Skip to main content

Synthesis and antioxidant properties of 2-(3-(hydroxyimino)methyl)-1H-indol-1-yl)acetamide derivatives

Abstract

Background

The main aim of this work was to synthesise a novel N-(substituted phenyl)-2-(3-(hydroxyimino) methyl)-1H-indol-1-yl) acetamide derivatives and evaluate their antioxidant activity. These compounds were prepared by a condensation reaction between 1H-indole carbaldehyde oxime and 2-chloro acetamide derivatives. The newly synthesised compound structures were characterised by FT-IR, 1H-NMR, mass spectroscopy and elemental analysis. Furthermore, the above-mentioned compounds were screened for antioxidant activity by using ferric reducing antioxidant power (FRAP) and 1,1-diphenyl-2-picrylhydrazyl (DPPH) methods.

Result

The antioxidant activity result reveals that most of the compounds were exhibiting considerable activity in both methods and the values are very closer to the standards. Among the synthesised compounds, compound 3j, 3a and 3k were shown remarkable activity at low concentration.

Conclusion

Compounds 3j, 3a and 3k were shown highest activity among the prepared analogues due to the attachment of halogens connected at the appropriate place in the phenyl ring. Hence, these substituted phenyl rings considered as a perfect side chain for the indole nucleus for the development of the new antioxidant agents.

Background

The indole framework is one of the most widely distributed heterocyclic nuclei in both natural and synthetic compounds in which benzene and pyrrole are fused in 2,3 positions [1]. The name indole is derived from the word indigo and oleum [2]. In 1986, Adolf von Bayer prepared indole from oxindole by a simple chemical reaction using zinc dust [3]. Indole and their derivatives are identified as a non-basic nitrogenous pharmacophore exhibiting a broad range of useful biological activities such as anti-inflammatory [4], anti-depressant [5], anti-fungal [6], anti-cancer [7], antihypertensive [8], antibiotic [9], anti-microbial agent [10], anti-viral [11], chelating agents [12], antimalarial [13], anti-HIV [14], anti-diabetic [15], anti-tuberculosis [16], insecticidal [17] and analgesic activity [18]. Because of the dynamic properties, they have been placed in a unique platform of nitrogenous heterocyclic compounds and enhance the interest of the scientist all over the world towards the preparation of the novel indole derivatives. Amide and their analogues are also found in many naturally occurring compounds and received much attention due to their high chemotherapeutic profile and easy way of developing a novel compound through the simple chemical reaction [19]. It has been prepared by the reaction of substituted acid with various aliphatic or aromatic amines. These derivatives are associated with a board spectrum of biological activities such as anti-fungal [20], insecticides [21], anticonvulsant [22], analgesic [23], anti-inflammatory [24], anti-tuberculosis [25] and anti-tumour [26] properties. As a result of a continuous search for the above-mentioned area, a series of novel N-(substituted phenyl)-2-(3-(hydroxyimino) methyl)-1H-indol-1-yl) acetamide derivatives were synthesised and evaluated their antioxidant activity by using ferric reducing antioxidant power (FRAP) and 1,1-diphenyl-2-picrylhydrazyl (DPPH) method. Most of the compounds were shown admirable anti-oxidant activity due to the presence of indole and amide pharmacophore.

Method

Chemistry

The melting points of prepared analogues were recorded by open capillary tube on an Electrothermal 9100 melting point apparatus and are uncorrected. Functional groups present in the compounds were confirmed by using IR spectra (KBr) were recorded on a Shimadzu FT-IR 8400S spectrophotometer between the ranges from 4000 cm−1 to 400 cm−1. 1H-NMR spectra were determined on a Bruker NMR spectrometer (500 MHz). 13C-NMR spectra were performed on a Bruker NMR spectrometer (100 MHz) and fully decoupled. Chemical shifts were reported in parts per million (ppm) using the deuterated solvent peak and tetramethylsilane as an internal standard. X-ray studies were determined by X Calibur diffractometer. The molecular weight of the compound was analysed by the Shimadzu mass spectrometer and element analysis was performed on Perkin Elmer 2400 CHN elemental analyser.

Preparation of 1H-indole-3-carbaldehyde [1]

An equimolar mixture of indole (0.001 mol), N, N-dimethyl formamide (0.001 mol) and 8.75 ml of phosphorous oxychloride was poured into the round bottom. The resulting solution was stirred magnetically for 5 h at 60 °C and transferred into 25 ml of 10% sodium carbonate solution. After complete addition, the mixture again stirred for 2 h at 100 °C. The separated product was washed with water, dried over anhydrous calcium chloride and recrystallised from hot ethanol [27].

Preparation of 1H-indole-3-carbaldehyde oxime [2]

1H-indole-3-carbaldehyde (1.66 mmol), sodium hydroxide (0.15 mol) and hydroxylamine hydrochloride (3.4 gm) were dissolved in 10 ml of ethanol and refluxed at 80 °C for 3 h. The progression of the reaction was monitored by TLC using the mixture of n-hexane and ethyl acetate as a mobile phase. After completion of the reaction, the separated product was washed with water and recrystallised from hot ethanol [28].

Preparation of N-(substituted phenyl)-2-(3-(hydroxyimino) methyl)-1H-indol-1-yl) acetamide derivatives: [3a–n]

A solution of 2-chloro acetamide derivatives (2 mmol) in 20 ml DMF, indole-3-carbaldehyde oxime [2] (2 mmol), potassium iodide (2 mmol) and potassium carbonate (2 mmol) were added and the resulting mixture was refluxed for 8 h at 80 °C. Then, the hot solution was poured into ice-cold water and the separated product was filtered, washed with water, dried over anhydrous calcium chloride and recrystallised from hot ethanol [29]. The scheme of the synthesis compounds were summarised in Fig. 1.

Fig. 1
figure 1

Synthetic route of various potent N-(substituted phenyl)-2-(3-(hydroxyimino)methyl)-1H-indol-1-yl) acetamide derivatives [3a–n]

Anti-oxidant activity

Ferric reducing antioxidant power (FRAP) method

The FRAP is a quantitative assay for evaluating the antioxidant potency of newly synthesised compounds [30]. The FRAP reagent was prepared by mixing of 2.5 ml of 10 μM ferric TPTZ, 2.5 ml of 20 μM FeCl3 and 25 ml of 0.3 μM of acetate buffer and maintain the pH 3.6. The mixture was prepared freshly and warmed at 37 °C before use. Here, the antioxidants react with ferric tripyridyl triazine complex (colourless) which turn to ferrous 2,4,6-tripyridyl-s-triazine (blue colour). To identify the antioxidant activity, different concentrations (50, 75, 100 μg/ml) of synthesised compounds in DMSO (1 ml) was mixed with FRAP reagent. The content was incubated at 50 °C for 20 min. The specified quantity of the solution was transferred into a cuvette and measured the absorbance at 593 nm using a Shimadzu ultraviolet spectrometer. Standard absorbance was also calibrated. The antioxidant power of acetamide derivatives at different concentration was determined through directly substituting the absorbance in the mentioned formula.

$$ \mathrm{FRAP}\ \mathrm{value}\ \mathrm{of}\ \mathrm{sample}\ \left(\upmu \mathrm{m}\right)=\frac{\mathrm{Absorbance}\ \left(\mathrm{sample}\right)\times \mathrm{FRAP}\ \mathrm{value}\ \mathrm{of}\ \mathrm{standard}\ \left(\upmu \mathrm{m}\right)\ }{\mathrm{Absorbance}\ \left(\mathrm{Standard}\right)} $$

DPPH radical scavenging method

DPPH is a stable free radical widely used to assess the radical scavenging activity of antioxidant components [31]. DPPH free radical has a stable violet colour in methanol and becomes colourless or yellow colour when paired with antioxidant or reducing agents. These radicals can accept the odd electron or hydrogen from the antioxidant and converted to a stable diamagnetic molecule (yellow). To identify the antioxidant activity, the solution of synthesised compounds has been prepared by dissolving in methanol (100 μg/ml). Simultaneously, a solution of DPPH was also prepared in another container had been shown maximum absorbance at 517 nm due to the presence of stable 1,1-diphenyl 2-picryl hydrazyl stable free radical (violet colour). Each test compound (4 ml) was added to 4 ml of DPPH solution and kept aside for 30 min at room temperature. Absorbance was measured at 517 nm using a Shimadzu ultraviolet spectrometer. Blank and standard absorbances were also calibrated. Antioxidant activity of acetamide derivatives was determined through directly substituting the absorbance in the mentioned formula.

$$ \mathrm{Percentage}\ \mathrm{inhibition}=\frac{\mathrm{Absorbance}\ \mathrm{of}\ \mathrm{blank}\hbox{-} \mathrm{Absorbance}\ \mathrm{of}\ \mathrm{test}}{\mathrm{Absorbance}\ \mathrm{of}\ \mathrm{blank}}\times 100 $$

Computational analysis

Computational science is a relatively new discipline that deals with the development and application of computational models and simulations, often coupled with high-performance computing, to solve the complex problem arising in drug design. Here, conformational analysis and geometric optimization of compound 3a have been performed by using Chemdraw ultra 12. Semi-empirical method PM3 is used for optimising the full geometry of the system using QA algorithm and unrestricted Hatee-Fock (UHF) method in conjunction with the 6-311++G(d,p) basis set [32].

X-ray crystallography

X-ray crystallography is a tool used to investigate the three-dimensional picture of the atomic and molecular structure of a crystal by using X-ray light, which has wavelengths of 1 Å. The beam of X-ray hits a crystal and causes the diffraction of light in particular directions; it has fed into the computer and finds out the position of every atom in the molecule. Crystallographic data were collected at 296 K, and the structure of crystal was solved by direct method using SHELXS-97 [33].

Results

Spectral data

Preparation of N-(2-chlorophenyl)-2-(3-((hydroxyimino)methyl)-1H-indol-1-yl)acetamide: [3a]

Molecular formula: C17H14N3O2Cl; yield: (77%); melting point: 229–230 °C; FT-IR (νmax/cm−1): 3046 (Ar-H str), 1506 (Ar-C str), 3317 (NH-str), 1646 (NH-bend), 1726 (C=O-str), 623 (Cl); 1H-NMR (500 MHz, CDCl3, δ ppm): δ 4.71 (s, 2H, CH2-H), δ 6.46 (d, 2H, J = 14.3 Hz, CH-H), δ 6.89 (t,1H, J = 9.1 Hz, Ar-H), δ 7.54 (t, 1H, J = 10.3 Hz, Ar-H), δ 7.64 (s, 4H, Ar-H) δ 7.74 (d, 1H, J = 12.1 Hz, Ar-H), δ 7.96 (d, 1H, J = 14.5 Hz, Ar-H), δ 8.31 (s, 1H, NH-H), δ 9.11 (s, 1H, NOH-H); 13C NMR (100 MHz, CDCl3, δ ppm): 43.9, 106.2, 110.1, 119.2, 122.8, 127.4, 128.1, 128.4, 128.4, 132.2, 134.5, 134.5, 136.1, 136.1, 141.7, 145.7, 166.4; ESI-MS (m/z, %): 327 (59) [M+], 329 (18) [M++2], 241 (26), 185 (46), 146 (23), 80 (100), 53 (40); anal. calcd for C17H14N3O2Cl (327), predicted: C, 62.30; H, 4.31; N, 12.82%. Found: C, 62.47; H, 4.16; N, 12.62%.

Preparation of N-(3-chlorophenyl)-2-(3-((hydroxyimino)methyl)-1H-indol-1-yl)acetamide: [3b]

Molecular formula: C17H14N3O2Cl; yield: (63%); melting point: 257–259 °C; FT-IR (νmax/cm−1): 3037 (Ar-H str), 1521 (Ar-C str), 3305 (NH-str), 1602 (NH-bend), 1757 (C=O-str), 724 (Cl); 1H-NMR (500 MHz, CDCl3, δ ppm): δ 4.83 (s, 2H, CH2-H), δ 6.69 (d, 2H, J = 14.3 Hz, CH-H), δ 7.06 (d,1H, J = 17.8 Hz, Ar-H), δ 7.29 (s, 1H, Ar-H), δ 7.37 (s, 4H, Ar-H), δ 7.57 (d, 1H, J = 14.7 Hz, Ar-H), δ 7.83 (s, 1H, Ar-H), δ 8.15 (s, 1H, NH-H), δ 9.12 (s, 1H, NOH-H); ESI-MS (m/z, %): 327 (49) [M+], 329 (17) [M++2], 267 (31), 216 (42), 173 (100), 146 (22); 80 (49), 39 (34); Anal. Calcd for C17H14N3O2Cl (327), predicted: C, 62.30; H, 4.31; N, 12.82%. Found: C, 62.35; H, 4.28; N, 12.91%.

Preparation of N-(4-chlorophenyl)-2-(3-((hydroxyimino)methyl)-1H-indol-1-yl)acetamide: [3c]

Molecular formula: C17H14N3O2Cl; yield: (89%); melting point: 211–213 °C; FT-IR (νmax/cm−1): 3061 (Ar-H str), 1538 (Ar-C str), 3369 (NH-str), 1646 (NH-bend), 1705 (C=O-str), 689 (Cl); 1H-NMR (500 MHz, CDCl3, δ ppm): δ 4.69 (s, 2H, CH2-H), δ 6.68 (d, 2H, J = 10.3 Hz, CH-H), δ 7.27 (s, 4H, Ar-H), δ 7.50 (d, 2H, J = 12.4 Hz, Ar-H) δ 7.88 (d, 2H, J = 14.1 Hz, Ar-H), δ 8.25 (s, 1H, NH-H), δ 9.10 (s, 1H, NOH-H); ESI-MS (m/z, %): 327 (57) [M+], 329 (21) [M++2], 267 (37), 201 (45), 159 (26), 115 (100), 77 (37), 39 (26); anal. calcd for C17H14N3O2Cl (327), predicted: C, 62.30; H, 4.31; N, 12.82%. Found: C, 62.52; H, 4.26; N, 12.84%.

Preparation of N-(2-bromophenyl)-2-(3-((hydroxyimino)methyl)-1H-indol-1-yl)acetamide: [3d]

Molecular formula: C17H14N3O2Br; yield: (72%); melting point: 283–287 °C; FT-IR (νmax/cm−1): 3043 (Ar-H str), 1532 (Ar-C str), 3326 (NH-str), 1628 (NH-bend), 1734 (C=O-str), 578 (Br); 1H-NMR (500 MHz, CDCl3, δ ppm): δ 4.55 (s, 2H, CH2-H), δ 6.53 (d, 2H, J = 14.3 Hz, CH-H), δ 6.76 (t,1H, J = 7.8 Hz, Ar-H), δ 7.23 (s, 5H, Ar-H), δ 7.41 (d, 1H, J = 10.8 Hz, Ar-H) δ 7.69 (d, 1H, J = 9.1 Hz, Ar-H), δ 8.09 (s, 1H, NH-H), δ 8.93 (s, 1H, NOH-H); ESI-MS (m/z, %): 371 (27) [M+], 373 (25) [M++2], 254 (47), 216 (30), 173 (63), 159 (43), 115 (100), 53 (30); anal. calcd for C17H14N3O2Br (371), predicted: C, 54.86; H, 3.79; N, 11.29%. Found: C, 54.92; H, 3.61; N, 11.08%.

Preparation of N-(3-bromophenyl)-2-(3-((hydroxyimino)methyl)-1H-indol-1-yl)acetamide: [3e]

Molecular formula: C17H14N3O2Br; yield: (59%); melting point: 226–230 °C; FT-IR (νmax/cm−1): 3012 (Ar-H str), 1505 (Ar-C str), 3365 (NH-str), 1638 (NH-bend), 1711 (C=O-str), 615 (Br); 1H-NMR (500 MHz, CDCl3, δ ppm): δ 4.45 (s, 2H, CH2-H), δ 6.57(d, 2H, J = 11.3 Hz, CH-H), δ 7.03 (s,1H, Ar-H), δ 7.15 (s, 1H, Ar-H), δ 7.35 (s, 4H, Ar-H), δ 7.50 (d, 1H, J = 12.1 Hz, Ar-H), δ 7.80 (s, 1H, Ar-H), δ 8.06 (s, 1H, NH-H), δ 9.15 (s, 1H, NOH-H); ESI-MS (m/z, %): 371 (29) [M+], 373 (27) [M++2], 254 (51), 201 (26), 159 (41), 115 (100), 90 (51), 44 (26); anal. calcd for C17H14N3O2Br (371), predicted: C, 54.86; H, 3.79; N, 11.29%. Found: C, 54.73; H, 3.55; N, 11.14%.

Preparation of N-(4-bromophenyl)-2-(3-((hydroxyimino)methyl)-1H-indol-1-yl)acetamide: [3f]

Molecular formula: C17H14N3O2Br; yield: (62%); melting point: 247–249 °C; FT-IR (νmax/cm−1): 3072 (Ar-H str), 1563 (Ar-C str), 3317(NH-str), 1620 (NH-bend), 1757 (C=O-str), 636 (Br); 1H-NMR (500 MHz, CDCl3, δ ppm): δ 4.79 (s, 2H, CH2-H), δ 6.75 (d, 2H, J = 14.1 Hz, CH-H), δ 7.07 (s, 4H, Ar-H), δ 7.34 (d, 2H, J = 13.3 Hz, Ar-H), δ 7.60 (d, 2H, J = 13.8 Hz, Ar-H), δ 8.23 (s, 1H, NH-H), δ 9.00 (s, 1H, NOH-H); ESI-MS (m/z, %): 371 (27) [M+], 373 (24) [M++2], 267 (35), 216 (62), 173 (26), 104 (100), 69 (55), 40 (41); anal. calcd for C17H14N3O2Br (371), predicted: C, 54.86; H, 3.79; N, 11.29%. Found: C, 54.80; H, 3.58; N, 11.32%.

Preparation of N-(2-flurophenyl)-2-(3-((hydroxyimino)methyl)-1H-indol-1-yl)acetamide: [3g]

Molecular formula: C17H14N3O2F; yield: (88%); melting point: 213–217 °C; FT-IR (νmax/cm−1): 3047 (Ar-H str), 1524 (Ar-C str), 3328(NH-str), 1641 (NH-bend), 1760 (C=O-str), 968 (F); 1H-NMR (500 MHz, CDCl3, δ ppm): δ 4.58 (s, 2H, CH2-H), δ 6.48 (d, 2H, J = 10.3 Hz, CH-H), δ 6.82-7.00 (m, 3H, Ar-H), δ 7.26 (s, 4H, Ar-H), δ 7.65 (t, 1H, J = 10.8 Hz, Ar-H), δ 8.04 (s, 1H, NH-H), δ 9.07 (s, 1H, NOH-H); ESI-MS (m/z, %): 311 (24) [M+], 254 (36), 216 (24), 203 (49), 137 (66), 96 (100), 41 (39); anal. calcd for C17H14N3O2F (311), predicted: C, 65.59; H, 4.53; N, 13.50%. Found: C, 65.40; H, 4.52; N, 13.46%.

Preparation of N-(3-flurophenyl)-2-(3-((hydroxyimino)methyl)-1H-indol-1-yl)acetamide: [3h]

Molecular formula: C17H14N3O2F; yield: (72%); melting point: 244–247 °C; FT-IR (νmax/cm−1): 3012 (Ar-H str), 1593 (Ar-C str), 3329(NH-str), 1611 (NH-bend), 1728 (C=O-str), 1038 (F); 1H-NMR (500 MHz, CDCl3, δ ppm): δ 4.74 (s, 2H, CH2-H), δ 6.63 (t, 1H, J = 8.4 Hz, Ar-H), δ 6.82 (d, 2H, J = 10.8 Hz, CH-H), δ 7.13 (s, 4H, Ar-H), δ 7.23-7.42 (m, 1H, Ar-H), δ 7.56 (d, 1H, J = 11.1 Hz, Ar-H), δ 7.81 (d, 1H, J = 14.3 Hz, Ar-H), δ 8.16 (s, 1H, NH-H), δ 8.85 (s, 1H, NOH-H); ESI-MS (m/z, %): 311 (34) [M+], 267 (24), 201 (53), 162 (34), 121 (60), 80 (100), 42 (27); anal. calcd for C17H14N3O2F (311), predicted: C, 65.59; H, 4.53; N, 13.50%. Found: C, 65.38; H, 4.47; N, 13.62%.

Preparation of N-(4-flurophenyl)-2-(3-((hydroxyimino)methyl)-1H-indol-1-yl)acetamide: [3i]

Molecular formula: C17H14N3O2F; yield: (90%); melting point: 211–213 °C; FT-IR (νmax/cm−1): 3097 (Ar-H str), 1502 (Ar-C str), 3311 (NH-str), 1624 (NH-bend), 1755 (C=O-str), 1137 (F); 1H-NMR (500 MHz, CDCl3, δ ppm): δ 4.72 (s, 2H, CH2-H), δ 6.37 (d, 2H, J = 13.3 Hz, CH-H), δ 6.76 (t, 2H, J = 12.8 Hz, Ar-H), δ 7.27 (s, 4H, Ar-H), δ 7.62 (t, 2H, J = 10.1 Hz, Ar-H), δ 7.99 (s, 1H, NH-H), δ 9.14 (s, 1H, NOH-H); ESI-MS (m/z, %): 311 (21) [M+], 254 (39), 216 (31), 159 (58), 115 (100), 77 (31), 41 (39); anal. calcd for C17H14N3O2F (311), predicted: C, 65.59; H, 4.53; N, 13.50%. Found: C, 65.47; H, 4.36; N, 12.55%.

Preparation of N-(2,6-dichlorophenyl)-2-(3-((hydroxyimino)methyl)-1H-indol-1-yl)acetamide: [3j]

Molecular formula: C17H13N3O2Cl2; yield: (78%); melting point: 225–229 °C; FT-IR (νmax/cm−1): 3071 (Ar-H str), 1536 (Ar-C str), 3328 (NH-str), 1612 (NH-bend), 1739 (C=O-str), 724 (Cl); 1H-NMR (500 MHz, CDCl3, δ ppm): δ 4.53 (s, 2H, CH2-H), δ 6.54 (d, 2H, J = 14.3 Hz, CH-H), δ 6.81 (d,1H, J = 14.1 Hz, Ar-H), δ 7.19 (d, 2H, J = 17.4 Hz, Ar-H), δ 7.59 (s, 4H, Ar-H) δ 8.21 (s, 1H, NH-H), δ 9.09 (s, 1H, NOH-H); ESI-MS (m/z, %): 361 (39) [M+], 363 (27) [M++2], 365 (15) [M++4] 288 (57), 201 (20), 159 (46), 115 (57), 90 (100), 44 (24); anal. calcd for C17H13N3O2Cl2 (361), predicted: C, 56.37; H, 3.62; N, 11.60%. Found: C, 56.17; H, 3.57; N, 11.62%.

Preparation of N-(2,6-dibromophenyl)-2-(3-((hydroxyimino)methyl)-1H-indol-1-yl)acetamide: [3k]

Molecular formula: C17H13N3O2Br2; yield: (62%); melting point: 198–201 °C; FT-IR (νmax/cm−1): 3053 (Ar-H str), 1517 (Ar-C str), 3325 (NH-str), 1618 (NH-bend), 1712 (C=O-str), 527 (Br); 1H-NMR (500 MHz, CDCl3, δ ppm): δ 4.55 (s, 2H, CH2-H), δ 6.57 (s, 1H, Ar-H), δ 6.89 (d, 2H, J = 14.8 Hz, CH-H), δ 7.54 (s, 4H, Ar-H), δ 7.90 (d, 2H, J = 16.2 Hz, Ar-H), δ 8.35 (s, 1H, NH-H), δ 9.07 (s, 1H, NOH-H); ESI-MS (m/z, %): 451 (16) [M+], 453 (34) [M++2], 318 (25), 216 (31), 173 (39), 159 (21), 115 (100), 77 (50), 54 (31); anal. calcd for C17H13N3O2Br2 (451), predicted: C, 45.26; H, 2.90; N, 9.31%. Found: C, 45.40; H, 2.75; N, 9.47%.

Preparation of 2-(3-((hydroxyimino)methyl)-1H-indol-1-yl)-N-O-tolylacetamide: [3l]

Molecular formula: C18H17N3O2; yield: (87%); melting point: 175–177 °C; FT-IR (νmax/cm−1): 3063 (Ar-H str), 1549 (Ar-C str), 3312 (NH-str), 1624 (NH-bend), 1720 (C=O-str); 1H-NMR (500 MHz, CDCl3, δ ppm): δ 2.46 (s, 3H, CH3-H), δ 4.75 (s, 2H, CH2-H), δ 6.43 (d, 2H, J = 11.3 Hz, CH-H), δ 6.59-6.64 (m,1H, Ar-H), δ 7.11 (d, 1H, J = 14.8 Hz, Ar-H), δ 7.32 (s, 1H, Ar-H), δ 7.52 (s, 4H, Ar-H), δ 7.85 (d, 1H, J = 16.1 Hz, Ar-H), δ 8.13 (s, 1H, NH-H), δ 9.03 (s, 1H, NOH-H); ESI-MS (m/z, %): 307 (20) [M+], 254 (30), 201 (19), 159 (39), 115 (100), 89 (20), 52 (39), 40 (25); anal. calcd for C18H17N3O2 (307), predicted: C, 69.61; H, 5.15; N, 14.33%. Found: C, 69.58; H, 5.19; N, 14.42%.

Preparation of 2-(3-((hydroxyimino)methyl)-1H-indol-1-yl)-N-m-tolylacetamide: [3m]

Molecular formula: C18H17N3O2; yield: (92%); melting point: 184–187 °C; FT-IR (νmax/cm−1): 3026 (Ar-H str), 1514 (Ar-C str), 3367 (NH-str), 1608 (NH-bend), 1704 (C=O-str); 1H-NMR (500 MHz, CDCl3, δ ppm): δ 2.70 (s, 3H, CH3-H), δ 4.79 (s, 2H, CH2-H), δ 6.41 (d, 3H, J = 13.5 Hz, CH-H), δ 7.02 (t,1H, J = 10.8 Hz, Ar-H), δ 7.44 (s, 4H, Ar-H), δ 7.67 (s, 1H, Ar-H), δ 7.87 (s, 1H, Ar-H), δ 8.36 (s, 1H, NH-H), δ 9.15 (s, 1H, NOH-H); ESI-MS (m/z, %): 307 (21) [M+], 267 (39), 242 (18), 216 (60), 159 (32), 115 (100), 77 (51), 39 (31); anal. calcd for C18H17N3O2 (307), predicted: C, 69.61; H, 5.15; N, 14.33%. Found: C, 69.29; H, 5.58; N, 14.70%.

Preparation of 2-(3-((hydroxyimino)methyl)-1H-indol-1-yl)-N-p-tolylacetamide: [3n]

Molecular formula: C18H17N3O2; yield: (81%); melting point: 193–197 °C; FT-IR (νmax/cm−1): 3072 (Ar-H str), 1583 (Ar-C str), 3305 (NH-str), 1623 (NH-bend), 1729 (C=O-str); 1H-NMR (500 MHz, CDCl3, δ ppm): δ 2.60 (s, 3H, CH3-H), δ 4.66 (s, 2H, CH2-H), δ 6.43 (d, 2H, J = 13.2 Hz, CH-H), δ 7.01 (d, 2H, J = 16.8 Hz, Ar-H), δ 7.50 (s, 4H, Ar-H), δ 7.79 (d, 2H, J = 17.4 Hz, Ar-H), δ 8.32 (s, 1H, NH-H), δ 9.13 (s, 1H, NOH-H); ESI-MS (m/z, %): 307 (18) [M+], 254 (26), 201 (18), 159 (26), 132 (35), 96 (100), 56 (60), 39 (24); anal. calcd for C18H17N3O2 (307), predicted: C, 69.61; H, 5.15; N, 14.33%. Found: C, 69.72; H, 5.16; N, 14.68%.

Anti-oxidant activity

FRAP method

The method measured the anti-oxidant potency of different N-(substituted phenyl)-2-(3-(hydroxyimino) methyl)-1H-indol-1-yl) acetamide derivative by reducing the Fe3+ into Fe2+ ion. Most of the compounds were shown admirable antioxidant activity. The antioxidant values of some of the tested acetamide derivatives are very closer to standard drugs. The anti-oxidant activity of synthesised compounds was shown in Table 1.

Table 1 The result of antioxidant activity of synthesised compounds using ferric reducing antioxidant power (FRAP)

DPPH method

DPPH is a perfect method to evaluate the antioxidant activity of newly synthesised compounds due to the short time required for the analysis and high reliability. The result reveals that decreases the absorbance due to the antioxidant molecule may supply the electron to the DPPH free radical. Most of the compounds were shown excellent antioxidant activity. The anti-oxidant activity of synthesised compounds was shown in Table 2.

Table 2 The result of antioxidant activity of synthesised compounds with percentage scavenging

Computational analysis

To gain a better understanding of the geometrical structure of the investigated compound, 3D molecular modelling studies have been done. It gives the knowledge of the molecular structure of the compound, geometric optimization and conformational analysis as reported in a recent study [34]. The optimised structure with the atom labelling of a compound 3a is represented in Fig. 2. Some selected bond lengths and angle are listed in Table 3.

Fig. 2
figure 2

Stereochemical structure of (E) N-(2-chlorophenyl)-2-(3-((hydroxyimino)methyl)-1H-indol-1-yl) acetamide [3a]

Table 3 Geometrical data of compound 3a

X-ray crystallography determination and refinement

Diffraction data of the compound 3a were collected from 7351 reflections using X Calibur diffractometer equipped with area detector and a graphite monochromator (λ = 0.83452) at 296 K. The dimension of the crystal employed for data collection was 0.30 × 0.28 × 0.28 mm at 30% probability of selected bond angle and bond length. The refinement was carried out by full-matrix least-squares using SHELXL 97. The different parameters, conditions and data collections of the refinement process of compound 3a were furnished in Table 4. The ORTEP view of the crystal bond angle and bond length was shown in Fig. 3 and Table 5.

Table 4 Crystal data and structural refinement of compound 3a
Fig. 3
figure 3

An ORTEP view of compound 3a at 30% probability level

Table 5 Crystal bond angle and bond length of compound 3a in ORTEP view

To bridge our theoretical computational results with experimental synthesis, we have selected (E) N-(2-chlorophenyl)-2-(3-(hydroxyimino)methyl)-1H-indol-1-yl)acetamide (3a). As shown in Fig. 2, the molecule structure of compound 3a consists of a N-(2-chlorophenyl) acetamide group linked with 1H-indole-3-carbaldehyde oxime. A comparison of optimised geometry and single-crystal XRD structure for compound 3a has been made to see the reliability of our used method. It can be seen that our calculated geometry and all the ring conformation using the B3LYP method match very well with the experiment. As per the 3D molecular structure of compound 3a, the observed bond length and bond angle around C(9)-N(1) and C(9)-N(1)-C(19) were 1.376 A° and 124.87° respectively. Similar kind of geometrical data were collected from the X-ray crystallography study. It is seen that no significant differences in bond length and bond angle were observed in the optimised and experimental structures. Therefore, the obtained results are in a good agreement with the experimental results and hence strongly support them. These studies have provided great insights into the chemical properties such as predicting reactivity, the most available site for substitution, as well as their bioactive conformation of (E) N-(2-chlorophenyl)-2-(3-(hydroxyimino)methyl)-1H-indol-1-yl) acetamide (3a). Comparison of the experimental and the calculated selected bond lengths and bond angles were mentioned in Table 6.

Table 6 Comparison of the selected value of experimental and calculated bond length, bond angle of compound 3a

Discussion

The route of synthesis of indole acetamide derivatives was depicted in the synthetic scheme outlines the preparation part of the synthetic analogues. Here, the key intermediate 1H-indole-3-carbaldehyde oxime was obtained by refluxing the mixture of 1H-indole-3-carbaldehyde and hydroxylamine in ethanol. The synthetic compounds N-(substituted phenyl)-2-(3-(hydroxyimino)methyl)-1H-indol-1-yl) acetamide derivatives were obtained from the interaction of 1H-indole-3-carbaldehyde oxime and 2-chloro acetamide derivatives. The FT-IR and 1H-NMR spectrum of compounds were shown peaks due to different functional groups and proton present in the synthesised compounds. In FT-IR spectrum, strong bands at the region of 623, 724, 689, 724, 578, 615, 636, 527, 968, 1038 and 1137 cm−1could be attributed to the chloro, bromo and fluro group respectively. The aryl ring was raised stretching peak in between 3097–3012 cm−1 and 1502–1593 cm−1. A strong absorption peak at 3369–3305 cm−1 and 1602–1646 cm−1 is due to the presence of the NH group, whereas a strong peak in the region of 1704–1760 cm−1 indicated that the carbonyl group exists in the synthesised compounds. The number of protons present in the analogues was identified by 1H-NMR spectroscopy from the chemical shift. The spectra showed a singlet at δ 2.34–2.82 ppm corresponding to a methyl proton (CH3-H); a singlet at δ 4.39–4.92 ppm corresponding to methylene proton (CH2-H) and a singlet at δ 6.31–6.89 ppm corresponding to CH proton; a singlet at δ 6.98–7.90 ppm corresponding to aromatic protons(Ar-H); a doublet at δ 6.37–8.04 ppm corresponding to aromatic protons (Ar-H); a triplet at δ 6.62–7.81 ppm corresponding to aromatic protons (Ar-H); a multiplet at δ 6.59–7.42 ppm corresponding to aromatic protons (Ar-H); a singlet at δ 8.00–8.39 ppm to amine (-NH-H) and a singlet at δ 8.79-9.21 ppm corresponding to oxime proton (-NOH-H). The mass and element of the synthetic compound were confirmed by the Shimadzu mass spectrometer and Perkin Elmer 2400 CHN elemental analyser. Finally, these synthesised compounds were screened for antioxidant activity using FRPH and DPPH methods. Most of the synthesised compounds were shown promising antioxidant activity by reducing ferric tripyridyl triazine complex and radical scavenging property. Some of the tested acetamide derivatives were found as a potent antioxidant activity as a standard drug. Synthesised compounds in which the halogenated compounds at ortho position were showing better antioxidant activities when compared to meta and para-substituted compounds. Therefore, compounds 3j, 3a, 3k and 3d were constituted with chloro, bromo at the ortho position of the phenyl ring had shown admirable antioxidant activity. Compounds 3b, 3c, 4b and 4c have been constituted with chloro and bromo at the meta/para position of the phenyl ring offered an interesting antioxidant activity, whereas fluro substituted compounds (3g, 3h, 3i) furnished a reasonable level of antioxidant activity. The result of the antioxidant activity of synthesised analogues was denoted in Table 1 and Table 2. The order of antioxidant potency of novel N-(substituted phenyl)-2-(3-(hydroxyimino) methyl)-1H-indol-1-yl) acetamide derivatives was found to be 3j>3a>3k>3d>3b>3c>3e>3f>3n>3m>3l>3g>3h>3i.

Conclusion

In this study, novel N-(substituted phenyl)-2-(3-(hydroxyimino)methyl)-1H-indol-1-yl) acetamide derivatives were synthesised and characterised by FT-IR, 1H-NMR, mass spectroscopy and elemental analysis. Moreover, the antioxidant activities of prepared indole acetamide derivatives were studied by using FRAP and DPPH methods. Compound 3j, 3a and 3k were shown highest activity among the prepared analogues due to the attachment of halogens connected at the appropriate place in the phenyl ring. Hence, these substituted phenyl rings considered as a perfect side chain for the indole nucleus for the development of the new antioxidant agents.

Availability of data and materials

Data and materials are available upon request.

Abbreviations

FT-IR:

Fourier transform infrared spectroscopy

TPTZ:

2,4,6-Tri(2-pyridyl)-s-triazine

1H-NMR:

Nuclear magnetic resonance spectroscopy

ESI-MS:

Electrospray ionisation mass spectrometry

FRAP:

Ferric reducing antioxidant power

DMSO:

Dimethyl sulfoxide

DMF:

Dimethyl formamide

TLC:

Thin-layer chromatography

DPPH:

1,1-Diphenyl-2-picrylhydrazyl

References

  1. Estevao MS, Carvalho LC, Ribeiro D, Couto D, Freitas M, Gomes A, Ferreira LM, Fernandes E, Marques MMB (2010) Antioxidant activity of unexplored indole derivatives: synthesis and screening. Eur J Med Chem 45(11): 4869-4878. http://doi.org/https://doi.org/10.1016/j.ejmech.2010.07.059

  2. Knepper K, Brase S (2003) Bartoli indole synthesis on solid supports. Org Lett 5(16):2829–2832 https://doi.org/https://doi.org/10.1021/ol034851y, https://doi.org/https://doi.org/10.1021/ol034851y

  3. Wales SM, Walker MM, Johnson JS (2013) Asymmetric synthesis of indole homo-Michael adducts via dynamic kinetic Friedel–crafts alkylation with cyclopropanes. Org Lett 15(10):2558–2561https://doi.org/https://doi.org/10.1021/ol4010646. https://doi.org/https://doi.org/10.1021/ol4010646

  4. Singh P, Prasher P, Dhillon P, Bhatti R (2015) Indole based peptidomimetics as anti-inflammatory and anti-hyperalgesic agents: dual inhibition of 5-LOX and COX-2 enzymes. Eur J Med Chem 5(97):104–123. https://doi.org/https://doi.org/10.1016/j.ejmech.2015.04.044

  5. Zajdel P, Marciniec K, Satala G, Canale V, Kos T, Partyka A, Jastrzębska-Więsek M, Wesołowska A, Basinska-Ziobroń A, Wojcikowski J, Daniel WA, Bojarski AJ, Popik P (2016) N1-Azinylsulfonyl-1H-indoles: 5-HT6 receptor antagonists with procognitive and antidepressant-like properties. Med Chem Lett 7(6):618–622. https://doi.org/https://doi.org/10.1021/acsmedchemlett.6b00056.

  6. Zhang M, Jia C, Gu Y, Mulholland N, Turner S, Beattie D, Zhang W, Yang G, Clough J (2017) Synthesis and antifungal activity of novel indole-replaced streptochlorin analogues. Eur J Med Chem 126(27):669–674 https://doi.org/https://doi.org/10.1016/j.ejmech.2016.12.001.

  7. Panathur N, Dalimba U, Koushik PV, Alvala M, Yogeeswari P, Sriram D, Kumar V (2013) Identification and characterization of novel indole based small molecules as anticancer agents through SIRT1 inhibition. Eur J Med Chem 69(1):125–138. https://doi.org/. https://doi.org/10.1016/j.ejmech.2013.08.018

    Article  CAS  PubMed  Google Scholar 

  8. Bandgar BP, Adsul LK, Chavan HV, Jalde SS, Shringare SN, Shaikh R, Meshram RJ, Gacche RN, Masand V (2012) Synthesis, biological evaluation, and docking studies of 3-(substituted)-aryl-5-(9-methyl-3-carbazole)-1H-2-pyrazolines as potent anti-inflammatory and antioxidant agents. Bioorg Med Chem Lett 22(18):5839–5844. https://doi.org/. https://doi.org/10.1016/j.bmcl.2012.07.080

    Article  CAS  PubMed  Google Scholar 

  9. Hu W, Yu H, Chen Y, Wang J (2003) Biological evaluation of an antibiotic DC-81–indole conjugate agent in human melanoma cell lines. Kaohsiung J Med Sci 19(1):6–12 https://doi.org/https://doi.org/10.1016/S1607-551X(09)70441-3.

  10. Lee YJ, Han YR, Park W, Nam SH, Oh KB, Lee HS (2010) Synthetic analogues of indole-containing natural products as inhibitors of sortase a and isocitrate lyase. Bioorg Med Chem Lett 20(23):6882–6885 https://doi.org/https://doi.org/10.1016/j.bmcl.2010.10.029

  11. Andreev IA, Manvar D, Barreca ML, Belov DS, Basu A, Sweeney NL, Ratmanova NK, Lukyanenko ER, Manfroni G, Cecchetti V, Frick DN, Altieri A, Kaushik-Basu N, Kurkin AV (2015) Discovery of the 2-phenyl-4,5,6,7-Tetrahydro-1H-indole as a novel anti-hepatitis C virus targeting scaffold. Eur J Med Chem 96(1):250–258. https://doi.org/ https://doi.org/10.1016/j.ejmech.2015.04.022

  12. Palmerini CA, Tartacca F, Mazzoni M, Granieri L, Goracci L, Scrascia A, Lepri S (2015) Synthesis of new indole-based bisphosphonates and evaluation of their chelating ability in PE/CA-PJ15 cells. Eur J Med Chem 102(1):403–412. https://doi.org/ https://doi.org/10.1016/j.ejmech.2015.08.019

  13. Yadav RR, Khan SI, Singh S, Khan IA, Vishwakarma RA, Bharate SB (2015) Synthesis, antimalarial and antitubercular activities of meridianin derivatives. Eur J Med Chem 98(1):160-169. https://doi.org/ https://doi.org/10.1016/j.ejmch.2015.05.020.

  14. Zhou G, Wu D, Hermel E, Balogh E, Gochin M (2010) Design, synthesis, and evaluation of indole compounds as novel inhibitors targeting Gp41. Bioorg Med Chem Lett 20(5): 1500-1503. https://doi.org/ https://doi.org/10.1016/j.bmcl.2010.01.111.

  15. Xu Q, Huang L, Liu J, Ma L, Chen T, Chen J, Peng F, Cao D, Yang Z, Qiu N, Qiu J, Wang G, Liang X, Peng A, Xiang M, Wei Y, Chen L (2012) Design, synthesis and biological evaluation of thiazole- and indole-based derivatives for the treatment of type II diabetes. Eur J Med Chem 52(1):70–81 https://doi.org/https://doi.org/10.1016/j.ejmech.2012.03.006.

  16. Soares A, Estevao MS, Marques MMB, Kovalishyn V, Latino DARS, Aires-de-Sousa J, Ramos J, Viveiros M, Martins F (2017) Synthesis and biological evaluation of hybrid 1,5- and 2,5-disubstituted indoles as potentially new antitubercular agents. Med Chem 13(5):439–447 https://doi.org/https://doi.org/10.2174/1573406413666170209144003.

  17. França PH, Barbosa DP, Silva DLD, Ribeiro EAN, Santana AEG, Santos BVO, Barbosa-Filho JM, Quintans JSS, Barreto RSS, Quintans-Júnior LJ, Araújo-Júnior JXD (2014) Indole alkaloids from marine sources as potential leads against infectious diseases Biomed Res Int 375423. https://doi.org/https://doi.org/10.1155/2014/375423.

  18. Guerra AS, Malta DJ, Laranjeira LP, Maia MB, Colaço NC, De Lima MC, Galdino SL, Pitta IR, Gonçalves-Silva T (2011) Anti-inflammatory and antinociceptive activities of indole-imidazolidine derivatives. Int Immunopharmacol 11(11):1816–1822 https://doi.org/https://doi.org/10.1016/j.intimp.2011.07.010.

  19. Zhen X, Peng Z, Zhao S, Han Y, Jin Q, Guan L (2015) Synthesis, potential anticonvulsant and antidepressant effects of 2-(5-methyl-2,3-dioxoindolin-1yl)acetamide derivatives. Acta Pharm Sin B 5(4):343–349. https://doi.org/ https://doi.org/10.1016/j.apsb.2015.01.008

  20. Bardiot D, Thevissen K, De Brucker K, Peeters A, Cos P, Taborda CP, McNaughton M, Maes L, Chaltin P, Cammue BP, Marchand A (2015) 2-(2-Oxo-morpholin-3-yl)-acetamide derivatives as broad-spectrum antifungal agents. J Med Chem 58(3):1502–1512 https://doi.org/https://doi.org/10.1021/jm501814x.

  21. Wu J, Song BA, Hu DY, Yue M, Yang S (2012) Design, synthesis and insecticidal activities of novel pyrazole amides containing hydrazone substructures. Pest Manag Sci 68:801–810 https://doi.org/https://doi.org/10.1002/ps.2329

  22. Shakya AK, Kamal M, Balaramnavar VM, Bardaweel SK, Naik RR, Saxena AK, Siddiqui HH (2016) Design, synthesis and evaluation of benzofuran-acetamide scaffold as potential anticonvulsant agent. Acta Pharma 66(3):353–372. https://doi.org/10.1515/acph-2016-0023

    Article  CAS  Google Scholar 

  23. Rani P, Pal D, Hegde RR, Hashim SR (2014) Anticancer, anti-inflammatory, and analgesic activities of synthesized 2-(substituted phenoxy) acetamide derivatives. Biomed Res Int 386473. https://doi.org/10.1155/2014/386473

  24. Koppireddi S, Komsani JR, Avula S, Pombala S, Vasamsetti S, Kotamraju S, Yadla R (2013) Novel 2-(2,4-dioxo-1,3-thiazolidin-5-yl)acetamides as antioxidant and/or anti-inflammatory compounds. Eur J Med Chem 66(1):305–313. https://doi.org/10.1016/j.ejmech.2013.06.005

    Article  CAS  PubMed  Google Scholar 

  25. De P, Yoya GK, Constant P, Bedos-Belval F, Duran H, Saffon N, Daffe M, Baltas M (2011) Design, synthesis, and biological evaluation of new cinnamic derivatives as antitubrculosis agent. J Med Chem 54(5):1449–1461 https://doi.org/https://doi.org/10.1021/jm101510d.

  26. Chiou CT, Lee WC, Liao JH, Cheng JJ, Lin LC, Chen CY, Song JS, Wu MH, Shia KS, Li WT (2015) Synthesis and evaluation of 3-ylideneoxindole acetamides as potent anticancer agents. Eur J Med Chem 98(1):1–12. https://doi.org/10.1016/j.ejmech.2015.04.062

    Article  CAS  PubMed  Google Scholar 

  27. Gopi C, Sastry VG, Dhanaraju MD (2016) Synthesis and spectroscopic characterisation of novel bioactive molecule of 3-(2-substituted)-1H-indol-3-yl)-1-(thiophen-2yl)prop-2-en-1-one chalcone derivatives as effective anti-oxidant and anti-microbial agents. Beni-Seuf Univ J Appl Sci 5:236–243. http://dx.doi.org/https://doi.org/10.1016/j.bjbas.2016.08.004

  28. Mashentseva AA, Seytembetov TS, Adekenov SM, Tuleuov BI, Loiko OP, Khalitova AI (2011) Synthesis and biological activity of the pinostrobin oxime complex compounds with some d-metals. Russ J Gen Chem 81(1):96–101 https://doi.org/https://doi.org/10.1134/S1070363211010142.

  29. Saeedi M, Goli F, Mahdavi M, Dehghan G, Faramarzic MA, Foroumadia A, Shafiee A (2014) Synthesis and biological investigation of some novel sulfonamide and amide derivatives containing coumarin moieties. Iran J Pharm Res 13(3):881–892

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Ma L, Xiao Y, Li C, Xie ZL, Li DD, Wang YT, Ma HT, Zhu HL, Wang MH, Ye YH (2013) Synthesis and antioxidant activity of novel mannich base of 1,3,4-oxadiazole derivatives possessing 1,4-benzodioxan. Bioorg Med Chem 21(21):6763–6770 https://doi.org/https://doi.org/10.1016/j.bmc.2013.08.002.

  31. Kumar A, Varadaraj BG, Singla RK (2013) Synthesis and evaluation of antioxidant activity of novel 3,5-disubstituted-2-pyrazolines. Bull Fac Pharm Cairo Univ 51(2:167–173 https://doi.org/https://doi.org/10.1016/j.bfopcu.2013.04.002.

  32. Dong XW, Wu H, Feng Y, Ma CH, Wang XY, Li YJ (2014) Syntheses, structures and luminescence of silver(I) sulfonate complexes with nitrogen-containing ligands. J Mol Stru 1074:516–521 https://doi.org/https://doi.org/10.1016/j.molstruc.2014.06.053.

  33. Yilmaz ZT, Odabasoglu HY, Senel P, Adimcilar V, Erdogan T, Ozdemir AD, Golcu A, Odabasoglu M, Buyukgungor O (2020) Investigations on crystal structure of a novel 3-((4,6-dimethylpyrimidin-2yl)amino)isobenzofuran-1(3H)-one, and related theoretical studies. Arb J Chem 13:5564–5580 https://doi.org/https://doi.org/10.1016/j.arabjc.2020.03.013.

  34. Kumar V, Upadhyay N, Manhas A (2015) Designing, syntheses, characterization, computatinal study and biological activities of silver-phenothiazine metal complex. J Mol Struct 1099:135–141. https://dx.doi.org/https://doi.org/10.1016/j.molstruc.2015.06.055

Download references

Acknowledgements

All authors wish to express the gratitude to GIET School of Pharmacy, Rajahmundry, Andhra Pradesh, India, for providing research facilities.

Funding

There is no external funding for this project. The authors of this manuscript contributed their money to meet the expenses.

Author information

Authors and Affiliations

Authors

Contributions

CG designed the study, synthesis, characterisation and computational analysis of the titled compounds. MDD has contributed to the major work in the anti-oxidant activity of the synthesised compounds, analysing the data and writing the manuscript in a journal format. All the authors read and approved the final manuscript.

Corresponding author

Correspondence to Chandravadivelu Gopi.

Ethics declarations

Ethics approval and consent to participate

Not applicable

Consent for publication

Not applicable

Competing interests

The authors declare that they have no competing interests.

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:.

Supplementary files

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

Gopi, C., Dhanaraju, M.D. Synthesis and antioxidant properties of 2-(3-(hydroxyimino)methyl)-1H-indol-1-yl)acetamide derivatives. Futur J Pharm Sci 6, 93 (2020). https://doi.org/10.1186/s43094-020-00090-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s43094-020-00090-6

Keywords