Synthesis and biological evaluation of novel carbazolyl glyoxamides as anticancer and antibacterial agents

Venkataramana Reddy P Oa, Mukund P. Tantaka, Reyna Valdezb, Rajnish Prakash Singhc, Okram Mukherjee Singhd, Rachna Sadana*b and Dalip Kumar*a
aDepartment of Chemistry, Birla Institute of Technology and Science, Pilani 333 031, Rajasthan, India. E-mail: dalipk@pilani.bits-pilani.ac.in; Fax: +91-1596-244183; Tel: +91-1596-515238
bDepartment of Natural Sciences, University of Houston – Downtown, Houston, TX 77002, USA. E-mail: sadanar@uhd.edu; Fax: +1-7132237476; Tel: +1-7132265562
cDepartment of Biological Sciences, Birla Institute of Technology and Science, Pilani 333031, Rajasthan, India
dDepartment of Chemistry, Manipur University, Canchipur, Imphal-795003, Manipur, India

Received 19th December 2015 , Accepted 13th January 2016

First published on 15th January 2016


Abstract

A new library of 24 carbazolyl glyoxamides 14a–x were designed and synthesized from glyoxalic acids and arylamines in the presence of HATU as a coupling reagent under MW irradiation. The synthesized carbazolyl glyoxamides were evaluated for their in vitro anticancer and antibacterial activities. Of the synthesized carbazolyl glyoxamides, compounds 14l and 14q exhibited the most potent cytotoxicity towards a breast cancer cell line with IC50 values of 9.3 and 9.8 μM, respectively. Further, caspase-3 assay for carbazolyl glyoxamides indicated that these compounds induced apoptotic cell death in Jurkat cells. Furthermore, some of the synthesized carbazolyl glyoxamides 14g, 14k, 14l and 14n exhibited comparable or even better antibacterial activity (MIC = 8–16 μg mL−1) than chloramphenicol against the selected bacterial strains.


Introduction

Indole alkaloids are an important class of natural products and medicinally important molecules, which elicit a wide range of biological activities through diverse mechanisms.1,2 Among the indole containing heterocycles, the benzo[b]indole system is a carbazole motif that is widely present in a variety of compounds obtained from natural and commercial sources with anticancer and/or antibacterial properties (Fig. 1).3–6 For example, Fujita et al. investigated carbazole-based hydrazone, HND-007 (1) and related compounds for their in vivo antitumor activity suppressing the growth of various cancer cell lines (IC50 ∼ 1.3–4.6 μM).7 Pyridine fused carbazole derivative, S16020 (2) exhibited potent cytotoxic effects (IC50 = 27.5 nM) by virtue of its DNA intercalative and topoisomerase inhibition properties.8–10 Multidrug resistant tumor cell lines are sensitive to S16020 and it is currently being evaluated in the clinical stages.11,12 Naturally occurring carbazole derivative, granulatimide (3) was isolated from the ascidian Didemnum granulatum and found to display anticancer and antibacterial activities.13 Caulfield and co-workers had patented carbazolyl chalcone 4 for its potential as a tubulin polymerization inhibitor (IC50 = 2 μM).14 Further, carbazole sulfonamide (5), exhibited significant cytotoxicity against leukemia cells (IC50 = 19 nM).15 Nakamura and co-workers reported the isolation of the carbazomycins A–F (6) from Streptoverticillium ehimense H 1051-MY 10 and these carbazole-based congeners were found to possess promising antibacterial and antifungal activities.16–18 The current interest in carbazoles for clinical applications arises mainly due to their high efficiencies against several types of diseases, limited toxic side effects, and complete lack of hematological toxicity.19 In addition to interesting and useful biological applications, carbazole derivatives are also used as organic materials due to their photorefractive, photoconductive, whole transporting and light-emitting properties.20
image file: c5ra27175d-f1.tif
Fig. 1 Carbazole analogues as anticancer and antibacterial agents.

Glyoxamide is an important structural unit found in many biologically active compounds and synthetic drug candidates,21,22 especially those with anticancer and antibacterial properties. For example, indibulin (7) destabilizes tubulin polymerization by arresting tumor cell growth at the G2/M phase. Indibulin is also active in multidrug resistant tumor cell lines and its oral formulation is currently being examined in clinical trials.23 Structurally similar, conscinamides A–C (8) containing an indolic enamide fragment, were isolated from marine sponge Coscinoderma sp and found to display antitumor activity against a human prostate cancer cell line (IC50 = 7.6 μg mL−1)24 and partial cytoprotection against HIV.25 More recently, Singh et al. have described the synthesis of different bis(indole)glyoxamides 9 as potent antibacterial candidates.26 In our continued efforts to find out biologically active indole-based molecules, recently, we have prepared α-cyano bis(indolyl)chalcones,27 2-arylamino-5-(3′-indolyl)-1,3,4-oxadi-azoles,28 5-(3′-indolyl)-1,3,4-thiadiazoles,29 2-arylamino-5-(3′-indolyl)-1,3,4-thiadiazoles30 and indolyl-1,2,4-triazoles31 as potent anticancer agents. Very recently, we have identified 2-(3′-indolyl)-N-arylthiazole-4-carboxamides as antibacterial and anticancer agents.32 Encouraging anticancer and antibacterial activities of glyoxamides and pivotal roles of carbazole scaffold in bioactive compounds prompted us to investigate their new analogues. In this paper, we report a series of 24 carbazolyl glyoxamides 14a–x by incorporating important scaffolds, glyoxamide and carbazole in a single molecule (Fig. 2).


image file: c5ra27175d-f2.tif
Fig. 2 Design of carbazolyl glyoxamides 14a–x.

Results and discussions

Synthesis and characterization

Carbazolyl glyoxamides 14a–x were synthesized from the reaction of carbazole 10 with ethyl chlorooxoacetate in the presence of anhydrous AlCl3 followed by ester hydrolysis using LiOH to afford glyoxalic acid 13 in good yield (Scheme 1). For the coupling of 13 with aryl/heteroaryl amines, we optimized the reaction conditions by varying temperatures, solvents, and reagents as illustrated in Table 1. Initial efforts by using thionyl chloride or oxalyl chloride failed to produce 14a (Table 1, entries 1–2). Subsequent efforts by employing well known carbodiimides including DCC, CDI and EDCI·HCl as coupling reagents under conventional as well as microwave (MW) irradiation conditions generated 14a in low yield (40%, Table 1 entries 3–6). Finally, the reaction of 13a and aryl/heteroarylamines in the presence of O-(7-azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (HATU) at 70 °C under MW irradiation led to 14a in 70% yield. Scope and generality of this developed protocol was further demonstrated by coupling glyoxalic acid 13 with various arylamines and a series of carbazolyl glyoxamides 14a–x was prepared in 70–91% yields. Structures of the newly synthesized glyoxamides were well characterized by using spectroscopic techniques including IR, NMR (1H and 13C) and HRMS. HPLC analysis of synthesized carbazolyl glyoxamides 14a–x indicated the purity of all the compounds was greater than 97%. In IR spectra of 14a–x, characteristic peaks at ∼1690 and 1655 cm−1 were assigned to the carbonyl and amide functionalities, respectively. Further, in 13C NMR spectra of 14a–x, the carbons of carbonyl and amide functionalities were resonated at ∼180 (CO) and ∼160 ppm (NHCO).
image file: c5ra27175d-s1.tif
Scheme 1 Reagents and conditions: (a) CH3I/C2H5I/4-ClC6H4CH2Cl, KOH, DMF, rt, 13 h; (b) Cl(CO)2OEt, AlCl3, DCM, 0 °C to rt, 3 h; (c) LiOH, THF[thin space (1/6-em)]:[thin space (1/6-em)]H2O (1[thin space (1/6-em)]:[thin space (1/6-em)]1), rt, 2 h; (d) R1NH2, HATU, DIPEA, DMF, 70 °C, 45 min, MW.
Table 1 Optimization of reaction conditions for the synthesis of carbazolyl glyoxamide 14aa

image file: c5ra27175d-u1.tif

Entry Reagent Solvent Base Temperature (°C) Conventional heating Microwave irradiation
Time (h) Yield (%) Time (min) Yield (%)
a RT: room temperature; NA: not attempted.
1 SOCl2 DCM TEA RT 12 Trace NA NA
2 (COCl)2 DCM TEA RT 15 Trace NA NA
3 DCC THF TEA RT 30 Trace NA NA
4 CDI THF TEA RT 20 Trace NA NA
5 EDCI·HCl/HOBt THF DIPEA RT 14 30 60 40
6 EDCI·HCl/HOBt DMF TEA 70 12 Trace 60 Trace
7 HATU THF DIPEA 60 15 Trace 60 Trace
8 HATU DMF DIPEA 70 12 40 45 70
9 HATU DMF TEA 70 12 20 45 40


Anticancer activity

Twenty four synthesized glyoxamides were evaluated for their anticancer activities towards human T lymphocyte (Jurkat), histiocytic lymphoma (U937), and breast (MCF-7 and MDA-MB-231) cancer cell lines (Table 2). Structure–activity relationship (SAR) studies of carbazolyl glyoxamides 14a–x were demonstrated by varying aryl/heteroarylamines and substitution at 9-NH position of carbazoles. Initial screening of compounds 14a–x at 10 μM concentration indicated that compounds 14i–m and 14q displayed about 50% cell survival (Table 2). N-Methyl and N-ethyl congeners of the carbazole were reported to possess significant cytotoxicity, therefore, derivatives 14r–x were synthesized.7,15 Unfortunately, analogs 14r–x exhibited low activity against the tested tumor cell lines. The IC50 values of selected carbazolyl glyoxamides 14i–m and 14q are summarized in Table 3. Compound 14i, bearing 4-chlorobenzyl and N,N′-dimethylaminophenyl moieties was found to show moderate activity. Cytotoxicity was retained by the replacement of a phenyl ring in 14a with heteroaryl moieties such as 6-quinolyl (14j) and 2-(5-methyl)thiazolyl (14k).
Table 2 In vitro cytotoxicity of carbazolyl glyoxamides 14a–xa

image file: c5ra27175d-u2.tif

Percentage cell survival (@10 μM)
Compound R R1 Jurkat U937 MCF-7 MDA-MB-231
a The activity data represent mean values ± SD of experiments conducted in triplicates at three independent times.
14a 4-ClC6H4CH2 C6H5 71.5 ± 10.7 88.5 ± 5.2 69.5 ± 7.8 82.3 ± 7.5
14b 4-ClC6H4CH2 4-CH3C6H4 70.2 ± 0.8 79.5 ± 3.4 67.1 ± 7.4 83.5 ± 6.8
14c 4-ClC6H4CH2 4-CH3OC6H4 71.3 ± 26.7 82.1 ± 2.2 74.6 ± 0.9 80.5 ± 5.5
14d 4-ClC6H4CH2 3-CH3OC6H4 71.3 ± 14.4 67.2 ± 8.5 70.9 ± 10.4 78.6 ± 6.8
14e 4-ClC6H4CH2 3,4-(CH3O)2C6H3 69.8 ± 14.7 85.5 ± 5.3 67.3 ± 4.6 72.5 ± 2.2
14f 4-ClC6H4CH2 3,4,5-(CH3O)3C6H2 64.5 ± 1.4 98.8 ± 2.9 73.7 ± 19.9 70.9 ± 5.2
14g 4-ClC6H4CH2 4-FC6H4 118.4 ± 26.4 83.2 ± 6.7 93.3 ± 5.6 88.9 ± 6.4
14h 4-ClC6H4CH2 4-Pyridyl 83.1 ± 19.6 59.1 ± 14.8 66.3 ± 5.4 90.2 ± 2.2
14i 4-ClC6H4CH2 4-(CH3)2N,N-C6H4 56.5 ± 6.9 78.8 ± 1.2 70.5 ± 5.8 62.8 ± 3.5
14j 4-ClC6H4CH2 6-Quinolyl 56.5 ± 1.6 60.3 ± 17.4 68.0 ± 7.0 63.5 ± 2.9
14k 4-ClC6H4CH2 2-(5-Methyl)thiazolyl 51.3 ± 0.5 62.2 ± 5.8 58.4 ± 6.6 64.8 ± 1.9
14l H C6H5 83.4 ± 41.0 64.4 ± 8.7 48.5 ± 4.0 75.2 ± 2.4
14m H 3,4,5-(CH3O)3C6H2 52.4 ± 5.9 67.4 ± 5.8 59.1 ± 13.3 61.3 ± 2.0
14n H 3-CH3OC6H4 60.7 ± 0.7 72.1 ± 6.9 59.8 ± 19.9 72.6 ± 8.3
14o H 4-CH3OC6H4 77.9 ± 9.4 85.6 ± 6.5 89.4 ± 17.3 84.9 ± 10.5
14p H 4-Pyridyl 94.9 ± 6.3 92.8 ± 5.2 60.5 ± 10.6 98.2 ± 5.4
14q H 3,4-(CH3O)2C6H3 60.5 ± 6.2 65.8 ± 8.5 50.0 ± 7.3 65.2 ± 4.5
14r CH3 C6H5 95.31 ± 7.2 109.7 ± 10.4 104.2 ± 6.7 94.7 ± 6.2
14s CH3 3,4,5-(CH3O)3C6H2 99.77 ± 6.6 107.5 ± 9.3 72.1 ± 5.4 92.3 ± 3.9
14t C2H5 C6H5 93.7 ± 5.3 91.3 ± 10.9 114.2 ± 11.3 94.5 ± 5.2
14u C2H5 3,4-(CH3O)2C6H3 92.3 ± 2.2 104.7 ± 9.5 106.9 ± 12.1 96.5 ± 6.6
14v C2H5 3,4,5-(CH3O)3C6H2 100.4 ± 10.8 106.9 ± 7.4 78.1 ± 8.8 88.8 ± 7.1
14w C2H5 4-(CH3)2N,N-C6H4 102.3 ± 9.7 93.6 ± 4.2 79.7 ± 7.6 89.9 ± 9.2
14x C2H5 6-Quinolyl 100.4 ± 10.8 95.7 ± 7.2 73.8 ± 8.5 83.9 ± 8.9
Control (negative) 100 ± 5.9 101.5 ± 9.5 100 ± 3.2 100 ± 2.2
Doxorubicin (positive) 21.2 ± 5.6 31.8 ± 1.1 38.2 ± 10.6 40.2 ± 3.5


Table 3 IC50 values (μM) of selected carbazolyl glyoxamidesa
S. No Jurkat U937 MCF-7 MDA-MB-231
a Bold value indicates IC50 > 10 μM; DX-1 = doxorubicin.
14i 10.5 ± 2.1 29.2 ± 3.9 23.5 ± 8.2 17.9 ± 5.7
14j 11.3 ± 3.6 15.1 ± 4.8 18.9 ± 3.8 18.7 ± 7.7
14k 10.2 ± 2.9 17.5 ± 8.2 12.2 ± 5.4 20.1 ± 8.4
14l 11.8 ± 3.3 18.3 ± 7.8 9.3 ± 4.3 31.2 ± 11.4
14m 12.1 ± 1.8 23.3 ± 10 11.5 ± 5.5 14.7 ± 5.4
14q 17.8 ± 3.2 29.2 ± 9.2 9.8 ± 2.8 18.5 ± 5.4
DX-1 0.25 ± 0.11 0.15 ± 0.05 0.35 ± 0.15 0.5 ± 0.20


Compound 14l with N–H free carbazole and N-phenyl glyoxamide was identified as the most active member of the series with IC50 values between 9.3 to 31.2 μM. Also, compound 14l was found to be 2–3 fold more cytotoxic towards MCF-7 (IC50 = 9.3 μM) and Jurkat (IC50 = 11.8 μM) cells. No significant change in activity was observed by the replacement of a phenyl group in compound 14l with a trimethoxyphenyl (14m) and dimethoxyphenyl (14q) groups except for 14q (IC50 = 9.8 μM; MCF-7). However, compound 14q was found to be equipotent (compounds 14l vs. 14q) against MCF-7 cell line (IC50 = 9.8 μM). From the structural variation it was realized that carbazole–N–H with its appended glyoxamide bearing phenyl, dimethoxyphenyl and trimethoxyphenyl units increases cytotoxicity.

To further characterize the mode of cellular death by carbazolyl glyoxamides, apoptosis induction studies for the selected compounds 14i–m and 14q were performed on Jurkat cells by the caspases 3/7 activation method. Caspases belonging to a family of cysteine proteases are known to play an essential role in apoptosis.33 Out of these caspases, caspase-3 is an effector caspase that cleaves multiple proteins in cells leading to apoptotic cell death. Therefore, activation of caspase 3 pathway is a hallmark of apoptosis and can be used in cellular assay to quantify activator. Of the carbazolyl glyoxamides tested, compounds 14i, 14k, 14l and 14q showed 4–5-fold enhancement in caspase level compared to the control (Fig. 3). These results imply that carbazolyl glyoxamides induced apoptosis in Jurkat cells via caspase-3-dependent pathway.


image file: c5ra27175d-f3.tif
Fig. 3 Carbazolyl glyoxamides 14i–m and 14q induced caspase activation in Jurkat cells.

Antibacterial activity

In light of interesting antibacterial activities of many carbazole containing natural and synthetic compounds, the newly synthesized carbazolyl glyoxamides 14a–x were screened for their antibacterial activity.27 All the compounds were tested for their in vitro antibacterial activities against Gram-positive bacteria including Staphylococcus aureus (MTCC 96) and Bacillus subtilis (MTCC 121), and Gram-negative bacteria including Escherichia coli (MTCC 1652) and Pseudomonas putida (MTCC102) with respect to chloramphenicol, a standard drug. The Minimum Inhibitory Concentrations (MICs) and zone of inhibition (ZOI) for compounds 14a–x were determined in vitro by the modified broth micro-dilution values method as given in Table 4. Compounds containing N-chlorobenzyl carbazole and C6H5 (14a), CH3C6H4 (14b) CH3OC6H4 (14c), (CH3O)3C6H2 (14f) and N,N′-(CH3)2C6H4 (14i) substituents in the glyoxamide fragment were found to display moderate activity against the tested bacterial strains. Interestingly, introduction of an electron-withdrawing fluoro group in the phenyl ring resulted in 14g endowed with potent antibacterial activity against all tested bacterial strains with MIC values ranging between 8 and 16 μg mL−1. Replacement of a N-phenyl ring in 14a with heteroaryl groups such as 4-pyridyl (14h) and 6-quinolyl (14j) led to inactive derivatives; except 14k bearing 2-(5-methyl)thiazolyl moiety exhibited comparable antibacterial activity against B. cereus and E. coli bacterial strains (ZOI = 16–19 mm; MIC = 16 μg mL−1). Compound 14l with carbazole N–H and a phenyl moiety on the amide part displayed improved activity when compared to the corresponding N-substituted carbazole 14a. Replacement of a phenyl ring in 14l by methoxyphenyl (14m–o and 14q) or 4-pyridyl (14p) moiety led to a decrease in the activity except for 14n (MIC = 8–16 μg mL−1). Alkylations of carbazole N–H resulted in compounds 14r–x with moderate activity against the tested bacterial strains.
Table 4 In vitro antibacterial activities of carbazolyl glyoxamides 14a–xa

image file: c5ra27175d-u3.tif

Compound R R1 Gram-positive bacteria Gram-negative bacteria
B. cereus S. aureus E. coli P. putida
ZOI MIC ZOI MIC ZOI MIC ZOI MIC
a ZOI (in mm) and MIC (in μg mL−1) values, bold values indicate comparable or even better antibacterial activity than chloramphenicol.
14a 4-ClC6H4CH2 C6H5 16 32 16 >32 14 64 15 32
14b 4-ClC6H4CH2 4-CH3C6H4 15 >32 16 64
14c 4-ClC6H4CH2 4-CH3OC6H4 16 32 15 32 17 >16 15 >16
14d 4-ClC6H4CH2 3-CH3OC6H4 17 16 14 >16
14e 4-ClC6H4CH2 3,4-(CH3O)2C6H3 15 >64 17 64 15 32 16 >16
14f 4-ClC6H4CH2 3,4,5-(CH3O)3C6H2 15 >32 17 >64 18 32 16 32
14g 4-ClC6H4CH2 4-FC6H4 18 16 18 >8 20 8 19 16
14h 4-ClC6H4CH2 4-pyridyl 15 128 17 >32 16 32 16 32
14i 4-ClC6H4CH2 4-(CH3)2N,N-C6H4 16 64 18 >32 14 32 16 64
14j 4-ClC6H4CH2 6-Quinolyl 15 >32 16 >128 15 32 18 64
14k 4-ClC6H4CH2 2-(5-Methyl)thiazolyl 16 16 19 >8 19 16 18 >16
14l H C6H5 17 16 18 >8 19 >8 17 8
14m H 3,4,5-(CH3O)3C6H2 15 >16 17 >16 16 32 16 64
14n H 3-CH3OC6H4 18 >8 19 16 16 16 18 8
14o H 4-CH3OC6H4 14 64 14 >64 17 128 15 >64
14p H 4-Pyridyl 15 32 16 >64 18 32 16 >32
14q H 3,4-(CH3O)2C6H3 16 128 18 >32 17 >16 14 >64
14r CH3 C6H5 13 >32 13 64 15 32 14 32
14s CH3 3,4,5-(CH3O)3C6H2 14 64 13 32 17 32 15 >32
14t C2H5 C6H5 15 >32 14 64 16 >16 17 >16
14u C2H5 3,4-(CH3O)2C6H3 14 64 16 >32 16 >16
14v C2H5 3,4,5-(CH3O)3C6H2 15 >16 15 >32 14 >64
14w C2H5 4-(CH3)2N,N-C6H4 15 >32 14 >64 16 32 16 32
14x C2H5 6-Quinolyl 12 32 14 32 13 >32 14 64
Chloramphenicol 21 32 21 16 22 16 21 16


Cell viability assay

Although antibacterial (ZOI & MIC) assays might show a potential to kill pathogenic micro-organisms, concentration vs. time curves provide more insight about the rate of antibacterial activity. To determine the rate of bactericidal activity of the two most active compounds 14g and 14l, time-kill studies were performed. The interactive time and the change in the number of microorganisms for compounds 14g and 14l is presented in Fig. 4A–D.
image file: c5ra27175d-f4.tif
Fig. 4 Cell viability assay (A–D) of 14g and 14l against selected bacteria.

The percent population reduction at different time interval was calculated to demonstrate the change of the population of microorganism respect to concentration dependent dose. The compound 14g showed high bacteriostatic effect against E. coli in the first hours of incubation.

Moreover, the highest reduction (85%) in E. coli population was observed at 8 h of incubation with compound 14g (Fig. 4A). Compound 14g was also showed about >70% reduction in viable cells of S. aureus (Fig. 4B). Compound 14l with free carbazole N–H and a phenyl moiety in amidic part led to significant inhibition of bacterial growth after 4 h of incubation. It is evident that within 8 h, compound 14l exhibited almost 80% reduction in the viability of P. putida (Fig. 4C). Similarly, bacteriostatic effect of compound 14l against S. aureus reveals about 70% reduction of viable cell at 8 h of incubation (Fig. 4D). Thus the results of present study revealed that 14g and 14l were capable of inhibiting the bacterial growth within few hours of initial interactions.

The toxicity of potent compounds 14i–m and 14q was evaluated using LDH (lactate dehydrogenase) assay. The LDH activity shows that all the tested compounds 14i–m and 14q exhibited lower toxicity than the standard drug, doxorubicin which justifies the potential use of these compounds as anti-bacterial agents (Fig. 5).


image file: c5ra27175d-f5.tif
Fig. 5 Cytotoxicity induced by carbazolyl glyoxamides in terms of LDH release.

From the structure–activity relationship (SAR) studies of carbazolyl glyoxamides it implies that a combination of carbazole N–H and glyoxamide unit possessing p-fluorophenyl and methoxyphenyl substituents are beneficial for the activity (Fig. 6). All the synthesized compounds demonstrated well to moderate cytotoxicity against a panel of cancer cell lines and excellent to moderate antibacterial activity towards tested bacterial strains. Particularly, compound 14l with carbazole N–H and phenyl moiety in glyoxamide part exhibited potent cytotoxicity and antibacterial activities. Exclusively, with carbazole N–H and dimethoxyphenyl moieties, compound 14q was found to be the most active against the tested cancer cell lines and less potent towards tested bacterial strains. However, compound 14g with N-chlorobenzylcarbazole and fluorophenyl units, and analogue 14n having carbazole N–H and 3-methoxyphenyl moieties, were the most active carbazolyl glyoxamides against the tested bacteria but exhibited low cytotoxicity towards tested cancer cell lines.


image file: c5ra27175d-f6.tif
Fig. 6 Structure–activity relationship of carbazolyl glyoxamides 14a–x.

Conclusions

In summary, we have synthesized various carbazolyl glyoxamides from readily available glyoxalic acids 13 and arylamines by employing HATU as a coupling reagent. Synthesized glyoxamides were assessed for their cytotoxicity which enabled us to identify 14l and 14q as the most potent compounds against MCF-7 cells with IC50 values of 9.3 μM and 9.8 μM, respectively. Preliminary mechanism of action studies indicated that carbazolyl glyoxamides induced apoptosis in Jurkat cells via caspase-3 and -7 activation. In addition, antibacterial activity evaluation led us to compounds 14g, 14k, l and 14n with significant potency against Gram-positive and Gram-negative bacteria (MIC = 8–16 μg mL−1 and ZOI = 16–20 mm). Antibacterial activities of potent compounds 14g, 14k, l and 14n were found to be comparable to the reference drug, chloramphenicol. Cell viability assay revealed that analogues 14g and 14l were capable of inhibiting the bacterial growth within few hours of initial interactions. Interesting activity results indicate that the identified potent carbazolyl glyoxamides 14g and 14l can be exploited further to develop either highly specific or potent antibacterial/anticancer agents, or if required both of these properties could be incorporated in the same molecule.

Experimental

General procedure for the synthesis of carbazolyl glyoxamides (14a–x)

To a 10 mL microwave tube was added carbazole glyoxalic acid 13 (0.275 mmol), HATU (0.12 g, 0.317 mmol), N,N-diisopropylethyl-amine (0.09 g, 0.687 mmol) and an appropriate aryl/heteroarylamine (0.303 mmol) in DMF (2 mL). The tube was sealed with a pressure cap and placed in the microwave cavity. The sample was irradiated for 45 min at 70 °C and then allowed to cool at room temperature. The residue was poured into ice-cold water (30 mL) and stirred for 20 min at room temperature. The solid so obtained was filtered, dried and purified by column chromatography on silica gel using ethylacetate[thin space (1/6-em)]:[thin space (1/6-em)]hexane (3[thin space (1/6-em)]:[thin space (1/6-em)]7) as eluent to give pure 14a–g, 14j–o and 14q–x in excellent yields. Some of the compounds (14h, i and 14p) were crystallized from acetone to obtain pure products in 70–91% yields.

2-(9-(4-Chlorobenzyl)-9H-carbazol-3-yl)-2-oxo-N-phenyl-acetamide (14a)

Yellow solid; yield 70%; mp: 191–193 °C; 1H NMR (400 MHz, CDCl3) δ 9.47 (d, J = 1.3 Hz, 1H), 9.15 (s, 1H), 8.58 (dd, J = 8.8, 1.7 Hz, 1H), 8.25 (dd, J = 7.6, 1.1 Hz, 1H), 7.78 (dd, J = 8.6, 1.0 Hz, 2H), 7.54–7.50 (m, 1H), 7.45 (dd, J = 8.3, 7.5 Hz, 3H), 7.41–7.36 (m, 3H), 7.26 (t, J = 2.6 Hz, 2H), 7.08 (d, J = 8.6 Hz, 2H), 5.55 (s, 2H); 13C NMR (101 MHz, CDCl3) δ 185.9, 160.0, 144.2, 141.2, 136.9, 134.6, 133.7, 129.8, 129.3, 129.2, 127.7, 127.0, 126.2, 125.2, 125.0, 123.6, 123.1, 121.1, 121.1, 120.0, 109.5, 108.8, 46.24; IR (KBr, ν, cm−1): 3335, 3090, 3052, 2916, 1682, 1653, 1589, 1520, 1435, 1307, 1250, 1134, 1011, 825, 795; anal. RP-HPLC tR = 4.641 min, purity 98.55%; HRMS (ESI+) calculated for C27H20ClN2O2 [M + H]+, 439.1213; found 439.1208 and 461.1025 [M + Na]+.

2-(9-(4-Chlorobenzyl)-9H-carbazol-3-yl)-2-oxo-N-(p-tolyl) acetamide (14b)

Yellow solid; yield 72%; mp: 172–173 °C; 1H NMR (400 MHz, CDCl3) δ 9.48 (d, J = 1.6 Hz, 1H), 9.09 (s, 1H), 8.58 (dd, J = 8.8, 1.7 Hz, 1H), 8.27–8.23 (m, 1H), 7.67–7.64 (m, 2H), 7.53–7.49 (m, 1H), 7.44–7.35 (m, 3H), 7.26 (t, J = 2.2 Hz, 3H), 7.24 (s, 1H), 7.09 (d, J = 8.6 Hz, 2H), 5.55 (s, 2H), 2.39 (s, 3H); 13C NMR (101 MHz, CDCl3) δ 186.0, 159.9, 144.2, 141.1, 134.9, 134.6, 134.3, 133.7, 129.9, 129.7, 129.2, 127.7, 126.9, 126.1, 125.1, 123.6, 123.1, 121.1, 121.0, 119.9, 109.5, 108.7, 46.2, 21.0; IR (KBr, ν, cm−1): 3325, 3094, 3055, 2916, 1682, 1651, 1620, 1582, 1520, 1443, 1327, 1265, 1149; anal. RP-HPLC tR = 5.317 min, purity 98.10%; HRMS (ESI+) calculated for C28H21ClN2O2 [M + H]+, 453.1369; found 453.1365 and 475.1183 [M + Na]+.

2-(9-(4-Chlorobenzyl)-9H-carbazol-3-yl)-N-(4-methoxyphenyl)-2-oxoacetamide (14c)

Yellow solid; yield 72%; mp: 168–170 °C; 1H NMR (400 MHz, CDCl3) δ 9.45 (d, J = 1.5 Hz, 1H), 9.04 (s, 1H), 8.56 (dd, J = 8.8, 1.7 Hz, 1H), 8.22 (d, J = 7.7 Hz, 1H), 7.69–7.65 (m, 2H), 7.51–7.47 (m, 1H), 7.42–7.32 (m, 4H), 7.25–7.23 (m, 1H), 7.06 (d, J = 8.5 Hz, 2H), 6.97–6.94 (m, 2H), 5.52 (s, 2H), 3.84 (s, 3H); 13C NMR (101 MHz, CDCl3) δ 186.1, 159.8, 157.0, 144.2, 141.1, 134.6, 133.7, 130.1, 129.7, 129.2, 127.7, 126.9, 126.1, 125.1, 123.6, 123.1, 121.6, 121.1, 121.0, 114.4, 109.5, 108.7, 55.5, 46.2; IR (KBr, ν, cm−1): 3348, 3055, 2924, 1682, 1643, 1620, 1582, 1528, 1443, 1327, 1250, 1149; anal. RP-HPLC tR = 4.368 min, purity 98.67%; HRMS (ESI+) calculated for C28H22ClN2O3 [M + H]+, 469.1319; found 469.1310 and 491.1129 [M + Na]+.

Acknowledgements

The authors acknowledge the financial support received from DBT (No. BCIL/NER-BPMC/2012/1549) New Delhi, India. Dr Rachna Sadana thanks to University of Houston-Downtown (UHD) for providing organized research and creative activity (ORCA) funds.

Notes and references

  1. J. Chen, S. Ahn, J. Wang, Y. Lu, J. T. Dalton, D. D. Miller and W. Li, J. Med. Chem., 2012, 55, 7285–7289 CrossRef CAS PubMed.
  2. S. A. Patil, R. Patil and D. D. Miller, Future Med. Chem., 2012, 4, 2085–2115 CrossRef CAS PubMed.
  3. A. W. Schmidt, K. R. Reddy and H.-J. Knölker, Chem. Rev., 2012, 112, 3193–3328 CrossRef CAS PubMed.
  4. A. Kamal, R. V. C. R. N. C. Shetti, M. J. Ramaiah, P. Swapna, K. S. Reddy, A. Mallareddy, M. P. N. Rao, M. Chourasia, G. N. Sastry, A. Juvekar, S. Zingde, P. Sarma, S. N. C. V. L. Pushpavalli and M. Pal-Bhadra, MedChemComm, 2011, 2, 780–788 RSC.
  5. B. P. Bandgar, L. K. Adsul, H. V. Chavan, S. S. Jalde, S. N. Shringare, R. Shaikh, R. J. Meshram, R. N. Gacche and V. Masand, Bioorg. Med. Chem. Lett., 2012, 22, 5839–5844 CrossRef CAS PubMed.
  6. F.-F. Zhang, L.-L. Gan and C.-H. Zhou, Bioorg. Med. Chem. Lett., 2010, 20, 1881–1884 CrossRef CAS PubMed.
  7. M. Ohira, Y. Iwasaki, C. Tanaka, M. Kuroki, N. Matsuo, T. Kitamura, M. Yukuhiro, H. Morimoto, N. Pang, B. Liu, T. Kiyono, M. Amemiya, K. Tanaka, K. Yoshida, N. Sugimoto, T. Ohshima and M. Fujita, Biochim. Biophys. Acta, 2015, 1850, 1676–1684 CrossRef CAS PubMed.
  8. C. M. Miller and F. O. McCarthy, RSC Adv., 2012, 2, 8883–8918 RSC.
  9. G. Vassal, J.-L. Merlin, M.-J. Terrier-Lacombe, J. Grill, F. Parker, C. Sainte-Rose, G. Aubert, J. Morizet, N. Sévenet, M.-G. Poullain, C. Lucas and C. Kalifa, Cancer Chemother. Pharmacol., 2003, 51, 385–394 CAS.
  10. S. Léonce, V. Perez, M.-R. Casabianca-Pignede, M. Anstett, E. Bisagni, A. Pierré and G. Atassi, Invest. New Drugs, 1996, 14, 169–180 CrossRef.
  11. N. Guilbaud, L. Kraus-Berthier, D. Saint-Dizier, M. Rouillon, M. Jan, M. Burbridge, A. Pierré and G. Atassi, Anti-Cancer Drugs, 1997, 8, 276–282 CrossRef CAS PubMed.
  12. L. Kraus-Berthier, N. Guilbaud, M. Jan, D. Saint-Dizier, M. Rouillon, M. Burbridge, A. Pierre and G. Atassi, Eur. J. Cancer, 1997, 33, 1881–1887 CrossRef CAS PubMed.
  13. E. Conchon, F. Anizon, B. Aboab, R. M. Golsteyn, S. Léonce, B. Pfeiffer and M. Prudhomme, Eur. J. Med. Chem., 2008, 43, 282–292 CrossRef CAS PubMed.
  14. T. Caulfield, M. Cherrier, C. Combeau and P. Mailliet, European Patent 1253141, 2002.
  15. L. Hu, Z.-r. Li, Y. Li, J. Qu, Y.-H. Ling, J.-d. Jiang and D. W. Boykin, J. Med. Chem., 2006, 49, 6273–6282 CrossRef CAS PubMed.
  16. S. Nakamura, K. Yamasaki, M. Kaneda, K. Watanabe, Y. Ueki, K. Ishimaru, R. Nomi, N. Yoshida and T. Nakajima, J. Antibiot., 1983, 36, 552–558 CrossRef.
  17. H.-J. Knölker, Top. Curr. Chem., 2005, 244, 115–148 Search PubMed.
  18. A. Głuszyńska, Eur. J. Med. Chem., 2015, 94, 405–426 CrossRef PubMed.
  19. C. Auclair, Arch. Biochem. Biophys., 1987, 259, 1–14 CrossRef CAS PubMed.
  20. Y. Tao, C. Yang and J. Qin, Chem. Soc. Rev., 2011, 40, 2943–2970 RSC.
  21. S. Álvarez, R. Álvarez, H. Khanwalkar, P. Germain, G. Lemaire, F. Rodríguez-Barrios, H. Gronemeyer and Á. R. de Lera, Bioorg. Med. Chem., 2009, 17, 4345–4359 CrossRef PubMed.
  22. L. Gupta, A. Talwar, Nishi, S. Palne, S. Gupta and P. M. S. Chauhan, Bioorg. Med. Chem. Lett., 2007, 17, 4075–4079 CrossRef CAS PubMed.
  23. A. Wienecke and G. Bacher, Cancer Res., 2009, 69, 171–177 CrossRef CAS PubMed.
  24. Y. Ma, K. Yakushijin, F. Miyake and D. Horne, Tetrahedron Lett., 2009, 50, 4343–4345 CrossRef CAS.
  25. H. R. Bokesch, L. K. Pannell, T. C. McKee and M. R. Boyd, Tetrahedron Lett., 2000, 41, 6305–6308 CrossRef CAS.
  26. P. Singh, P. Verma, B. Yadav and S. S. Komath, Bioorg. Med. Chem. Lett., 2011, 21, 3367–3372 CrossRef CAS PubMed.
  27. D. Kumar, N. Maruthi Kumar, M. P. Tantak, M. Ogura, E. Kusaka and T. Ito, Bioorg. Med. Chem. Lett., 2014, 24, 5170–5174 CrossRef CAS PubMed.
  28. M. P. Tantak, A. Kumar, B. Noel, K. Shah and D. Kumar, ChemMedChem, 2013, 8, 1468–1474 CrossRef CAS PubMed.
  29. D. Kumar, N. Maruthi Kumar, K.-H. Chang and K. Shah, Eur. J. Med. Chem., 2010, 45, 4664–4668 CrossRef CAS PubMed.
  30. D. Kumar, N. Maruthi Kumar, B. Noel and K. Shah, Eur. J. Med. Chem., 2012, 55, 432–438 CrossRef CAS PubMed.
  31. D. Kumar, N. Maruthi Kumar, K. H. Chang and K. Shah, Chem. Biol. Drug Des., 2011, 77, 182–188 CAS.
  32. M. P. Tantak, J. Wang, R. P. Singh, A. Kumar, K. Shah and D. Kumar, Bioorg. Med. Chem. Lett., 2015, 25, 4225–4231 CrossRef CAS PubMed.
  33. N. A. Thornberry, Br. Med. Bull., 1997, 53, 478–490 CrossRef CAS PubMed.

Footnote

Electronic supplementary information (ESI) available. See DOI: 10.1039/c5ra27175d

This journal is © The Royal Society of Chemistry 2016