Skip to main content

Synthesis, biological activity of newly designed sulfonamide based indole derivative as anti-microbial agent

Abstract

Background

In medicinal chemistry, indole and its derivative play an important role. Indole is gaining a lot of importance in medicinal chemistry due to its physiological activity which includes anticancer, antitubercular, antimicrobial, antiviral, antimalarial, anti-inflammatory activities, antileishmanial agents, anti-cholinesterase, and enzyme inhibitory. The spread of antimicrobial resistance becomes a threat to both humans and animals. Antimicrobial resistance has been declared in the top 10 global major health risks by WHO including reported data of 2020 of AMR with 3,106,002 confirmed infections in humans across 70 countries.

Result

In this present work some new sulfonamide-based indole derivatives were synthesized by using 1H-indole -2 carboxylic acid as a starting material. The structure of all synthesized sulfonamide-based indole derivatives was confirmed by 1H NMR and LCMS Spectroscopy.

Conclusion

All the synthesized compounds were screened for anti-microbial activity against Gram Positive Staphylococcus aureus, Bacillus megaterium, and Gram Negative Klebsiella pneumonia, Escherichia coli, Salmonellatyphiae, Shigella sp., Enterobacter aerogenes. Among gram-positive Staphylococcus aureus, and Bacillus megaterium. The compound shows activity against Staphylococcus aureus, and among all gram-negative bacteria against Klebsiella pneumonia shows good activity.

Background

Antimicrobial agents are essentially important because of massive microbial infections in today’s world [1, 2]. The spread of antimicrobial resistance becomes a threat to both humans and animals [3, 4]. Antimicrobial resistance has been declared in the top 10 global major health risks by WHO including reported data of 2020 of AMR with 3,106,02 confirmed infections in humans across 70 countries [5, 6]. When the microorganism broadens to capacity to defect drug design to kill microbes that time Antimicrobial resistance occurs [7, 8]. We can reduce antimicrobial infection by the proper use of antimicrobials, selecting the required antimicrobial drug regimen, and the proper dose of drugs [9, 10].

Heterocyclic chemistry plays an important role in drug design because a wide range of heterocyclic compounds are clinically active, one of them is “Indole” [11, 12]. Baeyar and knop first prepare indole in 1866 when they were studying the indigo dye [13]. To prepare Indole Baeyar reduced oxindole [14]. The formula of indole is “C8H7N” is also known as 1H-benzo[b] pyrrole [15]. The emergence of indole is a colorless crystalline solid with a melting point of 52 °C and a boiling point 253 °C at 762 mm [16]. Indole contains a six-membered benzene ring which is fused to a five-membered pyrrole ring containing nitrogen giving it a bicyclic structure [17]. Indole is gaining a lot of importance in medicinal chemistry due to their physiological activity which includes anti-cancer [18, 19], antitubercular [20, 21], antimicrobial [22, 23], antiviral [24, 25], antimalarial [26, 27], anti-inflammatory activities [28, 29], antileishmanial agents [30, 31], anti-cholinesterase [32, 33], enzyme inhibitory [34, 35]. The important medicinal activities of indole are shown in Fig. 1.

Fig. 1
figure 1

Different medicinal activity of indole

Sulfonamides act as important as biologically active in pharmaceuticals. The basic skeleton of sulfonamide moiety generates sulpha drugs or sulfa drugs [36]. The more potency showed by the sulfonamide functional group help in producing new action toward biologically active scaffolds [37]. The aim of having the best or more potent anti-microbial compounds were reported by substituting the chemistry of sulfonamidee [38] with indole 2-carboxylic acid which provides pharmacological properties and significant biological properties.

In 2022, Yuanyuan and co-workers synthesized para amino benzene sulfonamide indole hybrids and screened them for antibacterial activities against staphylococcus aureus and some of the compounds show good activity against staphylococcus aureus [39]. In 2022, S. jagadeesan and S. Karpagam designed a novel series of N-acyl substituted indole-based piperidine, thiazole, and tetrazoles, and after the synthesis, they screened compounds against Klebsiella and Escherichia coli and all compounds give excellent activity against both bacteria [40]. In 2021, Reem I. Al-wabli and co-workers synthesized new indole-triazole conjugates and screened them for antimicrobial resistance. All the assisted compounds were found as good antimicrobial agents [41]. Synthesis and antimicrobial activity of 3-(1H-indole-3-carbonyl)-2H-chromen-2-one derivatives done by Khadhar Navaz Umar Bashat etc. in 2021. Some of the compounds show potent antimicrobial activity [42]. In 2020, Andre Nehemine Bitombo reported indole alkaloids as a potent antimicrobial agents [43]. In this current work, we synthesized a derivative of Indole -2 carboxylic acid and all the compounds were evaluated for their antimicrobial activity against staphylococcus aureus, Pseudomonas aeruginosa, Escherichia coli, Klebsiella.

Here, we aim to facilitate the current research in designing innovative strategies for the discovery and development of good antimicrobial agents.

Method

Materials

All the chemicals were used without any further purification obtained from sigma Aldrich as well all the solvents were also purchased from sigma Aldrich. Thin layer chromatography (TLC) was performed by using Elutes the gradient of MeOH in MDC on silica gel plates (60F254, 0.2 mm thick, Merck). By taking tetra methyl silane (TMS) as internal standard 1H NMR spectra were carried out in CdCl3 solutions at 400 MHz by Bruker Advance II 400 NMR spectrometer. The δ value in 1 H NMR is calculated in ppm (parts per million).

LCMS data were recorded by WATERS instrument in which 0.15% formic acid in acetonitrile was used as the mobile phase.

Chemistry

As a precursor, commercially available N- unsubstituted indole 2-carboxylic acid (1) was used. Di-methylation at the indole site of N- unsubstituted indole 2-carboxylic acid(1) in presence of MeI and K2CO3 lead to the preparation of N-methylated indole ester(2) [44], which undergoes deprotection with lithium hydroxide water (LiOH.H2O) in presence of THF yields 1-methyl-1H-indole-2-carboxylic acid(3) [45]. 1-methyl-1H-indole-2-carboxylic acid(3) further reacts with commercially available reagent 4-boc pieridine amine(4) and a debocking reaction is carried out in presence of EDCl.HCl and HoBt with DIPEA which results (4-aminopiperidin-1-yl)(1-methyl-1H-indol-2-yl)methanone(5) [46].

In Scheme 1 synthesis route of compounds (6a-6o) was shown. All these compounds were derived from (4-aminopiperidin-1-yl) (1-methyl-1H-indol-2-yl) methanone(5) in the presence of various aldehydes via reductive amination in the presence of reducing agent NaBH4 and PTSA with ethanol. All the attached aldehydes are mentioned in Table 1. The synthetic route was shown to afford a series of substituted sulfonamide-based indole derivatives (7a-7o) with different substituents (6a-6o) that were reacted with methyl sulfonamide chloride in the presence of TEA to furnish the sulfonamide-based indole derivatives [47].

Scheme 1
figure 1

Synthetic route of Sulfonamide-based indole derivatives (R = Aldehyde used)

Table 1 Properties of compounds

Anti-microbial activity

The in vitro antimicrobial potential of new indole derivatives synthesized listed in Table 1 were primarily screened against model Gram-positive bacteria (Staphylococcus aureus and Bacillus megaterium) and Gram-negative bacteria (Escherichia coli, Klebsiella pneumoniae, Salmonella typhiae, Shigella sp., and Enterobacter aerogenes). The antibacterial activities of the tested compounds were evaluated by the agar well diffusion method. The solution of each compound is prepared in dimethyl sulfoxide (DMSO) at a final concentration of 0.005% (50 µg ml−1).

The Gram-positive and Gram-negative bacteria were sub-cultured in Mueller Hinton broth (MHB) and incubated overnight on a rotary shaker at 37 °C. Each one ml of overnight grown bacterial culture was inoculated in molten (cooled at 45 °C) Mueller Hinton agar and then poured into the sterile Petri dish. Plates were allowed to solidify and four wells were prepared using a sterile cup borer (6 mm in diameter) into each agar plate. Then, 50 μl of each compound was added to respective wells and the plates were pre-incubated at 4 °C in the refrigerator for 10 min to allow the diffusion of the compound into the agar. For every plate, DMSO was also kept as a negative control. Further, the plates were incubated at 37 °C overnight. The zone of growth inhibition around each well was observed to confirm antimicrobial activity and measured in mm (including the diameter of the wells). In order to determine the minimum inhibitory concentrations (MIC), the concentration was summarized by using agar well diffusion method as reported [48].

The SAR study exposed that the thiophene ring at the R position of moiety exhibits more potency toward the antimicrobial activity. In addition, the present study also showed that due to Mono-substitution on the N1 position of the sulfonamide group with heterocyclic or aromatic ring gives more potent antimicrobial compounds. Investigation performed in the current study by introducing different substituents into core indole. Structure–activity relationship was performed on different synthesized molecules and analyzed that compounds 7a and 7b showed good potency against Gram Negative bacteria and more potent towards Klebsiella pneumonia. Although compounds 7d and 7g are relevant but due to the different modes of attachment in both the compounds, the compounds 7d, and 7g showed almost the same potency against Gram-positive Staphylococcus aureus and compound 7g is more potent towards gram-negative Klebsiella pneumonia as compared with compound 7d. On comparing compounds 7j and 7l, the increased activity can be seen in 7j against gram-positive Staphylococcus aureus and all gram-negative bacteria due to the increase in no. of an electron-withdrawing group. As 7j is having more electron-withdrawing groups than 7l, thus it is more potent towards the anti-microbial organisms.

Result

The biological activity as expressed by the growth inhibition zone (in mm) of the tested microorganism is listed in Table 2. Data represented in Table 2 shows that the synthesized sulfonamide-based indole derivatives under investigation exhibited low antibacterial activity against Gram Positive Staphylococcus aureus but failed to inhibit the growth of Bacillus megaterium compared with standard Ciprofloxacin. All tested compounds showed the highest antibacterial activity towards Gram-Negative Klebsiella pneumonia whereas lower activity against other Gram-negative bacteria. The statistical diagram representing the anti-microbial activity of all compounds is shown in Fig. 2.

Table 2 Antibacterial activity of tested compounds as a zone of inhibition in mm and MIC (μg/mL−1) of synthesized compound
Fig. 2
figure 2

Statistical diagram of antimicrobial activity

Discussion

Preparation of N-methylated indole ester (2)

The synthesis route of desired compounds is illustrated in Scheme 1. In suspension of anhydrous potassium carbonate (3 eq.) and 1H-indole-2-carboxylic acid (1 eq.) methyl iodide (2.5 eq.) was added in dry DMF (10 vol.) with constant stirring at 80 °C, the reaction was then completed by performing TLC. Separation was done with 2–3 washings of ethyl acetate and water. When the temperature of the reaction mixture was allowed to come down to room temperature then ethyl acetate was used to extract the aqueous phase. Under the reduced pressure, the organic layer was dried over MgSO4, and as result we get beige colour solid, and the desired N-methylated indole ester (2) with 96–99% yield.

Preparation of 1-methyl-1H-indole-2-carboxylic acid (3)

For the desired 1-methyl-1H-indole-2-carboxylic acid (3), LiOH·H2O in water (3 eq.) was added to a solution of N-methylated indole ester (2) (1 eq.) in THF (3 ml) at room temperature. The ensuing residue was neutralized with 2 N HCl with constant stirring of 3 h and the solvent was completely evaporated under reduced pressure. To get desired corresponding acids with EtOAc (20 ml × 2), a solution was extracted by performing filtration, and then dried over Na2SO4 and excess solvent was evaporated under reduced pressure. We get brown solid residue with 85% yield.

Preparation of (4-aminopiperidin-1-yl)(1-methyl-1H-indol-2-yl)methanone (5)

In a round-bottomed flask first add reagent 4-amino bocpiperidine (4) (1.2 eq.) then add 1-methyl-1H-indole-2-carboxylic acid (3), in the presence of dry DMF. The colloid was allowed to stir overnight with the addition of a measured quantity of EDCl-HCl (1.5 eq.), HOBt (1.5 eq.), and DIPEA (2 eq.) at room temperature. Into a Separating funnel, pour the stirred solution which contains ethyl acetate and water. By using of separator funnel, the aqueous phase was separated as well as extracted three times. The collected residue was dried using Na2SO4. After filtration, the residue was concentrated in vacuo and further purified by using column chromatography (CH2Cl2:MeOH with a ratio of 9:1) to get the desired product (4-aminopiperidin-1-yl)(1-methyl-1H-indol-2-yl)methanone (5).

General procedure of compound (6a-6o)

The solution of (4-aminopiperidin-1-yl)(1-methyl-1H-indol-2-yl)methanone (5) (50 mg, 0.194 mmol) in MeOH (2 ml) was cooled at 0 °C. In the reaction mixture at 0 °C dropwise addition of TEA (0.582 mmol) was done. At 0 °C for 20 min. the reaction was allowed to stir for 1 h at 60 °C after the addition of aldehyde. To the ice-cold resulting mixture NaCNBH3 (0.582 mmol) was added and allowed to stir for 16 h at room temperature. After the dilution of the resulting mixture with ethyl acetate(50 ml), washed with sat. NaHCO3 solution and water. The organic layer was dried over sodium sulphate and under the vaccum crude product was extracted. Purification of this crude product was done by column chromatography using 12% MeOH in MDC to give off a white solid.

General procedure of compound (7a-7o)

The mixture of compound (6a-6o) with methanol (2 ml) was cooled down at 0 °C. After this reaction at 0 °C TEA (0.430 mmol)was added and stirred for 20 min. Then at room temperature reaction mixture was stirred for 3 h after the addition of methane sulfonyl chloride (0.253 mmol). Extraction of crude product was done under vacuum, column chromatography was used to purify and the product was eluted in 1–5% MeOH in MDC to obtain a white solid as the desired compound.

Methyl4-((N-(1-(1-methyl-1H-indole-2-carbonyl)piperidin-4-yl)methylsulfonamido)methyl)benzoate (7b)

1H NMR: δ 0.85–1.86 (4H, m, 0.90 (d, J = 13.4 Hz), 1.28 (d, J = 13.4 Hz), 1.30 (d, J = 13.4 Hz), 1.86 (d, J = 13.4 Hz)), 2.07–2.92 (8H, m, 2.07 (s), 2.13 (d, J = 15.6 Hz), 2.56 (d, J = 15.6 Hz), 2.90 (s)), 3.77–3.79 (4H, m, 3.77 (d, J = 15.2 Hz), 3.77 (d, J = 15.2 Hz), 3.78 (d, J = 15.2 Hz), 3.78 (d, J = 15.2 Hz)), 3.94 (3H, s), 4.13–4.15 (2H, 4.11 (t, J = 10.3 Hz), 4.11 (d, J = 6.7 Hz)), 4.50–4.67 (2H, 4.55 (s)), 6.54–7.37 (6H, m, 6.55 (d, J = 7.9 Hz), 7.16 (d, J = 8.1 Hz), 7.28 (d, J = 7.9 Hz), 7.34 (d, J = 8.1 Hz), 7.35 (d, J = 8.5 Hz)), 7.50–7.62 (2H, d, J = 8.5 Hz).LCMS m/z Cal. [M-H]+483.18 found [M-H]+483.23.

N-(1-(1-methyl-1H-indole-2-carbonyl)piperidin-4-yl)-N-(pyridin-4-ylmethyl)methanesulfonamide (7C)

1HNMR δ 1.29–1.91 (4H, m, 1.45 (d, J = 13.4 Hz), 1.90 (d, J = 13.4 Hz)), 3.16 (3H, s), 3.21–3.69 (4H, m, 3.53 (d, J = 15.2 Hz), 3.53 (d, J = 15.2 Hz)), 3.82 (3H, s), 4.00 (1H, t, J = 10.3 Hz), 4.92 (2H, s), 6.78 (1H, d, J = 1.0 Hz), 7.31–7.36 (5H, q, 7.31 (d, J = 8.0 Hz), 7.31 (t, J = 7.7 Hz), 7.35 (d, J = 8.0 Hz), 7.35 (d, J = 7.8 Hz), 7.36 (d, J = 8.0 Hz)), 7.66 (1H, d, J = 8.0 Hz), 8.73–8.77 (2H, d, 8.76 (d, J = 4.7 Hz), 8.76 (d, J = 1.9 Hz)). LCMS m/z Cal. [M-H]+426.17 found [M-H]+427.28.

N-((4-bromothiophen-3-yl)methyl)-N-(1-(1-methyl-1H-indole-2-carbonyl)piperidin-4-yl)methanesulfonamide (7d)

1H NMR: δ 1.61–1.94 (4H, m, 1.78 (d, J = 13.5 Hz), 1.87 (d, J = 13.5 Hz)), 2.98 (3H, s), 3.42–3.86 (4H, m, 3.45 (d, J = 15.0 Hz), 3.75 (d, J = 15.2 Hz)), 4.09 (3H, s), 4.40 (1H, t, J = 10.3 Hz), 6.61 (1H, d, J = 1.0 Hz), 7.18–7.67 (6H, m, 7.20 (d, J = 1.5 Hz), 7.30 (d, J = 1.5 Hz), 7.32 (d, J = 8.0 Hz), 7.33 (t, J = 7.7 Hz), 7.41 (d, J = 7.8 Hz), 7.65 (d, J = 8.0 Hz)). LCMS m/z Cal. [M + H]+511.04 found [M + H]+509.76.

N-(1-(1-methyl-1H-indole-2-carbonyl)piperidin-4-yl)-N-(pyridin-4-ylmethyl)methanesulfonamide (7e)

1HNMR: δ 1.30–2.52 (4H, m,1.47 (d, J = 13.4 Hz), 2.09 (d, J = 13.4 Hz)), 3.07 (3H, s), 3.25–3.69 (4H, m, 3.30 (d, J = 15.2 Hz), 3.30 (d, J = 15.2 Hz)), 3.86 (3H, s), 4.18 (1H, t, J = 10.3 Hz), 4.72 (2H, s), 6.86 (1H, d, J = 1.0 Hz), 7.19–7.77 (6H, m, 7.20 (d, J = 4.8 Hz), 7.31 (d, J = 8.0 Hz), 7.35 (t, J = 7.7 Hz), 7.39 (d, J = 7.8 Hz), 7.54 (d, J = 8.0 Hz)), 8.76 (2H, d, J = 4.8 Hz). LCMS m/z Cal. [M-H]+526.17 found [M-H]+526.82.

N-((4-bromothiophen-2-yl)methyl)-N-(1-(1-methyl-1H-indole-2-carbonyl)piperidin-4-yl)methanesulfonamide (7g)

1H NMR: δ 1.30–2.15 (15H, m,1.30 (d, J = 13.2 Hz), 1.43 (d, J = 13.2 Hz), 1.50 (d, J = 13.0 Hz), 1.51 (d, J = 12.9 Hz), 1. 51 (d, J = 12.9 Hz), 1.65 (d, J = 13.0 Hz), 1.68 (d, J = 13.1 Hz), 1.73 (d, J = 13.1 Hz), 1.76 (d, J = 13.4 Hz), 1.77 (d, J = 13.4 Hz), 1.89 (d, J = 13.4 Hz), 1.96 (d, J = 13.4 Hz), 1.99 (d, J = 13.6 Hz), 2.02 (d, J = 13.6 Hz), 2.02 (d, J = 9.6 Hz)), 2.93 (3H, s), 3.03 (3H, s), 3.28–3.55 (5H, q, 3.28 (d, J = 9.9 Hz), 3.28 (d, J = 15.2 Hz), 3.50 (d, J = 15.2 Hz), 3.55 (d, J = 15.2 Hz), 3.55 (d, J = 15.2 Hz)), 3.78 (1H, J = 7.4 Hz), 4.02 (1H, t, J = 10.3 Hz), 4.62 (2H, 4,62(s)), 7.05 (1H, d, J = 1.9 Hz), 7.67 (1H, d, J = 1.9 Hz). LCMS m/z Cal. [M-H]+511.04 found [M-H]+512.02.

N-(2,2-diphenylethyl)-N-(1-(1-methyl-1H-indole-2-carbonyl)piperidin-4-yl)methanesulfonamide (7h)

1H NMR: δ 1.23–1.29 (4H, m, 1.26 (d, J = 13.4 Hz), 1.29 (d, J = 13.4 Hz)), 1.64 (3H, s), 2.25–2.89 (4H, m, 2.57 (d, J = 15.2 Hz), 2.88 (d, J = 15.2 Hz)), 2.90–3.99 (5H, q, 3.86 (d, J = 6.1 Hz), 3.99 (s)), 4.00 (1H, tt, J = 10.3 Hz), 4.30 (1H, t, J = 6.1 Hz), 6.67 (1H, d, J = 0.9 Hz), 7.19–7.80 (14H, m, 7.19 (tt, J = 7.7, 1.3 Hz), 7.23 (d, J = 7.8 Hz), 7.30 (m, J = 7.7 Hz), 7.35 (m, J = 7.8 Hz), 7.37 (d, J = 8.0 Hz), 7.77 (d, J = 8.0 Hz), 7.79 (d, J = 7.8 Hz)). LCMS m/z Cal. [M-H]+515.22 found [M-H]+516.24.

N-(cyclohexylmethyl)-N-(1-(1-methyl-1H-indole-2-carbonyl)piperidin-4-yl)methanesulfonamide (7i)

1H NMR: δ 0.90–1.94 (10H, m, 0.92 (d, J = 12.1 Hz), 0.94 (d, J = 12.1 Hz), 1.25 (d, J = 12.8 Hz), 1.29 (d, J = 12.8 Hz), 1.37 (d, J = 13.3 Hz), 1.48 (d, J = 13.3 Hz)), 1.67–1.94 (5H, q, 1.68 (d, J = 13.5 Hz), 1.79 (t, J = 10.3 Hz), 1.85 (d, J = 13.5 Hz)), 2.92 (3H, s), 3.00–3.04 (4H, m, 3.028 (d, J = 15.2 Hz), 3.028 (d, J = 15.2 Hz)), 3.77–3.85 (5H, q, 3.77 (d, J = 5.7 Hz), 3.85 (s)), 4.00 (1H, tt, J = 10.3 Hz), 6.67 (1H, d, J = 1.0 Hz), 7.20–7.70 (4H, m, 7.21 (d J = 8.0 Hz), 7.33 (t, J = 7.7 Hz), 7.42 (d, J = 7.8 Hz), 7.69 (d, J = 8.0, 1.2, 0.5 Hz)). LCMS m/z Cal. [M + H] + 431.22 found [M + H] + 431.19.

N-(3,5-dichlorobenzyl)-N-(1-(1-methyl-1H-indole-2-carbonyl)piperidin-4-yl)methanesulfonamide (7j)

1HNMR: δ 0.90–1.87 (4H, m, 1.45 (d, J = 13.4 Hz), 1.64 (d, J = 13.4 Hz)), 2.92 (3H, s), 2.97–3.00 (4H, m, 2.98 (d, J = 15.2 Hz), 2,98 (d, J = 15.2 Hz)), 3.17 (3H, s), 3.82 (1H, t, J = 10.3 Hz), 4.40 (2H, s), 6.58 (1H, d, J = 1.0, 0.5 Hz), 7.18 (2H, d, J = 1.6 Hz), 7.30–7.66 (5H, q, 7.30 (t, J = 1.6 Hz), 7.34 (d, J = 8.0 Hz), 7.36 (t, J = 7.7 Hz), 7.41 (d, J = 7.8 Hz), 7.64 (d, J = 8.0 Hz)). LCMS m/z Cal. [M-H]+ 493.10 found [M-H]+494.10 (Additional file 1).

Conclusion

In Conclusion, Sulfonamide based indole derivatives are synthesized and discovered as antimicrobial agents. The biological assay showed that all the synthesized compounds showed good activity towards Gram Positive Staphylococcus aureus and inactive towards Bacillus megaterium. Among all Gram-negative bacterial organisms, all the synthesized compounds showed good activity towards Gram-Negative and are highly potent towards Klebsiella pneumonia. This study helps for further research in indole-containing derivatives.

Availability of data and materials

Data and materials are available upon request.

Abbreviations

AMR:

Antimicrobial resistance

WHO:

World Health Organization

MeI:

Methyl iodide

THF:

Tetra hydro furan

EDCl.HCl:

1-Ethyl-3-(3-dimethyl amineopropyl) carbodilimide

HoBt:

Hydroxy benzotrizole

DIPEA:

N,N-Diisopropylethylamine

PTSA:

P-toluen sulfonic acid

TLC:

Thin layer chromatography

MeOH:

Methanol

MDC:

Methylene chloride

TMS:

Tri methyl silane

NMR:

Nuclear magnetic resonance

PPM:

Parts per million

LCMS:

Liquid chromatography mass spectrometry

DMF:

Dimethylformamide

DCM:

Dichloromethane

TEA:

Triethanolamine

References

  1. Menazea AA, Eid MM, Ahmed MK (2020) Synthesis, characterization, and evaluation of antimicrobial activity of novel Chitosan/Tigecycline composite. Int J Biol Macromol 147:194–199. https://doi.org/10.1016/j.ijbiomac.2020.01.041

    Article  CAS  PubMed  Google Scholar 

  2. Annunziato G (2019) Strategies to overcome antimicrobial resistance (AMR) making use of non-essential target inhibitors: a review. Int J Mol Sci 20:5844. https://doi.org/10.3390/ijms20235844

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Khameneh B, Iranshahy M, Soheili V, Sedigheh B, Bazzaz F (2019) review on plant antimicrobials: a mechanistic viewpoint. Antimicrob Resist Infect Control 8:1–28. https://doi.org/10.1186/s13756-019-0559-6

    Article  Google Scholar 

  4. NasiriSovari S, Zobi F (2020) Recent studies on the antimicrobial activity of transition metal complexes of groups 6–12. Chemistry (Easton) 2:418–452. https://doi.org/10.3390/chemistry2020026

    Article  CAS  Google Scholar 

  5. WHO (2021) Global antimicrobial resistance and use surveillance system (GLASS) report 2021. http://www.who.int/glass/resources/publications/early-implementation-report-2020/en/. Accessed 2021.

  6. Ahmad N, Wee CE, Wai LK, Zin NM, Azmi F (2021) Biomimetic amphiphilic chitosan nanoparticles: synthesis, characterization and antimicrobial activity. Carbohydr Polym 254:117299. https://doi.org/10.1016/j.carbpol.2020.117299

    Article  CAS  PubMed  Google Scholar 

  7. Faruk A (2018) Antimicrobial activity of 1,3,4-thiadiazole derivatives: a recent review. J Appl Pharm Res 6:10–19. https://doi.org/10.18231/2348-0335.2018.0012

    Article  CAS  Google Scholar 

  8. Kakkar AK, Shafiq N, Singh G, Ray P, Gautam V, Agarwal R et al (2020) Antimicrobial stewardship programs in resource constrained environments: understanding and addressing the need of the systems. Front Public Heal. https://doi.org/10.3389/fpubh.2020.00140

    Article  Google Scholar 

  9. Manandhar S, Luitel S, Dahal RK (2019) In vitro antimicrobial activity of some medicinal plants against human pathogenic bacteria. J Trop Med. https://doi.org/10.1155/2019/1895340

    Article  PubMed  PubMed Central  Google Scholar 

  10. Bush K, Courvalin P, Dantas G, Davies J, Eisenstein B, Huovinen P et al (2011) Tackling antibiotic resistance. Nat Rev Microbiol 9:894–896. https://doi.org/10.1038/nrmicro2693

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Liu Z, Tang L, Zhu H, Xu T, Qiu C, Zheng S et al (2016) Design, synthesis, and structure-activity relationship study of novel indole-2-carboxamide derivatives as anti-inflammatory agents for the treatment of sepsis. J Med Chem 59:4637–4650. https://doi.org/10.1021/acs.jmedchem.5b02006

    Article  CAS  PubMed  Google Scholar 

  12. Kumar S, Ritika X (2020) A brief review of the biological potential of indole derivatives. Futur J Pharm Sci. https://doi.org/10.1186/s43094-020-00141-y

    Article  Google Scholar 

  13. Van Order RB, Lindwall H (1942) Indole. Chem Rev. https://doi.org/10.1021/cr60095a004

    Article  Google Scholar 

  14. Kumari A, Singh RK (2019) Medicinal chemistry of indole derivatives: Current to future therapeutic prospectives. Bioorg Chem 89:103021. https://doi.org/10.1016/j.bioorg.2019.103021

    Article  CAS  PubMed  Google Scholar 

  15. Jia Y, Wen X, Gong Y, Wang X (2020) Current scenario of indole derivatives with potential anti-drug-resistant cancer activity. Eur J Med Chem 200:112359. https://doi.org/10.1016/j.ejmech.2020.112359

    Article  CAS  PubMed  Google Scholar 

  16. Kaushik NK, Kaushik N, Attri P, Kumar N, Kim CH, Verma AK et al (2013) Biomedical importance of indoles. Molecules 18:6620–6662. https://doi.org/10.3390/molecules18066620

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Thanikachalam PV, Maurya RK, Garg V, Monga V (2019) An insight into the medicinal perspective of synthetic analogs of indole: a review. Eur J Med Chem 180:562–612. https://doi.org/10.1016/j.ejmech.2019.07.019

    Article  CAS  PubMed  Google Scholar 

  18. Dadashpour S, Emami S (2018) Indole in the target-based design of anticancer agents: a versatile scaffold with diverse mechanisms. Eur J Med Chem 150:9–29. https://doi.org/10.1016/j.ejmech.2018.02.065

    Article  CAS  PubMed  Google Scholar 

  19. Amr AEGE, Abdalla MM, Al-Omar MA, Elsayed EA (2017) Anti-ovarian and anti-breast cancers with dual topoisomerase ii/braf600e inhibitors activities of some substituted indole derivatives. Biomed Res 28:75–80

    Google Scholar 

  20. Tetali SR, Kunapaeddi E, Mailavaram RP, Singh V, Borah P, Deb PK et al (2020) Current advances in the clinical development of anti-tubercular agents. Tuberculosi 125:101989. https://doi.org/10.1016/j.tube.2020.101989

    Article  CAS  Google Scholar 

  21. Dogamanti A, Chiranjeevi P, Aamate VK, Vagolu SK, Sriram D, Balasubramanian S et al (2020) Indole-fused spirochromenes as potential anti-tubercular agents: design, synthesis and in vitro evaluation. Mol Divers. https://doi.org/10.1007/s11030-020-10108-z

    Article  PubMed  Google Scholar 

  22. Deswal S, Naveen TRK, GhuleVikas D, Lal K, Kumar A (2020) 5-Fluoro-1H-indole-2,3-dione-triazoles- synthesis, biological activity, molecular docking, and DFT study. J Mol Struct. https://doi.org/10.1016/j.molstruc.2020.127982

    Article  Google Scholar 

  23. Angelova VT, Pencheva T, Vassilev N, Simeonova R, Momekov G, Valcheva V (2019) New indole and indazole derivatives as potential antimycobacterial agents. Med Chem Res 28:485–497. https://doi.org/10.1007/s00044-019-02293-w

    Article  CAS  Google Scholar 

  24. Namasivayam V, Vanangamudi M, Kramer VG, Kurup S, Zhan P, Liu X et al (2019) The Journey of HIV-1 non-nucleoside reverse transcriptase inhibitors (NNRTIs) from lab to clinic. J Med Chem 62:4851–4883. https://doi.org/10.1021/acs.jmedchem.8b00843

    Article  CAS  PubMed  Google Scholar 

  25. Zhou G, Chu S, Nemati A, Huang C, Snyder BA, Ptak RG et al (2019) Investigation of the molecular characteristics of bisindole inhibitors as HIV-1 glycoprotein-41 fusion inhibitors. Eur J Med Chem 161:533–542. https://doi.org/10.1016/j.ejmech.2018.10.048

    Article  CAS  PubMed  Google Scholar 

  26. Elshemy HAH, Zaki MA, Mohamed EI, Khan SI, Lamie PF (2020) A multicomponent reaction to design antimalarial pyridyl-indole derivatives: synthesis, biological activities and molecular docking. Bioorg Chem. https://doi.org/10.1016/j.bioorg.2020.103673

    Article  PubMed  Google Scholar 

  27. Luthra T, Nayak AK, Bose S, Chakrabarti S, Gupta A, Sen S (2019) Indole based antimalarial compounds targeting the melatonin pathway: their design, synthesis and biological evaluation. Eur J Med Chem 168:11–27. https://doi.org/10.1016/j.ejmech.2019.02.019

    Article  CAS  PubMed  Google Scholar 

  28. Shaker AMM, Abdelall EKA, Abdellatif KRA, Abdel-Rahman HM (2020) Synthesis and biological evaluation of 2-(4-methylsulfonyl phenyl) indole derivatives: multi-target compounds with dual antimicrobial and anti-inflammatory activities. BMC Chem 14(1):1–15. https://doi.org/10.1186/s13065-020-00675-5

    Article  CAS  Google Scholar 

  29. Al-Ostoot FH, Geetha DV, Mohammed YHE, Akhileshwari P, Sridhar MA, Khanum SA (2019) Design-based synthesis, molecular docking analysis of an anti-inflammatory drug, and geometrical optimization and interaction energy studies of an indole acetamide derivative. J Mol Struct. https://doi.org/10.1016/j.molstruc.2019.127244

    Article  Google Scholar 

  30. Ashok P, Chander S, Smith TK, Prakash Singh R, Jha PN, Sankaranarayanan M (2019) Biological evaluation and structure activity relationship of 9-methyl-1-phenyl-9H-pyrido[3,4-b]indole derivatives as anti-leishmanial agents. Bioorg Chem 84:98–105. https://doi.org/10.1016/j.bioorg.2018.11.037

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Porwal S, Gupta S, Chauhan PMS (2017) gem-Dithioacetylated indole derivatives as novel antileishmanial agents. Bioorganic Med Chem Lett 27:4643–4646. https://doi.org/10.1016/j.bmcl.2017.09.018

    Article  CAS  Google Scholar 

  32. Bingul M, Ercan S, Boga M (2020) The design of novel 4,6-dimethoxyindole based hydrazide-hydrazones: molecular modeling, synthesis and anticholinesterase activity. J Mol Struct. https://doi.org/10.1016/j.molstruc.2020.128202

    Article  Google Scholar 

  33. Bingül M (2019) Synthesis and characterisation of novel 4,6-dimethoxyindole-7- and -2-thiosemicarbazone derivatives: biological evaluation as antioxidant and anticholinesterase candidates. J Chem Res 43:399–406. https://doi.org/10.1177/1747519819868386

    Article  CAS  Google Scholar 

  34. Fantacuzzi M, De Filippis B, Gallorini M, Ammazzalorso A, Giampietro L, Maccallini C et al (2020) Synthesis, biological evaluation, and docking study of indole aryl sulfonamides as aromatase inhibitors. Eur J Med Chem 185:111815. https://doi.org/10.1016/j.ejmech.2019.111815

    Article  CAS  PubMed  Google Scholar 

  35. Islam MS, Barakat A, Al-Majid AM, Ali M, Yousuf S, Iqbal Choudhary M et al (2018) Catalytic asymmetric synthesis of indole derivatives as novel α-glucosidase inhibitors in vitro. Bioorg Chem 79:350–354. https://doi.org/10.1016/j.bioorg.2018.05.004

    Article  CAS  PubMed  Google Scholar 

  36. Taha M, Imran S, Salahuddin M, Iqbal N, Rahim F, Uddin N et al (2021) Evaluation and docking of indole sulfonamide as a potent inhibitor of α-glucosidase enzyme in streptozotocin –induced diabetic albino wistar rats. Bioorg Chem. https://doi.org/10.1016/j.bioorg.2021.104808

    Article  PubMed  Google Scholar 

  37. Arshad M (2017) Synthesis, characterization, antimicrobial and computational studies of some sulfonamide derivatives possessing thiadiazole and indole nucleus. Eur J Pharm Med Res 4:511–517

    Google Scholar 

  38. Chohan ZH, Youssoufi MH, Jarrahpour A, Ben Hadda T (2010) Identification of antibacterial and antifungal pharmacophore sites for potent bacteria and fungi inhibition: indolenyl sulfonamide derivatives. Eur J Med Chem 45:1189–1199. https://doi.org/10.1016/j.ejmech.2009.11.029

    Article  CAS  PubMed  Google Scholar 

  39. Hu Y, Zhang L, Huang J, Wang T, Zhang J et al (2021) Novel schiff base-conjugated para aminobenzenesulfonamide indole hybrids as potentially muti-targeting blockers against Staphylococcus. Asian J Organ Chem 11:6–7. https://doi.org/10.1002/ajoc.202100737

    Article  CAS  Google Scholar 

  40. Jagadeesan S, Karpagam S (2023) Novel series of N-acyl substituted indole based piperazine, thiazole and tetrazoles as potential antibacterial, antifungal, antioxidant and cytotoxic agents, and their docking investigation as potential Mcl-1 inhibitors. J Mol Struct. https://doi.org/10.1016/j.molstruc.2022.134013

    Article  Google Scholar 

  41. Al-Wabli RI, Almomen AA, Almutairi MS, Keeton AB, Piazza GA, Attia MI (2020) New isatin–indole conjugates: synthesis, characterization, and a plausible mechanism of their in vitro antiproliferative activity. Drug Des Devel Ther 14:483–495. https://doi.org/10.2147/DDDT.S227862

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Umar Basha KN, Gnanamani S, Shanmugam P, Venugopal S, Murthy S, Ramasamy B (2021) Synthesis, antioxidant, and antimicrobial activity of 3-(1H-indole-3-carbonyl)-2H-chromen-2-ones. J Heterocycl Chem 58:2000–2008. https://doi.org/10.1002/jhet.4326

    Article  CAS  Google Scholar 

  43. Bitombo AN, Zintchem AAA, Atchade AdeT, Moni Ndedi EDF, Khan A, NgonoBikobo DS et al (2022) Antimicrobial and cytotoxic activities of indole alkaloids and other constituents from the stem barks of Rauvolfia caffra Sond (Apocynaceae). Nat Prod Res 36:1467–1475. https://doi.org/10.1080/14786419.2021.1891054

    Article  CAS  PubMed  Google Scholar 

  44. Dolušić E, Larrieu P, Blanc S, Sapunaric F, Norberg B, Moineaux L et al (2011) Indol-2-yl ethanones as novel indoleamine 2,3-dioxygenase (IDO) inhibitors. Bioorganic Med Chem 19:1550–1561. https://doi.org/10.1016/j.bmc.2010.12.032

    Article  CAS  Google Scholar 

  45. Thanigaimalai P, Konno S, Yamamoto T, Koiwai Y, Taguchi A, Takayama K et al (2013) Development of potent dipeptide-type SARS-CoV 3CL protease inhibitors with novel P3 scaffolds: design, synthesis, biological evaluation, and docking studies. Eur J Med Chem 68:372–384. https://doi.org/10.1016/j.ejmech.2013.07.037

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Tanuma SI, Katsuragi K, Oyama T, Yoshimori A, Shibasaki Y, Asawa Y et al (2020) Structural basis of beneficial design for effective nicotinamide phosphoribosyltransferase inhibitors. Molecules 25:1–15. https://doi.org/10.3390/molecules25163633

    Article  CAS  Google Scholar 

  47. Rubab L, Afroz S, Ahmad S, Hussain S, Nawaz I, Irfan A, Batool F, Kotwica-Mojzych K, Mojzych M (2022) An update on synthesis of coumarin sulfonamides as enzyme inhibitors and anticancer agents. Molecules 27:1604. https://doi.org/10.3390/molecules27051604

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Gonelimali FD, Lin J, Miao W, Xuan J, Charles F, Chen M et al (2018) Antimicrobial properties and mechanism of action of some plant extracts against food pathogens and spoilage microorganisms. Front Microbiol 9:1–9. https://doi.org/10.3389/fmicb.2018.01639

    Article  Google Scholar 

Download references

Acknowledgements

The authors are thankful to the Shri MM Patel College of Science and Research, Department of Chemistry, Kadi Sarva Vishwavidyalaya, India for providing necessary facilities. The authors thank especially Principal (Prof.) Keyur Shah, SMMPISR, Gandhinagar, Gujarat for providing lab research facilities, Dr. Shavi Rana and Ms. Kruti Patel for extending their support in whole research work.

Funding

The author declare that no funds, authorship and other support received from anyone.

Author information

Authors and Affiliations

Authors

Contributions

KA, Dr. TP Contributed to design of the research, Material preparation, writing the manuscript, editing, designed the table and figures done by KA, Antimicrobial activity was performed and written by Dr. RP. Sample characterization with spectroscopy was done by KA, Dr. TP. Dr. TP was involved in supervision of all work. All authors read and approved the final manuscirpt.

Corresponding author

Correspondence to Khushbu Agrawal.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

The authors declare no completing financial interest.

Competing interests

There are no competing interests to declare for all authors.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1

. Spectral data of NMR and LCMS Spectroscopy of Synthesised Compound.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Agrawal, K., Patel, T. & Patel, R. Synthesis, biological activity of newly designed sulfonamide based indole derivative as anti-microbial agent. Futur J Pharm Sci 9, 17 (2023). https://doi.org/10.1186/s43094-023-00466-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43094-023-00466-4

Keywords