Research Article | Volume: 10, Issue: 9, September, 2020

One-pot three-component synthesis of new triazolopyrimidine derivatives bearing indole moiety as antiproliferative agents

Mohamed A. A. Radwan Fahad M. Alminderej Hala E. M. Tolan Hanem M. Awad   

Open Access   

Published:  Sep 04, 2020

DOI: 10.7324/JAPS.2020.10902
Abstract

A new series of triazolopyrimidine derivatives was produced via three-component reactions of suitable aromatic or heteroaromatic carboxaldehyde, 3-amino-1,2,4-triazole, and 3-indolyl-3-oxopropanenitrile in triethylamine as a catalyst. The new compounds have been interpreted using elemental analysis, infrared, mass spectrometry, and nuclear magnetic resonance spectroscopy. Antiproliferative effects of the new compounds have been screened on four human cancer types and one human noncancerous type (retinal pigment ephitilial-1) via the 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide assay. Compounds 4a and 4h have moderate activity against the human colon cancer; most of the compounds were active toward human lung cancer; compounds 4i, 4h, and 4g were highly active on hormone-dependent human breast cancer, while compounds 4c, 4b, 4h, and 4e were the most active on the hormone-independent human breast. The results of this study offer a source for further investigation of selected triazolopyrimidine derivatives as antiproliferative agents.


Keyword:     Triazole pyrimidine indole multicomponent reaction antiproliferative.


Citation:

Radwan M, Alminderej F, Tolan H, Awad H. One-pot three-component synthesis of new triazolopyrimidine derivatives bearing indole moiety as antiproliferative agents. J Appl Pharm Sci, 2020;10(09):012-022.

Copyright: © The Author(s). This is an open-access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

HTML Full Text

INTRODUCTION

The construction of both scaffolds triazole and pyrimidine rings with a built-in indole moiety might have promising biological activity. Multicomponent reactions (MCRs) are a wide range of synthetic techniques that can be used to achieve this target candidate (Cioc et al., 2014; Dekamin et al., 2013; Liu et al., 2016; Peng et al., 2017; Ravichandiran et al., 2017; Rotstein et al., 2014). Compared to the direct synthesis method, the MCR method is appreciated for its cost-effectiveness, ease of set-up, and high production. Currently, many nitrogen heterocyclic structures (mainly indole units with multiple biological activities) are prepared by MCRs (Allen et al., 2017; Anand et al., 2017; Carbajales et al., 2017; Gribble, 2000). The unique indole backbone is considered to be the most widespread compound that is common in medical advances. Similarly, an indole is a substructure of many natural medical activities (Gribble, 2010). In addition, several indole compounds have attracted more consideration owing to their biological properties, such as their antimicrobial, anticancer, and antiviral activities (Bao et al., 2005; Endo et al., 2007; Sakemi and Sun, 1991; Yang et al., 2004). In such an indole, a framework, triazole (Duran et al., 2002; Gujjar et al., 2009; Holla et al., 2005; Sztanke et al., 2008) and pyrimidine (Kim et al., 2012; Selvaraj and Rajesh, 2016) ring, is well identified as the key structure of several therapeutic scaffolds as they display diverse bioactivities. Fused heterocycles are widely considered due to their share in the different steps of drug discovery, and they often function as basic central units in many therapeutic chemistry programs (Coxon et al., 2017; Taylor et al., 2014). Among them, the triazolopyrimidinone unit, a purine analog, is well known for displaying some biological activities, such as anti-inflammatory activity, ulcerogenic properties, serotonin antagonist, analgesic, antimicrobial, antifungal, cytotoxic, and antitumor activities (Boechat et al., 2012; Hafez et al., 2008; Huang et al., 2012; Lakamoska et al., 2009; Wang et al., 2017; Zhang et al., 2007; Zhao et al., 2007). Moreover, triazolopyrimidines have the greatest valuable construction base for the synthesis of bioactive drugs (Fig. 1) (Łakomska et al., 2013), which possess many pharmacological measures, such as antitumor (Marwaha et al., 2012), antimalarial (Luo et al., 2013), antimicrobial, (Abdel-Aziem et al., 2012), and anti-inflammatory activities (Khera et al., 2011), inhibition of the kinase insert domain-covering receptor (kill/death ratio kinase) (Ashour et al., 2013), and antifungal properties (Chen et al., 2008). They are known to show anticancer, anti-Alzheimer’s, antihypertensive, leishmanicidal cardiac stimulant, antipyretic, potential herbicidal, and anti-HBV activities (Gujjar et al., 2009). The previous biological history of triazolopyrimidine is evident in Trapidil (Fischer, 2008) and Cevipabulin (El-Gendy et al., 2008). Recently, triazolopyrimidines have been realized to modify AChE inhibition, amyloid βaggregation inhibition, anti-inflammation, and are noted as a probable anti-Alzheimer’s agent (Kumar et al., 2016). Generally, heterocyclic compounds have many applications (Adole et al., 2019, 2020; Bisht et al., 2018; Chobe et al., 2014; Kamble et al., 2016). As an extension of our work regarding the direction on the synthesis of bioactive indolyl molecules (El-Nezhawy et al., 2016, 2009a, 2009b; Fakhr et al., 2004, 2009; Ghorab et al., 2008; Muhammad et al., 2019; Radwan and El-Sherbiny, 2007; Radwan and Eman, 2009; Radwan et al., 2007; Radwan et al., 2009a, 2009b, 2020) (Fig. 2), we describe herein a multicomponent synthesis and antitumor estimation of the novel triazolopyrimidine-6-carbonitrile derivatives via the reaction of 3-aminotriazole, suitable aldehydes, and 3-cyanoacetyl indole.


MATERIALS AND METHODS

Chemicals were obtained from Merck and Sigma-Aldrich. Melting point (°C) was measured on the XT-5 microscopic apparatus. C, H, and N analyses and infrared (IR) spectra, which were measured using an iS10 spectrometer (ν in cm−1) using KBr disk, were carried out at Cairo University. MS (EI) m/z analysis was carried out via a Thermo Scientific DCQII. 1H and 13C nuclear magnetic resonance (NMR) were carried out in dimethyl sulfoxide (DMSO-d6) on a Bruker (400 MHz) Ascend - Magnets | Bruker spectrometer.

Figure 1. Biological activity of some of the triazolopyrimidine compounds.

[Click here to view]

Figure 2. Some of our previous and present works bearing indole moiety.

[Click here to view]

Synthesis of 7-substituted-5-(1H-indol-3-yl)-[1,2,4]triazolo[1,5-a]pyrimidine-6-carbonitrile 4a-i

General method

Triethylamine (0.5 mmol) was added to a mixture of aldehydes (1 mmol), 3-amino-1,2,4-triazole (1 mmol), and 3-cyanoacetyl-indole (1 mmol) in dimethyl-formamide (DMF) (5 ml), and heated at 120°C for about 10 hours (observed by thin layer chromatography). Then, the mixture was cooled and the precipitate was recrystallized from Ethanol/Dimethyl-Formamide (EtOH/DMF).

7-(4-Chlorophenyl)-5-(1H-indol-3-yl)-[1,2,4]triazolo[1,5-a]pyrimidine-6-carbonitrile (4a)

Yield 76%; mp 281°C–3°C. IR (KBr, cm−1): 3,312 (NH), 2,212 (CN). 1H NMR (DMSO-d6) δ/ppm: 7.23–7.27 (m, 2H, indole H-5, H-6), 7.51–7.53 (dd, J = 1.1, 8.4 Hz, 1H, indole H-7), 7.63–7.64 (d, J = 7.6 Hz, 2H, Ph), 8.03–8.04 (d, J = 7.6 Hz, 2H, Ph), 8.13 (dd, J = 1.1, 8.4 Hz, 1H, indole H4), 8.20 (s, 1H, triazole), 8.43 (s, 1H, indole-H2), 12.30 (brs, NH); 13C NMR (DMSO-d6) δ/ppm: 153.08, 149.44, 138.76, 136.72, 136.30, 131.34, 129.23, 126.04, 123.60, 122.46, 121.30, 117.38, 113.48, 112.49, 112.09; MS (EI) m/z %: 371 [M++1]. Anal. calcd. for C20H11ClN6: C, 64.78; H, 2.99; N, 22.67. Found: C, 64.89; H, 2.91; N, 22.61.

5-(1H-Indol-3-yl)-7-(4-nitrophenyl)-[1,2,4]triazolo[1,5-a]pyrimidine-6-carbonitrile (4b)

Yield 76%; mp 288°C–9°C. IR (KBr, cm−1): 3,232 (NH), 2,223 (CN). 1H NMR (DMSO-d6) δ/ppm: 7.23–7.28 (m, 2H, indole H5, H6), 7.52 (dd, J = 1.1, 8.6 Hz, 1H, indole H7), 8.14 (dd, J = 1.1, 8.4 Hz, 1H, indole H4), 8.16–8.20 ( dd, J = 1.1, 8.1 Hz, 2H, Ph), 8.31 (s, 1H, triazole), 8.36–8.38 (dd, J = 1.1, 8.1 Hz, 2H, Ph), 8.47 (s, 1H, indole-H2), 12.33 (brs, NH); 13C NMR (DMSO-d6) δ/ppm: 153.33, 147.66, 139.62, 135.90, 136.79, 132.13, 131.10, 125.95, 124.04, 122.62, 121.31, 116.76, 115.10, 113.42, 112.54; MS (EI) m/z (%) 383 [M++2]. Anal. calcd. for C20H11N7O2: C, 62.99; H, 2.91; N, 25.71. Found: C, 63.12; H, 2.93; N, 25.45.

7-(4-Bromophenyl)-5-(1H-indol-3-yl)-[1,2,4]triazolo[1,5-a]pyrimidine-6-carbonitrile (4c)

Yield 74%; mp 279°C–81°C. IR (KBr, cm−1): 3,232 (NH), 2,223 (CN). 1H NMR (DMSO-d6) δ/ppm: 7.23–7.26 (m, 2H, indole H5, H6), 7.50 (dd, J = 1.1, 8.3 Hz, 1H, indole H7), 7.62–7.64 (dd, J = 7.8 Hz, 2H, Ph), 8.02–8.03 (dd, J = 7.8 Hz, 2H, Ph), 8.12 (dd, J = 1.1, 8.3 Hz, 1H, indole H4), 8.21 (s, 1H, triazole), 8.44 (s, 1H, indole-H2), 12.28 (brs, NH); 13C NMR (DMSO-d6) δ/ppm: 151.72, 147.12, 137.74, 135.30, 132.18, 132.00, 131.64, 126.02, 125.85, 123.61, 122.45, 121.32, 117.40, 112.51, 112.12; MS (EI) m/z (%) 414 [M+, 4]; 3,286 (NH), 2,204 (CN). Anal. calcd. for C20H11BrN6: C, 57.85; H, 2.67; N, 20.24. Found: C, 57.90; H, 2.63; N, 20.19.

7-(2-Hydroxyphenyl)-5-(1H-indol-3-yl)-[1,2,4]triazolo[1,5-a]pyrimidine-6-carbonitrile (4d)

Yield 67%; mp 271°C–3°C. IR (KBr, cm−1): 3,210–3,324 (OH and NH), 2,210 (CN). 1H NMR (DMSO-d6) δ/ppm: 7.21–7.27 (m, 2H, indole H5, H6), 7.32–7.39 (m, 4H, Ph), 7.48 (dd, J = 1.1, 8.1 Hz, 1H, indole H7), 7.96–7.99 (dd, 2H, dd, J = 1.1, 8.1 Hz, 1H, indole H4), 8.23 (s, 1H, triazole), 8.41 (s, 1H, indole-H2), 12.19 (s, OH), 12.24 (s, NH); 13C NMR (DMSO-d6) δ/ppm: 155.12, 153.67, 148.36, 141.23, 137.87, 136.90, 135.41, 133.16, 131.27, 126.54, 123.69, 122.74, 121.77, 117.90, 113.56, 112.61, 112.13; MS (EI) m/z (%) 352 [M+]. Anal. calcd. for C20H12N6O: C, 68.18; H, 3.43; N, 23.85. Found: C, 68.25; H, 3.21; N, 23.81.

5-(1H-Indol-3-yl)-7-(2-nitrophenyl)-[1,2,4]triazolo[1,5-a]pyrimidine-6-carbonitrile (4e)

Yield 70%; mp 291°C–3°C. IR (KBr, cm−1): 3,202 (NH), 2,206 (CN). 1H NMR (DMSO-d6) δ/ppm: 7.29–7.36 (m, 2H, indole H5, H6), 7.47 (dd, J = 1.1, 8.2 Hz, 1H, indole H7), 7.73–7.78 (m, 2H, Ph), 7.96–7.98 (m, 2H, Ph), 8.02–8.03 (dd, J = 1.1, 8.2 Hz, 1H, indole H4), 8.24 (s, 1H, triazole), 8.40 (s, 1H, indole-H2), 12.24, (s, NH); 13C NMR (DMSO-d6) δ/ppm: 150.98, 146.82, 145.66, 138.65, 137.96, 135.92, 135.31, 132.36, 128.22, 127.02, 124.63, 123.47, 122.31, 118.37, 114.49, 112.11, 111.95. MS (EI) m/z (%) 384 [M++2]. Anal. calcd. for C20H11N7O2: C, 62.99; H, 2.91; N, 25.71. Found: C, 63.17; H, 2.90; N, 25.41.

5-(1H-Indol-3-yl)-7-(thiophen-2-yl)-[1,2,4]triazolo[1,5-a]pyrimidine-6-carbonitrile (4f)

Yield 64%; mp 266°C–8°C. IR (KBr, cm1): 3,245 (NH), 2,213 (CN). 1H NMR (DMSO-d6) δ/ppm: 7.20–7.23 (m, 2H, thiophene), 7.31–7.32 (m, 2H, indole H5, H6), 7.46 (dd, J = 1.1, 8.4 Hz, 1H, indole H7), 7.53 (s, 1H, thiophene), 7.99–8.03 (dd, J = 1.1, 8.4 Hz, 1H, indole H4), 8.26 (s, 1H, triazole), 8.45 (s, 1H, indole-H2), 12.28, (s, NH); 13C NMR (DMSO-d6) δ/ppm: 146.40, 137.76, 135.52, 134.38, 134.21, 129.51, 127.12 124.50, 123.31, 122.42, 117.14, 114.73, 113.44, 107.65; MS (EI) m/z (%) 342 [M+]. Anal. calcd. for C18H10N6S: C, 63.15; H, 2.94; N, 24.55; S, 9.36. Found: C, 63.21; H, 2.91; N, 24.52; S, 9.34.

7-(Furan-2-yl)-5-(1H-indol-3-yl)-[1,2,4]triazolo[1,5-a]pyrimidine-6-carbonitrile (4g)

Yield 64%; mp 269°C–71°C. IR (KBr, cm−1): 3,232 (NH), 2,214 (CN). 1H NMR (DMSO-d6) δ/ppm: 7.11(m, 1H, furan), 7.32–7.34 (m, 2H, indole H5, H6), 7.47 (dd, J = 1.1, 8.5 Hz, 1H, indole H7), 7.51(m, 1H, furan), 7.98 (m, 1H, furan), 8.02–8.03 (dd, J = 1.1, 8.5 Hz, 1H, indole H4), 8.22 (s, 1H, triazole), 8.43 (s, 1H, indole-H2), 12.17 (s, NH); 13C NMR (DMSO-d6) δ/ppm: 149.80, 147.64, 138.46, 136.12, 127.20, 124.50, 123.30, 121.32, 120.41, 118.74, 115.01, 114.64, 113.44, 106.72. MS (EI) m/z (%) 326 [M+]. Anal. calcd. for C18H10N6O: C, 66.25; H, 3.09; N, 25.75. Found C, 66.31; H, 3.01; N, 25.83.

5-(1H-Indol-3-yl)-7-(1H-pyrrol-2-yl)-[1,2,4]triazolo[1,5-a]pyrimidine-6-carbonitrile (4h)

Yield 67%; mp 258°C–61°C. IR (KBr, cm−1): 3,221 (NH), 2,216 (CN). 1H NMR (DMSO-d6) δ/ppm: 6.21–6.23 (m, 1H, pyrrole-H3), 6.51–6.52 (d, 1H, pyrrole-H4), 7.02–7.03 (d, 1H, pyrrole-H2), 7.31–7.34 (m, 2H, indole H5, H6), 7.43 (dd, J = 1.1, 8.6 Hz, 1H, indole H7), 8.01–8.02 (dd, J = 1.1, 8.6 Hz, 1H, indole H4), 8.20 (s, 1H, triazole), 8.41 (s, 1H, indole-H2), 11.64 (s, NH), 12.11 (s, NH); 13C NMR (DMSO-d6) δ/ppm: 145.97, 144.34, 137.86, 136.12, 127.13, 124.43, 123.30, 123.08, 122.11, 118.06, 114.36, 113.88, 111.20, 105.01; MS (EI) m/z (%) 327 [M++2]. Anal. calcd. for C18H11N7: C, 66.45; H, 3.41; N, 30.14. Found: C, 66.53; H, 3.37; N, 30.12.

5,7-di(1H-Indol-3-yl)-[1,2,4]triazolo[1,5-a]pyrimidine-6-carbonitrile (4i)

Yield 61%; mp 296°C–8°C. IR (KBr, cm−1): 3,236–3,265 (2NH), 2,206(CN). 1H NMR (DMSO-d6) δ/ppm: 7.31–7.35 (m, 4H, di-indole H5, H6), 7.48 (dd, J = 1.1, 8.1 Hz, 2H, di-indole H7), 8.01–8.02 (dd, J = 1.1, 8.1 Hz, 2H, di-indole H4), 8.18 (s, 1H, triazole), 8.31, 8.32 (ss, 2H, indole-H2), 12.21 (s, NH), 12.23 (s, NH); 13C NMR (DMSO-d6) δ/ppm: 152.06, 151.52 137.81, 137.74, 135.31, 132.33, 131.10, 128.24, 127.03, 124.95, 124.61, 123.47, 122.31, 118.39, 114.47, 112.23, 111.16; MS (EI) m/z (%) 375 [M+]. Anal. calcd. for C22H13N7: C, 70.39; H, 3.49; N, 26.12. Found: C, 70.45; H, 3.42; N, 26.10.

In vitro cell culture

The human colorectal carcinoma (HCT-116) and human breast adenocarcinoma (MCF-7), (MDA-MB-231), and (A549) cell lines were acquired from the American Type Culture Collection (ATCC, Rockville, MD) and preserved in the DMEM-F12 medium which was complemented with 10% heat-inactivated fetal bovine serum, 100 U/ml penicillin, and 100 U/ml streptomycin. The cells were grown at 37°C in a moistened atmosphere of 5% CO2.

3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide (MTT) antiproliferative assay

The antiproliferative activity on HCT-116, MCF-7, MDA-MB-231, and A549 human cancer cells and retinal pigment ephitilial-1 (RPE-1) human normal cells was assessed by the MTT assay, which depends on the reduction of the tetrazolium salt by mitochondrial dehydrogenases in feasible cells (Ashraf et al., 2017; Emam et al., 2017; Eman et al., 2017). Cells were first distributed in a 96-well sterile microplate (2 × 104 cells/well) and incubated at 37°C in DMSO with diverse concentrations of each established compound or doxorubicin for 48 hours in a serum-free medium. After incubation, the media were carefully discarded, and 40 μl of MTT (2.5 mg/ml) was added to individual wells and then incubated for an extra 4 hours. The purple formazan dye crystals were solubilized by adding 200 μl of DMSO. The absorbance was measured at 570 nm via a SpectraMax Paradigm multimode microplate reader. The comparative cell viability was stated as the mean ratio of viable cells related to the unprocessed control cells. All trials were conducted in triplicate and repeated on three diverse days. The values were signified as mean ± SD. The efficacious doses for 50% of the cancerous cells population (ED50) and their toxic doses in 50% of the noncancerous cells population were evaluated by Probit analysis by the Statistical Package for the Social Sciences (IBM Corp., Armonk, NY).

Scheme 1. Synthesis of triazolopyrimidine under different conditions of base, solvent, temperature, and time.

[Click here to view]


RESULTS AND DISCUSSION

Chemistry

The strategy in the presented work is the synthesis of a new series of triazolopyrimidine derivatives via an MCR of proper carboxaldehydes 1, 3-amino-1,2,4-triazole 2, and 3-indolyl-3-oxopropanenitrile 3 (Scheme 1 and Scheme 2).

The MCR was carried out in the presence of different solvents and bases to check the effect on the rate of reaction and to find out the standard optimized reaction conditions

When the MCR was completed in ethanol without a catalyst, despite heating for 12 hours (Table 1, entry 1), there was no product formation.

Subsequently, we catalyzed a reaction by triethylamine in different solvents (Table 1, entries 1–6). Then, we studied the reactions in various basic conditions (Table 1, entries 7–12). The reaction results showed that organic bases gave good results, while the yield of the reaction in the presence of inorganic strong bases, like KOH and NaOH, is not satisfactory. Surprisingly, K2CO3 did not furnish the product.In conclusion, the best condition for this reaction is to soak in DMF in 0.25 mol triethylamine for 10 hours at 120°C (Table 1, entry 6).

As a result of the overhead reaction, a possible mechanism was proposed for the construction of triazolopyrimidine. First, triethylamine activated the nucleophilic reaction of 3-indolyl-3-oxopropanenitrile 3 via hydrogen bonding to excite carboxaldehyde 1. The initiation of carboxaldehyde by H-bond enhances the electrophilicity of the carboxaldehyde and improves the building of a transitional A (Reddi et al., 2015). Intermediate A and 3-amino-1,2,4-triazole 2 then undergo Michael’s reaction to form intermediate B via intramolecular cyclization reaction to form a C–N bond. Subsequently, compound 4 was obtained by autoxidation (Scheme 3).

The structure of the newly synthesized compounds (4a–i) was identified via the interpretation of spectral studies and mass analysis (experimental units), e.g., the IR of compound 4a exposed bands at 3,312 and 2,212 cm−1 for the NH and cyano functions, respectively, along with the disappearance of NH2 and C=O groups. Furthermore, 1H NMR showed a broad peak (exchangeable D2O) at 12.27 ppm of the −NH group, a single peak at 8.20 ppm of the triazole proton, and aromatic signals at 7.23–8.43 ppm. Also, 13C NMR data and mass reinforced a proposed structure for 4a (m/z 370, M+).

Scheme 2. Reaction conditions: aldehyde 1 (1 mmol), 3-amino-1,2,4-triazole 2 (1 mmol), 3-cyanoacetyl indole 3 (1 mmol), and Et3N (0.25 mmol), in DMF at 120°C, 10 hours.

[Click here to view]

Table 1. Optimization of the reaction conditions for the synthesis of triazolopyrimidine derivatives 4a.

[Click here to view]

Scheme 3. Proposed mechanism of three-component reactions.

[Click here to view]

Antiproliferative screening

According to the revealed synthetic route, a series of the obtained triazolopyrimidines (4a4i) was selected for further investigation of their antiproliferative activities in vitro on four human cancer types along with their cytotoxic effects on one nontumorous human healthy type RPE-1 by the MTT test. The proportion of live cells was evaluated and correlated with the control and reference drug doxorubicin. All compounds repressed the five human cell types (Figs. 37). In the HCT-116, Table 2 and Figure 3 demonstrate that compounds 4a and 4h had close antiproliferative effects, and the remaining compounds had significantly fewer antiproliferative activities than doxorubicin. While, in the MCF-7, compounds 4i and 4h had a more potent effect, compound 4g had a comparable activity and the rest of the compounds had less antiproliferative properties relative to doxorubicin (Fig. 4 and Table 2). For MDA-MB-231 human breast cancer, compounds 4c and 4b had a more potent effect; compounds 4h and 4e had equipotent effects; and compounds 4d, 4a, 4g, 4f, and 4i had significantly less antiproliferative effects related to doxorubicin (Fig. 5 and Table 2). For A549 cancer, compounds 4d, 4i, 4e, 4f, 4g, and 4h had extra antiproliferative actions and compounds 4b, 4c, and 4a had insignificantly lesser antiproliferative effects than doxorubicin (Fig. 6 and Table 2). In the nontumorous human healthy normal cells (RPE-1), compounds 4f, 4g, and 4e were more toxic; three compounds 4a, 4i, and 4d had insignificant toxic effects; and three compounds 4c, 4b, and, 4h had significant toxic effects related to the reference drug.

These results indicated that all the prepared candidates are effective anticancer compounds in human lung cancer, except for compound 4a, which had significantly less activity; compound 4h had an effective antiproliferative activity on all cancerous types and had the least toxic effect on the noncancerous cells; and compound 4i had the best antiproliferative effect on both MCF-7 and A549 cancerous types, while having a weak antiproliferative effect on both HCT-116 and MDA-MB-231 cancerous types; compounds 4i, 4h, and 4g are efficient antiproliferative medicines on hormone-dependent contrary to the independent human breast cancer.

Figure 3. Dose-dependent antiproliferative effects of 4a4i compounds on HCT-116 cancer cells via the MTT assay.

[Click here to view]

Figure 4. Dose-dependent antiproliferative effects of 4a-4i compounds on MCF-7 cancer cells via the MTT assay.

[Click here to view]

Figure 5. Dose-dependent antiproliferative effects of 4a4i compounds on MDA-MB-231 cancer cells via the MTT assay.

[Click here to view]

Figure 6. Dose-dependent antiproliferative effects of 4a4i compounds on A549 cancer cells via the MTT assay.

[Click here to view]

Figure 7. Dose-dependent toxic effects of 4a4i compounds on RPE-1 human normal cells via the MTT assay.

[Click here to view]

Table 3. TI of the compounds’ activities on the four cancer types relative to the normal cells according to the results in Table 2.

[Click here to view]

Table 2. EC50 of the compounds on the four human cancer types and the TD50 on the noncancerous human normal cells via the MTT assay.

[Click here to view]

The therapeutic index (TI) of the compounds, which was calculated by dividing their efficacious dose for 50% of the cancerous cells population (ED50) over their toxic dose in 50% of the noncancerous cells population, is presented in Table 3 (Hatem et al., 2019). From Table 3, it is obtained that the TI ranged from 1.7 to 3.1 compared to 2.8 for the reference drug on human colon cancer. In addition, TI ranged from 1.6 to 6.1 relative to 3.3 for the reference drug on the hormone-dependent human breast adenocarcinoma. While the TI ranged from 1.5 to 3.8 relative to 2.5 for the reference drug on the hormone-independent human breast adenocarcinoma. In addition, TI ranged from 1.8 to 3.1 relative to 2.3 for human lung cancer reference drugs. These results indicate that all synthetic compounds have the same efficacy and safety, or better in some compounds, than the standard drug doxorubicin.

Structure activity relationships discussion

In general, triazolopyrimidine derivatives have a function of promoting biological activity. In this work, all synthetic compounds have a built-in triazolopyrimidine unit with an important indole moiety at the 5-site. The compound 4a with 4-chlorophenyl at 7-position has the greatest antiproliferative activity against HCT-116 (EC50 = 24.5 μM), followed by compound 4h (EC50 = 24.9), which comprises a pyrrole unit at 7-site. While with MCF-7 cells, 4i (EC50 = 10.6 μM), which contains the bi-indole rings, compound 4h (EC50 = 18.5 μM), which contains a pyrrole ring at 7-position, and compound 4g (EC50 = 20.4 μM), which has a furan ring at 7-site are anticancer applicants on hormone-dependent instead of on hormone-independent human breast cancer. For, MDA-MB-231, compound 4c (EC50 = 18.3 μM), which was substituted with 4-bromophenyl at position 7, and compound 4b (EC50 = 21.3 μM), which was substituted with 4-nitrophenyl at position 7, had more effect; compound 4h (EC50 = 25.5 μM), with pyrrole unit at the 7-site, and compound 4e (EC50 = 25.5 μM), substituted with 2-nitrophenyl at 7-position, had equipotent effects; compared to doxorubicin, compound 4i (EC50 = 32.4 μM), with the bi-indole rings, had a significantly lesser antiproliferative effect. In the A549 cancer type, compound 4d (EC50 = 21.8 μM), substituted with 2-nitrophenyl at 7-position, had more antiproliferative activity; compared to doxorubicin, compound 4a (EC50 = 31.7 μM), substituted with 4-chlorophenyl at 7-position, had insignificantly lesser antiproliferative effects. Moreover, these results indicate that all synthetic compounds have the same efficacy and safety, or better in some compounds, than the standard drug doxorubicin.


CONCLUSION

The conventional three-component reactions have been used to construct a new series of triazolopyrimidine derivatives with built-in indole moiety. Compared to other traditional methods, this method is advanced in its cost-effectiveness, is easy to set-up, and results in high product quality. Antiproliferative activity of the new compounds has been examined toward four different human cancer cells and one human healthy cell line. Compounds 4a and 4h are active against the human colon cancer; all triazolopyrimidines are active toward MCF-7; 4i, 4h, and 4g are effective anticancer applicants on hormone-dependent instead of hormone-independent MCF-7. Consequently, it was found that the triazolopyrimidine derivatives could be studied for further biological investigation. Moreover, the multicomponent method is promising in the synthesis of many additional heterocyclic compounds.


ACKNOWLEDGMENTS

We express our appreciation to the National Research Centre and Qassim University for the services provided.


CONFLICT OF INTEREST

The authors declare that there is no conflict of interest.


FINANCIAL SUPPORT AND SPONSORSHIP

None.


REFERENCES

Abdel-Aziem A, El-Gendy MS, Abdelhamid AO. Synthesis and antimicrobial activities of pyrido [2, 3-d] pyrimidine, pyridotriazolopyrimidine, triazolopyrimidine, and pyrido [2, 3-d: 6, 5d’] dipyrimidine derivatives. Eur J Chem, 2012; 3:455–60. CrossRef

Adole VA, More RA, Jagdale BS, Pawar TB, Chobe SS. Efficient synthesis, antibacterial, antifungal, antioxidant and cytotoxicity study of 2-(2-hydrazineyl) thiazole derivatives. ChemistrySelect, 2020; 5:2778–6. CrossRef

Adole VA, Pawar TB, Koli PB, Jagdale BS. Exploration of catalytic performance of nano-La2O3 as an efficient catalyst for dihydropyrimidinone/thione synthesis and gas sensing. J Nanostruct Chem, 2019; 9:61–76. CrossRef

Allen EE, Zhu C, Panek JS, Schaus SE. Multicomponent condensation reactions via ortho-quinone methides. Org Lett, 2017; 19:1878–81. CrossRef

Anand D, Yadav PK, Patel OPS. Antileishmanial activity of pyrazolopyridine derivatives and their potential as an adjunct therapy with miltefosine. J Med Chem, 2017; 60:1041–59. CrossRef

Ashour HM, Shaaban OG, Rizk OH, El-Ashmawy IM. Synthesis and biological evaluation of thieno [2′,3′:4,5]pyrimido[1,2-b][1,2,4]triazines and thieno[2,3-d] [1,2,4]triazolo[1,5-a]pyrimidines as anti- inflammatory and analgesic agents. Eur J Med Chem, 2013; 62:341–51. CrossRef

Ashraf SH, Mohamed FM, Hanem MA, Taghrid SH. Synthesis and antitumor activity of some new pyrazolo [1, 5-a] pyrimidines. Chin Chem Lett, 2017; 28:388–93; doi:10.1016/j.cclet.2016.10.022 CrossRef

Bao B, Sun Q, Yao X, Hong J, Lee CO, Sim, CJ, Im KS, Jung JH. Cytotoxic bisindole alkaloids from a marine sponge Spongosorites sp. J Nat Prod, 2005; 68:711–5. CrossRef

Bisht GS, Pandey AM, Chaudhari MB, Agalave SG, Kanyal A, Karmodiya K, Gnanaprakasam B. Ru-Catalyzed dehydrogenative synthesis of antimalarial arylidene oxindoles. Org Biomol Chem, 2018; 16:7223–9. CrossRef

Boechat N, Pinheiro LCS, Silva TS, Aguiar ACC, Carvalho AS, Bastos MM, Costa CCP, Pinheiro S, Pinto AC, Mendonça JS, Dutra KDB, Valverde AL, Santos-Filho OA, Ceravolo IP, Krettli AU. New trifluoromethyl triazolopyrimidines as anti-plasmodium falciparum agents. Molecules, 2012; 17:8285–302. CrossRef

Carbajales C, Jun-ichi SJ, Marzaro G. Multicomponent assembly of the kinesin spindle protein inhibitor CPUYJ039 and analogues as antimitotic agents. ACS Comb Sci, 2017; 19:153–60. CrossRef

Chen Q, Zhu XL, Jiang LL, Liu ZM, Yang GF. Synthesis, antifungal activity and CoMFA analysis of novel 1,2,4-triazolo[1,5-a]pyrimidine derivatives. Eur J Med Chem, 2008; 43:595–603. CrossRef

Chobe SS, Adole VA, Deshmukh KP, Pawar TB, Jagdale BS. Poly (ethylene glycol)(PEG-400): a green approach towards the synthesis of novel pyrazolo [3, 4-d] pyrimidin-6-amines derivatives and their antimicrobial screening. Arch Appl Sci Res, 2014; 6:61–6.

Cioc RC, Ruijter E, Orru RVA. Multicomponent reactions: advanced tools for sustainable organic synthesis. Green Chem, 2014; 16:2958–75. CrossRef

Coxon CR, Anscombe E, Harnor SJ, Martin MP, Carbain B, Golding BT, Hardcastle IR, Harlow LK, Korolchuk S, Matheson CJ, Newell DR, Noble MEM, Sivaprakasam M, Tudhope SJ, Turner DM, Wang LZ, Wedge SR, Wong C, Griffin RJ, Endicott JA, Cano C. Cyclin-dependent kinase (CDK) inhibitors: structure–activity relationships and insights into the CDK-2 selectivity of 6-Substituted 2-Arylaminopurines. J Med Chem, 2017; 1746–67. CrossRef

Dekamin MG, Eslami M, Maleki A. Potassium phthalimide-N-oxyl: a novel, efficient, and simple organocatalyst for the one-pot three-component synthesis of various 2-amino-4H-chromene derivatives in water. Tetrahedron, 2013; 69:1074–85. CrossRef

Duran A, Dogan HN, Rollas H. Synthesis and preliminary anticancer activity of new 1,4-Dihydro-3-(3-hydroxy-2-naphthyl)-4-substituted-5H-1,2,4-triazoline-5-thiones. Farmaco, 2002; 57:559–64. CrossRef

El-Gendy MMA, Shaaban M, Shaaban KA, El-Bondkly AM, Laatsch H. Essramycin: a first triazolopyrimidine antibiotic isolated from nature. J Antibiot, 2008; 61(3):149–57. CrossRef

El-Nezhawy A, Eweas A, Radwan MA, El-Naggar T. Synthesis and molecular docking studies of novel 2-phenyl-4-substituted oxazole derivatives as potential anti-cancer agents. J Heterocycl Chem, 2016; 53:271–9. CrossRef

El-Nezhawy A, Gaballah S, Radwan MA. Studying the reactivity of (phthalazin-1(2H)-on-2-yl)methyl trichloroacetimidate towards different C- and O-nucleophiles. Tetrahedron Lett, 2009a; 50:6646–50. CrossRef

El-Nezhawy A, Radwan MA, Gaballah S. Synthesis of chiral N-(2-(1-oxophthalazin-2(1H)-yl) ethanoyl)-α-amino acid derivatives as antitumor agents. Arkivoc, 2009b; 12:119–30. CrossRef

Emam AN, Loutfy SA, Mostafa AA, Awad HM, Mohamed MB. Cyto-toxicity, biocompatibility and cellular response of carbon dots–plasmonic based nano-hybrids for bioimaging. RSC Adv, 2017; 7:23502–14; doi:10.1039/C7RA01423F. CrossRef

Eman MF, Wael AE, Ashraf MM, Walaa IE, Hanem MA. Synthesis and anticancer activity of new 1-thia-4-azaspiro[4.5]decane, their derived thiazolopyrimidine and 1,3,4-thiadiazole thioglycosides. Molecules, 2017; 22:170–3; doi:10.3390/molecules22010170 CrossRef

Endo T, Tsuda M, Fromont J, Jun’ichi K. Hyrtinadine A, a bis-indole alkaloid from a marine sponge. J Nat Prod, 2007; 70:423–4. CrossRef

Fakhr I, Hamdy N, Radwan MA, Ahmed Y. Synthesis of new bioactive benzothiophene derivatives. Egypt J Chem, 2004; 47:201–15.

Fakhr IM, Radwan MA, el-Batran S, Abd el-Salam OM, el-Shenawy SM. Synthesis and pharmacological evaluation of 2-substituted benzo[b]thiophenes as anti-inflammatory and analgesic agents. Eur J Med Chem, 2009; 44:1718–25. CrossRef

Fischer G. Recent progress in 1,2,4-triazolo[1,5-a]pyrimidine chemistry. Adv Heterocycl Chem, 2008; 95:143–219. CrossRef

Ghorab M, Radwan MA, Taha N, Amin N, Shehab M, Faker I. Dapson in heterocyclic chemistry, part II: antimicrobial and antitumor activities of some novel sulfone biscompounds containing biologically active thioureido, carbamothioate, quinazoline, imidazolidine, and thiazole moieties. Phosphorus Sulfur Silicon Relat Elem, 2008; 183:2906–17. CrossRef

Gribble GW. Heterocyclic scaffolds II: reactions and applications of indoles. Springer, Berlin, Germany, .

Gribble GW. Recent developments in indole ring synthesis—methodology and applications. J Chem Soc Perkin 1, 2000; 7:1045–75. CrossRef

Gujjar R, Marwaha A, White J, White L, Creason S, Shackleford DM, Baldwin J, Charman WN, Buckner FS, Charman S, Rathod PK, Phillips MA. Identification of a metabolically stable triazolopyrimidine based dihydroorotate dehydrogenase inhibitor with antimalarial activity in mice. J Med Chem, 2009; 52:1864–72. CrossRef

Hafez HN, Abbas H-AS, El-Gazzar A-RBA. Synthesis and evaluation of analgesic, anti-inflammatory and ulcerogenic activities of some triazolo- and 2-pyrazolyl-pyrido[2,3-d]-pyrimidines. Acta Pharm, 2008; 58:359–78. CrossRef

Hatem AA, Hanem MA, Mohamed M, Fahd AN, Ali S, Alqahtani A, Ahmed M, Rashad AS. Synthesis and biological evaluation of 4-(1H-1,2,4- triazol-1-yl)benzoic acid hybrids as anticancer agents. RSC Adv, 2019; 9:19065–74; doi:10.1039/c9ra03151k CrossRef

Holla BS, Mahalinga M, Karthikeyen MS, Poojary B, Akberali PM, Kumari NS. Synthesis, characterization and antimicrobial activity of some substituted 1,2,3-triazoles. Eur J Med Chem, 2005; 40:1173–8. CrossRef

Huang LH, Zheng YF, Lu YZ, Song CJ, Wang YG, Yu B, Hong-Liu M. Synthesis and biological evaluation of novel steroidal[17,16-d][1,2,4]triazolo[1,5- a]pyrimidines. Steroids, 2012; 77:710–5. CrossRef

Kamble RD, Meshram RJ, Hese SV, More RA, Kamble SS, Gacche RN, Dawane BS. Synthesis and in silico investigation of thiazoles bearing pyrazoles derivatives as anti-inflammatory agents. Comput Biol Chem, 2016; 61:86–96. CrossRef

Khera MJ, Cliffe IA, Marthur T, Prakash O. Synthesis and in vitro activity of novel 1,2,4-triazolo[4,3-a]pyrimidine oxazolidinone antibacterial agents. Bioorg Med Chem Lett, 2011; 21:2887–9. CrossRef

Kim TW, Yoo BW, Lee JK, Kim JH, Lee K, Chi YH, Lee JY. Synthesis and antihypertensive activity of pyrimidin-4(3H)-one derivatives as losartan analogue for new angiotensin II receptor type 1 (AT1) antagonists. Bioorg Med Chem Lett, 2012; 22:1649–54. CrossRef

Kumar J, Meena P, Singh A, Jameel E, Maqbool M, Mobashir M, Shandilya A, Tiwari M, Hoda N, Jayaram B. Synthesis and screening of triazolopyrimidine scaffold as multi-functional agents for Alzheimer’s disease therapies. Eur J Med Chem, 2016; 119:260–77; doi:10.1016/j.ejmech.2016.04.053 CrossRef

Lakamoska I. Molecular structure and antitumor activity of platinum(II) complexes containing purine analogs. Inorg Chim Acta Rev, 2009; 362:669–81. CrossRef

Łakomska I, Fandzloch M, Muziol T, Lis T, Jezierska J. Synthesis, characterization and antitumor properties of two highly cytotoxic ruthenium(III) complexes with bulky triazolopyrimidine ligands. Dalton Trans, 2013; 42:6219–26. CrossRef

Liu C, Zhou L, Jiang D, Gu Y. Multicomponent reactions of aldo-X bifunctional reagent α-oxoketene dithioacetals and indoles or amines: divergent synthesis of dihydrocoumarins, quinolines, furans, and pyrroles. Asian J Org Chem, 2016; 5:367–72. CrossRef

Luo Y, Zhang S, Liu ZJ, Chen W, Fu J, Zeng Q-F, Zhu H-L. Synthesis and antimicrobical evaluation of a novel class of 1,3,4-thiadiazole: derivatives bearing 1,2,4-triazolo[1,5-a]pyrimidine moiety. Eur J Med Chem, 2013; 64:54–61. CrossRef

Marwaha A, White J, El Mazouni F, Creason S, Kokkonda S, Buckner F, Rathod P. Bioisosteric transformations and permutations in the triazolopyrimidine scaffold to identify the minimum pharmacophore required for inhibitory activity against plasmodium falciparum dihydroorotate dehydrogenase. J Med Chem, 2012; 55:7425–37. CrossRef

Muhammad Z, Radwan MA, Farghaly T, Gaber H, Elaasser M. Synthesis and antitumor activity of novel[1,2,4,5]tetrazepino[6,7-b]indole derivatives: marine natural product hyrtioreticuline C and D analogues. Mini Rev Med Chem, 2019; 19:79–86. CrossRef

Peng J, Gao Y, Zhu C, Liu B, Gao Y, Hu M, Wu W, Jiang H. Synthesis of polysubstituted 3-amino pyrroles via palladium-catalyzed multicomponent reaction. J Org Chem, 2017; 82:3581–88. CrossRef

Radwan MA, Abbas E. Synthesis of some pyridine, thiopyrimidine, and isoxazoline derivatives based on the pyrrole moiety. Monatsh Chem, 2009; 140:229–33. CrossRef

Radwan MA, Alminderej FM, Awad HM. One-pot multicomponent synthesis and cytotoxic evaluation of novel 7-substituted-5-(1H-indol-3-yl)tetrazolo[1,5-a]pyrimidine-6-carbonitrile. Molecules, 2020; 25:255; doi:10.3390/molecules25020255. CrossRef

Radwan MA, El-Sherbiny M. Synthesis and antitumor activity of indolylpyrimidines: marine natural product meridianin D analogues. Bioorg Med Chem, 2007; 15:1206–11. CrossRef

Radwan MA, Ragab EA, Sabry NM, El-Shenawy SM. Synthesis and biological evaluation of new 3-substituted indole derivatives as potential anti-inflammatory and analgesic agents. Bioorg Med Chem, 2007; 15:3832–41. CrossRef

Radwan MA, Ragab EA, Shaaban MR, El-Nezhawy AO. Application of (2Z)-3-dimethylamino-2-(1H-indole-3-carbonyl) acrylonitrile in the synthesis of novel 3-heteroarylindoles: condensed meridianine analogs. ARKIVOC, 2009a; 2009(7):281–91. CrossRef

Radwan MAA, Shehab M, El-Shenawy S. Synthesis and biological evaluation of 5-substituted benzo[b]thiophene derivatives as anti-inflammatory agents. Monatsh Chem, 2009b; 140:445–50. CrossRef

Ravichandiran P, Lai B, Gu Y. Aldo-X bifunctional building blocks for the synthesis of heterocycles. Chem Rec, 2017; 17:142–83. CrossRef

Reddi MNK, Park H, Lee HR, Suh H, Kim I. Efficient, solvent-free, multi-component method for organic-base-catalyzed synthesis of βphosphonomalonates. ACS Comb Sci, 2015; 17:691–97; doi:10.1021/acscombsci.5b00109 CrossRef

Rotstein BH, Zaretsky S, Rai V, Yudi AK. Small heterocycles in multicomponent reactions. Chem Rev, 2014; 114:8323–59. CrossRef

Sakemi S, Sun HH. Nortopsentins A, B, and C. Cytotoxic and antifungal imidazolediylbis[indoles] from the sponge Spongosorites ruetzleri. J Org Chem, 1991; 56:4304–7. CrossRef

Selvaraj MR, Rajesh S. Synthetic chemistry of pyrimidines and fused pyrimidines: a review. Syn Commun, 2016; 46:645–72. CrossRef

Sztanke K, Tuzimski T, Rzymowska J. Synthesis, determination of the lipophilicity, anticancer and antimicrobial properties of some fused 1,2,4-triazole derivatives. Eur J Med Chem, 2008; 43:404–19. CrossRef

Taylor RD, MacCoss M, Lawson ADG. Rings in drugs, Miniperspective. J Med Chem, 2014; 57(14):5845–59. CrossRef

Wang S, Zhao LJ, Zheng YC, Shen DD, Miao EF, Qiao XP, Zhao LJ, Liu Y, Huang R, Yu B, Liu HM. Design, synthesis and biological evaluation of [1,2,4]triazolo[1,5-a]pyrimidines as potent lysine specific demethylase 1 (LSD1/KDM1A) inhibitors. Eur J Med Chem, 2017; 125:940–51. CrossRef

Yang CG, Huang H, Jiang B. Progress in studies of novel marine bis (indole) alkaloids. Curr Org Chem, 2004; 8:1691–720. CrossRef

Zhang N, Ayral-Kaloustian S, Nguyen T, Afragola J, Hernandez R, Lucas J, Gibbons J, Beyer C. Synthesis and SAR of [1,2,4]Triazolo[1,5-a]pyrimidines, a class of anticancer agents with a unique mechanism of tubulin inhibition. J Med Chem, 2007; 50:319–27. CrossRef

Zhao XL, Zhao YF, Guo SC, Song HS, Wang D, Gong P. Synthesis and antitumor activities of novel [1,2,4]triazolo[1,5-a]pyrimidines. Molecules, 2007; 12:1136–46. CrossRef

Reference

Abdel-Aziem A, El-Gendy MS, Abdelhamid AO. Synthesis and antimicrobial activities of pyrido [2, 3-d] pyrimidine, pyridotriazolopyrimidine, triazolopyrimidine, and pyrido [2, 3-d: 6, 5d'] dipyrimidine derivatives. Eur J Chem, 2012; 3:455-60. https://doi.org/10.5155/eurjchem.3.4.455-460.683

Adole VA, More RA, Jagdale BS, Pawar TB, Chobe SS. Efficient synthesis, antibacterial, antifungal, antioxidant and cytotoxicity study of 2-(2-hydrazineyl) thiazole derivatives. ChemistrySelect, 2020; 5:2778-6. https://doi.org/10.1002/slct.201904609

Adole VA, Pawar TB, Koli PB, Jagdale BS. Exploration of catalytic performance of nano-La2O3 as an efficient catalyst for dihydropyrimidinone/thione synthesis and gas sensing. J Nanostruct Chem, 2019; 9:61-76. https://doi.org/10.1007/s40097-019-0298-5

Allen EE, Zhu C, Panek JS, Schaus SE. Multicomponent condensation reactions via ortho-quinone methides. Org Lett, 2017; 19:1878-81. https://doi.org/10.1021/acs.orglett.7b00647

Anand D, Yadav PK, Patel OPS. Antileishmanial activity of pyrazolopyridine derivatives and their potential as an adjunct therapy with miltefosine. J Med Chem, 2017; 60:1041-59. https://doi.org/10.1021/acs.jmedchem.6b01447

Ashour HM, Shaaban OG, Rizk OH, El-Ashmawy IM. Synthesis and biological evaluation of thieno [2′,3′:4,5]pyrimido[1,2-b] [1,2,4]triazines and thieno[2,3-d] [1,2,4]triazolo[1,5-a]pyrimidines as anti-inflammatory and analgesic agents. Eur J Med Chem, 2013; 62:341-51. https://doi.org/10.1016/j.ejmech.2012.12.003

Ashraf SH, Mohamed FM, Hanem MA, Taghrid SH. Synthesis and antitumor activity of some new pyrazolo [1, 5-a] pyrimidines. Chin Chem Lett, 2017; 28:388-93; doi:10.1016/j.cclet.2016.10.022 https://doi.org/10.1016/j.cclet.2016.10.022

Bao B, Sun Q, Yao X, Hong J, Lee CO, Sim, CJ, Im KS, Jung JH. Cytotoxic bisindole alkaloids from a marine sponge Spongosorites sp. J Nat Prod, 2005; 68:711-5. https://doi.org/10.1021/np049577a

Bisht GS, Pandey AM, Chaudhari MB, Agalave SG, Kanyal A, Karmodiya K, Gnanaprakasam B. Ru-Catalyzed dehydrogenative synthesis of antimalarial arylidene oxindoles. Org Biomol Chem, 2018; 16:7223-9. https://doi.org/10.1039/C8OB01852A

Boechat N, Pinheiro LCS, Silva TS, Aguiar ACC, Carvalho AS, Bastos MM, Costa CCP, Pinheiro S, Pinto AC, Mendonça JS, Dutra KDB, Valverde AL, Santos-Filho OA, Ceravolo IP, Krettli AU. New trifluoromethyl triazolopyrimidines as anti-plasmodium falciparum agents. Molecules, 2012; 17:8285-302. https://doi.org/10.3390/molecules17078285

Carbajales C, Jun-ichi SJ, Marzaro G. Multicomponent assembly of the kinesin spindle protein inhibitor CPUYJ039 and analogues as antimitotic agents. ACS Comb Sci, 2017; 19:153-60. https://doi.org/10.1021/acscombsci.6b00166

Chen Q, Zhu XL, Jiang LL, Liu ZM, Yang GF. Synthesis, antifungal activity and CoMFA analysis of novel 1,2,4-triazolo[1,5-a] pyrimidine derivatives. Eur J Med Chem, 2008; 43:595-603. https://doi.org/10.1016/j.ejmech.2007.04.021

Chobe SS, Adole VA, Deshmukh KP, Pawar TB, Jagdale BS. Poly (ethylene glycol)(PEG-400): a green approach towards the synthesis of novel pyrazolo [3, 4-d] pyrimidin-6-amines derivatives and their antimicrobial screening. Arch Appl Sci Res, 2014; 6:61-6.

Cioc RC, Ruijter E, Orru RVA. Multicomponent reactions: advanced tools for sustainable organic synthesis. Green Chem, 2014; 16:2958-75. https://doi.org/10.1039/C4GC00013G

Coxon CR, Anscombe E, Harnor SJ, Martin MP, Carbain B, Golding BT, Hardcastle IR, Harlow LK, Korolchuk S, Matheson CJ, Newell DR, Noble MEM, Sivaprakasam M, Tudhope SJ, Turner DM, Wang LZ, Wedge SR, Wong C, Griffin RJ, Endicott JA, Cano C. Cyclin-dependent kinase (CDK) inhibitors: structure-activity relationships and insights into the CDK-2 selectivity of 6-Substituted 2-Arylaminopurines. J Med Chem, 2017; 1746-67. https://doi.org/10.1021/acs.jmedchem.6b01254

Dekamin MG, Eslami M, Maleki A. Potassium phthalimide-N-oxyl: a novel, efficient, and simple organocatalyst for the one-pot three-component synthesis of various 2-amino-4H-chromene derivatives in water. Tetrahedron, 2013; 69:1074-85. https://doi.org/10.1016/j.tet.2012.11.068

Duran A, Dogan HN, Rollas H. Synthesis and preliminary anticancer activity of new 1,4-Dihydro-3-(3-hydroxy-2-naphthyl)-4- substituted-5H-1,2,4-triazoline-5-thiones. Farmaco, 2002; 57:559-64. https://doi.org/10.1016/S0014-827X(02)01248-X

El-Gendy MMA, Shaaban M, Shaaban KA, El-Bondkly AM, Laatsch H. Essramycin: a first triazolopyrimidine antibiotic isolated from nature. J Antibiot, 2008; 61(3):149-57. https://doi.org/10.1038/ja.2008.124

El-Nezhawy A, Eweas A, Radwan MA, El-Naggar T. Synthesis and molecular docking studies of novel 2-phenyl-4-substituted oxazole derivatives as potential anti-cancer agents. J Heterocycl Chem, 2016; 53:271-9. https://doi.org/10.1002/jhet.2422

El-Nezhawy A, Gaballah S, Radwan MA. Studying the reactivity of (phthalazin-1(2H)-on-2-yl)methyl trichloroacetimidate towards different C- and O-nucleophiles. Tetrahedron Lett, 2009a; 50:6646-50. https://doi.org/10.1016/j.tetlet.2009.09.068

El-Nezhawy A, Radwan MA, Gaballah S. Synthesis of chiral N-(2-(1-oxophthalazin-2(1H)-yl) ethanoyl)-α-amino acid derivatives as antitumor agents. Arkivoc, 2009b; 12:119-30. https://doi.org/10.3998/ark.5550190.0010.c10

Emam AN, Loutfy SA, Mostafa AA, Awad HM, Mohamed MB. Cyto-toxicity, biocompatibility and cellular response of carbon dots- plasmonic based nano-hybrids for bioimaging. RSC Adv, 2017; 7:23502- 14; doi:10.1039/C7RA01423F. https://doi.org/10.1039/C7RA01423F

Eman MF, Wael AE, Ashraf MM, Walaa IE, Hanem MA. Synthesis and anticancer activity of new 1-thia-4-azaspiro[4.5]decane, their derived thiazolopyrimidine and 1,3,4-thiadiazole thioglycosides. Molecules, 2017; 22:170-3; doi:10.3390/molecules22010170 https://doi.org/10.3390/molecules22010170

Endo T, Tsuda M, Fromont J, Jun'ichi K. Hyrtinadine A, a bis-indole alkaloid from a marine sponge. J Nat Prod, 2007; 70:423-4. https://doi.org/10.1021/np060420n

Fakhr I, Hamdy N, Radwan MA, Ahmed Y. Synthesis of new bioactive benzothiophene derivatives. Egypt J Chem, 2004; 47:201-15.

Fakhr IM, Radwan MA, el-Batran S, Abd el-Salam OM, el- Shenawy SM. Synthesis and pharmacological evaluation of 2-substituted benzo[b]thiophenes as anti-inflammatory and analgesic agents. Eur J Med Chem, 2009; 44:1718-25. https://doi.org/10.1016/j.ejmech.2008.02.034

Fischer G. Recent progress in 1,2,4-triazolo[1,5-a]pyrimidine chemistry. Adv Heterocycl Chem, 2008; 95:143-219. https://doi.org/10.1016/S0065-2725(07)95003-5

Ghorab M, Radwan MA, Taha N, Amin N, Shehab M, Faker I. Dapson in heterocyclic chemistry, part II: antimicrobial and antitumor activities of some novel sulfone biscompounds containing biologically active thioureido, carbamothioate, quinazoline, imidazolidine, and thiazole moieties. Phosphorus Sulfur Silicon Relat Elem, 2008; 183:2906-17. https://doi.org/10.1080/10426500802505424

Gribble GW. Heterocyclic scaffolds II: reactions and applications of indoles. Springer, Berlin, Germany, .

Gribble GW. Recent developments in indole ring synthesis- methodology and applications. J Chem Soc Perkin 1, 2000; 7:1045-75. https://doi.org/10.1039/a909834h

Gujjar R, Marwaha A, White J, White L, Creason S, Shackleford DM, Baldwin J, Charman WN, Buckner FS, Charman S, Rathod PK, Phillips MA. Identification of a metabolically stable triazolopyrimidine based dihydroorotate dehydrogenase inhibitor with antimalarial activity in mice. J Med Chem, 2009; 52:1864-72. https://doi.org/10.1021/jm801343r

Hafez HN, Abbas H-AS, El-Gazzar A-RBA. Synthesis and evaluation of analgesic, anti-inflammatory and ulcerogenic activities of some triazolo- and 2-pyrazolyl-pyrido[2,3-d]-pyrimidines. Acta Pharm, 2008; 58:359-78. https://doi.org/10.2478/v10007-008-0024-1

Hatem AA, Hanem MA, Mohamed M, Fahd AN, Ali S, Alqahtani A, Ahmed M, Rashad AS. Synthesis and biological evaluation of 4-(1H-1,2,4- triazol-1-yl)benzoic acid hybrids as anticancer agents. RSC Adv, 2019; 9:19065-74; doi:10.1039/c9ra03151k https://doi.org/10.1039/C9RA03151K

Holla BS, Mahalinga M, Karthikeyen MS, Poojary B, Akberali PM, Kumari NS. Synthesis, characterization and antimicrobial activity of some substituted 1,2,3-triazoles. Eur J Med Chem, 2005; 40:1173-8. https://doi.org/10.1016/j.ejmech.2005.02.013

Huang LH, Zheng YF, Lu YZ, Song CJ, Wang YG, Yu B, Hong- Liu M. Synthesis and biological evaluation of novel steroidal[17,16-d] [1,2,4]triazolo[1,5- a]pyrimidines. Steroids, 2012; 77:710-5. https://doi.org/10.1016/j.steroids.2012.03.002

Kamble RD, Meshram RJ, Hese SV, More RA, Kamble SS, Gacche RN, Dawane BS. Synthesis and in silico investigation of thiazoles bearing pyrazoles derivatives as anti-inflammatory agents. Comput Biol Chem, 2016; 61:86-96. https://doi.org/10.1016/j.compbiolchem.2016.01.007

Khera MJ, Cliffe IA, Marthur T, Prakash O. Synthesis and in vitro activity of novel 1,2,4-triazolo[4,3-a]pyrimidine oxazolidinone antibacterial agents. Bioorg Med Chem Lett, 2011; 21:2887-9. https://doi.org/10.1016/j.bmcl.2011.03.075

Kim TW, Yoo BW, Lee JK, Kim JH, Lee K, Chi YH, Lee JY. Synthesis and antihypertensive activity of pyrimidin-4(3H)-one derivatives as losartan analogue for new angiotensin II receptor type 1 (AT1) antagonists. Bioorg Med Chem Lett, 2012; 22:1649-54. https://doi.org/10.1016/j.bmcl.2011.12.116

Kumar J, Meena P, Singh A, Jameel E, Maqbool M, Mobashir M, Shandilya A, Tiwari M, Hoda N, Jayaram B. Synthesis and screening of triazolopyrimidine scaffold as multi-functional agents for Alzheimer's disease therapies. Eur J Med Chem, 2016; 119:260-77; doi:10.1016/j. ejmech.2016.04.053 https://doi.org/10.1016/j.ejmech.2016.04.053

Lakamoska I. Molecular structure and antitumor activity of platinum(II) complexes containing purine analogs. Inorg Chim Acta Rev, 2009; 362:669-81. https://doi.org/10.1016/j.ica.2008.02.030

Łakomska I, Fandzloch M, Muziol T, Lis T, Jezierska J. Synthesis, characterization and antitumor properties of two highly cytotoxic ruthenium(III) complexes with bulky triazolopyrimidine ligands. Dalton Trans, 2013; 42:6219-26. https://doi.org/10.1039/c2dt32216a

Liu C, Zhou L, Jiang D, Gu Y. Multicomponent reactions of aldo-X bifunctional reagent α-oxoketene dithioacetals and indoles or amines: divergent synthesis of dihydrocoumarins, quinolines, furans, and pyrroles. Asian J Org Chem, 2016; 5:367-72. https://doi.org/10.1002/ajoc.201500497

Luo Y, Zhang S, Liu ZJ, Chen W, Fu J, Zeng Q-F, Zhu H-L. Synthesis and antimicrobical evaluation of a novel class of 1,3,4-thiadiazole: derivatives bearing 1,2,4-triazolo[1,5-a]pyrimidine moiety. Eur J Med Chem, 2013; 64:54-61. https://doi.org/10.1016/j.ejmech.2013.04.014

Marwaha A, White J, El Mazouni F, Creason S, Kokkonda S, Buckner F, Rathod P. Bioisosteric transformations and permutations in the triazolopyrimidine scaffold to identify the minimum pharmacophore required for inhibitory activity against plasmodium falciparum dihydroorotate dehydrogenase. J Med Chem, 2012; 55:7425-37. https://doi.org/10.1021/jm300351w

Muhammad Z, Radwan MA, Farghaly T, Gaber H, Elaasser M. Synthesis and antitumor activity of novel[1,2,4,5]tetrazepino[6,7-b]indole derivatives: marine natural product hyrtioreticuline C and D analogues. Mini Rev Med Chem, 2019; 19:79-86. https://doi.org/10.2174/1389557518666180724094244

Peng J, Gao Y, Zhu C, Liu B, Gao Y, Hu M, Wu W, Jiang H. Synthesis of polysubstituted 3-amino pyrroles via palladium-catalyzed multicomponent reaction. J Org Chem, 2017; 82:3581-88. https://doi.org/10.1021/acs.joc.7b00098

Radwan MA, Abbas E. Synthesis of some pyridine, thiopyrimidine, and isoxazoline derivatives based on the pyrrole moiety. Monatsh Chem, 2009; 140:229-33. https://doi.org/10.1007/s00706-008-0061-y

Radwan MA, Alminderej FM, Awad HM. One-pot multicomponent synthesis and cytotoxic evaluation of novel 7-substituted- 5-(1H-indol-3-yl)tetrazolo[1,5-a]pyrimidine-6-carbonitrile. Molecules, 2020; 25:255; doi:10.3390/molecules25020255. https://doi.org/10.3390/molecules25020255

Radwan MA, El-Sherbiny M. Synthesis and antitumor activity of indolylpyrimidines: marine natural product meridianin D analogues. Bioorg Med Chem, 2007; 15:1206-11. https://doi.org/10.1016/j.bmc.2006.11.023

Radwan MA, Ragab EA, Sabry NM, El-Shenawy SM. Synthesis and biological evaluation of new 3-substituted indole derivatives as potential anti-inflammatory and analgesic agents. Bioorg Med Chem, 2007; 15:3832-41. https://doi.org/10.1016/j.bmc.2007.03.024

Radwan MA, Ragab EA, Shaaban MR, El-Nezhawy AO. Application of (2Z)-3-dimethylamino-2-(1H-indole-3-carbonyl) acrylonitrile in the synthesis of novel 3-heteroarylindoles: condensed meridianine analogs. ARKIVOC, 2009a; 2009(7):281-91. https://doi.org/10.3998/ark.5550190.0010.727

Radwan MAA, Shehab M, El-Shenawy S. Synthesis and biological evaluation of 5-substituted benzo[b]thiophene derivatives as anti-inflammatory agents. Monatsh Chem, 2009b; 140:445-50. https://doi.org/10.1007/s00706-008-0067-5

Ravichandiran P, Lai B, Gu Y. Aldo-X bifunctional building blocks for the synthesis of heterocycles. Chem Rec, 2017; 17:142-83. https://doi.org/10.1002/tcr.201600042

Reddi MNK, Park H, Lee HR, Suh H, Kim I. Efficient, solvent-free, multi-component method for organic-base-catalyzed synthesis of β-phosphonomalonates. ACS Comb Sci, 2015; 17:691-97; doi:10.1021/ acscombsci.5b00109 https://doi.org/10.1021/acscombsci.5b00109

Rotstein BH, Zaretsky S, Rai V, Yudi AK. Small heterocycles in multicomponent reactions. Chem Rev, 2014; 114:8323-59. https://doi.org/10.1021/cr400615v

Sakemi S, Sun HH. Nortopsentins A, B, and C. Cytotoxic and antifungal imidazolediylbis[indoles] from the sponge Spongosorites ruetzleri. J Org Chem, 1991; 56:4304-7. https://doi.org/10.1021/jo00013a044

Selvaraj MR, Rajesh S. Synthetic chemistry of pyrimidines and fused pyrimidines: a review. Syn Commun, 2016; 46:645-72. https://doi.org/10.1080/00397911.2016.1165254

Sztanke K, Tuzimski T, Rzymowska J. Synthesis, determination of the lipophilicity, anticancer and antimicrobial properties of some fused 1,2,4-triazole derivatives. Eur J Med Chem, 2008; 43:404-19. https://doi.org/10.1016/j.ejmech.2007.03.033

Taylor RD, MacCoss M, Lawson ADG. Rings in drugs, Miniperspective. J Med Chem, 2014; 57(14):5845-59. https://doi.org/10.1021/jm4017625

Wang S, Zhao LJ, Zheng YC, Shen DD, Miao EF, Qiao XP, Zhao LJ, Liu Y, Huang R, Yu B, Liu HM. Design, synthesis and biological evaluation of [1,2,4]triazolo[1,5-a]pyrimidines as potent lysine specific demethylase 1 (LSD1/KDM1A) inhibitors. Eur J Med Chem, 2017; 125:940-51. https://doi.org/10.1016/j.ejmech.2016.10.021

Yang CG, Huang H, Jiang B. Progress in studies of novel marine bis (indole) alkaloids. Curr Org Chem, 2004; 8:1691-720. https://doi.org/10.2174/1385272043369656

Zhang N, Ayral-Kaloustian S, Nguyen T, Afragola J, Hernandez R, Lucas J, Gibbons J, Beyer C. Synthesis and SAR of [1,2,4]Triazolo[1,5-a] pyrimidines, a class of anticancer agents with a unique mechanism of tubulin inhibition. J Med Chem, 2007; 50:319-27. https://doi.org/10.1021/jm060717i

Zhao XL, Zhao YF, Guo SC, Song HS, Wang D, Gong P. Synthesis and antitumor activities of novel [1,2,4]triazolo[1,5-a] pyrimidines. Molecules, 2007; 12:1136-46. https://doi.org/10.3390/12051136

Article Metrics
600 Views 101 Downloads 701 Total

Year

Month

Related Search

By author names