Next Article in Journal
Differential Effects of Viscum album Preparations on the Maturation and Activation of Human Dendritic Cells and CD4+ T Cell Responses
Next Article in Special Issue
Antiproliferative Activity and Cellular Uptake of Evodiamine and Rutaecarpine Based on 3D Tumor Models
Previous Article in Journal
Simultaneous Determination of Eight Alkaloids in Rat Plasma by UHPLC-MS/MS after Oral Administration of Coptis deltoidea C. Y. Cheng et Hsiao and Coptis chinensis Franch
Previous Article in Special Issue
Novel (E)-β-Farnesene Analogues Containing 2-Nitroiminohexahydro-1,3,5-triazine: Synthesis and Biological Activity Evaluation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

New Potential Antimalarial Agents: Design, Synthesis and Biological Evaluation of Some Novel Quinoline Derivatives as Antimalarial Agents

by
Ibrahim Ali M. Radini
1,
Tarek M. Y. Elsheikh
2,3,
Emad M. El-Telbani
1,4 and
Rizk E. Khidre
1,4,*
1
Chemistry Department, Faculty of Science, Jazan University, Jazan 2097, Saudi Arabia
2
Biology Department, Faculty of Science, Jazan University, Jazan 2097, Saudi Arabia
3
Department of Zoology, Faculty of Science, Al-Azhar University, Cairo 11675, Egypt
4
Chemical Industries Division, National Research Centre, Dokki 12622, Giza, Egypt
*
Author to whom correspondence should be addressed.
Molecules 2016, 21(7), 909; https://doi.org/10.3390/molecules21070909
Submission received: 26 May 2016 / Revised: 3 July 2016 / Accepted: 8 July 2016 / Published: 14 July 2016
(This article belongs to the Special Issue Drug Design and Discovery: Principles and Applications)

Abstract

:
A novel series of dihydropyrimidines (DHPMs) 4aj; 2-oxopyran-3-carboxylate 7a,b; 1-amino-1,2-dihydropyridine-3-carboxylate 8; and 1,3,4-oxadiazole derivatives 12 with quinolinyl residues have been synthesized in fairly good yields. The structure of the newly synthesized compounds was elucidated on the basis of analytical and spectral analyses. In vitro antimalarial evaluation of the synthesized quinoline derivatives against Plasmodium falciparum revealed them to possess moderate to high antimalarial activities, with IC50 values ranging from 0.014–5.87 μg/mL. Compounds 4b,g,i and 12 showed excellent antimalarial activity against to Plasmodium falciparum compared with the antimalarial agent chloroquine (CQ).

Graphical Abstract

1. Introduction

Malaria is one of the principal diseases of the developing countries, particularly in Africa, Asia and South America. According to a World Health Organization (WHO) report, there are between 300 million and 500 million cases of malaria worldwide annually and more than one million people die from that disease, most of them are children under the age of five years [1,2]. Among five typically recognized Plasmodium species causing this disease in humans, Plasmodium falciparum is responsible for about 95% of worldwide malaria and has a mortality rate of 1%–3%, and Plasmodium vivax for most morbidity, additionally representing a reservoir of latent infection that hampers current control and future elimination efforts [3,4,5,6]. Due to the toxic side effects and the risk of developing resistance after prolonged treatment with aminoquinolines and their derivatives (Figure 1) which are nowadays used as antimalarial agents, the growth of and increasing resistance [7,8] of the malaria parasite Plasmodium falciparum to known antimalarial agents demands a continuous effort to develop new antimalarial agents especially, as an effective vaccine for malaria is not available.
In addition, quinoline-based fused heterocyclic systems are found to possess potential antimicrobial [9,10], antimalarial [11,12], anti-inflammatory [13,14], antitumor [15], and anti-parasitic activity [16].
Currently there are only limited safe drugs for the treatment of the disease, however, the design of new chemical agents, specifically affecting these targets, could lead to the availability of better drugs to treat malaria. Based on the above information and in continuation with our previous work [11,15,17,18], quinoline-based antimalarials that would not induce resistance, we have designed and prepared several quinolines compounds and screened for their antimalarial activities. Hopefully, these compounds will be active on the CQ-resistant strain FcB1 and could lead to the availability of better drugs to treat malaria.

2. Results

2.1. Chemistry

Dihydropyrimidin-2(1H)-ones (DHPMs) and their derivatives have received much attention because they are important substructures in both biologically active compounds and several marine alkaloids involving DHPM core units [19,20,21]. A facile three-component Biginelli’s one-pot cyclo-condensation reaction takes place between the quinolinealdehydes namely, tetrazolo-[1,5-a]quinoline-4-carbaldehyde (1a), 7-methyltetrazolo[1,5-a]quinoline-4-carbaldehyde (1b) [22,23], 2-oxo-1,2-dihydroquinoline-3-carbaldehyde (1c) [24]; ethyl acetoacetate (2a) or acetylacetone (2b); and (thio)urea 3 in ethanol in the presence of a catalytic amount of hydrochloric acid at reflux temperature to yield dihydropyrimidine (DHPMs) 4aj in good yield [25,26] (Scheme 1 and Experimental Section).
The structure of products 4aj has been confirmed by both analytical and spectral analyses. The presence of a single proton at a range of δ = 5.41–5.91 ppm corresponding to H-4 of DHPMs in addition to the two NH groups at δ = 7.13–9.24 ppm and 9.29–11.79 ppm supported the suggested DHPMs structures. Also, molecular weight determination (MS) confirmed their structures. (cf. Scheme 1 and Experimental Section).
The synthetic strategies adopted to obtain the target 8 and 12 are somewhat long and linear with few common intermediates. To this aim, the chalcones derivatives 6a,b, which were prepared by reaction of 2-(piperidin-1-yl)quinoline-3-carbaldehyde (1d) [27] with methyl ketones 5a,b, were reacted with ethyl cyanoacetate in ethanol at room temperature to give pyran-3-carboxylate derivatives 7a,b in fairly good yield (Scheme 2). The structures of compounds 7 were established by both analytical and spectral analyses. The IR spectra show two absorption bands at 1690–1682 cm−1 and 1743–1736 cm−1 for the ester and lactone carbonyl groups, respectively. In addition, the 1H-NMR shows the pyran H-5 at δ 7.33–7.39 ppm and other protons in their expected locations. N-Nucleophilic addition reaction of hydrazine at the lactonic carbonyl group of 7a, gave 1,2-dihydropyridine-3-carboxylate derivative 8. The IR spectra showed absence of the lactonic carbonyl group perilously appeared in the parent 7 and the appearance of new bands at ν 3383, 3182 cm−1 due to NH2 function and 1H-NMR showed a singlet signal at δ 5.41 ppm attributed to amino group.
2-Chloroquinoline-3-carboxylic acid was prepared by oxidation of 1e using silver nitrate in the presence of sodium hydroxide [28]. Esterification of the carboxylic acid derivative 9 using absolute ethanol and sulfuric acid afforded the ester derivative 10, in a good yield, followed by subsequent hydrazinolysis in boiling ethanol to afford 2-chloroquinoline-3-carbohydrazide 11. The later compound 11 was subjected to react with carbon disulfide in ethanol in the presence of KOH under reflux followed by acidification by using diluted hydrochloric acid to give 5-(2-chloro-quinolin-3-yl)-1,3,4-oxadiazole-2-thiol (12). The IR spectrum showed the presence of the absorption band at 2500 cm−1 due to S-H function, in addition 13C-NMR revealed signal at δC 164.54 (C2-1,3,4-oxadiazole) ppm indicates that 12 exists in the thiol form (cf. Scheme 3 and Experimental Section).

2.2. Antimalarial Evaluation

Seventeen quinoline derivatives were evaluated in vitro against P. falciparum. The results of the antimalarial screening are presented in Table 1, Table 2 and Table 3. The basic measurement of antimalarial activity used in this study was the reduction in number of parasitized cells in the test cultures compared to control at 36–48 h of incubation. Compounds exhibiting IC50 P. falciparum >5 μg/mL was considered inactive. If the IC50 is between 0.5 and 5 μg/mL, the compound is classified as moderately active. If the IC50 is <0.5 μg/mL, the compound is classified as active.
The percentages of inhibition plasmodial parasite were recorded in Table 1, Table 2 and Table 3. The data revealed that the highest percent of inhibition (100%) was recorded by 4b, 4g, 4i, 6a, 7b and 8b at the concentrations 5.0, 1.25, 2.50, 5.0, 5.0, and 2.50 μg/mL, respectively. Also, 4b, 4c, 4gi, 8a, 10, 11 and 12 showed inhibition more than 75% of plasmodial parasite at the concentrations 1.25, 5.0, 0.625, 5.0, 0.625, 2.50, 5.0, 5.0 and 2.50 μg/mL, respectively. At the lowest concentrations 0.312, 0.156 and 0.078 μg/mL, 4b, 4g, 4i showed significant effect against parasite where the inhibition percentage were 43.3% ± 0.89%, 24.4% ± 0.54%, and 34.8% ± 1.16%.
Median inhibitory concentrations (IC50) of synthesized compounds against P. falciparum in vitro are summarized in Table 3. The IC50 values for these compounds were in the range 0.014–15.87 μg/mL. Two compounds exhibited IC50 values more than 5 μg/mL against P. falciparum. Twelve compounds (4a, 4c, 4f, 4h, 4j, 6a, 7a,b, 8a,b, 10, 11) showed IC50 values between 0.5 μg/mL and 5 μg/mL, and considered a moderately active. Four compounds (4b, 4j, 4i and 12) were classified as active with IC50 (0.46, 0.30, 0.014 and 0.46 μg/mL) compared with chloroquine 0.49 μg/mL.

2.3. Structure Activity Relationship

Structure-activity relationship (SAR) revealed that pyrimidine-2-thione moieties incorporating tetrazolo, methyltetrazolo or quinolinone substituents at position 4 along with an acetyl or ester group at position 5 (compounds 4b, 4g, 4i) have greater potency than the corresponding pyrimidine compounds with the same substituents except compound 4d which has a lowest effect (IC50 = 15.87 μg/mL). A combination of the three substituents (quinolinone, acetyl, and methyl) at positions 4, 5, and 6 of the pyrimidine-2-thione has the best activity and greater (IC50 = 0.041 μg/mL) than the corresponding pyrimidine, notably 4h (IC50 = 2.21 μg/mL). 2-Chloroquinoline moiety containing a 1,3,4-oxadiazole residue at position 3 (compound 12) has greater activity (IC50 = 0.46 μg/mL) than the corresponding compounds where ethyl ester, or carbohydrazide groups present at position 3 in compounds 10 and 11 (IC50 =1.91 μg/mL and 2.21 μg/mL, respectively).

3. Experimental Section

3.1. General Information

Melting points were determined on digital MFB-595 instrument (Gallenkamp London, UK) using open capillary tubes and are uncorrected. IR spectra were recorded on a FTIR 440 spectrometer (Shimadzu, Tokyo, Japan) using KBr pellets. Mass spectra were obtained on a Qp-2010 plus mass spectrometer (Shimadzu) at 70 eV. 1H-NMR and 13C-NMR spectra were recorded on a model Ultra Shield-NMR spectrometer (500 MHz or 400 MHz, Bruker, Coventry, UK) in DMSO-d6 using tetramethylsilane (TMS) as an internal standard at the College of Science, King Khalid University, Saudi Arabia; chemical shifts are reported as δ ppm units. The elemental analyses (% C, H, N) were done at the Microanalytical Center, Cairo University, Cairo, Egypt. Solvents were dried by standard techniques. The monitoring of the progress of all reactions and homogeneity of the synthesized compounds was carried out and was run using thin layer chromatography (TLC) aluminum sheets silica gel 60 F254 (Merck, Darmstadt, Germany).

3.2. Synthesis

3.2.1. General Procedure for the Synthesis of Dihydropyrimidines (DHPMs) 4aj

A mixture of the appropriate aromatic aldehyde 1ac (10 mmol), ethyl acetoacetate (2a) (or acetylacetone (2b)) (10 mmol), urea (3a) (or thiourea (3b)) (10 mmol) in ethanol (50 mL) in the presence of a catalytic amount of hydrochloric acid was refluxed for 8–12 h (TLC). The precipitated crude was filtered off, washed with ethanol and recrystallized from acetic acid to give pure crystals of DHPMs 4aj.
Ethyl-6-methyl-2-oxo-4-(tetrazolo[1,5-a]quinolin-4-yl)-1,2,3,4-tetrahydropyrimidine-5-carboxylate (4a) Yellow crystals, m.p. 278–280 °C; IR (cm−1): ν 3256 and 3122 (2N–H str.), 1725 (C=O), 1693 (C=O), 1610 (C=N), 1556 (C=C); 1H-NMR (DMSO-d6): δH 0.94 (t, 3H, J = 6.6 Hz, CH3), 2.29 (s, 3H, CH3), 3.99 (q, 2H, J = 6.6 Hz, CH2), 5.85 (s, 1H, CH), 7.74 (s, 1H, D2O exchangeable, NH), 7.79–8.59 (m, 5H, quinoline-H), 9.49 (s, 1H, D2O exchangeable, NH) ppm; 13C-NMR (DMSO-d6): δC 13.88, 18.06, 52.19, 59.07, 95.22, 116.11, 123.56, 128.33, 128.60, 129.33, 129.58, 130.29, 131.29, 146.29, 150.33 (quinoline-C), 151.33 (CONH), 165.01 (C=O, ester) ppm; EI-Ms: m/z (%): 353 [M+ + 1]. Anal. Calcd for C17H16N6O3 (352.35.): C, 57.95; H, 4.58; N, 23.85; Found C, 57.80; H, 4.60; N, 23.70.
Ethyl-6-methyl-4-(tetrazolo[1,5-a]quinolin-4-yl)-2-thioxo-1,2,3,4-tetrahydropyrimidine-5-carboxylate (4b) Yellow crystals, m.p. 263–265 °C; IR (cm−1): ν 3275 and 3146 (2NH), 1700 (C=O), 1690 (C=O), 1612 (C=N), 1588 (C=C); 1H-NMR (DMSO-d6): δH 1.02 (t, 3H, J = 6.77 Hz, CH3), 2.20 (s, 3H, CH3), 4.10 (q, 2H, J = 6.77 Hz, CH2), 5.80 (s, 1H, CH), 7.74 (s, 1H, D2O exchangeable, NH), 7.79–8.59 (m, 5H, quinoline-H), 10.39 (s, 1H, D2O exchangeable, NH) ppm; 13C-NMR (DMSO-d6): δC 13.88, 18.06, 52.19, 59.07, 95.22, 116.11, 123.56, 128.33, 128.60, 129.33, 129.58, 130.29, 131.29, 146.29, 150.33 (quinoline-C), 151.33 (CONH), 165.01 (C=O, ester) ppm; EI-Ms: m/z (%): 368 [M+, 35]; Anal. Calcd for C17H16N6O2S (368.41): C, 55.42; H, 4.38; N, 22.81; Found C, 55.60; H, 4.50; N, 22.90.
5-Acetyl-6-methyl-4-(tetrazolo[1,5-a]quinolin-4-yl)-3,4-dihydropyrimidin-2(1H)-one (4c) Yellow crystals, m.p. ˃300 °C; IR (cm−1): ν 3350 and 3294 (2NH), 1687 (C=O),1651 (C=O), 1622 (C=N), 1565 (C=C); 1H-NMR (DMSO-d6): δH 2.18 (s, 3H, CH3), 2.30 (s, 3H, CH3), 5.65 (s, 1H, CH), 7.20–7.80 (m, 5H, quinoline-H), 8.01 (s, 1H, D2O exchangeable, NH), 9.29 (s,1H, D2O exchangeable, NH) ppm; 13C-NMR (DMSO-d6): δC 17.35, 29.88, 49.50, 106.95, 116.87, 118.89, 121.94, 128.11, 130.22, 133.04, 134.65, 138.09, 149.12, 150.27 (quinoline-C), 152.26 (C=O), 193.22 (C=O) ppm; EI-Ms: m/z (%): 322 [M+, 23]; Anal. Calcd for C16H14N6O2 (322.32): C, 59.62; H, 4.38; N, 26.06; Found C, 59.80; H, 4.20; N, 26.20.
1-(6-Methyl-4-(tetrazolo[1,5-a]quinolin-4-yl)-2-thiooxo-1,2,3,4-tetrahydropyrimidin-5-yl)ethanone (4d) Yellow crystals, m.p. 280–282 °C; IR (cm−1): ν 3373, 3172 (2NH), 1685 (C=O), 1628 (C=N), 1588 (C=C); 1H-NMR (DMSO-d6): δH 2.23 (s, 3H, CH3), 2.39 (s, 3H, CH3), 5.57 (s, 1H, CH), 7.18–7.77 (m, 5H, quinoline-H), 7.95 (s, 1H, D2O exchangeable, NH), 9.44 (s,1H, D2O exchangeable, NH) ppm; 13C-NMR (DMSO-d6): δC 17.88, 29.70, 49.67, 106.17, 114.83, 118.71, 127.40, 128.17, 130.40, 132.46, 135.76, 138.23, 146.39, 151.22 (quinoline-C), 161.98 (C=O), 174.60 (C=S) ppm; EI-Ms: m/z (%): 338 [M+, 25]; Anal. Calcd for C16H14N6OS (338.39): C, 56.79; H, 4.17; N, 24.84; Found C, 56.80; H, 4.30; N, 24.70.
Ethyl-6-methyl-4-(7-methyltetrazolo[1,5-a]quinolin-4-yl)-2-oxo-1,2,3,4-tetrahydropyrimidine-5-carboxylate (4e) Yellow crystals, m.p. 293 °C; IR (cm−1): ν 3325, 3271 (2NH), 1720 (CO, ester), 1675 (C=O), 1610 (C=N), 1596 (C=C); 1H-NMR (DMSO-d6): δH 1.19 (t, 3H, J = 6.50 Hz, CH3), 2.33 (s, 3H, CH3), 2.55 (s, 3H, CH3), 4.20 (q, 2H, J = 6.50 Hz, CH2), 5.91 (s, 1H, CH), 7.13 (s, 1H, D2O exchangeable, NH), 7.77 (d, 1H, J = 8.5 Hz, quinoline-H), 7.97 (d, 1H, J = 8.5 Hz, quinoline-H), 8.19 (s, 1H, quinoline-H), 8.46 (s, 1H, quinoline-H), 9.55 (s, 1H, D2O exchangeable, NH); 13C-NMR (DMSO-d6): δC 13.91, 18.03, 20.84, 52.11, 62.49, 59.07, 95.22, 123.63, 127.45, 128.45, 128.96, 129.31, 129.51, 132.62, 138.10, 145.70 (quinoline-C), 151.38 (CONH), 157.66 (C=O, ester) ppm; EI-Ms: m/z (%): 367 [M+ + 1, 15], Anal. Calcd for C18H18N6O3 (366.37): C, 59.01; H, 4.95; N, 22.94; Found C, 59.20; H, 4.90; N, 22.90.
Ethyl-6-methyl-4-(7-methyltetrazolo[1,5-a]quinolin-4-yl)-2-thioxo-1,2,3,4-tetrahydropyrimidine-5-carboxylate (4f) Yellow crystals, m.p. 289 °C; IR (cm−1): ν 3325, 3271 (2NH), 1710 (C=O, ester), 1683 (C=O), 1624 (C=N), 1596 (C=C); 1H-NMR (DMSO-d6): δH 1.24 (t, 3H, J = 6.55 Hz, CH3), 2.33 (s, 3H, CH3), 2.55 (s, 3H, CH3), 4.10 (q, 2H, J = 6.55 Hz, CH2), 5.85 (s, 1H, CH), 7.80 (s, 1H, D2O exchangeable, NH), 7.82 (d, 1H, J = 8.7 Hz, quinoline-H), 8.01 (d, 1H, J = 8.7 Hz, quinoline-H), 8.11 (s,1H, quinoline-H), 8.50 (s, 1H, quinoline-H), 9.59 (s, 1H, D2O exchangeable, NH); 13C-NMR (DMSO-d6): δC 13.81, 17.30, 20.84, 51.98, 59.42, 115.97, 123.67, 127.40, 128.66, 128.96, 129.06, 129.22, 130.84, 132.82, 138.27, 146.59 (quinoline-C), 157.66 (C=O, ester), 174.55 (C=S) ppm; EI-Ms: m/z (%): 382 [M+, 23]; Anal. Calcd for C18H18N6O2S (382.44.): C, 56.53; H, 4.74; N, 21.97; Found C, 56.50; H, 4.80; N, 21.80.
1-(6-Methyl-4-(7-methyltetrazolo[1,5-a]quinolin-4-yl)-2-thioxo-1,2,3,4-tetrahydropyrimidine-5-ethanone (4g) Yellow crystals, m.p. 280–282 °C; IR (cm−1): ν 3290, 3215 (2NH), 1700 (C=O), 1655 (C=O), 1614 (C=N), 1586 (C=C); 1H-NMR (DMSO-d6): δH 2.33 (s, 3H, CH3), 2.40 (s, 3H, CH3), 2.60 (s, 3H, CH3), 5.77 (s, 1H, CH), 7.49 (s, 1H, D2O exchangeable, NH), 7.77 (d, 1H, J = 9 Hz, quinoline-H), 7.99 (d, 1H, J = 9 Hz, quinoline-H), 8.10 (s, 1H, quinoline-H), 8.50 (s, 1H, quinoline-H), 9.44 (s, 1H, D2O exchangeable, NH); 13C-NMR (DMSO-d6): δC 17.81, 20.84, 29.18, 50.98, 106.17, 114.83, 118.71, 128.17, 130.40, 132.46, 135.76, 136.4, 138.23, 146.39, 151.22 (quinoline-C), 174.60 (C=S), 192.20 (C=O) ppm; EI-Ms: m/z (%): 352 [M+, 45]; Anal. Calcd for C17H16N6OS (352.41.): C, 57.94; H, 4.58; N, 23.85; Found C, 57.80; H, 4.70; N, 23.80.
3-(5-Acetyl-6-methyl-2-oxo-1,2,3,4-tetrahydropyridin-4yl)quinolin-2(1H)-one (4h) Yellow crystals, m.p. ˃300 °C; IR (cm−1): ν 3301, 3224, 3180 (3NH), 1700 (C=O), 1649 (C=O), 1618 (C=N), 1583 (C=C); 1H-NMR (DMSO-d6): δH 2.16 (s, 3H, CH3), 2.35 (s, 3H, CH3), 5.44 (s, 1H, CH), 7.16–8.04 (m, 4H, quinoline-H), 8.32 (s, 1H, quinoline-H), 9.24 (s,1H, D2O exchangeable, NH), 11.97 (s, 1H, D2O exchangeable, NH), 12.31 (s, 1H, D2O exchangeable, NH); 13C-NMR (DMSO-d6): δC 18.35, 29.91, 49.25, 106.95, 114.87, 118.89, 121.94, 128.11, 130.22, 133.04, 134.65, 138.09, 149.12 (quinoline-C), 152.26 (C=O), 161.32 (C=O), 194.22 (C=O) ppm; EI-Ms: m/z (%): 297 [M+]; Anal. Calcd for C16H15N3O3 (297.31.): C, 64.64; H, 5.09; N, 14.13; Found C, 64.50; H, 5.20; N, 14.20.
3-(5-Acetyl-6-methyl-2-thioxo-1,2,3,4-tetrahydropyrimidin-4yl)quinolin-2(1H)-one (4i) Yellow crystals, m.p. ˃300 °C; IR (cm−1): ν 3299, 3234, 3172 (3NH), 1651 (C=O), 1610 (C=N), 1590 (C=C); 1H-NMR (DMSO-d6): δH 2.20 (s, 3H, CH3), 2.30 (s, 3H, CH3), 5.50 (s, 1H, CH), 7.16–7.95 (m, 4H, quinoline-H), 8.20 (s, 1H, quinoline-H), 9.11 (s,1H, D2O exchangeable, NH), 11.82 (s, 1H, D2O exchangeable, NH), 12.20 (s, 1H, D2O exchangeable, NH); 13C-NMR (DMSO-d6): δC 17.19, 49.78, 59.42, 104.66, 114.83, 118.71, 127.40, 128.17, 129.23, 130.40, 132.46, 135.76, 138.23, 146.39 (quinoline-C), 164.67 (C=O), 174.50 (C=S) ppm; EI-Ms: m/z (%): 313 [M+]; Anal. Calcd for C16H15N3O2S (313.37): C, 61.37; H, 4.82; N, 13.41; Found C, 61.20; H, 4.70; N, 13.20.
Ethyl-6-methyl-4-(2-oxo-1,2-dihydroquinolin-3-yl)-2-thioxo-1,2,3,4-tetrahydropyrimidine-5-carboxylate (4j) Yellow crystals, m.p. ˃300 °C; IR (cm−1): ν 3377, 3224, 3186 (3NH), 1780 (C=O, ester), 1660 (C=O), 16118 (C=N), 1566 (C=C); 1H-NMR (DMSO-d6): δH 1.08 (t, 3H, J = 7 Hz, CH3), 2.36 (s, 3H, CH3), 4.01 (q, 2H, J = 7 Hz, CH2), 5.41 (s, 1H, CH), 7.17 (dd, 1H, J = 1 Hz, J = 8 Hz, quinoline-H), 7.49 (dd, 1H, J = 1.35 Hz, J = 7.25 Hz, quinoline-H), 7.53 (dd, 1H, J = 1.5 Hz, J = 9 Hz, quinoline-H), 7.71 (dd, 1H, J = 2 Hz, J = 8.2 Hz), 7.72 ( s, 1H, quinoline-H), 9.01 (s, 1H, D2O exchangeable, NH), 10.33 (s, 1H, D2O exchangeable, NH), 11.90 (s, 1H, D2O exchangeable, NH); 13C-NMR (DMSO-d6): δC 14.01, 17.19, 49.78, 59.42, 97.97, 114.83, 118.71, 127.40, 128.17, 130.40, 132.46, 135.76, 138.23, 146.39 (quinoline-C), 160.80 (C=O, ester) ,164.98 (C=O), 174.64 (C=S) ppm; EI-Ms: m/z (%): 343.95 [M+, 35], Anal. Calcd for C17H17N3O3S (343.40.): C, 59.46; H, 4.99; N, 12.24; Found C, 59.50; H, 4.80; N, 12.20.

3.2.2. General Procedure for the Synthesis of Chalcones 6a,b

To a stirred solution of acetophenone (5a) or 2-acetylthiophene (5b) (10 mmol) in alcoholic NaOH solution (5%, 25 mL) at 0–5 °C a solution of 2-(piperidin-1-yl) quinoline-3-carbaldehyde (1e) (2.40 g, 10 mmol) was added gradually. Stirring was continued for 24 h at r.t. the resulting precipitate was filtrated, washed with EtOH (10 mL), and dried, and crystallized from ethanol.
1-Phen.yl-3-[2-(piperidin-1-yl)quinolin-3-yl]prop-2-en-1-one (6a). Yellow crystals, m.p. 127–129 °C, IR (cm−1): ν 1654 (C=O), 1585 (C=C); 1H-NMR (CDCl3): δH 1.62–1.77 (m, 6H, CH2-piperidinyl), 3.30–3.34 (m, 4H, CH2-piperidinyl), 6.94 (d, 1H, J = 18.90 Hz, CH=CH), 7.40–7.79 (m, 10H, quinoline-H & Ar-H & CH=CH), 8.14 (s, 1H, quinoline-H); EI-Ms: m/z (%): 342 [M+, 60]; Anal. Calcd for C23H22N2O (342.43.): C, 80.67; H, 6.48; N, 8.18; Found C, 80.50; H, 6.60; N, 8.20.
3-[2-(Piperidin-1-yl)quinolin-3-yl]-1-thiophene-2-yl)prop-2-en-1-one (6b). Yellow crystals, m.p. 162–164 °C (EtOH); IR (cm−1): ν 1649 (C=O), 1595 (C=C); 1H-NMR (CDCl3): δH 1.66–1.80 (m, 6H, CH2-piperidinyl), 3.34–3.36 (m, 4H, CH2-piperidinyl), 7.19–7.92 (m, 8H, quinoline-H , thiophene-H, CH=CH), 8.02 (d, 1H, J = 19.2 Hz, CH=CH), 8.21 (s, 1H, quinoline-H); 13C-NMR (CDCl3): δC 24.59, 25.88, 51.83, 122.17, 122.94, 124.32, 127.36, 128.31, 128.38, 130.49, 131.84.132.19, 132.95, 133.99, 137.26, 141.80, 145.48, 160.61(thiophene-C & quinoline-C), 181.97 (C=O) ppm; EI-Ms: m/z (%): 348 [M+, 74], Anal. Calcd for C21H20N2OS (348.46.): C, 72.38; H, 5.79; N, 8.04; Found C, 72.40; H, 5.70; N, 8.10.

3.2.3. General Procedure for the Synthesis of 7a,b

A mixture of 6a,b (10 mmol) and ethyl cyanoacetate (1.13 mL, 10 mmole) in absolute ethanol (20 mL) in the presence of a few drops of piperidine was stirred at room temperature for 6 h. The solid formed was filtered off, washed with ethanol, dried and crystallized from acetic acid.
Ethyl-2-oxo-6-phenyl-4-[2-(piperidin-1-yl)quinolin-3-yl]-2H-pyran-3-carboxylate (7a). Colorless crystals, m.p. 174–176 °C; IR (cm−1): ν 1736 (C=O), 1681 (C=O), 1595 (C=C); 1H-NMR (CDCl3): δH 1.38 (t, 3H, J = 7.5 Hz, CH3), 1.69–1.89 (m, 6H, CH2-piperidinyl), 3.53–3.57 (m, 4H, CH2-piperidinyl), 4.35 (q, 2H, J = 7.5 Hz, CH2), 7.39 (s, 1H, pyran H-5), 7.52–7.93 (m, 9H, Ar-H & quinoline-H), 8.06 (s, 1H, quinoline-H); 13C-NMR (CDCl3): δC 14.05, 24.45, 26.33, 52.66, 62.68, 115.20, 116.09, 125.17, 125.75, 127.38, 127.56, 127.72, 128.07, 128.17, 128.31, 128.88, 129.47, 133.74, 133.81, 134.74, 136.21, 136.33, 146.71, 161.52 (thiophene-C & quinoline-C), 165.56 (C=O), 196.49 (CO, ester) ppm; EI-Ms: m/z (%): 356 [M+ + 2, 100]; Anal. Calcd for C28H26N2O4 (454.52.): C, 73.99; H, 5.77; N, 6.16; Found C, 73.90; H, 5.70; N, 6.10.
Ethyl-2-oxo-4-[2-(piperidin-1-yl)quinolin-3yl]-6-(thiophen-2-yl)-2H-pyran-3-carboxylate (7b). Colorless crystals, m.p. 154–156 °C; IR (cm−1): ν 1743 (CO), 1662 (C=O), 1596 (C=C). 1H-NMR (CDCl3): δH 1.34 (t, 3H, J = 6.95 Hz, CH3), 1.65–1.80 (m, 6H, CH2-piperidinyl), 3.50–3.59 (m, 4H, CH2-piperidinyl), 4.30 (q, 2H, J = 6.95 Hz, CH2), 7.33 (s, 1H, pyran H-5), 7.40–8.06 (m, 7H, quinoline-H & thiophene-H), 8.20 (s, 1H, quinoline-H); EI-Ms: m/z (%): 461 [M+ + 1, 78], Anal. Calcd for C26H24N2O4S (460.54.): C, 67.81; H, 5.25; N, 6.08; Found C, 67.80; H, 5.400; N, 6.20.
Ethyl-1-amino-2-oxo-6-phenyl-4-(2-(piperidin-1-yl)quinolin-3-yl)-1,2-dihydropyridine-3-carboxylate (8). A mixture of 7b (4.6 g, 10 mmol) and hydrazine hydrate (10 mmol) in absolute ethanol (20 mL) was stirred at refluxed temperature for 8 h. The formed solid was filtered off, washed with ethanol, dried and crystallized from acetic acid to give as colorless crystals, m.p. 184–186 °C, (80%, yield) IR (cm−1): ν 3383, 3182 (NH2), 1690 (C=O), 1656 (C=O), 1588 (C=C); 1H-NMR (DMSO-d6): δH 1.19 (t, 3H, J = 6.67 Hz, CH3), 1.73–1.84 (m, 6H, CH2-piperidinyl), 3.33–3.43 (m, 4H, CH2-piperidinyl), 4.44 (q, 2H, J = 6.67 Hz, CH2), 5.41 (s, 2H, D2O exchangeable, NH2), 6.70 (s, 1H, pyran H-4), 7.59–7.92 (m, 9H, Ar-H & quinoline-H), 8.79 (s, 1H, quinoline-H); 13C-NMR (DMSO-d6): δC 18.52, 23.42, 26.34, 52.67, 56.00, 125.47, 126.77, 126.97, 127.01, 127.27, 127.73,127.87, 127.93, 128.79, 128.83, 129.88, 131.11, 132,19, 134.28, 136.09 ,137.74, 139.34, 144.87, 145.40 (Ar-C & quinoline-C), 150.26 (C=O), 165.66 (C=O) ppm; EI-Ms: m/z (%): 468 [M+, 25], Anal. Calcd for C28H28N4O3 (468.54): C, 71.78; H, 6.02; N, 11.96; Found C, 71.57; H, 6; N, 11.98.
Ethyl-2-chloroquinolin-3- carboxylate (10). To a solution of 2-chloroquinoline-3-carboxylic acid (9, 2.07 g, 10 mmol) in absolute ethanol (50 mL), 5 drops of conc. H2SO4 were added and the reaction mixture was heated under reflux for 8 h. The solution was poured onto crushed ice water; the precipitate was filtered, washed with water, dried and recrystallized from ethanol to afford 10. White crystals, 82% yield, m.p. 295–297 °C; IR (cm−1): ν 1716 (C=O), 1611 (C=N), 1596 (C=C); 1H-NMR (CDCl3): δH 1.3 (t, 3H, J = 7.25 Hz, CH3), 4.34 (q, 2H, J = 7.25 Hz, CH2), 7.38–8.12 (m, 4H, quinoline-H), 8.54 (s, 1H, quinoline-H) ppm; EI-Ms: m/z (%): 235 [M+, 100], 237 [M+ + 2, 33]; Anal. Calcd for C12H10ClNO2 (235.67): C, 61.16; H, 4.28; N, 5.94; Found C, 61.30; H, 4.40; N, 5.80.
2-Chloroquinoline-3-carbohydrazide (11). A solution of ester derivative 10 (2.35 g, 10 mmol,) and hydrazine hydrate (10 mL) in absolute ethanol (50 mL) was refluxed for 12 h. The solution was cooled to room temperature, poured onto cooled water, the resulting precipitate was filtered, washed with water, (3 × 20 mL) dried and recrystallized from ethanol to give 11 as a white crystals in 76% yield, m.p. ˃300 °C; IR (cm−1): ν 3409, 3309 (NH2), 3135 (NH), 1652 (C=O), 1611 (C=N), 1585 (C=C), 1H-NMR (CDCl3): δH 4.60 (s, brs., 2H, D2O exchangeable, NH2), 7.48–7.54 (dd, 1H, J = 1.60 Hz, J = 8.99 Hz, quinoline-H), 7.70–7.75 (m, 1H, quinoline-H), 8.01–8.06 (dd, 1H, J = 2.1 Hz, J = 8.6 Hz, quinoline-H), 8.85 (s, 1H, quinoline-H), 9.25 (s, 1H, D2O exchangeable, NH) ppm; EI-Ms: m/z (%): 223 [M+ + 2, 26], 221 [M+, 9]; Anal. Calcd for C10H8ClN3O (221.64): C, 54.19; H, 3.64; N, 18.96; Found C, 54.30; H, 3.50; N, 18.80.
5-(2-Chloroquinolin-3-yl)-1,3,4-oxadiazole-2)-thiol (12). To a solution of 11 (2.21 g, 10 mmol) in ethanol (10 mL) was added carbon disulfide (50 mmol) and potassium hydroxide (0.65 g, 10 mmol) at 0 °C. The resulting solution was refluxed for 4 h. The solvent was evaporated and the residue dissolved in water and acidified with a diluted solution of HCl. The resulting precipitate was filtered, washed with water, dried and recrystallized from ethanol to afford compound 12. Yield 78%, Colorless crystals, m.p. 278–280 °C, IR (cm−1): 2500 cm−1 (-SH, stretching), 1612 (C=N), 1598 (C=C); 1H-NMR (DMSO-d6): δH 7.38–7.42 (dd, 1H, J = 2.1 Hz, J = 8.6 Hz, quinoline-H), 7.505–7.522 (d, 1H, J = 8 Hz, quinoline-H), 7.75–7.79 (dd, 1H, J = 1.5 Hz, J = 7 Hz quinoline-H), 8.03–8.04 (dd, 1H, J = 1.5 Hz, J = 8 Hz, quinoline-H), 8.96 (s, 1H, quinoline-H) 13.4 (s, 1H, SH); 13C-NMR (DMSO-d6): δC 115.98, 116.14, 116.30, 117.56, 119.18, 123.67, 123.83, 123.99, 130.17, 130.33, 133.85, 134.01, 139.38, 146.36, 146.52, 163.94, 164.54 (C2-1,3,4-oxadiazole) ppm; EI-Ms: m/z (%): 265 [M+ + 2, 11], 263 [M+, 33]; Anal. Calcd for C11H6ClN3OS (263.70): C, 50.10; H, 2.29; N, 15.39; Found C, 50.30; H, 2.40; N, 15.20.

3.3. Anti-Plasmodial Assay

Blood sample infected with the malaria parasite P. falciparum strain was obtained from King Fahd Hospital in Jazan. Parasitemia was measured by examining 1500 red cells with microscopic inspection of Giemsa-stained thin blood smears and is reported as the percent of parasitized erythrocytes. The parasitisma in sample was 3.3%. In vitro experiments were conducted to measure inhibition of parasite growth after incubation of human parasitized red blood cells in media of Roswell Park Memorial Institute (RPMI 1640) and fetal calf serum. Briefly, this procedure involved preparing stock solutions of each compound in dimethyl sulfoxide (DMSO). The stock solutions were diluted to provide test solutions having concentrations in the range 0.078–5.0 μg/mL. Test solutions were transferred to 96-well test plates containing parasitized red blood cells with 3.3% parasitaemia. Each sample was evaluated in triplicate and the test plate was incubated for 48 h at 37 °C. After incubation, quantification of parasites was achieved by optical microscopy on blood smears from each well [29]. The blood films were fixed with methanol alcohol and transferred into a stand and immersed in Giemsa’s stain for 30 min, then washed with tape water and kept to dry until microscopic examination. The ring and schizont forms were counted. Inhibition percentage was calculated using the following formula: Parasite number of control well-Parasite number of treated well/Parasite number of control well ×100.

3.4. Statistical Analysis

Statistical analysis of the data was carried out according to the method of Lentner [30]. IC50 values were calculated using multiple linear regressions [31].

4. Conclusions

A series of new quinoline derivatives has been synthesized starting from tetrazoloquinoline-3-carbaldehyde, 2-oxo-1,2-dihydroquinoline-3-carbaldehyde, 2-chloroquinoline-3-carbaldehyde, and 2-(piperidin-1-yl)quinoline-3-carbaldehyde. In vitro antimalarial evaluation of the synthesized compounds showed moderate to high antimalarial activities, with IC50 values ranging from 0.014–15.87 μg/mL. The presence of acetyl groups along with 2-thiooxo-1,2,3,4-tetrahydropyrimidine and 1,3,5-oxadiazole residues incorporating quinoline moieties is responsible for increasing the antimalarial activity compared with classical antimalarial agents (chloroquine).

Acknowledgments

The authors are thankful to the Deanship of Scientific Research, Jazan University, Saudi Arabia for the financial support via project No: 1434-G5-19.

Author Contributions

Research design: Rizk E. Khidre; Emad M. El-Telbani, Ibrahim Ali M. Radini Literature review: Rizk E. Khidre; Emad M. El-Telbani, and Ibrahim Ali M. Radini Conducted experiments, Tarek M.Y. Elsheikh Conducted antimalarial: Rizk E. Khidre; Emad M. El-Telbani, Ibrahim Ali M. Radini; Data analysis: Rizk E. Khidre; Emad M. El-Telbani, Ibrahim Ali M. Radini i; Manuscript writing (wrote and contributed): Rizk E. Khidre; Emad M. El-Telbani, Ibrahim Ali M. Radini and Tarek M.Y. Elsheikh. The Authors disclose no actual or potential conflict of interest, and have approved the article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. World Health Organization. WHO Expert Committee on Malaria: Twentieth Report, Technical Report 892. 2000. Available online: http://www.who.int/iris/handle/10665/42247#sthash.rpqlEh0j.dpuf (accessed on 12 July 2016).
  2. World Health Organization. World Malaria Report; WHO: Geneva, Switzerland, 2012; Available online: http://www.who.int/malaria/publications/world_malaria_report_2012/en/ (accessed on 12 July 2016).
  3. Marques, M.M.; Costa, M.R.F.; Santana Filho, F.S.; Vieira, J.L.F.; Nascimento, M.T.S.; Brasil, L.W.; Nogueira, F.; Silveira, H.; Reyes-Lecca, R.B.; Monteiro, W.M.; et al. Plasmodium vivax Chloroquine Resistance and Anemia in the Western Brazilian Amazon. Antimicrob. Agents Chemother. 2013, 58, 342–347. [Google Scholar] [CrossRef] [PubMed]
  4. Cruz, L.R.; Spangenberg, T.; Lacerda, M.V.G.; Wells, T.N.C. Malaria in South America: A drug discovery perspective. Malaria J. 2013, 12, 168–180. [Google Scholar] [CrossRef] [PubMed]
  5. Gama, B.E.; Lacerda, M.V.G.; Daniel-Ribeiro, C.T.; Ferreira-da-Cruz, M.F.F. Chemoresistance of Plasmodium falciparum and Plasmodium vivax parasites in Brazil: Consequences on disease morbidity and control. Mem. Inst. Oswaldo Cruz. 2011, 106, 159–166. [Google Scholar] [CrossRef] [PubMed]
  6. Haynes, J.D.; Diggs, C.L.; Hines, F.A.; Desjardins, R.E. Culture of human malaria parasites Plasmodium falciparum. Nature 1976, 263, 767–769. [Google Scholar] [CrossRef] [PubMed]
  7. White, N.J. Antimalarial drug resistance. J. Clin. Investig. 2004, 113, 1084–1092. [Google Scholar] [CrossRef] [PubMed]
  8. Widyawaruyanti, A.; Devi, A.P.; Fatria1, N.; Tumewu, L.; Tantular, I.S.; Hafid, A.F. In vitro antimalarial activity screening of several Indonesian plants using hrp2 assay. Int. J. Pharm. Pharm. Sci. 2014, 6, 125–128. [Google Scholar]
  9. Wells, C.D.; Cegielski, J.P.; Nelson, L.J.; Laserson, K.F.; Holtz, T.H.; Finlay, A.; Castro, K.G.; Weyer, K. HIV infection and multidrug-resistant tuberculosis-the perfect storm. J. Infect. Dis. 2007, 196, S68–S107. [Google Scholar] [CrossRef] [PubMed]
  10. Khan, M.W.; Alam, M.J.; Rashid, M.A.; Chowdhury, R. A new structural alternative in benzo[b]furans for antimicrobial activity. Bioorg. Med. Chem. 2005, 13, 4796–4805. [Google Scholar] [CrossRef] [PubMed]
  11. Abdel-Wahab, B.F.; Khidre, R.E.; Farahat, A.A.; El-Ahl, A.A.S. 2-Chloroquinoline-3-carbaldehydes: Synthesis, reactions and applications. Arkivoc 2012, I, 211–276. [Google Scholar] [CrossRef]
  12. Dominguez, J.; Basante, W.; Charris, J.; Riggione, F. Synthesis and activity of some quinolone derivatives against Plasmodium falciparum in vitro. Fermaco 1996, 51, 407–412. [Google Scholar]
  13. Ali, M.M.; Rajanna, K.C.; Prakash, P.K.S. An Efficient and Facile Synthesis of 2-Chloro-3-formyl Quinolines from Acetanilides in Micellar Media by Vilsmeier-Haack Cyclisation. Synlett 2001, 2, 251–253. [Google Scholar]
  14. Pokalwar, R.U.; Hangarge, R.V.; Maske, P.V.; Shingare, M.S. Synthesis and antibacterial activities of α-hydroxyphosphonates and α-acetyloxyphosphonates derived from 2-chloroquinoline-3-carbaldehyde. Arkivoc 2006, 11, 196–204. [Google Scholar]
  15. Khidre, R.E.; Abu-Hashem, A.A.; El-Shazly, M. Synthesis and anti-microbial activity of some 1-substituted amino-4,6-dimethyl-2-oxo-pyridine-3-carbonitrile derivatives. Eur. J. Med. Chem. 2011, 46, 5057–5064. [Google Scholar] [CrossRef] [PubMed]
  16. Kaur, K.; Jain, M.; Reddy, R.P.; Jain, R. Quinolines and structurally related heterocycles as antimalarials. Eur. J. Med. Chem. 2010, 45, 3245–3264. [Google Scholar] [CrossRef] [PubMed]
  17. Abdou, W.M.; Kamel, A.A.; Khidre, R.E.; Geronikaki, A.; Ekonomopoulou, M.T. Synthesis of 5- and 6-N-heterocyclic methylenebisphosphonate derivatives of cytogenetic activity in normal human lymphocyte cultures. Chem. Biol. Drug. Des. 2012, 79, 719–730. [Google Scholar] [CrossRef] [PubMed]
  18. Abdou, W.M.; Shaddy, A.A.; Khidre, R.E.; Awad, G.E.A. Synthesis and Antimicrobial Evaluation of Newly Synthesized N, S-Bisphosphonate Derivatives. J. Heterocycl. Chem. 2015. [Google Scholar] [CrossRef]
  19. Tayebee, R.; Amini, M.M.; Ghadamgahi, M.; Armaghan, M. H5PW10V2O40/Pip-SBA-15: A novel reusable organic-inorganic hybrid material as potent Lewis acid catalyst for one-pot solvent-free synthesis of 3,4-dihydropyrimidinones. J. Mol. Catal. Chem. 2013, 366, 266–274. [Google Scholar]
  20. Biginelli, P. Aldehyde-urea derivatives of aceto-and oxaloacetic acids. Gazz. Chim. Ital. 1893, 23, 360–369. [Google Scholar]
  21. Hu, E.H.; Sidler, D.R.; Dolling, U.-H. Unprecedented catalytic three component one-pot condensation reaction: An efficient synthesis of 5-alkoxycarbonyl-4-aryl-3,4-dihydropyrimidin-2(1H)-ones. J. Org. Chem. 1998, 63, 3454–3457. [Google Scholar] [CrossRef]
  22. Abdou, W.M.; Khidre, R.E.; Kamel, A.A. Elaborating on Efficient Anti-Proliferation Agents of Cancer Cells and Anti-Inflammatory-Based N-Bisphosphonic Acids. Arch. Pharm. Chem. Life Sci. 2012, 345, 123–136. [Google Scholar] [CrossRef] [PubMed]
  23. Meth-Cohn, O.; Narine, B.; Tarnowski, B. A versatile new synthesis of quinolines and related fused pyridines, Part 5. The synthesis of 2-chloroquinoline-3-carbaldehydes. J. Chem. Soc. Perkin Trans. 1 1981, 9, 1520–1530. [Google Scholar]
  24. Liu, Z.C.; Wang, B.D.; Li, B.; Wang, Q.; Yang, Z.Y.; Li, T.R.; Li, Y. Crystal structures, DNA-binding and cytotoxic activities studies of Cu(II) complexes with 2-oxo-quinoline-3-carbaldehyde Schiff-bases. Eur. J. Med. Chem. 2010, 45, 5353–5361. [Google Scholar] [CrossRef] [PubMed]
  25. Kaur, B.; Kaur, R. Synthesis of fused quinazolinethiones and their S-alkyl/aryl derivatives. Arkivoc 2007, 15, 315–323. [Google Scholar]
  26. Abelman, M.M.; Smith, S.C.; James, D.R. Cyclic ketones and substituted α-keto acids as alternative substrates for novel Biginelli-like scaffold syntheses. Tetrahedron Lett. 2003, 44, 4559–4562. [Google Scholar] [CrossRef]
  27. Obaid, A.; Sandhya, B.; Suresh, K.; Rajiv, K.; Md Quamrul, H. Design, Synthesis Chalcones/Amines and Evaluation of Novel 2-piperidinyl Quinoline as Potential Antidepressant Agents Lett. Drug Des. Discov. 2013, 10, 75–85. [Google Scholar]
  28. Rao, K.R.; Bhanumathi, N.; Sattur, P.B. Synthesis of novel quino[2,3-b][1,5]benzodiazepin-12-ones. J. Heterocycl. Chem. 1991, 28, 1339–1340. [Google Scholar] [CrossRef]
  29. Silva, L.F.R.; Montoia, A.; Amorim, R.C.N.; Melo, M.R.; Henrique, M.C.; Nunomura, S.M.; Costa, M.R.F.; Andrade Neto, V.F.; Costa, D.S.; Dantas, G.; et al. Comparative in vitro and in vivo antimalarial activity of the indole alkaloids ellipticine, olivacine, cryptolepine and a synthetic cryptolepine analog. Phytomedicine 2012, 20, 71–76. [Google Scholar] [CrossRef] [PubMed]
  30. Lentner, C.; Lentner, C.; Wink, A. Student’s t-distribution tables. In Geigy Scientific Tables Vol. 2. International Medical and Pharmaceutical Information; Ciba-Geigy Limited: Basel, Switzerland, 1982. [Google Scholar]
  31. Finney, D.J. Probit Analysis, 3rd ed.; Cambridge University Press: Cambridge, UK, 1971. [Google Scholar]
  • Sample Availability: Samples of the compounds 4, 7, 8, and 12 are available from the authors.
Figure 1. Aminoquinoline and their derivatives as antimalarial agents.
Figure 1. Aminoquinoline and their derivatives as antimalarial agents.
Molecules 21 00909 g001
Scheme 1. Synthesis of dihydropyridine (DHPMs) 4aj.
Scheme 1. Synthesis of dihydropyridine (DHPMs) 4aj.
Molecules 21 00909 sch001

CompoundR1R2XYield %CompoundR1R2XYield %
4a Molecules 21 00909 i001OEtO894f Molecules 21 00909 i002OEtS88
4b Molecules 21 00909 i003OEtS864g Molecules 21 00909 i004MeS86
4c Molecules 21 00909 i005MeO874h Molecules 21 00909 i006MeO85
4d Molecules 21 00909 i007MeS854i Molecules 21 00909 i008MeS89
4e Molecules 21 00909 i009OEtO884j Molecules 21 00909 i010OEtS85
Scheme 2. Synthesis of quinolinyl 1,2-dihydropyridine 8.
Scheme 2. Synthesis of quinolinyl 1,2-dihydropyridine 8.
Molecules 21 00909 sch002
Scheme 3. Synthesis of quinoliny 1,3,4-oxadiazole 12.
Scheme 3. Synthesis of quinoliny 1,3,4-oxadiazole 12.
Molecules 21 00909 sch003
Table 1. % Parasite inhibition of Plasmodium falciparum by some quinolinyl derivatives 412.
Table 1. % Parasite inhibition of Plasmodium falciparum by some quinolinyl derivatives 412.
Compound% Mean Parasite Inhibition Per 1500 RBC
Concentrations (μg/mL)
0.6251.252.55
RangeMean ± SERangeMean ± SERangeMean ± SERangeMean ± SE
4a2.0–7.85.7 ± 1.858.2–18.214.1 ± 3.0231.3–41.836.6 ± 3.0347.1–53.151.0 ± 1.93
4c14.2–15.614.7 ± 0.4226.5–36.335.3 ± 3.0954.5–62.757.4 ± 2.6385.7–92.189.5 ± 1.96
4d2.0–5.43.7 ± 0.980.0–3.61.8 ± 1.036.1–7.87.1 ± 0.4914.3–18.116.1 ± 1.11
4e0.0–2.01.3 ± 0.632.0–7.24.4 ± 1.5113.7–14.214.1 ± 0.2324.4–35.229.5 ± 3.12
4f9.8–12.210.9 ± 0.6927.4–29.128.3 ± 0.4932.7–43.137.5 ± 3.0269.1–78.472.9 ± 2.79
4h20.4–23.621.8 ± 0.9324.5–35.231.4 ± 3.0569.1–75.572.3 ± 1.8480–83.782.0 ± 1.07
4j1.9–7.23.7 ± 1.750.0–5.42.5 ± 1.5719.6–20.420.0 ± 0.2349–56.851.6 ± 2.58
6b42.8–43.643.2 ± 0.2352.7–62.756.2 ± 3.2655.1–69.161.0 ± 4.18100–100100 ± 00
7a27.4–29.129.1 ± 0.9237.3–45.542.6 ± 2.6364.7–67.366.4 ± 0.8666.7–73.571.0 ± 2.14
7b34.5–35.334.8 ± 0.2554.5–55.154.8 ± 0.1778.4–89.183.0 ± 3.17100–100100 ± 00
835.3–50.142.3 ± 4.363.3–76.470.1 ± 3.8100–100100 ± 00--
1011.8–25.517.2 ± 4.2143.1–6051.4 ± 4.8867.3–74.570.3 ± 2.1684.3–84.384.3 ± 00
115.9–18.212.8 ± 3.6239.2–4944.6 ± 2.8663.2–74.570.1 ± 35075.5–83.680.0 ± 2.51
1235.3–45.441.8 ± 3.2759.2–70.964.3 ± 3.4674.5–78.276.7 ± 1.1386.3–91.889.7 ± 1.70
CQ59.9–63.861.8 ± 1.1375.9–78.176.7 ± 0.67100–100100 ± 0.0--
Table 2. % Parasite inhibition of Plasmodium falciparum (% Mean Parasite Inhibition per 1500 RBC) of compounds 4b,g,i.
Table 2. % Parasite inhibition of Plasmodium falciparum (% Mean Parasite Inhibition per 1500 RBC) of compounds 4b,g,i.
Concentrations (μg/mL)Compound
4b4g4iCQ
0.078RangeNPNP32.7–36.7NP
Mean ± SENPNP34.8 ± 1.16NP
0.156RangeNP23.5–25.4NP28.9–33.7
Mean ± SENP24.4 ± 0.54NP31.8 ± 1.44
0.312Range41.8–44.956.3–61.263.6–71.443.3–46.4
Mean ± SE43.3 ± 0.8958.7 ± 1.4167.8 ± 2.2844.9 ± 0.90
0.625Range55.1–70.182.3–89.774.5–86.259.9–63.8
Mean ± SE63.3 ± 4.3887.0 ± 2.3579.4 ± 3.561.8 ± 1.13
1.25Range73.5–82.3100–10085.7–94.175.9–78.1
Mean ± SE76.7 ± 2.78100 ± 0.089.9 ± 2.5976.7 ± 0.67
2.5Range89.1–95.9-100–100100–100
Mean ± SE91.7 ± 2.11-100 ± 0.0100 ± 0.0
5Range100–100---
Mean ± SE100 ± 0.0---
NP = Not performed.
Table 3. IC50 values of some derivatives compound of chloroquine against Plasmodium falciparum.
Table 3. IC50 values of some derivatives compound of chloroquine against Plasmodium falciparum.
CompoundIC50 (μg/mL)SlopeR2
4a4.5910.320.929
4b0.4610.430.772
4c2.3916.280.968
4d15.873.170.932
4e8.106.550.997
4f3.2913.340.977
4g0.3061.620.791
4h2.2114.00.831
4i0.01423.480.733
4j4.9611.670.975
6a1.1312.410.965
7a2.099.110.790
7b1.0614.140.889
80.7629.790.971
101.9113.270.782
112.2113.590.767
120.469.620.834
CQ0.4927.010.927

Share and Cite

MDPI and ACS Style

Radini, I.A.M.; Elsheikh, T.M.Y.; El-Telbani, E.M.; Khidre, R.E. New Potential Antimalarial Agents: Design, Synthesis and Biological Evaluation of Some Novel Quinoline Derivatives as Antimalarial Agents. Molecules 2016, 21, 909. https://doi.org/10.3390/molecules21070909

AMA Style

Radini IAM, Elsheikh TMY, El-Telbani EM, Khidre RE. New Potential Antimalarial Agents: Design, Synthesis and Biological Evaluation of Some Novel Quinoline Derivatives as Antimalarial Agents. Molecules. 2016; 21(7):909. https://doi.org/10.3390/molecules21070909

Chicago/Turabian Style

Radini, Ibrahim Ali M., Tarek M. Y. Elsheikh, Emad M. El-Telbani, and Rizk E. Khidre. 2016. "New Potential Antimalarial Agents: Design, Synthesis and Biological Evaluation of Some Novel Quinoline Derivatives as Antimalarial Agents" Molecules 21, no. 7: 909. https://doi.org/10.3390/molecules21070909

Article Metrics

Back to TopTop