397
Views
1
CrossRef citations to date
0
Altmetric
Research Article

A novel synthesis and antimicrobial activity of 1-[(Substituted-phenyl) sulfonyl]pyrrolidin-2-ones

, &
Pages 82-86 | Received 24 Feb 2007, Accepted 28 Mar 2007, Published online: 04 Oct 2008

Abstract

Novel cyclization of 4-(substituted-phenylsulfonamido)butanoic acids to their corresponding 1-[(substituted-phenyl)sulfonyl]pyrrolidin-2-ones was successfully achieved by using polyphosphate ester (PPE). The reaction time was considerably reduced with corresponding increase in the yields, when polyphosphate ester (PPE) was used in combination with 4-(N,N-dimethylamino)pyridine (DMAP). All the synthesized compounds were screened for their antimicrobial activity. Minimum Inhibitory Concentration (MIC) values of synthesized compounds were also determined, and were found to be in the range of 0.09–1.0 mg.

Introduction

The N-substitutedpyrrolidin-2-ones and the pyrrolidinone moiety in the structures of several bioactive compounds have been reported as anti-HIV, antitumor and antifungal agents [Citation1]. They have also been reported for other promising therapeutic applications such as; inhibitors of influenza virus [Citation2], potent and selective potassium channel openers [Citation3], neuroprotective [Citation4] and anti-hypertensive [Citation5] agents. Substituted pyrrolidin-2-ones are also important because of their use as intermediates for the synthesis of γ- amino acids [Citation6] and pyrrolidines [Citation7].

A number of methods have been reported in the literature Citation1-5Citation8Citation9 for the synthesis of substituted pyrrolidin-2-ones. Herein, we report an interesting, cost effective and synthetically useful method for the preparation of N-substitutedpyrrolidin-2-ones (Scheme ).

Scheme 1 Synthesis of the 1-[(substituted-phenyl) sulfonyl] pyrrolidin-2-ones

Scheme 1 Synthesis of the 1-[(substituted-phenyl) sulfonyl] pyrrolidin-2-ones

The synthesized compounds were screened for their antimicrobial activity (Tables I – III).

Table I.  Antibacterial Activity of the Synthesized Compounds 3a–g.

Table II.  Minimum Inhibitory Concentrations (MIC) of the Synthesized Compounds 3a–g in mg/mL.

Table III.  Antifungal Activity of 3a–d and 3f and Inhibition Zones (%).

Materials and methods

General

Melting points were determined on a Gallenkamp digital melting point apparatus and are uncorrected. IR spectra were recorded in KBr disc on a FT-IR model FTS 3000 MX spectrometer. Elemental analysis was performed on a Carlo Erba 1106 elemental analyzer. 1H NMR (300, 400 and 500 MHz) spectra were recorded on a Bruker NMR spectrophotometer. The chemical shifts of proton signals are in parts per million (ppm) downfield from tetramethylsilane (TMS) as internal standard. EI-MS spectra were recorded on MAT 312 and MAT 311A mass spectrometer. Thin layer chromatography (TLC) was performed on pre-coated silica gel 60 F254 aluminum sheets (Merck).

General procedure for the synthesis of 1-[(substituted-phenyl)sulfonyl]pyrrolidin-2-ones (3a–g)

Method A: In this method polyphosphate ester (PPE) was used for the dehydrative cyclization of 4-(substituted-phenylsulfonamido)butanoic acids 2a–g to 1-substituted-pyrrolidin-2-ones 3a–g (Scheme ). A mixture of 4-(substituted-phenylsulfonamido)butanoic acid (2) (1.0 mmol) and polyphosphate ester (2.5 mL) was stirred, under anhydrous conditions, at room temperature for 25–30 h. After completion of the reaction the mixture was a treated with saturated solution of aqueous sodium bicarbonate (25 mL), and extracted with chloroform (3 × 15 mL). The combined extract was washed with brine, water and dried over sodium sulfate (anhydrous). The solvent was distilled off using a rotary evaporator. The oily product was crystallized with absolute ethanol, filtered and recrystallized from chloroform and ethanol (1:5). The compounds 3a–g were purified by preparative TLC on silica plates, using pet. ether: ethylacetate (4:1) as eluent.

Method B: In this method 4-(4-substituted-phenylsulfonamido)butanoic acid (2) (1.0 mmol), polyphosphate ester (PPE)(2.5 mL) and a catalytic amount of DMAP (25 mg) in dry CHCl3 (2 mL), were stirred at room temperature for 15–20 h. The remaining procedure for the synthesis of 1-[(substituted-phenyl)sulfonyl]pyrrolidin-2-ones (3a–g), followed that described in method A. The reaction time was reduced with a corresponding increase in yield, as indicated with each case.

1-[(2-Chlorophenyl)sulfonyl]pyrrolidin-2-one (3a)

Reaction time (method A) 27 h, yield 81%; (method B) 16 h, yield = 89%, m.p = 149°–151°C, UV (λmax, CH3OH, nm): 269, 247. IR (νmax, KBr, cm− 1): 3089 (CH(Ar), 1739 (C = O), 1376, 1163 (SO2). 1H NMR (300 MHz, CDCl3): δ(ppm): 2.36–2.12 (m, 2H, CH2), 2.46 (t, J = 7.0 Hz, 2H, COCH2), 3.88 (t, J = 7.0, 2H, NCH2), 7.63–7.69 (m, 2H, ArH), 7.74 (d, 1H, ArH, J = 7.2 Hz), 7.95 (d, 1H, ArH, J = 7.2 Hz). 13C NMR (75 MHz, CDCl3): (19.1, 32.6, 48.5, 129.3, 129.9, 136.3, 145.8, 173.4. EI(MS; m/z (rel. int. %): 260 (M++1), 195 (69), 175 (100), 112 (14), 111 (45), 76 (26), 75 (9). HRMS (EI, 80 eV): m/z calculated for C10H10NO3SCl: 259.007 Found: 259.009.

1-[(3-Chlorophenyl)sulfonyl]pyrrolidin-2-one (3b)

Reaction time (method A) 29 h, yield 79%; (method B) 18 h, yield = 92%, m.p = 141°–142°C, UV (λmax, CH3OH, nm): 268, 255. IR (νmax, KBr, cm− 1): 3069 (CH(Ar), 1736 (C = O), 1375, 1163 (SO2). 1H NMR (300 MHz, CDCl3): δ(ppm): 2.33–2.08 (m, 2H, CH2), 2.51 (t, J = 7.0 Hz, 2H, COCH2), 3.89 (t, J = 7.0, 2H, NCH2), 7.69 (d, 1H, ArH, J = 6.0 Hz), 7.79 (bs, 1H, ArH), 7.91 (s, 1H, ArH), 7.93–7.96 (m, 1H, ArH). 13C NMR (75 MHz, CDCl3): (18.7, 35.6, 49.5, 129.3, 130.3, 137.1, 145.0, 173.0. EI(MS; m/z (rel. int. %): 260 (M++1), 195 (49), 175 (100), 112 (20), 111 (40), 76 (21), 75 (28). HRMS (EI, 80 eV): m/z calculated for C10H10NO3SCl: 259.007 Found: 259.003.

1-[(4-Chlorophenyl)sulfonyl]pyrrolidin-2-one (3c)

Reaction time (method A) 28 h, yield 78%; (method B) 17 h, yield = 86%, m.p = 153°–155°C, UV (λmax, CH3OH, nm): 266, 242. IR (νmax, KBr, cm− 1): 3055 (CH(Ar), 1735 (C = O), 1375, 1166 (SO2). 1H NMR (250 MHz, CDCl3): δ(ppm): 2.34–2.02 (m, 2H, CH2), 2.40 (t, J = 7.0 Hz, 2H, COCH2), 3.87 (t, J = 7.0, 2H, NCH2), 7.89 (d, J = 8.2 Hz, 2H, ArH), 7.96 (d, J = 8.2 Hz, 2H, ArH). 13C NMR (63 MHz, CDCl3): (18.4, 32.4, 47.5, 128.3, 129.9, 135.3, 145.4, 173.6. EI(MS; m/z (rel. int. %): 260 (M++1), 195 (62), 175 (100), 112 (15), 111 (39), 76 (16), 75 (29). HRMS (EI, 80 eV): m/z calculated for C10H10NO3SCl: 259.007 Found: 259.004.

1-[(4-Methylphenyl)sulfonyl]pyrrolidin-2-one (3d)

Reaction time (method A) 27 h, yield 81%; (method B) 17 h, yield 85%; m.p 145°–147°C. UV (λmax, CH3OH, nm): 261, 244. IR (νmax, KBr, cm− 1): 3057 (CH(Ar), 1728 (C = O), 1352, 1166 (SO2). 1H NMR (500 MHz, Acetone-d6): δ(ppm): 2.05–2.12 (m, 2H, CH2), 2.38 (t, J = 8.0 Hz, 2H, COCH2), 2.43 (s, 3H, CH3), 3.90 (t, J = 7.0, 2H, NCH2), 7.42 (d, J = 8.2 Hz, 2H, ArH), 7.88 (d, J = 8.3 Hz, 2H, ArH). 13C NMR (75 MHz, CDCl3): (18.2, 21.7, 32.2, 47.3, 128.0, 129.7, 135.1, 145.2, 173.4. EI(MS; m/z (rel. int. %): 241 (M++2), 240 (M++1), 177 (3), 176 (39), 175 (90), 174 (90), 157 (3), 156 (6), 155 (62), 139 (16), 121 (29), 120 (89), 119 (4), 118 (4), 93 (2), 92 (29), 91 (100), 89 (17), 65 (54). Anal. Calcd for C11H13NO3S: (239.2899) C, 55.21; H, 5.48; N, 5.85; S, 13.41. Found: C, 55.54; H, 5.29; N, 5.66; S, 13.70%.

1-[(4-Methoxyphenyl)sulfonyl]pyrrolidin-2-one (3e)

Reaction time (method A) 25 h, yield 85%; (method B) 15 h, yield 91%; m.p 151–152oC. UV (λmax, CH3OH, nm): 291, 247. IR (νmax, KBr, cm− 1): 3069 (CH(Ar), 1731 (C = O), 1355, 1161 (SO2). 1H NMR (500 MHz, Acetone-d6): δ(ppm): 2.00–2.03 (m, 2H, CH2), 2.37 (t, J = 7.0 Hz, 2H, CH2CO), 3.80 (t, J = 7.0 Hz, 2H, CH2), 3.86 (s, 3H, OCH3), 7.26 (d, J = 8.2 Hz, 2H, ArH), 7.86 (d, J = 8.2 Hz, 2H, ArH). 13C NMR (63 MHz, CDCl3): δ(ppm): 18.0, 32.6, 47.2, 57.1, 128.0, 129.2, 135.0, 145.1, 173.1. EI(MS; m/z (rel. int. %): 256 (M++1), 192 (54), 172 (16), 171 (100), 109 (4), 108 (4), 107 (39), 76 (29), 65 (54). Anal. Calcd for C11H13NO4S: (255.2993) C, 51.75; H, 5.13; N, 5.49; S, 12.56. Found: C, 51.60; H, 4.98; N, 5.68; S, 12.33%.

1-[(4-Acetamidophenyl)sulfonyl]pyrrolidin-2-one (3f)

Reaction time (method A) 27 hours, yield = 72%; (method B) 19 hours, yield 77%; m.p 155°–156°C. UV (λm*ax, CH3OH, nm): 299, 245. IR (νmax, KBr, cm− 1): 3286 (NH), 1725 (C = O), 1678 (C = O), 1362, 1155 (SO2). 1H NMR (500 MHz, Acetone-d6): δ(ppm): 2.01–2.03 (m, 2H, CH2), 2.38 (t, J = 7.0 Hz, 2H, COCH2), 2.48 (s, 3H, CH3), 3.83 (t, J = 6.8, 2H, NCH2), 7.73 (d, J = 8.0 Hz, 2H, ArH), 7.86 (d, J = 8.2 Hz, 2H, ArH). EI(MS; m/z (rel. int. %): 284 (M++2), 220 (62), 198 (39), 183 (100), 155 (29), 75 (4), 65 (16). Anal. Calcd for C12H14N2O4S: (282.315) C, 51.05; H, 5.10; N, 9.92; S, 11.36. Found: C, 51.36; H, 5.37; N, 9.70; S, 11.29.

1-[(4-Nitrophenyl)sulfonyl]pyrrolidin-2-one (3g)

Reaction time (method A): 30 h, yield 65%; (method B) 20 h, yield 72%; m.p 161°–163°C. UV (λmax, CH3OH, nm): 309, 246. IR (νmax, KBr, cm− 1): 3066 (CH(Ar), 1739 (C = O), 1375, 1166 (SO2). 1H NMR (250 MHz, CDCl3): δ(ppm): 2.01–2.04 (m, 2H, CH2), 2.48 (t, J = 7.3 Hz, 2H, COCH2), 3.92 (t, J = 7.2, 2H, NCH2), 7.88 (d, J = 8.2 Hz, 2H, ArH), 8.48 (d, J = 8.0 Hz, 2H, ArH). 13C NMR (63 MHz, CDCl3): (19.1, 32.5, 49.2, 128.8, 129.8, 135.7, 147.2, 176.5. HRMS (EI, 80 eV): m/z calculated for C10H10NO5S: 270.2604. Found: 270.2648.

Results and discussion

Chemistry

In the present work, 1-[(substitutedphenyl)sulfonyl]pyrrolidin-2-ones 3a–g were prepared from 4-(substitutedphenylsulfonamido)butanoic acids 2a–g in the presence of polyphosphate ester (PPE). The reaction provided 1-[(substituted-phenyl)sulfonyl]pyrrolidin-2-ones 3a–g in excellent yield (65–85%) by simple stirring at room temperature (method A).A second method (method B) was also employed for the synthesis of 1-[(substituted-phenyl)sulfonyl]pyrrolidin-2-ones 3a–g in 72–92% yields. In this method polyphosphate ester (PPE) and 4-(N, N-dimethylamino)pyridine (DMAP) in chloroform (2 mL), were used for the dehydrative cyclization of 4-(Substitutedphenylsulfonamido)butanoic acids 2a–g to the corresponding 1-[(Substituted-phenyl)sulfonyl]pyrrolidin-2-ones 3a–g. It is worth noting here that the reaction time was reduced from 30 to 15 hours. Polyphosphate ester (PPE) and 4-(substitutedphenylsulfonamido)butanoic acids 2a–g were prepared by reported methods [Citation10,Citation11]. The synthesized compounds were characterized by elemental / HRMS, UV, IR, 1H NMR, 13C NMR and mass spectral data.

Antimicrobial activity

The synthesized compounds were tested in vitro for their antibacterial activity against Staphylococcus aureus, Bacillus subtillus, Escherichia coli, Pseudomonas aeruginosa and Micrococcus luteus bacteria by the agar well diffusion method [Citation12]. DMSO was used as a control solvent and, chloramphenicol and cefixime as standard drugs. After 24 h incubation at 37°C, the zone of inhibition was measured in mm. The results are listed in . The results showed that all compounds were active against E. coli. It is worth noting here that compound 3a exhibited significant activity against E. coli (17.6 mm) and B. subtillus (16.15 mm). The other compounds showed moderate to low activity. The structure-activity relationship (SAR) shows that the presence of the chloro group at the 2-position (ortho) of the phenyl substituent enhanced the antibacterial action of the compounds. The minimum Inhibitory Concentration (MIC) values for 3a–g were also determined by the agar well diffusion method [Citation12], and the results are shown in . Five selected compounds 3a–d and 3f were screened in vitro for their antifungal activity against six species using the agar plate technique [Citation13]. The linear growth of the fungus was obtained by measuring the diameter of the fungal colony after seven days. The amount of growth inhibition in each case was calculated as percentage inhibition. The results shown in , indicated that compounds 3a and 3f exhibited significant activity (90%) against Aspergillus flavus and Candida albicans. respectively. It is worth noting that compound 3a exhibited significant (maximum) antibacterial and antifungal activities, possibly due to the presence of chloro group at the 2-position (ortho) of the phenyl substituent, in addition to the sulfonamido moiety.

Conclusions

We have developed an efficient, simple and cost effective method for the synthesis of 1-[(substituted-phenyl)sulfonyl]pyrrolidin-2-ones by using polyphosphate ester (PPE). It is worth noting that reaction times have been reduced with a corresponding increase in the yields, when PPE was used in combination with DMAP (method B). To the best of our knowledge from the existing literature, the reported method for the synthesis of substituted pyrrolidin-2-ones is novel. The antimicrobial studies showed that compound 3a exhibited significant (maximum) antibacterial and antifungal activities.

References

  • Coutrot P, Claudel S, Didierjean C, Grison C. Stereoselective synthesis and glycosidase inhibitory activity of 3,4-dihydroxy-pyrrolidin-2-one, 3,4-dihydroxy-piperidin-2-one and 1,2-dihydroxy-pyrrolizidin-3-one. Bioorg Med Chem Lett 2006; 16: 417–420
  • Brouillette WJ, Bajpai SN, Ali SM, Velu SE, Atigadda VR, Lommer BS, Finely JB, Luo M, Air GM. Pyrrolidinobenzoic acid inhibitors of influenza virus neuraminidase: Modifications of essential pyrrolidinone ring substituents. Bioorg Med Chem 2003; 11: 2739–2749
  • Liang PH, Hsin LW, Cheng CY. N-Arylated pyrrolidin-2-ones and morpholin-3-ones as potassium channel openers. Bioorg Med Chem 2002; 10: 3267–3276
  • Moglioni AG, Brousse BN, Larena AA, Moltrasio GY, Ortuno RM. Stereoselective synthesis of cyclobutyl GABA analogues and related compounds from ( − )–(S)-verbenone. Tetrahedron: Asymm 2002; 13: 451–454
  • Kulig K, Holzgrabe U, Malawska B. Stereocontrolled synthesis of the enantiomers of 1-[2-hydroxy-3-(4-phenyl-1-piperazinyl)-propyl]-pyrrolidin-2-one. Tetrahedron: Asymm 2001; 12: 2533–2536
  • Corey EJ, Zhang FY. Enantioselective Michael addition of nitromethane to r,â-enones catalyzed by chiral quaternary ammonium salts. A simple synthesis of (R)-Baclofen. Org Lett 2000; 15: 4257–4259
  • Xia Q, Ganem B. Asymmetric total synthesis of ( − )-r-Kainic acid using an enantioselective, metal promoted ene cyclization. Org Lett 2001; 3: 485–487
  • Domingos JLO, Lima EC, Dias AG, Costa RR. Stereoselective preparation of pyrrolidin-2-ones from a Z-enoate derived from D-(+)-mannitol. Tetrahedron: Asymm 2001; 15: 2313–2314, and references cited therein
  • Lesniak S, Pasternak B. Cyclisation at very high temperature. Thermal transformations of N-alkyl and N,N-dialkyl cinnamic amides into pyrrolidin-2-ones under FVT conditions. Tetrahedron Lett 2005; 46: 3093–3095
  • Fieser FL, Fieser M. Reagents for organic synthesis. John Wiley and Sons, Inc, USA 1967; 892, and references cited therein
  • Furniss BS, Hannaford AJ, Rogers V, Smith PWG, Tatchell AR. Vogel's textbook of practical organic chemistry4th Ed. Longman, London 1978; 1135
  • Carron RA, Marran JM, Monero L, Femazndozalgo DominguezAA. Plants Medicinal Phyotherap 1987; 21: 195
  • Horsfall JG. Bot Rev 1945; 11: 419

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.