1 mL DMSO to obtain concentrations of 0.125, 0.250, 0.750, 1.25, 1.75, 2.25 and 2.5 mg/mL which equivalent to 6.25, 12.5, 37.5, 62.5, 87.5, 112.5 and 125 μg/50, respectively. 50 μL of each of the later concentrations were used to assess antibacterial susceptibility and minimum inhibitory concentrations (MICs) by filling the growth media wells with it accompanied by 24-h incubation at 37 °C and growth inhibition monitor in. DMSO did not display any inhibition of bacterial growth.

3.6. Analysis of Results

SPSS software (version 20, IBM, Armonk, NY, USA, www.ibm.com/software/analytics/spss) was used to evaluate the effects of the antimicrobial susceptibility study. Mean values and standard deviations for the inhibition zone diameters were determined. The findings were presented as average values ± SD. Statistically standard deviation, variations are calculated relative to normal antibiotics and the amount of dispersion between them.

4. Conclusions

2-Chloroacetamides are versatile intermediates in organic synthesis. The reported synthesis method is based primarily on chloroacetylation of amines by chloroacetyl chloride. The simple replacement of a chlorine atom allowed us to prepare many acetamide derivatives through the reaction with amines and para-tosyl sodium salt. A series of new acetamide derivatives were successfully synthesized by adding primary and secondary amines to chloroacetyl chloride 15. With the help of microwave irradiation, we have been able to synthesize seven compounds in an attempt to increase yields and reduce the reaction time. Moderate to good yields and reduction of the reaction time from 2–3 h to a few minutes were achieved. The application of the compounds 2228 against Gram-positive and Gram-negative bacterial species demonstrated encouraging relatively good antibacterial potency in comparison with the used reference antibiotics. Isolates of two of the tested species, namely, E. coli and P. mirabilis displaced higher susceptibility than S. pyogenes. The results show that among synthetic compounds 2228 compounds 22 and 24 present the most important antibacterial properties bearing para-chlorophenyl moiety in the acetamide 2-position of moiety 1. We have successfully developed a synthetic method has proven to be a fast, environmentally friendly technique with moderate to good performance in microwave irradiation, and high acceleration of reaction rates has been achieved in the presence of Et3N as a base.

Supplementary Materials

The following are available online. Table S1, Table S2, and Table S3.

Author Contributions

Conceptualization, A.S.H.A.; methodology, S.S.A., S.S.A.; validation, A.S.H.A., S.S.A.; formal analysis A.S.H.A., S.S.A.; investigation, A.S.H.A.; resources, S.S.A.; data curation S.S.A,; writing—original draft preparation, A.S.H.A.; writing—review and editing A.S.H.A; visualization, A.S.H.A.; supervision, A.S.H.A.; project administration, A.S.H.A.; funding acquisition, A.S.H.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available in article and Supplementary Material.

Acknowledgments

The authors thank the Department of Chemistry, College of Applied Science, Samarra University, Iraq, for supporting facilities. The authors also thank Maroof S. Juma for technical assistance and Faesal G. Hussein for helpful comments.

Conflicts of Interest

The authors declare no conflict of interest.

Sample Availability

Samples of the compounds are not available from the authors.

References

  1. Lu, H.; Zhou, X.; Wang, L.; Jin, L. Synthesis and Antibacterial Evaluation of N-phenylacetamide Derivatives Containing 4-Arylthiazole Moieties. Molecules 2020, 25, 1772. [Google Scholar] [CrossRef] [PubMed]
  2. Ahmed, H.E.; Ihmaid, S.; Omar, A.M.; Shehata, A.M.; Rateb, H.S.; Zayed, M.F.; Ahmed, S.; Elaasser, M.M. Design, synthesis, molecular docking of new lipophilic acetamide derivatives affording potential anticancer and antimicrobial agents. Bioorg. Chem. 2018, 76, 332–342. [Google Scholar] [CrossRef] [PubMed]
  3. Ghorab, M.M.; Alqahtani, A.S.; Soliman, A.M.; Askar, A.A. Novel N-(Substituted) Thioacetamide Quinazolinone Benzenesulfonamides as Antimicrobial Agents. Int. J. Nanomed. 2020, 15, 3161–3180. [Google Scholar] [CrossRef] [PubMed]
  4. Kaplancıklı, Z.A.; Altintop, M.D.; Turan-Zitouni, G.; Ozdemir, A.; Ozic, R.; Akalın, G. Synthesis, antimicrobial activity and cytotoxicity of novel oxadiazole derivatives. J. Enzym. Inhib. Med. Chem. 2011, 27, 51–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Altintop, M.D.; Abu Mohsen, U.; Okay, Y.; Demirel, R.; Kaplancikli, Z.A. Synthesis and antimicrobial activity of benzimidazole-base acetamide derivatives. Turk. J. Pharm. Sci. 2015, 12, 29–38. [Google Scholar]
  6. Zogota, R.; Kinena, L.; Withers-Martinez, C.; Blackman, M.J.; Bobrovs, R.; Pantelejevs, T.; Kanepe-Lapsa, I.; Ozola, V.; Jaudzems, K.; Suna, E.; et al. Peptidomimetic plasmepsin inhibitors with potent anti-malarial activity and selectivity against cathepsin D. Eur. J. Med. Chem. 2019, 163, 344–352. [Google Scholar] [CrossRef]
  7. Li, L.; Zhao, P.; Hu, J.; Liu, J.; Liu, Y.; Wang, Z.; Xia, Y.; Dai, Y.; Chen, L. Synthesis, in vitro and in vivo antitumor activity of scopoletin-cinnamic acid hybrids. Eur. J. Med. Chem. 2015, 93, 300–307. [Google Scholar] [CrossRef]
  8. Naim, M.J.; Alam, M.J.; Nawaz, F.; Naidu, V.G.M.; Aaghaz, S.; Sahu, M.; Alam, O. Synthesis, molecular docking and anti-diabetic evaluation of 2,4-thiazolidinedione based amide derivatives. Bioorg. Chem. 2017, 73, 24–36. [Google Scholar] [CrossRef]
  9. Tanwar, B.; Kumar, A.; Yogeeswari, P.; Sriram, D.; Chakraborti, A.K. Design, development of new synthetic methodology, and biological evaluation of substituted quinolines as new anti-tubercular leads. Bioorg. Med. Chem. Lett. 2016, 26, 5960–5966. [Google Scholar] [CrossRef]
  10. Kouatly, O.; Geronikaki, A.; Kamoutsis, C.; Hadjipavlou-Litina, D.; Eleftheriou, P. Adamantane derivatives of thiazolyl-N-substituted amide, as possible non-steroidal anti-inflammatory agents. Eur. J. Med. Chem. 2009, 44, 1198–1204. [Google Scholar] [CrossRef]
  11. Wang, M.; Song, X.; Jiang, J.; Xia, J.; Li, M. Binary amide-containing tung-oil-based Ca/Zn stabilizers: Effects on thermal stability and plasticization performance of poly(vinyl chloride) and mechanism of thermal stabilization. Polym. Degrad. Stab. 2017, 143, 106–117. [Google Scholar] [CrossRef]
  12. Coleman, E.A. Applied Plastics Engineering Handbook, Plastics Additives, 2nd ed.; Elsevier: Amsterdam, The Netherlands, 2017; pp. 489–500. [Google Scholar]
  13. Uppu, D.S.; Samaddar, S.; Ghosh, C.; Paramanandham, K.; Shome, B.R.; Haldar, J. Amide side chain amphiphilic polymers disrupt surface established bacterial bio-films and protect mice from chronic Acinetobacter baumannii infection. Biomaterials 2016, 74, 131–143. [Google Scholar] [CrossRef] [PubMed]
  14. Kojima, N.; Maruko, D. Cleaning Flux, Cleaning Solder Paste, and Solder Joint. U.S. Patent 10,259,083, 16 April 2019. [Google Scholar]
  15. Ribeiro, R.F.; Pardini, L.C.; Alves, N.P.; Júnior, C.A.R.B. Thermal Stabilization study of polyacrylonitrile fiber obtained by extrusion. Polímeros 2015, 25, 523–530. [Google Scholar] [CrossRef] [Green Version]
  16. Zannikos, F.; Lois, E.; Stournas, S. Desulfurization of petroleum fractions by oxidation and solvent extraction. Fuel Process. Technol. 1995, 42, 35–45. [Google Scholar] [CrossRef]
  17. Zarganes-Tzitzikas, T.; Neochoritis, C.G.; Dömling, A. Atorvastatin (Lipitor) by MCR. ACS Med. Chem. Lett. 2019, 10, 389–392. [Google Scholar] [CrossRef] [Green Version]
  18. Fabbri, L.M.; Calverley, P.M.; Izquierdo-Alonso, J.L.; Bundschuh, D.S.; Brose, M.; Martinez, F.J.; Rabe, K.F. Roflumilast in moderate-to-severe chronic obstructive pulmonary disease treated with longacting bronchodilators: Two randomised clinical trials. Lancet 2009, 374, 695–703. [Google Scholar] [CrossRef]
  19. Gupta, K.; Mitra, S.; Kazal, S.; Saroa, R.; Ahuja, V.; Goel, P. IV paracetamol as an adjunct to patient-controlled epidural analgesia with levobupivacaine and fentanyl in labour: A randomized controlled study. Br. J. Anaesth. 2016, 117, 617–622. [Google Scholar] [CrossRef] [Green Version]
  20. Karaman, M.; Budak, H.; Ciftci, M. Amoxicillin and gentamicin antibiotics treatment adversely influence the fertility and morphology through decreasing the Dazl gene expression level and increasing the oxidative stress. Arch. Physiol. Biochem. 2018, 125, 447–455. [Google Scholar] [CrossRef]
  21. Kilic, M.; Seyhan, T.O.; Sungur, M.O.; Ekiz, N.; Bastu, E.; Senturk, M. The effects of subfascial wound versus epidural levobu-pivacaine infusion on postoperative pain following hysterectomy. Minerva Anestesiol. 2014, 80, 769–778. [Google Scholar]
  22. Siddiqui, A.M.; Sattigeri, J.A.; Javed, K.; Shafi, S.; Shamim, M.; Singhal, S.; Malik, Z.M. Design, synthesis and biological evaluation of spiropyrimidinetriones oxazolidinone derivatives as antibacterial agents. Bioorg. Med. Chem. Lett. 2018, 28, 1198–1206. [Google Scholar] [CrossRef]
  23. Al-Abdullah, E.S.; Al-Tuwaijri, H.M.; Hassan, H.M.; Al-Alshaikh, M.A.; Habib, E.-S.E.; El-Emam, A.A. Synthesis, Antimicrobial and Hypoglycemic Activities of Novel N-(1-Adamantyl)carbothioamide Derivatives. Molecules 2015, 20, 8125–8143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Wyrzykiewicz, E.; Wendzonka, M.; Kedzia, B. Synthesis and antimicrobial activity of new (E)-4-[piperidino (4′-methylpiperidino-, morpholino-) N-alkoxy]stilbenes. Eur. J. Med. Chem. 2006, 41, 519–525. [Google Scholar] [CrossRef] [PubMed]
  25. Arslan, S.; Loğoğlu, E.; Öktemer, A. Antimicrobial activity studies on some piperidine and pyrrolidine substituted halogenobenzene derivatives. J. Enzym. Inhib. Med. Chem. 2006, 21, 211–214. [Google Scholar] [CrossRef] [PubMed]
  26. Kim, H.Y.; Lee, J.S.; Cha, J.H.; Pae, A.N.; Cho, Y.S.; Chang, M.H.; Koh, H.Y. Synthesis and in vitro activity of new methylenepiperidinyl and methylenepyrrolidinyl oxazolidinone antibacterial agents. Bioorg. Med. Chem. Lett. 2003, 13, 2227–2230. [Google Scholar] [CrossRef]
  27. Kamiński, K.; Obniska, J.; Wiklik, B.; Atamanyuk, D. Synthesis and anticonvulsant properties of new acetamide derivatives of phthalimide, and its saturated cyclohexane and norbornene analogs. Eur. J. Med. Chem. 2011, 46, 4634–4641. [Google Scholar] [CrossRef]
  28. Kumar, K.N.; Sreeramamurthy, K.; Palle, S.; Mukkanti, K.; Das, P. Dithiocarbamate and DBU-promoted amide bond formation under microwave condition. Tetrahedron Lett. 2010, 51, 899–902. [Google Scholar] [CrossRef]
  29. Nakamoto, K.; Tsukada, I.; Tanaka, K.; Matsukura, M.; Haneda, T.; Inoue, S.; Murai, N.; Abe, S.; Ueda, N.; Miyazaki, M.; et al. Synthesis and evaluation of novel antifungal agents-quinoline and pyridine amide derivatives. Bioorg. Med. Chem. Lett. 2010, 20, 4624–4626. [Google Scholar] [CrossRef]
  30. Tang, J.; Wang, B.; Wu, T.; Wan, J.; Tu, Z.; Njire, M.; Wan, B.; Franzblauc, S.G.; Zhang, T.; Lu, X.; et al. Design, Synthesis, and Biological Evaluation of Pyrazolo[1,5-a]pyridine-3-carboxamides as Novel Antitubercular Agents. ACS Med. Chem. Lett. 2015, 6, 814–818. [Google Scholar] [CrossRef] [Green Version]
  31. Oliver, K.C. Microwave dielectric heating in synthetic organic chemistry. Chem. Soc. Rev. 2008, 37, 1127–1139. [Google Scholar] [CrossRef]
  32. Alsamarrai, A.S.H.; Abdulla, N.H.; Aldoori, M.K. Synthesis and Characterization of yl)-N-(Substituted Phenyl) Acetamides Derivatives Anticipated to Inhibit HIV-1 Activity. Pharm. Phytopharm. Res. (eIJPPR) 2018, 8, 7–11. [Google Scholar]
  33. Chari, M.A.; Syamasundar, K. Polymer (PVP) supported ferric chloride: An efficient and recyclable heterogeneous catalyst for high yield synthesis of 1,5-benzodiazepine derivatives under solvent free conditions and microwave irradiation. Catal. Commun. 2005, 6, 67–70. [Google Scholar] [CrossRef]
  34. De la Hoz, A.; Diaz-Ortiz, A.; Moreno, A. Microwaves in organic synthesis. Thermal and non-thermal microwave effects. Chem. Soc. Rev. 2005, 34, 164–178. [Google Scholar] [CrossRef] [PubMed]
  35. Özden, S.; Atabey, D.; Yıldız, S.; Göker, H. Synthesis and potent antimicrobial activity of some novel methyl or ethyl 1H-benzimidazole-5-carboxylates derivatives carrying amide or amidine groups. Bioorg. Med. Chem. 2005, 13, 1587–1597. [Google Scholar] [CrossRef] [PubMed]
  36. Andrews, J.M. BSAC standardized disc susceptibility testing method (version 4). J. Antimicrob. Chemother. 2005, 56, 60–76. [Google Scholar] [CrossRef] [Green Version]
  37. Clinical and Laboratory Standards Institute. M100. Performance Standards for Antibacterial Testing, 27th ed.; CLSI: Wayne, PA, USA, 2017. [Google Scholar]
  38. Trivedi, M.K.; Branton, A. Antimicrobial Susceptibility of Proteus mirabilis: Impact of Biofield Energy Treatment. J. Microb. Biochem. Technol. 2015, 8, 25–29. [Google Scholar] [CrossRef] [Green Version]
  39. Giovanetti, E.; Prenna, M.; Repetto, A.; Biavasco, F.; Romagnoli, M.; Ripa, S.; Varaldo, P.E. Susceptibility of Streptococcus pyogenes from throat cultures to macrolide antibiotics and influence of collection criteria. Clin. Microbiol. Infect. 1997, 3, 58–62. [Google Scholar] [CrossRef]
  40. Benouda, A.; Sibile, S.; Ziane, Y.; Elouennass, M.; Dahani, K.; Hassani, A. Place of Streptococcus pyogenes in the throat infection in Morocco and overview of its susceptibility to antibiotics. Pathol. Biol. 2009, 57, 76–80. [Google Scholar] [CrossRef]
  41. Nakae, M.; Murai, T.; Kaneko, Y.; Mitsuhashi, S. Drug Resistance in Streptococcus pyogenes Isolated in Japan (1974–1975). Antimicrob. Agents Chemother. 1977, 12, 427–428. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Silva-Costa, C.; Ramiirez, M.; Melo-Crisino, J. Portuguense by a diversification of T and emm types among streptococcus pyogenes in Portugal. Antimicrob agents Chemther. Surveillance group for the study of respiratory pathogens. Rapid Invers. Preval. Macrolide Resist. Phenotype Paralleled 2005, 49, 2109–2111. [Google Scholar]
  43. Camara, M.; Dieng, A.; Boye, C.S.B. Antibiotic Susceptibility ofStreptococcus PyogenesIsolated from Respiratory Tract Infections in Dakar, Senegal. Microbiol. Insights 2013, 6, 71–75. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Li, L.; Zhao, Y.; Zhou, H.; Ning, A.; Zhang, F.; Kent, Z.Z. Synthesis of pyridinium N-chloramines for antibacterial applications. Tetrahedron Lett. 2017, 58, 321–325. [Google Scholar] [CrossRef]
  45. Hoelzer, K.; Cummings, K.J.; Warnick, L.D.; Schukken, Y.H.; Siler, J.D.; Gröhn, Y.T.; Davis, M.A.; Besser, T.E.; Wiedmann, M. Agar Disk Diffusion and Automated Microbroth Dilution Produce Similar Antimicrobial Susceptibility Testing Results forSalmonellaSerotypes Newport, Typhimurium, and 4,5,12:i-, But Differ in Economic Cost. Foodborne Pathog. Dis. 2011, 8, 1281–1288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. The most widely used drugs in medicine.
Figure 1. The most widely used drugs in medicine.
Molecules 26 00533 g001
Figure 2. Some biologically active molecules containing pyridines.
Figure 2. Some biologically active molecules containing pyridines.
Molecules 26 00533 g002
Scheme 1. Synthetic pathway to compounds 2228.
Scheme 1. Synthetic pathway to compounds 2228.
Molecules 26 00533 sch001
Figure 3. Antibacterial susceptibility of species E. coli given as MICs values in µg/mL for compounds 2228.
Figure 3. Antibacterial susceptibility of species E. coli given as MICs values in µg/mL for compounds 2228.
Molecules 26 00533 g003
Figure 4. Antibacterial susceptibility of species P. mirabilis given as MICs values in µg/mL for compounds 2228.
Figure 4. Antibacterial susceptibility of species P. mirabilis given as MICs values in µg/mL for compounds 2228.
Molecules 26 00533 g004
Figure 5. Antibacterial susceptibility of species S. pyogenes given as MICs values in µg/mL for compounds 2228.
Figure 5. Antibacterial susceptibility of species S. pyogenes given as MICs values in µg/mL for compounds 2228.
Molecules 26 00533 g005
Figure 6. Antibacterial susceptibility of some species bacteria given as MICs values in µg/mL for referenced antibiotics.
Figure 6. Antibacterial susceptibility of some species bacteria given as MICs values in µg/mL for referenced antibiotics.
Molecules 26 00533 g006
Table 1. Melting points, colours, solvents for recrystallization, and yields of the compounds 1628.
Table 1. Melting points, colours, solvents for recrystallization, and yields of the compounds 1628.
Comp. No.Recryst. SolventColorM. P.
°C
Yield% of Conventional MethodYield% of Microwave Method
16n-HexaneGray143–14578
17EthanolWhite202–20385
18EthanolBrown276–27860
19n-Hexane:methanol
1:3
Brown196–19883
20Ethyl acetate:acetone
1:1
White251–25380
21n-HexaneBrown198–20090
22n-HexaneGray183–1852450
23EthanolYellow188–1895585
24n-Hexane:ethanol
1:3
White197–1985578
25Ethyl acetate:ethanol
1:1
White170–1723090
26Methanolwhite204–2075378
27n-Hexane:methanol
1:1
White251–2536076
28EthanolBrown212–2135062
Table 2. 1H-NMR data of compounds 2228.
Table 2. 1H-NMR data of compounds 2228.
StructureChemical Shift
(δ) ppm
Signal FeaturesNo. of ProtonsType of Protons
227.20–6.75
6.32
3.36 and 1.75
3.30
dd, J2,3 5.25 Hz, J2,6 1.30 Hz
s
2m
s
4H
1H
8H
2H
aromatic
NH
pyrrolidinyl proton
-CH2CO-
237.75–7.42
4.20
2.85 and 1.90
2.35
dd, J2,3 5.35 Hz, J2,6 1.30 Hz
s
2m
s
4H
2H
8H
3H
tolyl
-CH2CO-
pyrrolidinyl proton
CH3
247.20–6.70
6.35
3.20
2.90 and 1.45
dd, J2,3 5.0 Hz, J2,6 1.45 Hz
s
s
2 m
4H
1H
2H
10H
aromatic
NH
-CH2CO-
piperidinyl proton
257.75–7.42
4.15
3.70 and 2.50
2.40
dd, J2,3 5.26 Hz, J2,6 1.4 Hz
s
2t
s
4H
2H
8H
3H
tolyl protons
-CH2CO
morpholinyl protons
CH3
2610.65
9.0
8.41–7.10
7.70–7.35
4.20
2.35
s
s
dd, J4,5 5.31 Hz, J5,6 5.25 Hz
dd, J2,3 5.25 Hz, J2,6 1.5 Hz
s
s
1H
1H
3H
4H
2H
3H
NH
pyridinyl proton
pyridinyl protons
tolyl protons
CH2
CH3
279.90
8.41–7.91
7.65–6.40
4.25
2.32
s
dd, J2,3 5,31 Hz., J2,6 1.45 Hz
dd, J2,3 5.20, J2,6 1.5 Hz
s
s
1H
4H
4H
2H
3H
NH
pyridinyl protons
tolyl protons
CH2
CH3
2810.40
8.30–7.40
7.60–7.45
4.25
2.45–2.35
s
2s
dd, J2,3 5.26 Hz, J2,6 1.55 Hz
s
3s
1H
2H
4H
2H
9H
NH
pyridinyl proton
tolyl protons
CH2
3CH3
Table 3. Inhibition zone diameters (mm) of reference antibiotics and synthesized compounds 22 and 24.
Table 3. Inhibition zone diameters (mm) of reference antibiotics and synthesized compounds 22 and 24.
AmxAziDoxyCepAm2224
20 µg15 µg30 µg30 µg25 µg37.5 µg/mL37.5 µg/mL
E. coli12.317.019.013.516.07.75.8
P. mirabilis12.811.013.011.07.514.212.9
S. pyogenes5.86.57.37.66.415.013.0
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Alsamarrai, A.S.H.; Abdulghani, S.S. Microwave-Assisted Synthesis, Structural Characterization and Assessment of the Antibacterial Activity of Some New Aminopyridine, Pyrrolidine, Piperidine and Morpholine Acetamides. Molecules 2021, 26, 533. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules26030533

AMA Style

Alsamarrai ASH, Abdulghani SS. Microwave-Assisted Synthesis, Structural Characterization and Assessment of the Antibacterial Activity of Some New Aminopyridine, Pyrrolidine, Piperidine and Morpholine Acetamides. Molecules. 2021; 26(3):533. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules26030533

Chicago/Turabian Style

Alsamarrai, Abdulmajeed S. H., and Saba S. Abdulghani. 2021. "Microwave-Assisted Synthesis, Structural Characterization and Assessment of the Antibacterial Activity of Some New Aminopyridine, Pyrrolidine, Piperidine and Morpholine Acetamides" Molecules 26, no. 3: 533. https://0-doi-org.brum.beds.ac.uk/10.3390/molecules26030533

Article Metrics

Back to TopTop