Next Article in Journal
Encapsulation of Hemp (Cannabis sativa L.) Essential Oils into Nanoemulsions for Potential Therapeutic Applications: Assessment of Cytotoxicological Profiles
Previous Article in Journal
Contribution of Dynamic Rheology Coupled to FTIR and Raman Spectroscopies to the Real-Time Shaping Ability of a Hyperbranched Polycarbosilane
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Application of Quinoline Ring in Structural Modification of Natural Products

1
Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, China
2
Department of Pharmacy, Shenyang Medical College, Shenyang 110034, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2023, 28(18), 6478; https://doi.org/10.3390/molecules28186478
Submission received: 11 August 2023 / Revised: 4 September 2023 / Accepted: 5 September 2023 / Published: 6 September 2023
(This article belongs to the Section Medicinal Chemistry)

Abstract

:
Natural compounds are rich in pharmacological properties that are a hot topic in pharmaceutical research. The quinoline ring plays important roles in many biological processes in heterocycles. Many pharmacological compounds, including saquinavir and chloroquine, have been marketed as quinoline molecules with good anti-viral and anti-parasitic properties. Therefore, in this review, we summarize the medicinal chemistry of quinoline-modified natural product quinoline derivatives that were developed by several research teams in the past 10 years and find that these compounds have inhibitory effects on bacteria, viruses, parasites, inflammation, cancer, Alzheimer’s disease, and others.

1. Introduction

Active molecules of natural products have always been an important source of drug leads due to their diverse chemical structures and extensive pharmacological activities. According to statistics, from January 1981 to September 2019, the FDA has approved 1881 new drugs, of which about half are directly or indirectly derived from natural compounds [1]. Quinoline, also known as benzo[b]pyridine, is a nitrogen-containing heterocyclic aromatic molecule with a weak tertiary base that can form salts with acids and perform electrophilic substitution reactions and reactions resembling those of pyridine and benzene. Quinolines have numerous biological effects, such as antibacterial [2,3,4], antifungal [5], antituberculosis [6], antiprotozoal [7,8], antineoplastic [9], anti-viral [10], anti-cholesterol medications [11], analgesics [12], anti-disease Alzheimer’s pharmaceuticals [13], and more.
In fact, quinoline drugs are widely used in the pharmaceutical industry. Many drugs contain quinoline rings. For example, quinine (Figure 1) is one of the natural products is present in the bark of cinchona, which has been used to treat malaria. Camptothecin (Figure 1) is a quinoline alkaloid extracted from Camptotheca acuminata, and its analogue, topotecan (Figure 1), are effective antitumor drugs. And other chemical drugs, such as dibucaine (Figure 1) as a local anesthetic, montelukast (Figure 1) for asthma, aripiprazole (Figure 1) as an antipsychotic, vesnarinonez (Figure 1) as a cardiac agent, etc., all contain quinoline structure. Creating novel homologs with enhanced biological activity and fewer potentially harmful side effects has been a goal for many years. Emphasizing the biological activities of the quinolones, we will highlight some recent results regarding the development of novel quinoline-natural product hybrids in this study.
Figure 1. The chemical structures of introduction.
Figure 1. The chemical structures of introduction.
Molecules 28 06478 g001

2. Method

Published articles, network databases (PubMed, Science Direct, SCI Finder, CNKI), and clinical trial websites https://clinicaltrials.gov/ (accessed on 31 September 2022) related to natural products and quinoline derivatives) were included in the discussion. The 94 studies that met the inclusion criteria were chosen for discussion. It is possible to categorize natural quinoline derivative compounds according to their biological activity by screening. The entire text is divided into sections based on how biologically active their derivatives are, namely, in the following categories: inhibition of bacteria, viruses, parasites, inflammation, cancer, Alzheimer’s disease, etc. The biological activities of quinoline derivatives were discussed in detail. It is worth noting that most derivatives exhibit enhanced biological activity and reduced cytotoxicity compared to lead compounds. Quinoline has many advantages and can be widely used in the synthesis of natural product derivatives to enhance the properties of drug bulks.

3. Pharmacological Activities

3.1. Anti-Alzheimer’s Disease Activity

Coumarin (chemical formula (CF): C9H6O2, molecular weight (MW): 146.14, 2H-1-benzopyran-2-one) is a phenolic compound widely found in orchids, legumes, Umbelliferae, Compositae, Rutaceae, and other plants. Duarte and colleagues [14] synthesized quinoline-substituted compounds 14 (Figure 2) (Table 1) at the seventh position of coumarin and determined their AChE/BChE activity regulation. Compound 2a, containing a strong electron-donating group (methoxy) at position 7, were against both enzymes (AChE IC50 = 194 μM and BChE IC50 = 255 μM), being the only representative dual compound of the entire series. However, compound 2b, which has the same substitution pattern at position 7 and a different aminoquinoline at position 3, showed selective activity against the AChE enzyme (AChE IC50 = 181 μM, selectivity ratio > 2.75). Compound 3, with the same amidoquinoline as compound 2b at position 3 and an electron-withdrawing atom (chloride atom) at position 7 of the coumarin scaffold, was the only selective BChE inhibitor of the entire series (BChE IC50 = 146 μM, selectivity ratio < 0.29). The most active and selective AChE compound among all others was compound 4, which had an electron-donating group (diethylamine) at position 7 and a 6-quinoline derivative at position 3 (AChE IC50 = 159 μM, selectivity ratio > 3.13). This result was intriguing because other electron-donating groups such as methyl and methoxy groups, compounds 1 and 2c, respectively, present at position 7, showed results that were the opposite of this trend, suggesting that the diethylamine substituents at position 7 are only marginally important for the AChE binding affinity. Compound 4 is also an iron chelator (100 μM Fe chelation = 72.87%), forming a well-defined stacking contact with Phe330 and interacting with Tyr121 residues via hydrogen bonding.
Wang and colleagues [15] designed and synthesized 2-arylethenylquinoline derivatives 5 and 6 (Figure 1) (Table 1) against AChE and BChE. The results showed that compounds 5 and 6 had moderate ChE inhibitory activity (IC50 of compound 5 AChE and BChE were 64.0 ± 0.1 and 0.2 ± 0.1 μM, respectively; and the IC50 of compound 6 AChE and BChE were 68.3 ± 0.1 and 1.0 ± 0.1 μM, respectively). These two compounds showed high selectivity for BChE; however, their inhibitory activity against ChE was significantly weaker than that of positive control tacrine. Even the most energetic compound 5 was 640- and 7-fold weaker than tacrine against AChE and BChE, respectively.
Galantamine (CF: C17H22ClNO3, MW:323.8145, (4aS,6R,8aS)-4a,5,9,10,11,12-Hexahydro-3-methoxy-11-methyl-6H-benzofuro[3a,3,2-ef][2]benzazepin-6-ol) was initially isolated and extracted from the bulbs of snowdrops. However, due to the scarcity of the extraction species and the high cost of the extraction, numerous businesses throughout the world started producing galantamine by chemical synthesis. Ţînţaş and colleagues [16] synthesized dihydroquinoline galanthamine derivatives 7ab (Figure 2) (Table 1) and evaluated their in vitro inhibitory activity on AChE in the human body. Compound 7a could not be assessed since it was insoluble in water; however, compound 7b had an IC50 larger than 10 μM. Additionally, its activity was significantly lower than that of the parent galanthamine.
Caffeic acid (CA, CF: C9H8O4, MW: 180.15, 3-(3,4-dihydroxyphenyl)- (9CI, ACI)) is derived from the whole plant of Solidago virga-urea L.var.leiocarpa, the fruit of Crataegus pinnatifida Bge., and others. Benchekroun and colleagues [17] synthesized a caffeic acid derivative 8 (Figure 2) (Table 1) and tested its antioxidant capacity. The experimental findings showed compound 8 showed 25% or more significant neuroprotection against H2O2 damage at 10 μM.
Chromones (CF: C9H6O2, MW: 146.1427, 2,3-Benzo-4-pyrone), scientific name benzo-γ-pyrone, are widely present in plants, and some are colored substances, so their matrixes are called chromone. Shah and colleagues [18] synthesized chromone quinoline derivatives 9ae and 10ae (Figure 2) (Table 1) and evaluated their cholinesterase inhibitory activity. The screening of quinolinyl-chromone derivatives 9ae and 10ae for ChE revealed that most are selective BChE inhibitors. The active group connected to the quinoline acyl chalcone derivative has significant AChE inhibitory activity. The most effective molecule against BChE among 2,6-dimethyl quinoline derivatives is 9e (2,3-dihydrobenzo[b]1,4-dioxin-6-yl as Ar substituent) with an IC50 of 0.56 ± 0.02 μM. The exceptional inhibitory potential of compound 9e may be attributed to the presence of highly electronegative oxygen atoms (1,4-dioxane) and a tiny functional group (-CH3). The chemical 10e likewise contains 1,4-dioxane, but it has a methoxy (-OCH3) functional group in its basic structure, which may limit its inhibitory effectiveness against BChE. Compounds 10a and 10b inhibit BChE significantly, with IC50 of 0.94 ± 0.04 μM and 0.73 ± 0.03 μM, respectively. These compounds have 5-methylfuran-2-yl (for 10a) and 2,5-dimethylfuran-3-yl (for 10b) substitutions at the -Ar position of the quinolinyl ring. When the experimental data from both series were examined, a modest decrease in the inhibitory potential was observed in the presence of a 6-methoxy group at the quinolinyl moiety. To investigate the putative mechanism of ChEs inhibition, detailed kinetic investigations of the most effective derivatives were conducted. The results indicated that compound 9e could interact with both the catalytic anionic site (CAS) and the peripheral anionic site (PAS) of BChE.
Figure 2. The chemical structures of anti-Alzheimer’s disease compounds 110.
Figure 2. The chemical structures of anti-Alzheimer’s disease compounds 110.
Molecules 28 06478 g002
Table 1. Quinoline derivatives with anti-Alzheimer’s disease activity.
Table 1. Quinoline derivatives with anti-Alzheimer’s disease activity.
Compd.Activity TargetOrigin Ref
1AChE IC50 = 324.88 μMAChEsynthetic [14]
2aAChE IC50 = 194 μM
BChE IC50 = 255 μM
ACHE
BCHE
synthetic[14]
2bAChE IC50 = 181.72 μM, selectivity ratio > 2.75ACHEsynthetic[14]
2cAChE IC50 > 500 μM
BChE IC50 > 500 μM
-synthetic[14]
2dAChE IC50 > 500 μM
BChE IC50 > 500 μM
-synthetic[14]
3BChE IC50 = 146.74 μM, selectivity ratio < 0.29BCHEsynthetic [14]
4AChE IC50 = 159.53 μM, selectivity ratio > 3.13ACHEsynthetic [14]
5AChE IC50 = 64.0 μM
BChE IC50 = 0.2 μM
ACHE
BCHE
synthetic [15]
6AChE IC50 = 68.3 μM
BChE IC50 = 1.0 μM
ACHE
BCHE
synthetic [15]
7a--synthetic [16]
7bAChE IC50 > 10 μMACHEsynthetic [16]
825% H2O2 damage at 10 μM.-synthetic [17]
9aAChE IC50 = 2.99 μM
BChE IC50 = 0.91 μM
ACHE
BCHE
synthetic [18]
9bAChE IC50 = 0.32 μM
BChE IC50 = 0.90 μM
ACHE
BCHE
synthetic [18]
9cAChE IC50 = 3.99 μM
BChE IC50 = 0.64 μM
ACHE
BCHE
synthetic [18]
9dAChE IC50 = 3.45 μM
BChE IC50 = 0.63 μM
ACHE
BCHE
synthetic [18]
9eAChE IC50 = 44.46 μM
BChE IC50 = 0.56 μM
ACHE
BCHE
synthetic [18]
10aAChE IC50 = 43.36 μM
BChE IC50 = 0.94 μM
ACHE
BCHE
synthetic [18]
10bAChE IC50 = 46.42 μM
BChE IC50 = 0.73 μM
ACHE
BCHE
synthetic [18]
10cAChE IC50 = 3.91 μM
BChE IC50 = 2.15 μM
ACHE
BCHE
synthetic [18]
10dAChE IC50 = 3.36 μM
BChE IC50 = 2.36 μM
ACHE
BCHE
synthetic [18]
10eAChE IC50 = 40.29 μM
BChE IC50 = 1.32 μM
ACHE
BCHE
synthetic [18]
Summary: Coumarin, 2-aryl, and Chromones-linked quinoline derivatives can enhance their AChE/BChE activity. Among them, compounds 9b and 9e showed inhibitory effects on AchE and BchE, with IC50 values of 0.32 and 0.56 μM, respectively. When the 6 position of Chromones is methyl, the activity is significantly higher than that of methoxy; the activity is most obvious when the R group is furan. In addition, coumarin–quinoline derivatives 3 and 4 showed inhibitory effects on AchE and BchE, with IC50 values of 146.74 and 153.53 μM, respectively; therefore, the Chromones-linked quinoline derivatives have the value of further research.

3.2. Anti-Osteoporosis Activity

Oleanolic acid (OA, CF: C30H48O3, MW: 456.70, (3β)-3-Hydroxyolean-12-en-28-oic acid) is a kind of pentacyclic triterpenoid obtained by separating and extracting from the fruit of the whole herb of the gentian family, the genus of the genus Radix, or Ligustrum lucidum. It exists in free form and glycoside in polyphenols in plants. Li and colleagues [19] tested the inhibitory activity of OA derivatives 11 (Figure 3) (Table 2) in the formation of TRAP-positive, osteoclast-like multinucleated cells (OCLs) induced by the effect of 1a, 25-dihydroxy vitamin D3 [1a,25(OH)2D3] effect. The results showed that compound 11 exhibited good activity at 20 μM (OCL% = 73.0%), which was better than OA at 20 μM. Encouragingly, compound 11 showed moderate activity even at 2 μM (OCL% = 18.9%).
Pregnenolone (CF: C21H32O2, MW: 316.48, 3β-Hydroxy-5-pregnen-20-one), a naturally occurring endogenous steroid, is well-known as one of the biosynthetic precursors of steroid hormones. Maurya and colleagues [20] synthesized pregnenolone derivatives 12ab (Figure 3) (Table 2) and tested its osteogenic effect. Compared with untreated control cells, compounds 12a and 12b significantly increased ALP activity. Among the compounds studied, compound 12a showed the greatest osteogenic effect according to the ALP activity evaluation. At the concentration of 1 pM and 100 pM the ALP activity increased by 100% and 85%, respectively. In addition, studies have shown that compound 12a can significantly increase the formation of mineral nodules, and compound 12a upregulates the expression of osteogenic markers BMP-2, RUNX-2, and OCN.
Figure 3. The chemical structures of anti-osteoporosis compounds 1112.
Figure 3. The chemical structures of anti-osteoporosis compounds 1112.
Molecules 28 06478 g003
Table 2. Quinoline derivatives with anti-osteoporosis activity.
Table 2. Quinoline derivatives with anti-osteoporosis activity.
Compd.Activity TargetOrigin Ref
112 μM OCL% = 18.9%
20 μM OCL% = 73.0%
-synthetic [19]
12a1 pM ALP% = 100%
100 pM ALP% = 85%
ALP, BMP-2, RUNX-2, OCNsynthetic [20]
12b1 pM ALP% = 70%
100 pM ALP% = 60%
ALPsynthetic [20]
Summary: The introduction of quinoline by oleanolic acid only showed moderate activity. The introduction of quinoline by pregnenolone still showed strong ALP activity at a concentration of 1 pM, and there was some research on its mechanism. The anti-osteoporosis activity of compound 12a has a certain research value.

3.3. Anti-Viral Activity

Andrographolide (CF: C20H30O5, MW: 350.45, 3-[2-[Decahydro-6-hydroyx-5-(hydroxymethyl)-5,8a-dimethyl-2-methylene-1-naphthalenylethylidene]dihydro-4-hydroxy-2(3H)-furanone), is derived from the leaves of Andrographis paniculata. Li and colleagues [21] synthesized andrographolide quinoline derivative 1314 (Figure 4) (Table 3) and tested their anti-Zika virus activity. The results show that the EC50 of compound 13 is 1.3 μM. SI > 16 (CC50 values of SNB-19 and Vero cell lines are 22.7 and 20.9 μM). EC50 of compound 14 is 4.5 μM. SI > 19 (CC50s of SNB-19 and Vero cell lines are 88.7 and 85.0 μM). In conclusion, compounds 13 and 14 have good anti-ZIKV virus activity.
Baltina and colleagues [22] tested the anti-ZIKV virus activity of glycyrrhizic acid derivatives 15ab (Figure 4) (Table 3) and found that these two compounds 15ab did not have good anti-Zika virus activity, the IC50 value of compound 15a is less than 30 μM.
Wang and colleagues [23] tested the glycyrrhetinic acid quinoline derivative 16 (Figure 4) (Table 3) and tested its anti-HBV activity. Compound 16 inhibits HBV DNA replication with IC50 of 15.30 μM (SI > 111.0), showing strong anti-HBV activity.
Figure 4. The chemical structures of anti-viral compounds 1316.
Figure 4. The chemical structures of anti-viral compounds 1316.
Molecules 28 06478 g004
Table 3. Quinoline derivatives with anti-viral activity.
Table 3. Quinoline derivatives with anti-viral activity.
Compd.Activity TargetOrigin Ref
13anti-Zika virus EC50 = 1.3 μM-synthetic [21]
14anti-Zika virus EC50 = 4.5 μM -synthetic[21]
15aanti-Zika virus IC50 < 30 μMZIKV NS2B-NS3 proteasesynthetic [22]
15banti-Zika virus IC50 < 30 μMZIKV NS2B-NS3 proteasesynthetic [22]
16anti-HBV IC50 = 15.30 μM-synthetic [23]

3.4. Anti-Hyperglycemic Activity

Lupeol (CF: C30H50O, MW: 426.7174, (3β)-Lup-20(29)-en-3-ol) is a compound found in the epidermis of lupin seeds, fig latex, and rubber. Reddy and colleagues [24] synthesized lupeol quinoline derivative 17 (Figure 5) (Table 4) and measured its anti-lipid activity. The hypolipidemic activity of derivative 17 was screened in mice at a dose of 50 mg/kg body weight. Blood cholesterol decreased by 27% (p < 0.05). Compound 17 showed good cholesterol-lowering effectiveness. Unfortunately, compound 17 reduces HDL-C.
Figure 5. The chemical structures of anti-hyperglycemic compound 17.
Figure 5. The chemical structures of anti-hyperglycemic compound 17.
Molecules 28 06478 g005
Table 4. Quinoline derivatives with anti-hyperglycemic activity.
Table 4. Quinoline derivatives with anti-hyperglycemic activity.
Compd.Activity TargetOrigin Ref
17Blood cholesterol decreased 27%triglyceridessynthetic [24]

3.5. Anti-Inflammatory Activity

Nam and colleagues [25] synthesized a series of resveratrol derivatives 1822 (Figure 6) (Table 5) and measured their anti-inflammatory activities. The iEI value of compounds 1822 is higher than that of the parent compound (iEI values of compounds 1822 are 3.75, 3.18, 4.84, 2.11, and 4.27, respectively).
Glycyrrhetinic acid (GA, CF: C30H46O4, MW: 470.69, 3β-hydroxy-11-oxo-18βH-Olean-12-en-30-oic acid) is a well-known pentacyclic triterpene extracted from liquorice root. Bian and colleagues [26] synthesized quinoline glycyrrhetinic acid derivative 23 (Figure 6) (Table 5) and measured its cytotoxicity and anti-inflammatory activities. Because of its low cytotoxicity, compound 23 was chosen for further investigation of anti-inflammatory effects. The inhibitory effect of quinoline compound 23 in glycyrrhetinic acid structure on IL-6 was significantly stronger than that of glycyrrhetinic acid.
Bakuchiol (CF: C18H24O, MW: 256.383, 4-(3,7-Dimethyl-3-vinylocta-1,6-dien-1-yl)phenol) is a natural plant component found in plant seeds. Ma and colleagues [27] synthesized quinoline bakuchiol derivatives 2425 (Figure 6) (Table 5) and evaluated their anti-inflammatory activity in vitro. Adding quinoline structure did not increase BAK activity but considerably lowered its toxicity. Compound 24 had a moderate inhibitory effect on NO, but compound 25 had a strong inhibitory effect. Compounds 2425 slightly inhibited IL-6 (p < 0.05 vs. LPS group) but did not affect TNF-a.
Figure 6. The chemical structures of anti-inflammatory compounds 1825.
Figure 6. The chemical structures of anti-inflammatory compounds 1825.
Molecules 28 06478 g006
Table 5. Quinoline derivatives with anti-inflammatory activity.
Table 5. Quinoline derivatives with anti-inflammatory activity.
Compd.Activity TargetOrigin Ref
18iEI = 3.75IL-1β, IL-6synthetic [25]
19iEI = 3.18IL-1β, IL-6synthetic [25]
20iEI = 4.84IL-1β, IL-6synthetic [25]
21iEI = 2.11IL-1β, IL-6synthetic [25]
22iEI = 4.27IL-1β, IL-6synthetic [25]
23IL-6 stronger than that of glycyrrhetinic acid.IL-6, TNF-α, NO, iNOS, COX-2synthetic[26]
24moderate inhibitory effect on NOErythroid 2-related factor 2, Heme oxygenase-1synthetic[27]
25strong inhibitory effect on NOErythroid 2-related factor 2, Heme oxygenase-1synthetic[27]

3.6. Antithrombotic Activity

Isosteviol (CF: C20H30O3, MW: 318.4504, (4α,8β,13β)-13-Methyl-16-oxo-17-norkauran-18-oic acid) has the structural characteristics of tetracyclic diterpenoids. Chen and colleagues [28] synthesized an isosteviol–quinoline derivative 26 (Figure 7) (Table 6) and evaluated it in vitro FXa inhibition. The modification impact may not be optimum due to the Ki value of 26 on FXa being 4.177 μM, which is lower than the positive control for antithrombotic activity (Ki = 3.4 nM).
Figure 7. The chemical structures of antithrombotic compound 26.
Figure 7. The chemical structures of antithrombotic compound 26.
Molecules 28 06478 g007
Table 6. Quinoline derivatives with antithrombotic activity.
Table 6. Quinoline derivatives with antithrombotic activity.
Compd.Activity TargetOrigin Ref
26antithrombotic Ki = 3.4 nMFXasynthetic [28]

3.7. Anti-Parasitic Activity

Cinchonasine (CF: C20H24N2O2, MW: 324.417, (8S,9R)-6′-Methoxy-cinchona-9-ol sulfate dihydrate) is the main alkaloid in the bark of the cinchona tree and its congeners. The structure of cinchonasine includes a quinoline ring. Leverrier and his colleagues [29] evaluated the anti-parasitic activity of cinchona–alkaloid derivatives 27ac (Figure 8) (Table 7). According to the results, compounds 27ac had good anti-T. brucei activity (IC50s of compound 27ac are 0.37, 0.39, and 0.40 μg/mL, respectively), and good resistance to anti-L. mexicana activity (IC50s of compound 27ac are 3.86, 3.39, and 3.45 μg/mL, respectively).
Isatin (CF: C8H5NO2, MW: 147.13, 2,3-Indolinedione) is an orange-red single co-prism crystal. Nisha and his colleagues [30] synthesized the isatin–quinoline derivatives 28ac (Figure 8) (Table 7) and determined their anti-trichomonas activity. Compounds 28ac among them have effective anti-trichomonas action. At 50 μM, the growth inhibition of 28ac was 98%, 100%, and 100%, respectively. 28a and 28c have IC50s of 22.2 and 11.3 μM, respectively. 28a28c had no cytotoxicity to PC-3 cells.
Licorice chalcone A (CF: C21H22O4, MW: 338.4, ((2E)-3-[5-(1,1-dimethylprop-2-en-1-yl)-4-hydroxy-2-methoxyphenyl]-1-(4-hydroxyphenyl)prop-2-en-1-one)) is derived from the roots of Glycyrrhiza glabra and G. inflata. Coa and colleagues [31] synthesized quinoline–chalcone derivatives 29af and quinoline–chromone derivatives 30ac (Figure 8) (Table 7), and evaluated their activity against Leishmania (Viannia) panamensis. Compounds 29af and 30ac demonstrated activity against Leishmania (V) panamensis, while compounds 29be, and 30ab demonstrated activity against Trypanosoma cruzi with EC50 values less than 18 μg/mL. Compound 29f was the most active compound against Leishmania (V) panamensis and Trypanosoma cruzi, with EC50s of 6.11 ± 0.26 and 4.09 ± 0.24 μg/mL. All hybrid compounds outperformed the anti-leishmanial drug meglumine antimoniate. Compounds 29d and 30b outperformed benznidazole, the current anti-trypanosomal drug; however, these compounds were toxic to mammalian U-937 cells.
Figure 8. The chemical structures of anti-parasitic compounds 2730.
Figure 8. The chemical structures of anti-parasitic compounds 2730.
Molecules 28 06478 g008
Matrine (CF: C15H24N2O, MW: 248.37, (7aS,13aR,13bR,13cS)-dodecahydro-1H,5H,10H-dipyrido[2,1-f:3′,2′,1′-ij][1,6]naphthyridin-10-one) is an alkaloid extracted from the dried roots, plants, and fruits of the legume Sophora flavescens by ethanol and other organic solvents. Huang and colleagues [32] synthesized quinoline–matrine derivatives 31au (Figure 9) (Table 7) and evaluated its acaricidal and insecticidal activities. Interestingly, all quinoline–matrine derivatives (save compound 31c) outperformed their antecedents in terms of acaricidal activity. In particular, compounds 31g, 31m, and 31r showed the most promising acaricidal action (the MR of compounds 31g, 31m, and 31r at 72 h are 37.1%, 37.1%, and 36.1%, respectively). It is worth noting that the introduction of chlorine atoms at the C-21 position of compound 31aj is important for the acaricidal activity (the MR of compounds 31d (including 21-CH3), 31f (including 21-F), 31h (including 21-Br), 31i (including 21-NH2), and 31j (including 21-OH) at 72 h are 29.5%, 29.6%, 26.6%, 26.0%, and 26.9%, respectively; the MR of compound 31g (containing 21-Cl) at 72 h was 37.1%). The insertion of chlorine atoms on the benzoyl oxy group of compound 31k is required for acaricidal activity in compounds 31ko. Compound 31r (having 3-Br on the benzoyl amino group) has a higher acaricidal efficacy than compounds 31pt (31p, 31q, and 31s containing chlorine atoms on the benzoyl amino group). The findings suggested that these compounds may have antithrombotic hormone-like properties. All quinoline–matrine derivatives (31au) were more effective against insects than their antecedents. Compounds 31dg, 31i, 31p, and 31s showed the most effective activity (the FMR of compounds 31dg, 31i, 31p, and 31s are 65.5%, 62.1%, 65.5%, 69.0%, 62.1%, 65.5%, and 65.5%, respectively). The location of the methyl group is critical for insecticidal efficacy in compounds 31bd. Compounds 31b (containing 19-CH3) and 31d (containing 21-CH3) have FMRs of 58.6% and 65.5%, respectively; compound 31c (containing 20-CH3) has an FMR of 44.8%. For compounds 31aj, the quinoline segment of 31a is the modified position, and the introduction of an appropriate group on the quinoline segment of 31a can lead to more effective derivatives, such as compounds 31d (containing 21-CH3; FMR: 65.5%), 31e (containing 19-OCH3; FMR: 62.1%), 31f (containing 21-F; FMR: 65.5%), 31g (containing 21-Cl; FMR: 69.0%), and 31i (containing 21-NH2; FMR: 62.1%). Although the derivative 31ko was created by inserting various benzoyl groups into compound 31j, it did not have the same insecticidal action as compound 31a. When R2 is a 3- or 4-chlorine atom, the related compounds 31p and 31s have higher insecticidal action than 31a. Compound 31u (21-NHCH3CO groups; 72 h FMR: 33.2%) showed more activity than compound 31i (21-NH2 groups; 72 h FMR: 26.0%).
Wang and colleagues [33] synthesized 4-hydroxy-11-oxo-11H-chromoene [2,3-g] quinoline-3-carboxylic acid ethyl ester 32ad (Figure 9) (Table 7) and tested their anti-coccidian activity. Compounds 32ad demonstrated anticoccidial activity against Eimeria tenella with ACIs of 147, 123, 133, and 148, respectively.
Figure 9. The chemical structures of anti-parasitic compounds 3132.
Figure 9. The chemical structures of anti-parasitic compounds 3132.
Molecules 28 06478 g009
Roussaki and colleagues [34] synthesized quinolinone–chalcone derivatives 3336 (Figure 10) (Table 7) and evaluated their biological activities against mammalian T. brucei and Leishmania infantis. Among them, the most effective is compound 33b (IC50 = 2.6 ± 0.1 μM), followed by compounds 33g, 33c, 33f, and 33d (decreasing potency), all of which contain electron-donating substituents on the B ring of the chalcone group. The data analysis shows that the position and number of these groups contribute to the anti-parasitic properties of the compounds. For the most effective trypanosome agent, compound 33b, the electron-donating methyl substituent is located at the 2 position of the B ring, and its isomer, compound 33a, contains a methyl group at the 4 position, which does not affect the growth of trypanosome at a concentration of up to 10 μM. Similarly, compound 33d containing a single methoxy group at the 3 position of the B ring has lethality (IC50 = 6.5 ± 0.1 μM). In contrast, the isomer chalcone containing the 4-methoxy group does not affect the growth of the parasite. The presence of two electron-abandoned substituents resulted in a slight increase in trypanosomal activity as follows: Compound 33f contains two methoxy groups at the 3 and 4 positions of the B ring, with an IC50 value of 4.9 ± 0.2 μM, while compound 33c has an IC50 value of 4.9 ± 0.1 μM, and its 3 and 4 positions contain a methoxy group and a hydroxyl group, respectively. The remaining insecticide chalcone, compound 33g, has a (di-tert-butyl) phenol substitute. Although it is the second most effective anti-trypanosomal structure in the chalcone series (IC50 = 3.3 ± 0.1 μM), it shows high cytotoxicity (IC50 ≈ 26 μM). For compounds 33b and 33f, analogues containing alkyl substituents on the amide nitrogen of the heterocyclic ring of the quinolinone molecule (compounds 35ad) were synthesized. N-ethyl analogues 35a and 35b did not show growth inhibition against Brucella in the blood. Compounds 35c and 35d, N-benzyl analogues of chalcones, were less active against Brucella than non-alkyl compounds, although higher than N-ethyl analogues. These results indicate that the hydrogen of heterocyclic amide groups is important in the mechanism of these compounds. Similarly, by adding electron-withdrawing groups (-COOH, -CF3, and -NO2) (compounds 33h, 33i, and 33j) to the B ring of chalcone or extending the conjugated system between quinolinone and chalcone patterns (compounds 34a and 34b), the quinolinone–chalcone structure was changed in other parts. There was no effect on the growth of trypanosomes at a concentration of 10 μM. The a,b-unsaturated carbonyl system was modified by synthesizing the pyrazoline analogues 36a and 36b, significantly increasing anti-parasitic activity against B. haemolytic. The IC50s of these two compounds were lower than that of the reference drug nifurtimox, so they were the most active against B. haemolytic among all the tested compounds in this work. Pyrazoline 36a (IC50 = 1.46 ± 0.1 μM) can be considered a promising anti-parasitic compound because it has no cytotoxicity to THP1 cells. When a series of quinolinone–chalcone hybrids were screened against the astigmatic stage in larval cells, structures (compounds 3334, 35ab) were shown to affect the growth of parasites. The most potent compounds were 33g, 33a, and 33h, with IC50s of 1.3 ± 0.1, 2.1 ± 0.6, and 3.1 ± 1.0 μM, respectively. Interestingly, the structure–activity relationship against L. infantum observed using compound series significantly differs from that against T. brucei, and many differences are directly opposite. For example, the shift of the methyl substituent from the 4 position (compound 33a) to the 2 position (compound 33b) or the methoxy group from the 4 position (compound 33e) to the 3 position (compound 33d) results in a significant decrease in insecticidal activity, which is different from the case of Brucella virus. In addition, the electronic properties of the substituents do not seem to play an important role in anti-Lishmaniasis activity as they do for Brucellosis as follows: Compound 33h with electron-withdrawing group trifluoromethyl (methyl isomer) at the 4 position of the B ring is the third most effective agent for infantile disease in this series, and derivatives containing-NO2 and-COOH (compounds 33i and 33j) also affect infantile disease amastigotes. Why these conflicting SAR differences occur is unclear. This may reflect the following different locations of these parasites in mammalian hosts: Brucella is an extracellular pathogen found in the blood of the host, and larvae can invade and grow in the host’s macrophages, residing in an acidic compartment. In addition, this may only be due to the concentration range used in the screening as follows: many leishmaniasis compounds have IC50s > 10 μM, which is the highest level used to combat Brucella. The N-alkylation of heterocyclic amide groups led to compounds with lower or slightly better anti-Lishmaniasis activity than non-alkylated analogues, indicating that the N-H group may be the key to anti-Lishmaniasis activity, which is also true in anti-filarial activity. Compared with chalcone analogue 9, pyrazoline 36a has lower activity against infantile lymphoma, while pyrazoline 36b, an analogue of chalcone 5, is the most active anti-Lishman disease agent among all tested compounds, with an IC50 value of 0.71 μM. Notably, pyrazoline 36b showed the best anti-parasitic activity against both parasites. In order to evaluate their effects on mammalian cells, cytotoxicity tests were performed on differentiated THP-1 macrophages with all compounds. Among the most effective agents (IC50 < 10 μM), compounds 33a, 33b, 33d, 33f, 33h, and 36a had no growth inhibitory effect on THP-1 cells at concentrations of 30 μM (33b, 33d, and 33f) or 50 μM (33a, 33h, and 36a), while the IC50s of compounds 33c and 33g were about 20 μM. Due to problems related to the solubility of compounds in DMSO and other common solvents, the exact IC50 cytotoxicity data cannot be determined. A comparison of efficacy against parasites and mammalian strains showed that pyrazoline 36a and chalcone 5 were the most effective trypanosome preparations, while chalcone analogue 36a was an interesting guide structure for L. infantum. In summary, compounds 36a and 36b have obvious anti-parasitic activity against T. brucei blood in vitro. Although pyrazoline 36b showed the best activity against both parasites and was the most effective compound among all the parasites tested in this work, its high cytotoxicity to mammalian cells prohibits further consideration as a lead compound. In contrast, pyrazoline 36a should be considered a drug to induce trypanosomiasis because of its high anti-parasitic activity and no cytotoxicity.
Pan and colleagues [35] synthesized coumarin derivatives 3738 (Figure 10) (Table 7) and tested the nematicidal activity of these derivatives. Compounds 37 and 38 showed significant broad spectrum nematicidal activity. (The LC50s for M. incognita, Ditylenchus destructor, Bursaphelenchus mucronatus, B. xylophilus, and Aphelenchoides besseyi of compound 37 were are 64.0, 52.9, 97.9, 103.2, and 95.2 μmol/L, respectively. The LC50s for M. incognita, Ditylenchus destructor, Bursaphelenchus mucronatus, B. xylophilus, and Aphelenchoides besseyi of compound 38 were 42.4, 68.0, 77.8, 145.5, and 120.7 μmol/L, respectively).
Fraxinellone (CF: C14H16O3, MW: 232.275, 7-dimethyl-3a,4,5,6-tetrahydro-2-benzofuran-1(3H)-one) is isolated from the root bark of the Brassica plant Dictamnus dasycarpus. Guo and colleagues [36] synthesized fraxinellone–quinoline derivative 39 (Figure 10) (Table 7) and tested the in vivo insecticidal activity of isolated Mycobacterium pre-3 instar larvae. Compound 39 showed more promising insecticidal activity than positive contrast (the M. separata on leaves mortality rate for 10 days, 25 days, and 35 days are 20.7%, 48.1%, and 63.0%, respectively).
Guo and colleagues [37] synthesized usnea acid quinoline derivative 40 (Figure 10) (Table 7) and tested its anti-T. gondii activity. Compound 40 had lower cytotoxicity and a higher selectivity index, indicating that its activity was superior to that of the lead drug and positive control drugs. The results of tachyzoite content assessment in mice abdomen showed that the compound 40 inhibition rate of abdominal tachyzoites in mice reached 55.2% (p < 0.01), 58.3% (p < 0.01), and 64.6% (p < 0.001), respectively, the numbers of tachyzoites were significantly reduced, respectively. At the same concentration, (+)-usnic acid and 40 inhibited tachyzoites more effectively than the positive control; moreover, compound 40 has better anti-T. The activity of Toxoplasma gondii is higher than the natural product (+)-usnic acid. The toxicity of compound 40 was further investigated by measuring the ALT and AST levels in the serum of KM mice. Compared with the normal group, the serum ALT level of the Toxoplasma-infected mice was significantly increased (p < 0.05). The serum ALT levels were significantly lower (p < 0.01) in the compound 40-treated group than in the (+)-usnic acid-treated group.
Dihydroartemisinin (CF: C15H24O5, MW: 284.35, (4S,5R,8S,9R,10S,12R,13R)-1,5,9-trimethyl-11,14,15,16-tetraoxatetracyclo hexadecan-10-ol) is an artemisinin derivative that has a powerful and rapid killing impact on the red internal stage of Plasmodium and can reduce clinical attacks and symptoms swiftly. Deng and colleagues [38] synthesized dihydroartemisinin quinoline derivatives 4142 (Figure 10) (Table 7) and evaluated their anti-Toxoplasma activity. Compounds 41 and 42ac were more effective against Toxoplasma than dihydroartemisinin (The selectivity index of compounds 41a and 42ac was 0.84, 1.02, 0.43, and 1.02).
Figure 10. The chemical structures of anti-parasitic compounds 3342.
Figure 10. The chemical structures of anti-parasitic compounds 3342.
Molecules 28 06478 g010
Table 7. Quinoline derivatives with anti-parasitic activity.
Table 7. Quinoline derivatives with anti-parasitic activity.
Compd.Activity TargetOrigin Ref
27aanti-T. brucei
IC50 = 0.37 μg/mL
anti-L. Mexicana
IC50 = 3.86 μg/mL
-synthetic [29]
27banti-T. brucei
IC50 = 0.39 μg/mL
anti-L. Mexicana
IC50 = 3.39 μg/mL
-synthetic [29]
27canti-T. brucei
IC50 = 0.40 μg/mL
anti-L. Mexicana
IC50 = 3.45 μg/mL
-synthetic [29]
28aanti-trichomonas
IC50 = 22.2 μM
-synthetic [30]
28b--synthetic [30]
28canti-trichomonas
IC50 = 11.3 μM
-synthetic[30]
29aLeishmanicidal
EC50 = 11.79 μg/mL
Trypanocidal
EC50 = 35.08 μg/mL
-synthetic[30]
29bLeishmanicidal
EC50 = 6.24 μg/mL
Trypanocidal
EC50 = 17.62 μg/mL
-synthetic[31]
29cLeishmanicidal
EC50 = 12.37 μg/mL
Trypanocidal
EC50 = 15.79 μg/mL
-synthetic[31]
29dLeishmanicidal
EC50 = 8.53 μg/mL
Trypanocidal
EC50 = 37.61 μg/mL
-synthetic[31]
29eLeishmanicidal
EC50 = 16.41 μg/mL
Trypanocidal
EC50 = 15.12 μg/mL
-synthetic[31]
29fLeishmanicidal
EC50 = 22.0 μg/mL
Trypanocidal
EC50 = 54.95 μg/mL
-synthetic[31]
30aLeishmanicidal
EC50 = 6.11 μg/mL
Trypanocidal
EC50 = 4.09 μg/mL
-synthetic[31]
30bLeishmanicidal
EC50 = 16.18 μg/mL
Trypanocidal
EC50 > 20 μg/mL
-synthetic[31]
30cLeishmanicidal
EC50 = 2.36 μg/mL
Trypanocidal
EC50 > 2 μg/mL
-synthetic[31]
31aCorrected mortality rate
48 h FMR 8.7%
72 h FMR 32.7%
-synthetic[32]
31bCorrected mortality rate
48 h FMR 7.8%
72 h FMR 28.0%
-synthetic[32]
31cCorrected mortality rate
48 h FMR 2.8%
72 h FMR 19.2%
-synthetic[32]
31dCorrected mortality rate
48 h FMR 7.5%
72 h FMR 29.5%
-synthetic[32]
31eCorrected mortality rate
48 h FMR 6.9%
72 h FMR 24.4%
-synthetic[32]
31fCorrected mortality rate
48 h FMR 6.9%
72 h FMR 29.6%
-synthetic[32]
31gCorrected mortality rate
48 h FMR 7.7%
72 h FMR 37.1%
-synthetic[32]
31hCorrected mortality rate
48 h FMR 6.3%
72 h FMR 26.6%
-synthetic[32]
31iCorrected mortality rate
48 h FMR 5.0%
72 h FMR 26.0%
-synthetic[32]
31jCorrected mortality rate
48 h FMR 6.5%
72 h FMR 26.9%
-synthetic[32]
31kCorrected mortality rate
48 h FMR 3.8%
72 h FMR 26.5%
-synthetic[32]
31lCorrected mortality rate
48 h FMR 2.9%
72 h FMR 20.6%
-synthetic[32]
31mCorrected mortality rate
48 h FMR 15.5%
72 h FMR 37.1%
-synthetic[32]
31nCorrected mortality rate
48 h FMR 7.2%
72 h FMR 30.7%
-synthetic[32]
31oCorrected mortality rate
48 h FMR 5.9%
72 h FMR 28.7%
-synthetic[32]
31pCorrected mortality rate
48 h FMR 9.6%
72 h FMR 30.9%
-synthetic[32]
31qCorrected mortality rate
48 h FMR 3.2%
72 h FMR 22.5%
-synthetic[32]
31rCorrected mortality rate
48 h FMR 9.5%
72 h FMR 36.1%
-synthetic[32]
31sCorrected mortality rate
48 h FMR 6.4%
72 h FMR 26.7%
-synthetic[32]
31tCorrected mortality rate
48 h FMR 4.8%
72 h FMR 26.6%
-synthetic[32]
31uCorrected mortality rate
48 h FMR 8.6%
72 h FMR 33.2%
-synthetic[32]
32aACI = 147-synthetic[33]
32bACI = 123-synthetic[33]
32cACI = 133-synthetic[33]
32dACI = 148-synthetic[33]
33aT. brucei
IC50 > 10 μM
L. infantum
IC50 = 2.1 μM
FRDsynthetic[34]
33bT. brucei
IC50 = 2.6 μM
L. infantum
IC50 > 50 μM
FRDsynthetic[34]
33cT. brucei
IC50 = 4.9 μM
L. infantum
IC50 = 11.5 μM
FRDsynthetic[34]
33dT. brucei
IC50 = 6.5 μM
L. infantum
IC50 = 28.4 μM
FRDsynthetic[34]
33eT. brucei
IC50 > 10 μM
L. infantum
IC50 = 12.7 μM
FRDsynthetic[34]
33fT. brucei
IC50 = 4.9 μM
L. infantum
IC50 = 7.5 μM
FRDsynthetic[34]
33gT. brucei
IC50 = 3.3 μM
L. infantum
IC50 = 1.3 μM
FRDsynthetic[34]
33hT. brucei
IC50 > 10 μM
L. infantum
IC50 = 3.1 μM
FRDsynthetic[34]
33iT. brucei
IC50 > 10 μM
L. infantum
IC50 = 26.9 μM
FRDsynthetic[34]
33jT. brucei
IC50 > 10 μM
L. infantum
IC50 = 18.4 μM
FRDsynthetic[34]
34aT. brucei
IC50 > 10 μM
L. infantum
IC50 > 50 μM
FRDsynthetic[34]
34bT. brucei
IC50 > 10 μM
L. infantum
IC50 = 20.0 μM
FRDsynthetic[34]
35aT. brucei
IC50 > 10 μM
L. infantum
IC50 = 24.8 μM
FRDsynthetic[34]
35bT. brucei
IC50 > 10 μM
L. infantum
IC50 > 50 μM
FRDsynthetic[34]
35cT. brucei
IC50 = 6.17 μM
L. infantum
IC50 > 25 μM
FRDsynthetic[34]
35dT. brucei
IC50 = 5.68 μM
L. infantum
IC50 > 25 μM
FRDsynthetic[34]
36aT. brucei
IC50 = 1.46 μM
L. infantum
IC50 = 13.46 μM
FRDsynthetic[34]
36bT. brucei
IC50 = 1.43 μM
L. infantum
IC50 = 0.71 μM
FRDsynthetic[34]
37M. incognita
LC50 = 64.0 μmol/L
Ditylenchus destructor
LC50 = 52.9 μmol/L
Bursaphelenchus mucronatus
LC50 = 97.9 μmol/L
B. xylophilus
LC50 = 103.2 μmol/L
Aphelenchoides besseyi
LC50 = 95.2 μmol/L
-synthetic[35]
38M. incognita
LC50 = 42.4 μmol/L
Ditylenchus destructor
LC50 = 68.0 μmol/L
Bursaphelenchus mucronatus
LC50 = 77.8 μmol/L
B. xylophilus
LC50 = 145.5 μmol/L
Aphelenchoides besseyi
LC50 = 120.7 μmol/L
-synthetic[35]
39M. separata on leaves mortality rate
10 days 20.7%
25 days 48.1%
35 days 63.0%
-synthetic[36]
40inhibition rate of abdominal tachyzoites in mice
55.2% (p < 0.1), 58.3% (p < 0.01)
64.6% (p < 0.001)
-synthetic[37]
41selectivity index 0.84TgCDPK1synthetic[38]
42aselectivity index 1.02TgCDPK1synthetic[38]
42bselectivity index 0.43TgCDPK1synthetic[38]
42cselectivity index 1.02TgCDPK1synthetic[38]
Summary: The anti-T.brucei activity of Cinchonasine derivative 27a was the strongest, and the IC50 value was 0.37 μg/mL. The anti-L.mexicana activity of derivative 27b was the strongest, and the IC50 value was 3.39 μg/mL.
The EC50 values of compound 29f against Leishmania (V) panamensis and Trypanosoma cruzi were 6.11 and 4.09 μg/mL, respectively.
The quinolinone–chalcone derivatives 36a and 36b showed the strongest inhibitory effect on T.brucei, with IC50 values of 1.46 and 1.43 μM. 36a has no obvious cytotoxicity to THP1 cells and is an anti-parasitic compound with development value. Compounds 42a and 42c were more effective against Toxoplasma than dihydroartemisinin (The selectivity index of compounds 42a was 1.02). The anti-parasitic activity of natural products was enhanced by adding quinoline to natural products; however, the mechanism of action of these compounds has not been studied, so these compounds need further study.

3.8. Antimalarial Activity

Artemisinin (CF: C15H22O5, MW: 282.34, (3R,5aS,6R,8aS,9R,10S,12R,12aR)-Decahydro-3,6,9-trimethyl-3,12-oxo-12H-pyrano[4,3-j]-1,2-Benzodixepin-10-one) is mostly derived by direct extraction from Artemisia annua or by extracting artemisinic acid from Artemisia annua and then semi-synthetically obtaining it. Lombard and colleagues [39] synthesized two quinoline–artemisinin derivatives 43 and 44 (Figure 11) (Table 8) and evaluated their antimalarial activity. The in vivo anti-plasma parasite activity of hybrid dimers 43 and 44 was stronger than the positive control, with in vitro IC50s of 8.7 and 29.5 μM against the 3D7 strain, respectively.
Raj and colleagues [40] synthesized a series of piperazine-coupled 7-chloroquinoline–isatin derivatives 45ae (Figure 11) (Table 8), and their antimalarial and anti-chloroquine activities against Plasmodium falciparum were evaluated. However, compounds 45ae have shown strong antimalarial activity with IC50s of 1.12, 1.17, 0.27, 0.81, and 0.36 μM, respectively. Unfortunately, all of the compounds had lower activities than the positive control.
Raj and colleagues [41] performed the synthesis and antimalarial activity of a 1H-1,2,3-triazole-tethered 7-chloroquinoline–isatin derivatives 46aj (Figure 11) (Table 8). Compounds 46aj have longer alkyl chains between the 7-chloroquinoline and isatin groups and have better anti-plasma action. Unfortunately, the IC50s of the tested compounds 46aj were all greater than 1 μM, which was less effective than the positive control or artemisinin.
Nisha and colleagues [42] synthesized a series of β-amino alcohol-linked 4-aminoquinoline–indigo derivatives 47ac (Figure 11) (Table 8) and evaluated their activity against CQ-resistant P. falciparum W2. The most active compounds were 47bc, with IC50s of 11.8 and 13.5 μM, respectively.
Videnović and colleagues [43] synthesized 4-amino-7-chloroquinoline (4,7-ACQ) bile salt derivative 48 (Figure 11) (Table 8) and determined its antimalarial activity. Compound 48 demonstrated the highest antimalarial activity, with a 95% inhibition rate and a Ki of 0.103 μM.
Figure 11. The chemical structures of antimalarial and anticancer compounds 4348.
Figure 11. The chemical structures of antimalarial and anticancer compounds 4348.
Molecules 28 06478 g011
Leverrier and colleagues [44] synthesized cinchona–alkaloid derivatives 4950 (Figure 12) (Table 8) and determined their in vitro activity against Trypanosoma brucei. Compounds 49ab and 50ab emerged as the most promising anti-Trypanosoma brucei due to their low cytotoxicity and IC50s of 1.47, 0.64, 0.69, and 0.61 μM, respectively.
1,2,3,4-Tetrahydro-β-carbolines (THβCs) (CF: C11H12N2, MW: 172.226, 1,2,3,4-tetrahydro-9h-pyrido[3,4-b]indole) represent a class of privileged structural motifs found in many pharmacologically active natural compounds possessing potential anticancer and antimalarial activities. Sharma and colleagues [45] synthesized 1H-1,2,3-triazole/hydrazole-integrated tetrahydro-β-carboline-4-aminoquinoline compounds 51af, 52ah, and 53ab (Figure 12) (Table 8) and evaluated their activity against the chloroquine-resistant (CQ) strain of Plasmodium falciparum W2. Although all compounds were not as active as the positive control medication, including the quinoline nucleus considerably increased the antimalarial efficacy of the THC nucleus. The activity of the synthesized conjugates depends on the nature of the substitution at the C-1 position of the THC nucleus, the type of function introduced as a linker, and the length of the alkyl chain, according to the structure–activity relationship (SAR). An analysis of the SAR of 1H-1,2,3-triazole tethered THC-4-aminoquinoline conjugates revealed an increase in the antimalarial activity with the introduction of flexible alkyl chains on the aminoquinoline core, as evidenced by the scaffolds 51cf being more active than compounds 51ab (The IC50s of compounds 51ab are 4.02 μM, 9.28 μM). The antimalarial activity of the aliphatic hydrazide-linked conjugates 52ah increased with increasing alkyl chain length, although the type of the substituent at the C-1 position of THC did not appear to alter the activity profile. Interestingly, replacing the alkyl chain with an aryl core enhanced antimalarial activity, as seen by compound 53a, which has an IC50 of 0.61 μM. The antimalarial activity of the -carboline and quinolinyl precursors used in this synthesis was also investigated. Compounds 53ab (acyl hydrazide precursors) and 51c, 51e (1H-12,3-triazole-like precursors) were tested on mammalian Vero cells to determine if the observed activity was due to inherent antimalarial activity or cytotoxicity. The 1H-1,2,3-triazole-linked conjugates 51c and 51e were clearly non-cytotoxic, with SI > 300, whereas the acyl hydrazide-linked compounds 53ab were somewhat cytotoxic to mammalian Vero cells. The most potent and non-cytotoxic compound 51e exhibited the best properties, including a C-1 unsubstituted THC core, a 1H-1,2,3-triazole core, and propyl as a flexible linker, with an IC50 of 0.50 μM and a selectivity index of 495.76.
Vinindwa and colleagues [46] synthesized chalcone–quinoline derivatives 54ao (Figure 12) (Table 8) and evaluated their antimalarial activity. With IC50s ranging from 0.10 to 4.45 μM, all compounds showed potent activity against the Plasmodium falciparum NF54 sensitive strain. The fluorine-substituted molecular derivatives 54d, 54h, and 54n displayed higher activity than the unsubstituted molecular derivative 54a (IC50 = 1.67 μM), suggesting the relevance of the electronic effect imparted by the more electronegative fluorine atoms. Other halogens with similar trends included bromine 54c (IC50 = 0.10 μM), chlorine 54e (IC50 = 0.10 μM), and methoxy 54f (IC50 = 0.11 μM). The most active compounds were 54c, 54e, and 54f, with IC50s of 0.10, 0.10, and 0.11 μM, respectively. The inhibitory properties of the compounds against the multidrug-resistant K1 strain of Plasmodium falciparum were investigated further. Compound 54f’s resistivity index RI = 5.36 (IC50 = 0.59 μM) was roughly double that of the positive control, which was the leading compound in this study. Compounds 54c and 54e, on the other hand, displayed poor activity against the K1 strain, with IC50s of 2.97 and 6 μM, respectively. Compounds with three methylene (n = 3) as linkers had higher efficacy against Plasmodium falciparum than compounds with two methylene (n = 2). Compared to compound 54i (n = 2), the addition of an extra CH2 group (n = 3) in compound 54f enhanced its activity by nearly five times, while compound 54d (n = 2 IC50 = 0.28 μM) increased by two times. Furthermore, practically all molecular derivatives 54bf and 54gj with three methylene linkers are the most active in the series, with IC50s ranging from 0.10 to 0.86 μM. Compounds 54b and 54l, which have methyl groups at the para position and have the same activity (The IC50s were 0.37 and 0.39 μM, respectively), show no effect on activity with smaller polarity. Compounds 54k and 54m have poor activity and contain 2-furanyl and ferrocenyl aromatic rings, demonstrating the relevance of chalcone units in the overall activity of the molecule. Most compounds were insoluble except for compounds 54b, 54i, 54j, 54l, and 54o, with IC50 values less than 5 μM.
Flavonoids are secondary plant metabolites that are frequently found in nature. Flavonoids are a yellow pigment formed from a core of flavonoids (2-phenylchromones), which includes flavonoid isomers and their hydrogenation and reduction products, such as C6-C3-C6. Rodrigues and colleagues [47] synthesized the flavonoid quinoline derivative 55 (Figure 12) (Table 8) and determined its antimalarial activity. Compound 55 was tested in vitro for antimalarial activity against the chloroquine-resistant Plasmodium falciparum W2 strain. Compound 55 had moderate antimalarial activity, with an IC50 of 5.17 μM.
Figure 12. The chemical structures of antimalarial compounds 4955.
Figure 12. The chemical structures of antimalarial compounds 4955.
Molecules 28 06478 g012
Table 8. Quinoline derivatives with antimalarial activity.
Table 8. Quinoline derivatives with antimalarial activity.
Compd.Activity Origin Ref
43Anti-plasma parasite
IC50 = 8.7 μM
synthetic[39]
44Anti-plasma parasite
IC50 = 29.5 μM
synthetic[39]
45aAntimalarial activity
IC50 = 1.12 μM
synthetic[40]
45bAntimalarial activity
IC50 = 1.17 μM
synthetic[40]
45cAntimalarial activity
IC50 = 0.27 μM
synthetic[40]
45dAntimalarial activity
IC50 = 0.81 μM
synthetic[40]
45eAntimalarial activity
IC50 = 0.36 μM
synthetic[40]
46aAntimalarial activity
IC50 > 5 μM
synthetic[41]
46bAntimalarial activity
IC50 > 5 μM
synthetic[41]
46cAntimalarial activity
IC50 > 5 μM
synthetic[41]
46dAntimalarial activity
IC50 > 5 μM
synthetic[41]
46eAntimalarial activity
IC50 > 5 μM
synthetic[41]
46fAntimalarial activity
IC50 = 3.07 μM
synthetic[41]
46gAntimalarial activity
IC50 = 2.30 μM
synthetic[41]
46hAntimalarial activity
IC50 = 1.37 μM
synthetic[41]
46iAntimalarial activity
IC50 = 1.73 μM
synthetic[41]
46jAntimalarial activity
IC50 = 1.63 μM
synthetic[41]
47aAntimalarial activity
IC50 = 24.9 nM
synthetic[42]
47bAntimalarial activity
IC50 = 11.8 nM
synthetic[42]
47cAntimalarial activity
IC50 = 13.5 nM
synthetic[42]
48Antimalarial activity
Ki = 0.103 μM
synthetic[43]
49aAnti-Trypanosoma brucei
IC50 = 1.47 μM
synthetic[44]
49bAnti-Trypanosoma brucei
IC50 = 0.64 μM
synthetic[44]
50aAnti-Trypanosoma brucei
IC50 = 0.69 μM
synthetic[44]
50bAnti-Trypanosoma brucei
IC50 = 0.61 μM
synthetic[44]
51aAntimalarial activity
IC50 = 9.28 μM
synthetic[45]
51bAntimalarial activity
IC50 = 4.02 μM
synthetic[45]
51cAntimalarial activity
IC50 = 0.86 μM
synthetic[45]
51dAntimalarial activity
IC50 = 0.93 μM
synthetic[45]
51eAntimalarial activity
IC50 = 0.49 μM
synthetic[45]
51fAntimalarial activity
IC50 = 1.37 μM
synthetic[45]
52aAntimalarial activity
IC50 = 4.4 μM
synthetic[45]
52bAntimalarial activity
IC50 = 1.73 μM
synthetic[45]
52cAntimalarial activity
IC50 = 5.0 μM
synthetic[45]
52dAntimalarial activity
IC50 = 3.1 μM
synthetic[45]
52eAntimalarial activity
IC50 = 2.0 μM
synthetic[45]
52fAntimalarial activity
IC50 = 3.1 μM
synthetic[45]
52gAntimalarial activity
IC50 = 3.4 μM
synthetic[45]
52hAntimalarial activity
IC50 = 2.2 μM
synthetic[45]
53aAntimalarial activity
IC50 = 1.8 μM
synthetic[45]
53bAntimalarial activity
IC50 = 0.61 μM
synthetic[45]
54aAntimalarial activity
IC50 = 0.45 μM
synthetic[46]
54bAntimalarial activity
IC50 = 0.37 μM
synthetic[46]
54cAntimalarial activity
IC50 = 0.10 μM
synthetic[46]
54dAntimalarial activity
IC50 = 0.28 μM
synthetic[46]
54eAntimalarial activity
IC50 = 0.10 μM
synthetic[46]
54fAntimalarial activity
IC50 = 0.11 μM
synthetic[46]
54gAntimalarial activity
IC50 = 0.32 μM
synthetic[46]
54hAntimalarial activity
IC50 = 0.86 μM
synthetic[46]
54iAntimalarial activity
IC50 = 0.49 μM
synthetic[46]
54jAntimalarial activity
IC50 = 0.50 μM
synthetic[46]
54kAntimalarial activity
IC50 = 4.45 μM
synthetic[46]
54lAntimalarial activity
IC50 = 0.39 μM
synthetic[46]
54mAntimalarial activity
IC50 = 1.53 μM
synthetic[46]
54nAntimalarial activity
IC50 = 0.57 μM
synthetic[46]
54oAntimalarial activity
IC50 = 0.69 μM
synthetic[46]
55Antimalarial activity
IC50 = 5.17 μM
synthetic[47]
Summary: Quinoline has always been a key scaffold for antimalarial research. Many antimalarial drugs, such as chloroquine and primaquine, contain a quinoline structure. With the increasing resistance of Plasmodium falciparum, there is an urgent need to develop new strategies and new antimalarial compounds to overcome the growing resistance. These include drug combination, drug reuse, and the use of chemical sensitizers (resistance reversal agents) and the development of new analogues, both of which involve the synthesis of new quinoline analogues.
7-chloroquinoline–isatin derivatives 45c showed strong antimalarial activity with IC50 of 0.27 μM. Compounds 54c and 54e also showed significant antimalarial activity with IC50 of 0.10 μM.
The introduction of quinoline by isatin and chalcone can significantly increase antimalarial activity. Unfortunately, the antimalarial mechanism of these compounds has not been studied.

3.9. Antibacterial Activity

Paul and colleagues [48] synthesized a sulfur-linked quinoline derivative 5657 (Figure 13) (Table 9) and preliminarily evaluated its in vitro antibacterial activity against Gram-positive Staphylococcus aureus (ATCC 11632) and Gram-negative Escherichia coli (ATCC 25922), and antifungal activity against Candida albicans (ATCC 90028). The results showed that most compounds had moderate to good antibacterial and antifungal activities (6.25–50.0 μg/mL). Compounds 56bh and 57ab have moderate to good activity, and the inhibition zone is equivalent to that of the positive control against Escherichia coli. Compared with the positive control, compounds 56a, 56c, 56d, 56e, 56f, 56g, 56h, and 57a showed moderate activity against S.aureus. In the antifungal activity evaluation, compared with the positive control, compounds 56a, 56c, 56e, 56f, 56g, 56h, 57a, and 57b showed moderate activity against Candida albicans. Nitro, brominated, or chlorinated compounds have comparable activity to standard drugs.
Patel and colleagues [49] synthesized a series of novel 7-hydroxy-9- (furo [2,3-b] quinoline-2-yl) 6H-benzo [c] coumarin derivatives 58al (Figure 13) (Table 9) against two Gram-positive bacteria Staphylococcus aureus (MTCC 96) and Bacillus subtilis (MTCC 441) and two Gram-negative bacteria Escherichia coli (MTCC 443) and Salmonella (MTCC 98) in vitro antibacterial activity. The antifungal activities of Candida albicans (MTCC 227) and Aspergillus niger (MTCC 282) were also evaluated in vitro. Ampicillin, chloramphenicol, and norfloxacin were used as standard antimicrobial agents. Glibenclamide and nystatin were used as standard antifungal agents. Compared with standard antimicrobial agents, all compounds are active against Gram-negative bacteria and fungi. Upon evaluating the antimicrobial activity data, it was observed that compounds 58c, 58h, and 58k (MIC = 200 μg/mL) showed good activity compared to ampicillin (MIC = 250 μg/mL) against Gram-positive bacteria B. subtilis. The compounds 58b, 58d, 58e, 58f, 58g, and 58l (MIC = 250 μg/mL) exerted equipotent activity against Gram-positive bacteria B. subtilis. against S. aureus, Compounds 58j and 58l (MIC = 100 μg/mL) and 58d, 58e, and 58k (MIC = 125 μg/mL) exhibited moderate activity compared to ampicillin (MIC = 250 μg/mL) against Gram-positive bacteria S. aureus. Compounds 58b and 58g (MIC = 200 μg/mL) showed better activity compared to ampicillin (MIC = 250 μg/mL) against Gram-positive bacteria S. aureus. Compounds 58a, 58c, 58h, and 58i (MIC = 250 μg/mL) were found equipotent to ampicillin (MIC = 250 μg/mL) against Gram-positive bacteria S. aureus. Compounds 58c, 58d, 58f, and 58j (MIC = 62.5 μg/mL) exhibited outstanding activity compared to ampicillin (MIC = 100 μg/mL) against Gram-negative bacteria E. coli and S. typhi, respectively. The compounds 58a, 58f, 58g, 58h, and 58k (MIC = 100 μg/mL) and compounds 58a, 58c, 58d, and 58j (MIC = 100 μg/mL) were found equipotent compared to ampicillin (MIC = 100 μg/mL) against E. coli and S. typhi, respectively. Compound 58i and 58g (MIC = 200 μg/mL) and compounds 58c, 58f, and 58h (MIC = 250 μg/mL) were found to be more active against C. albicans compared to griseofulvin (MIC = 500 μg/mL). Compounds 58a and 58b (MIC = 500 μg/mL) were found equipotent to griseofulvin (MIC = 500 μg/mL) against C. albicans. It is perceived from the antimicrobial data that almost all the tested derivatives 58al was found to be potent against the Gram-positive bacterial strains. Among all the tested compounds, the compounds 58c, 58d, 58f, and 58j were found to be more efficient members of the series. Most synthesized compounds were active against Gram-positive bacteria viz. Bacillus subtilis (MTCC 441) and Staphylococcus aureus (MTCC 96), Gram-negative bacteria viz. Escherichia coli (MTCC 443) and Salmonella typhi (MTCC 98). Some of the synthesized compounds were found sufficiently potent to inhibit fungal pathogen viz. Candida albicans (MTCC 227).
Curcumin (CF: C21H20O6, MW: 368.39, (1E,6E)-1,7-bis(4-hydroxy-3-methoxyphenyl)hepta-1,6-diene-3,5-dione) is a diketone compound extracted from the rhizomes of some plants in the Zingiberaceae and Araceae, and is a very rare pigment with diketone structure in the plant kingdom. Subhedar and colleagues [50] synthesized quinoline–curcumin derivatives 5960 (Figure 13) (Table 9), and evaluated their anti-tuberculosis activity against MTB and M. bovis BCG in vitro. Rifampicin as a positive control. Overall, the synthesized compounds showed excellent selectivity against M. bovis BCG compared to the MTB strain. Among the synthesized quinolinyl monocarbonyl curcumin analogues, compounds 59a, 59c, and 60ac with MIC90 range of 7.8–27.9 μg/mL were active against dormant MTB strains, while compounds 59bd, 60ac, and 60d with MIC90 range of 2.7–27.2 μg/mL were active against dormant M. bovis BCG strains were active. The biological evaluation results reveal that, the activity was considerably affected by introducing various substituents on the quinoline ring and N-methylation of piperidinone scaffold. From the compounds 59ad, compound 59a and 59c showed moderate antitubercular activity with MIC90 value 25.5 and 27.9 µg/mL, respectively, against dormant MTB strain. The remaining analogues from the series does not display significant antitubercular activity against dormant MTB strain with MIC90 values > 30 µg/mL. N-Methylpiperidinone-based analogues 60ad, particularly, compounds 60a (MIC90 value 26.5 µg/mL) and 60b (MIC90 value 20.0 µg/mL), showed good to moderate antitubercular activity against dormant MTB strain. In particular, compound 60c showed excellent antitubercular activity against dormant MTB strain with MIC90 value 7.8 µg/mL. Hence, among all the synthesized analogues, the only compounds 59a, 59c, and 60ac showed moderate to excellent antitubercular activity against dormant MTB strain. From the analogues 59ad, compound 59b showed excellent antitubercular activity with MIC90 value 2.7 µg/mL against dormant M. bovis BCG strain. Compound 59c showed moderate antitubercular activity with MIC90 value 27.2 µg/mL against dormant M. bovis BCG strain. Compound 59d showed promising antitubercular activity against dormant M. bovis BCG strain with MIC90 value 9.2 µg/mL. From the series 60ad, compound 60a showed excellent antitubercular activity with a MIC90 value 7.3 µg/mL against dormant M. bovis BCG strain. Compound 60b and 60d showed good to moderate antitubercular activity with MIC90 value 15.4 and 21.5 µg/mL, respectively, against dormant M. bovis BCG strain. Compound 60c showed promising antitubercular activity against dormant M. bovis BCG strain with MIC90 value 9.4 µg/mL. In short, quinoline-based monocarbonyl curcumin analogues 59bd, 60ad showed good to excellent antitubercular activity against dormant M. bovis BCG strain.
Aurone (CF: C15H10O2, MW: 222.24, 2-Benzylidene-3(2H)-benzofuranone) is a heterocyclic compound of the flavonoid family with a benzofuran moiety linked to a benzylidene group at position C-2. Campaniço and colleagues [51] synthesized azaaurones derivative 61 (Figure 13) (Table 9) and evaluated its anti-mycobacterial MDR- and XDR-TB activity. Compound 61 showed excellent activity against clinically isolated MDR and XDR-TB with MIC99 values of 0.649 and 0.736 μM, respectively.
Kumar and colleagues [52] synthesized aurones quinoline derivatives 62af (Figure 13) (Table 9) and evaluated their antibacterial and antifungal activities against Bacillus subtilis, Staphylococcus aureus, Klebsiella pneumoniae, Aspergillus fumigatus, Candida albicans, and Fusarium oxysporum. All test compounds showed antibacterial activity against Gram-positive test strains; however, only compounds 62c and 62e showed antibacterial activity against Gram-negative test strains (Klebsiella pneumoniae); both MIC values are 0.625 mg/mL. In addition, only compounds 62ab, 62d, and 62f showed activity against the acid-fast microbial strain (MIC range of 0.625–0.078 mg/mL). This different antimicrobial behavior may be due to the differences in their composition and cell membrane structure in different microbial strains since the peptidoglycan layer of Gram-positive bacteria and the phospholipid membrane of Gram-negative bacteria interact differently with different antimicrobial agent molecules.
Sabatini and colleagues [53] synthesized the quinoline derivatives 6364 (Figure 13) (Table 9) from flavonoids and evaluated the function of EPI. The results showed that compounds 63 and 64 exhibited good antibacterial activity (SA-1199B inhibited EtBr efflux > 65% at 50 μM concentration).
Wang and colleagues [54] synthesized 3-(iso)quinolinyl-4-chromenone derivatives 6566 (Figure 13) (Table 9) and evaluated their antifungal activity. Bioassay data showed that 3-quinolinyl-4-chromenone 65 showed significant in vitro activity against Sclerotinia sclerotiorum, Vibrio marcescens, and grey mould with EC50 values of 3.65, 2.61, and 2.32 mg/L, respectively. 3-isoquinolinyl-4-chrome none 66 showed excellent in vitro activity against Sclerotinia sclerotiorum with an EC50 value of 1.94 mg/L, which was close to the commercial fungicide chlorothalonil (EC50 = 1.57 mg/L), but lower than Boscard (EC50 = 0.67 mg/L). For V. mali and B. cinerea, the activity of 3-isoquinolinyl-4-chrome none 66 (EC50 = 1.56, 1.54 mg/L) was significantly higher than that of chlorothalonil (EC50 = 11.24, 2.92 mg/L). In addition, in vivo experiments showed that compounds 65 and 66 showed 88.24% and 94.12% inhibition against grey mould at 50 mg/L, compared to 76.47% and 97.06% inhibition by the positive controls chlorothalonil and boscalid, respectively. Physiological and biochemical studies suggest that the main mechanism of action of compounds 65 and 66 on S. sclerotiorum and B. cinerea may involve altering the morphology and increasing the permeability of cell membranes of A. aurantium.
Figure 13. The chemical structures of antibacterial compounds 5666.
Figure 13. The chemical structures of antibacterial compounds 5666.
Molecules 28 06478 g013
Gogoi and colleagues [55] synthesized the A- and D-ring-fused steroid quinolines derivatives 67aj (Figure 14) (Table 9) and evaluated their antibacterial and antifungal activities. Cystine was used as a standard drug against fungi, and gentamicin sulfate was used against bacteria. The results showed that compound 67a did not exert potent inhibitory activity against all bacterial strains tested. Methoxy derivative 67c showed an inhibitory effect against bacterial strains Pseudomonas aeruginosa, while compounds 67e, 67gh, and 67j showed significant inhibition against bacterial strains Staphylococcus aureus and Bacillus subtilis. These results suggest that the substituents in the quinoline molecules of compounds 67a and 67f, as well as the backbone structure, play an important role in the inhibitory activity of bacterial strains. For the fungal strain A. niger, only the tested compound 67a showed a strong inhibition (MIC = 22 μg/mL), while compounds 67e and 67gh showed only moderate inhibition. In contrast, most of the tested compounds 67eh and 67j showed a strong inhibition of the growth of the fungal strain C. albicans (MIC values arranged 13–19 μg/mL). Compounds 67b, 67d, and 67i were not effective against any of the tested strains. Furthermore, the determination of MICs and MFCs of the active compounds showed that compound 67e showed maximum activity against most of the tested strains with 21 μg/mL for S. aureus, 19 μg/mL for B. subtilis, 23 μg/mL for C. albicans and 28 μg/mL for C. niger. It is evident from the data that compounds 67c and 67f against Pseudomonas aeruginosa showed very good antibacterial activity, almost similar to the standard drug gentamicin sulfate. Similarly, 67e, 67gh, and 67j showed inhibitory activity against the bacterial strains Bacillus subtilis (MIC values arranged 12–22 μg/mL) and Staphylococcus aureus (MIC values arranged 10–23 μg/mL), indicating that these compounds are promising antimicrobial compounds for further investigation.
Balaji and colleagues [56] synthesized a series of quinoline–coumarin derivatives 68ag (Figure 14) (Table 9) and evaluated the in vitro antibacterial activity against Gram (+) and Gram (−) bacteria, such as Escherichia coli, Pseudomonas aeruginosa, Staphylococcus aureus, Meloidogyne litoralis, and Bacillus subtilis. Compounds possessing methyl, methoxy, and fused aryl rings, such as 68c, 68d, and 68g, at C-8 of quinoline ring showed better activity than their standard drug streptomycin against Escherichia coli (all MIC values are 6.25 μg/mL). Similarly, compounds 68d and 68g showed better activity against P. aeruginosa (both MIC values are 6.25 μg/mL). Compound 68f with bromine at C-6 showed better activity against M. litoralis (MIC = 6.25 μg/mL), whereas 68a with methyl at C-6 showed better activity against S. aureus (MIC = 12.5 μg/mL). No compound has good activity against B. subtilis with the standard drug streptomycin, respectively. DPPH (1,1-diphenyl-2-picryl-hydrazil) radical scavenging method has been chosen to evaluate the antioxidant potential of the compounds 68ag compared with that of commercial antioxidant butylated hydroxytoluene (BHT). The results in percentage are expressed as of absorbance decrease at 517 nm, and the absorbance of DPPH solution in the absence of compounds. The values revealed that the radical scavenging activity of 7-(2-chloroquinolin-4-yloxy)-4-methyl-2H-hromen-2-one on DPPH radicals increases with the increase in concentration. Compounds possessing chloro, bromo substituents at C-6 (68e,f), showed maximum activity at a concentration of 1000 μg/mL. The radical scavenging activity of compounds possessing methyl at C-7 (68b) exhibited less potent than the standard. In summary, these compounds have been subjected to antimicrobial screening against a panel of human pathogens, and most of them are found to be more active than the standard drugs. In addition, antioxidant activity for compound 68e shows a moderate 78% of inhibition. The binding energy value of synthesized compounds is less than the standard antimalarial drugs like chloroquine and amodiaquine.
Khatkar and colleagues [57] synthesized a quinoline ferulic acid derivative 69 (Figure 14) (Table 9). The synthesized compound was evaluated in vitro for its antibacterial activity against various Gram-positive and Gram-negative bacterial and fungal strains. As a result, compound 69 was found to be most effective against B. subtilis with a pMICbs value of 2.01.
Figure 14. The chemical structures of antibacterial compounds 6769.
Figure 14. The chemical structures of antibacterial compounds 6769.
Molecules 28 06478 g014
Maddela and colleagues [58] designed and synthesized the isatin–quinoline derivative compound 70 (Figure 15) (Table 9) and evaluated its antitubercular activity against Mycobacterium tuberculosis. Compound 70 had a MIC of 0.09 mg/L and showed good inhibitory activity compared to the standard drug isoniazid.
Tabbi and colleagues [59] synthesized new adamantane-containing chalcones derivatives 7173 (Figure 15) (Table 9) and evaluated their resistance against Enterococcus faecalis 29212, Pseudomonas aeruginosa ATCC 27853, strong antibacterial activity against Escherichia coli and interesting antifungal activity against Candida albicans ATCC 90030. Compounds 7173 were also tested for anti-Candida activity. All compounds were found to have the same activity as ketoconazole against C. glabrata ATCC 90030 (MIC = 200 μg/mL). However, the compounds had no significant anti-Candida activity against C. krusei ATCC 6258 (MIC = 100 μg/mL).
Pan and colleagues [60] synthesized hydroxycoumarin quinoline derivatives 74ab (Figure 15) (Table 9) to evaluate their antifungal activity. Compound 74b exhibited potent growth inhibition against all tested fungi. (The inhibition rates of compound 74b against A. alternate, A. solani, B. cinerea, and F.oxysporum were 57.6%, 79.0%, 72.9%, and 89.6%, respectively). Compound 74a exhibited moderate activity. The inhibition rates of compound 74a against A. alternate, A. solani, B. cinerea, and F. oxysporum were 49.3%, 61.2%, 63.3%, and 56.4%, respectively. In contrast, compound 74b exhibited selective antifungal activity against A. alternata and A. solani, which were alternatively present.
Naphthoquinone (CF: C10H6O2, MW: 158.15, Naphthalene-1,4-dione) is an organic substance, and theoretically, there are 6 kinds of naphthoquinone, of which only 1,4-, 1,2-, and 2,6- can be obtained stably. 1,4-Naphthoquinone, also known as α-naphthoquinone, is used to make dyes, medicines, and fungicides. Kalt and colleagues [61] synthesized 1,4-naphthoquinonequinoline derivative 75 (Figure 15) (Table 9) and tested its anti-mycobacterial activity. The results showed that compound 75 had a slight inhibitory effect against mycobacteria with a MIC of 8 μg/mL.
Lasiokaurin (molecular formula: C22H30O7, MW: 406.47, (1α,6β,7α,14R)-1-(Acetyloxy)-7,20-epoxy-6,7,14-trihydroxykaur-16-en-15-one) is a diterpene compound present in the leaves of the Lamiaceae plant, Isodon trichocarpus Kudo, and the leaves of Rabdosia japonica (Burm.f.) Hara. Li and colleagues [62] synthesized quinoline-based lasiokaurin derivative 76 (Figure 15) (Table 9) and performed antibacterial tests. The results showed that compound 76 exhibited the most promising antibacterial activity with MICs of 2.0 and 1.0 μg/mL against the Gram-positive bacteria Staphylococcus aureus and Bacillus subtilis, respectively.
Figure 15. The chemical structures of antibacterial compound 7076.
Figure 15. The chemical structures of antibacterial compound 7076.
Molecules 28 06478 g015
Table 9. Quinoline derivatives with antibacterial activity.
Table 9. Quinoline derivatives with antibacterial activity.
Compd.Activity Origin Ref
56aE. coli
MIC = 25.0 μg/mL
S. aureus
MIC = 12.5 μg/mL
C. albicans
MIC = 12.5 μg/mL
synthetic[48]
56bE. coli
MIC = 12.5 μg/mL
S. aureus
MIC = 25.0 μg/mL
C. albicans
MIC = 25.0 μg/mL
synthetic[48]
56cE. coli
MIC = 6.25 μg/mL
S. aureus
MIC = 12.5 μg/mL
C. albicans
MIC = 12.5 μg/mL
synthetic[48]
56dE. coli
MIC = 12.5 μg/mL
S. aureus
MIC = 12.5 μg/mL
C. albicans
MIC = 25.0 μg/mL
synthetic[48]
56eE. coli
MIC = 6.25 μg/mL
S. aureus
MIC = 12.5 μg/mL
C. albicans
MIC = 12.5 μg/mL
synthetic[48]
56fE. coli
MIC = 12.5 μg/mL
S. aureus
MIC = 12.5 μg/mL
C. albicans
MIC = 12.5 μg/mL
synthetic[48]
56gE. coli
MIC = 12.5 μg/mL
S. aureus
MIC = 12.5 μg/mL
C. albicans
MIC = 12.5 μg/mL
synthetic[48]
56hE. coli
MIC = 12.5 μg/mL
S. aureus
MIC = 25.0 μg/mL
C. albicans
MIC = 25.0 μg/mL
synthetic[48]
57aE. coli
MIC = 12.5 μg/mL
S. aureus
MIC = 12.5 μg/mL
C. albicans
MIC = 12.5 μg/mL
synthetic[48]
57bE. coli
MIC = 12.5 μg/mL
S. aureus
MIC = 25.0 μg/mL
C. albicans
MIC = 12.5 μg/mL
synthetic[48]
58aB.s.
MIC = 500 μg/mL
S.a.
MIC = 250 μg/mL
E.c.
MIC = 100 μg/mL
S.t.
MIC = 100 μg/mL
A.n.
MIC = 500 μg/mL
C.a.
MIC = 500 μg/mL
synthetic[49]
58bB.s.
MIC = 250 μg/mL
S.a.
MIC = 200 μg/mL
E.c.
MIC = 500 μg/mL
S.t.
MIC = 250 μg/mL
A.n.
MIC = 200 μg/mL
C.a.
MIC = 500 μg/mL
synthetic[49]
58cB.s.
MIC = 200 μg/mL
S.a.
MIC = 250 μg/mL
E.c.
MIC = 62.5 μg/mL
S.t.
MIC = 100 μg/mL
A.n.
MIC = 250 μg/mL
C.a.
MIC = 250 μg/mL
synthetic[49]
58dB.s.
MIC = 250 μg/mL
S.a.
MIC = 125 μg/mL
E.c.
MIC = 62.5 μg/mL
S.t.
MIC = 100 μg/mL
A.n.
MIC = 500 μg/mL
C.a.
MIC = 1000 μg/mL
synthetic[49]
58eB.s.
MIC = 250 μg/mL
S.a.
MIC = 125 μg/mL
E.c.
MIC = 200 μg/mL
S.t.
MIC = 250 μg/mL
A.n.
MIC = 500 μg/mL
C.a.
MIC = 1000 μg/mL
synthetic[49]
58fB.s.
MIC = 250 μg/mL
S.a.
MIC = 500 μg/mL
E.c.
MIC = 100 μg/mL
S.t.
MIC = 62.5 μg/mL
A.n.
MIC = 1000 μg/mL
C.a.
MIC = 250 μg/mL
synthetic[49]
58gB.s.
MIC = 250 μg/mL
S.a.
MIC = 200 μg/mL
E.c.
MIC = 100 μg/mL
S.t.
MIC = 200 μg/mL
A.n.
MIC = 1000 μg/mL
C.a.
MIC = 200 μg/mL
synthetic[49]
58hB.s.
MIC = 200 μg/mL
S.a.
MIC = 250 μg/mL
E.c.
MIC = 100 μg/mL
S.t.
MIC = 200 μg/mL
A.n.
MIC = 500 μg/mL
C.a.
MIC = 250 μg/mL
synthetic[49]
58iB.s.
MIC = 500 μg/mL
S.a.
MIC = 250 μg/mL
E.c.
MIC = 250 μg/mL
S.t.
MIC = 250 μg/mL
A.n.
MIC = 1000 μg/mL
C.a.
MIC = 200 μg/mL
synthetic[49]
58jB.s.
MIC = 500 μg/mL
S.a.
MIC = 100 μg/mL
E.c.
MIC = 62.5 μg/mL
S.t.
MIC = 100 μg/mL
A.n.
MIC = 1000 μg/mL
C.a.
MIC > 1000 μg/mL
synthetic[49]
58kB.s.
MIC = 200 μg/mL
S.a.
MIC = 125 μg/mL
E.c.
MIC = 100 μg/mL
S.t.
MIC = 200 μg/mL
A.n.
MIC = 1000 μg/mL
C.a.
MIC = 1000 μg/mL
synthetic[49]
58lB.s.
MIC = 250 μg/mL
S.a.
MIC = 100 μg/mL
E.c.
MIC = 200 μg/mL
S.t.
MIC = 250 μg/mL
A.n.
MIC > 1000 μg/mL
C.a.
MIC = 1000 μg/mL
synthetic[49]
59aMTB
MIC50 = 8.7 μg/mL
MIC90 = 25.5 μg/mL
M. Bovis BCG
MIC50 = 12.9 μg/mL
MIC90 > 30 μg/mL
synthetic[50]
59bMTB
MIC50 > 30 μg/mL
MIC90 > 30 μg/mL
M. Bovis BCG
MIC50 = 2.5 μg/mL
MIC90 > 30 μg/mL
synthetic[50]
59cMTB
MIC50 = 2.8 μg/mL
MIC90 > 30 μg/mL
M. Bovis BCG
MIC50 = 1.4 μg/mL
MIC90 = 2.7 μg/mL
synthetic[50]
59dMTB
MIC50 = 27.8 μg/mL
MIC90 > 30 μg/mL
M. Bovis BCG
MIC50 = 19.7 μg/mL
MIC90 > 30 μg/mL
synthetic[50]
60aMTB
MIC50 = 6.7 μg/mL
MIC90 = 26.5 μg/mL
M. Bovis BCG
MIC50 = 6.0 μg/mL
MIC90 = 7.3 μg/mL
synthetic[50]
60bMTB
MIC50 = 7.7 μg/mL
MIC90 = 20.0 μg/mL
M. Bovis BCG
MIC50 = 5.8 μg/mL
MIC90 = 15.4 μg/mL
synthetic[50]
60cMTB
MIC50 = 2.3 μg/mL
MIC90 = 7.8 μg/mL
M. Bovis BCG
MIC50 = 5.8 μg/mL
MIC90 = 9.4 μg/mL
synthetic[50]
60dMTB
MIC50 > 30 μg/mL
MIC90 > 30 μg/mL
M. Bovis BCG
MIC50 = 3.3 μg/mL
MIC90 = 21.5 μg/mL
synthetic[50]
61MDR
MIC99 = 0.649 μM
XDR-TB
MIC99 = 0.736 μM
synthetic[51]
62aB. subtilis
MIC = 0.020 mg/mL
S. aureus
MIC = 1.25 mg/mL
M. smegmatis
MIC = 0.625 mg/mL
F. oxysporum
MIC = 0.625 mg/mL
synthetic[52]
62bB. subtilis
MIC = 1.25 mg/mL
S. aureus
MIC = 2.5 mg/mL
synthetic[52]
62cB. subtilis
MIC = 1.25 mg/mL
S. aureus
MIC = 1.25 mg/mL
K. pneumoniae
MIC = 0.625 mg/mL
synthetic[52]
62dB. subtilis
MIC = 1.25 mg/mL
S. aureus
MIC = 1.25 mg/mL
M. smegmatis
MIC = 0.625 mg/mL
synthetic[52]
62eB. subtilis
MIC = 1.25 mg/mL
S. aureus
MIC = 1.25 mg/mL
M. smegmatis
MIC = 0.625 mg/mL
C. albicans
MIC = 0.156 mg/mL
Klebsiella pneumoniae
MIC = 0.625 mg/mL
synthetic[52]
62fB. subtilis
MIC = 1.25 mg/mL
S. aureus
MIC = 1.25 mg/mL
M. smegmatis
MIC = 0.625 mg/mL
synthetic[52]
63inhibited EtBr efflux > 65% at 50 μMsynthetic[53]
64inhibited EtBr efflux > 65% at 50 μMsynthetic[53]
65Sclerotinia sclerotiorum
EC50 = 1.57 mg/L
synthetic[54]
66V. mali
EC50 = 1.56 mg/L
B. cinerea
EC50 = 1.54 mg/L
synthetic[54]
67a-synthetic[55]
67b-synthetic[55]
67cPseudomonas aeruginosa
MIC = 18 μg/mL
synthetic[55]
67d-synthetic[55]
67eStaphylococcus aureus
MIC = 21 μg/mL
Bacillus subtilis
MIC = 19 μg/mL
synthetic[55]
67fPseudomonas aeruginosa
MIC = 19 μg/mL
synthetic[55]
67gStaphylococcus aureus
MIC = 28 μg/mL
Bacillus subtilis
MIC = 26 μg/mL
synthetic[55]
67hStaphylococcus aureus
MIC = 40 μg/mL
Bacillus subtilis
MIC = 24 μg/mL
synthetic[55]
67i-synthetic[55]
67jStaphylococcusAureus
MIC = 24 μg/mL
Bacillus subtilis
MIC = 40 μg/mL
synthetic[55]
68aEscherichia coli
MIC = 12.5 μg/mL
Pseudomonas aeruginosa
MIC = 100 μg/mL
Staphylococcus aureus
MIC = 12.5 μg/mL
synthetic[56]
68bPseudomonas aeruginosa
MIC = 12.5 μg/mL
synthetic[56]
68cEscherichia coli
MIC = 6.25 μg/mL
Meloidogyne litoralis
MIC = 100 μg/mL
synthetic[56]
68dEscherichia coli
MIC = 6.25 μg/mL
Pseudomonas aeruginosa
MIC = 6.25 μg/mL
Bacillus subtilis
MIC = 100 μg/mL
synthetic[56]
68eEscherichia coli
MIC = 100 μg/mL
Pseudomonas aeruginosa
MIC = 12.5 μg/mL
Meloidogyne litoralis
MIC = 100 μg/mL
Staphylococcus aureus
MIC = 50 μg/mL
synthetic[56]
68fPseudomonas aeruginosa
MIC = 50 μg/mL
Meloidogyne litoralis
MIC = 6.25 μg/mL
synthetic[56]
68gEscherichia coli
MIC = 6.25 μg/mL
Pseudomonas aeruginosa
MIC = 6.25 μg/mL
Staphylococcus aureus
MIC = 50 μg/mL
synthetic[56]
69B. subtilis pMICbs = 2.01synthetic[57]
70Mycobacterium tuberculosis
MIC = 0.09 mg/L
synthetic[58]
71aC. glabrata ATCC 90030 MIC = 200 μg/mLsynthetic[59]
71bC. glabrata ATCC 90030 MIC = 200 μg/mLsynthetic[59]
72C. glabrata ATCC 90030 MIC = 200 μg/mLsynthetic[59]
73C. glabrata ATCC 90030 MIC = 200 μg/mLsynthetic[59]
74aA. alternate
inhibition rate = 49.3%
A. solani
inhibition rate = 61.2%
B. cinerea
inhibition rate = 63.3%
F. oxysporum
inhibition rate = 56.4%
synthetic[60]
74bA. alternate
inhibition rate = 57.6%
A. solani
inhibition rate = 79.0%
B. cinerea
inhibition rate = 72.9%
F. oxysporum
inhibition rate = 89.6%
synthetic[60]
75Mycobacteria MIC = 8 μg/mLsynthetic[61]
76Staphylococcus aureus
MIC = 2.0 μg/mL
Bacillus subtilis
MIC = 1.0 μg/mL
synthetic[62]
Summary: Quinoline-based molecules have been found to be very effective in inhibiting microbial pathogens. Among the drugs of quinoline scaffolds, fluoroquinolone antibiotics, represented by ciprofloxacin, are a large class of antibiotics. In addition, bedaquiline is a diarylquinoline-based drug that has been used to treat multidrug-resistant tuberculosis (MDR-TB). In order to adapt to environmental changes, especially the use of antibiotics, bacteria have developed a variety of mechanisms to resist various adverse conditions. Therefore, bacterial infection has once again evolved into a serious threat worldwide. The increasing number of multidrug-resistant microbial strains and new advances in untreatable infections make the treatment of bacterial infections difficult.
Compound 62e showed excellent antitubercular activity with MIC value 0.156 µg/mL against dormant C. albicans strain. Compound 70 against Mycobacterium tuberculosis showed the strongest inhibitory activity with a MIC value of 0.09 μg/mL. Compound 76 exhibited the most promising antibacterial activity with MICs of 2.0 and 1.0 μg/mL against the Gram-positive Bacteria Staphylococcus aureus and Bacillus subtilis, respectively.
These results suggest that the introduction of quinoline on the basis of aurone, isatin, and lasiokaurin can enhance antibacterial activity. However, there is no obvious rule between the introduced sites and the substituents on quinoline; moreover, the antibacterial mechanism of these compounds has not been studied.

3.10. Anticancer Activity

Lombard and colleagues [39] synthesized two quinoline–coumarin derivatives 4344 (Figure 11) (Table 10) and evaluated their anticancer activity against kidney cancer (TK10), melanoma (UACC62), and breast cancer (MCF7) cell lines, etoposide was used as a positive control. The results of the five-dose cancer screening showed that mixed dimer 44 was less active, and its anticancer activity was classified as against renal (TGI = 18.5 μM) and melanoma (TGI = 17.43 μM) cell linesmoderate. The breast (MCF7) cell line showed higher sensitivity to dimer 44 with a TGI of 2.92 μM, so the activity of dimer 44 could be classified as potent for this cell line. Dimer 44 was 2-fold (TGI, 18.5 vs. 43.33 μM), 15-fold (TGI, 2.92 vs. 43.52 μM), and 5.7-fold (TGI, 17.43 vs. >100 μM) activity than etoposide against TK10, UACC62, and MCF7, respectively. The synthetic dimer exhibited moderate to potent anticancer activity against the cell lines studied and inhibited the growth of all three cell lines at very low concentrations (GI50 values in the range of 0.03–0.08 μM). Compound 43 was able to inhibit the growth of all three cell lines at 10 μM, while compound 44 could inhibit the growth of the UACC62 cell line and the other two cell lines at 100 μM only at 10 μM. Very low LC50 and LC100 values were obtained for both compounds; moreover, these two compounds have very low toxicity to normal cells.
Tabbi and colleagues [59] synthesized the adamantane chalcone–quinoline derivatives 71ab (Figure 15) (Table 10) and evaluated their in vitro anticancer activity against human pancreatic cancer cells Mia Paka 2. The growth inhibitory activities of compounds 71a and 71b were 85% and 77%. Thus, compounds 71ab possess some anticancer activity.
Abonia and colleagues [63] synthesized novel quinoline-2-ketochalcone derivative 77 (Figure 16) (Table 10). In vitro antitumor assays showed that compound 77 exhibited high activity in the samples selected and evaluated by NCI. In particular, compound 77 showed the most significant activity against 50 human tumor cell lines with GI50 values of 1.0 μM, with HCT-116 (colon, GI50 = 0.131 μM) and LOX IMVI (melanoma, GI50 = 0.134 μM) being the most susceptible strains.
Podophyllotoxin (CF: C22H22O8, MW: 414.41, 1,3,3a,4,9,9a-hexahydro-9-hydroxy-6,7-(methylenedioxy)-4-(3′,4′,5′-trimethoxyphenyl)benz[f]isobenzofuran-3-one), also known as podafilol, is a non-alkaloid lignin-like toxin. Kamal and colleagues [64] synthesized the onychotoxin–quinoline derivatives 78ab and 79ab (Figure 16) (Table 10) and evaluated their higher activity against A549, A375, MCF-7, HT-29, and ACHN, and the positive control drugs etoposide and doxorubicin than they themselves. The IC50 values of compound 78a against A549, A375, MCF-7, HT-29, and ACHN were 15.4 μM, 14.5 μM, 13.8 μM, 12.3 μM, and 10.8 μM, respectively. The IC50 values of compound 78b against A549, A375, MCF-7, HT-29, and ACHN were 13.4 μM, 7.7 μM, 11.2 μM, 7.75 μM, and 15.7 μM, respectively. The IC50 values of compound 79a against A549, A375, MCF-7, HT-29, and ACHN were 10.6 μM, 10.3 μM, 8.6 μM, 11.8 μM, and 10.7 μM, respectively. The IC50 values of compound 79b against A549, A375, MCF-7, HT-29, and ACHN were 7.7 μM, 6.8 μM, 2.2 μM, 8.9 μM, and 9.46 μM, respectively.
Ring-A 3,4-seco-cycloartane-type triterpenes (CF: C30H44O2, MW: 468.67, (3R,3aR,5aS,6aR,6bR,9aR,10aS,10bS)-3-[(1R)-1,5-Dimethyl-4-hexen-1yl]tetradecahydro-3a) mainly exist in the gardenia plants of Rubi ceae. Pudhom and colleagues [65] synthesized 3,4-eco-cycloartane-type triterpene quinoline derivative 80 (Figure 16) (Table 10) and evaluated the effect on angiogenesis. The inhibition rate of compound 80 ranged between 50 and 60%.
Kamal and colleagues [66] synthesized 4b-sulfonamide and 4b-[(40-sulfonamide)benzamide] conjugates of podophyllotoxin 81 (Figure 16) (Table 10) and evaluated its effect on anticancer activity. The results showed that the inhibition A549 activity of compound 81 was more potent than that of the positive control drugs doxorubicin and etoposide (the GI50 of compound 81 was 2.51 μM).
Zhao and colleagues [67] synthesized 4′-demethylghostatin (DMEP) quinoline derivatives 8283 (Figure 16) (Table 10) and evaluated their anticancer activity. The inhibitory activity of compound 82 on HepG2, HeLa, A549, and BGC-823 was stronger than that of itself and the positive control etoposide. The IC50 values of compound 82 against HepG2, HeLa, A549, BGC-823, and HL-7702 were 16.03 μM, 0.60 μM, 10.05 μM, 17.41 μM, and 41.77 μM, respectively. The inhibitory activity of compound 83 on HepG2 and A549 is stronger than that of itself and the positive control. The inhibitory activity of compound 83 on HeLa and BGC-823 is not as good as its own. The IC50 values of compound 90 against HepG2, HeLa, A549, BGC-823, and HL-7702 were 9.18 μM, 20.53 μM, 19.20 μM, 28.81 μM, and 20.09 μM, respectively. Compounds 8283 are both on HL-7702 was more toxic than itself.
Ayan and colleagues [68] synthesized a derivative of the aminosteroidal E-37P-quinoline derivative 84 (Figure 16) (Table 10) and evaluated their anticancer activity. The results showed that compound 84, a 5a-androstane-3a,17b-diol derivative with a quinoline nucleus at the end of the piperazine-proline side-chain at position 2b and an ethinyl at position 17a, showed very good antiproliferative activity among the five cancer cell lines studied. The IC50 values of compound 84 for HL-60, MCF-7, T-47D, LNCaP, and WEHI-3 were 0.1, 0.1, 0.1, 2.0, and 1.1 μM, respectively. Furthermore, compound 84 weakly inhibited the two representative liver enzymes, CYP3A4 and CYP2D6, indicating a low risk of drug-drug interactions.
Cui and colleagues [69] synthesized quinoline-like estrone-17-hydrazone 85 (Figure 16) (Table 10) and assayed its activities against the proliferation of HeLa, HT-29, Bel 7404, and SGC 7901, respectively. The results showed that compound 85 had a quinoline structure on the side chain of 17 and showed a better effect. The antiproliferative activity against test cells in vitro was higher than that of the positive control drug cisplatin. In particular, compound 85 showed excellent antiproliferative activity against SGC 7901 in vitro with an IC50 value of 1 μM.
Berberine (CF: C20H18NO4+, MW: 336.36, 16,17-dimethoxy-5,7-dioxa-13-azoniapentacyclo [11.8.0.02,10.04,8.015,20] henicosa-1(13),2,4(8),9,14,16,18,20-octaene), a quaternary alkaloid isolated from the traditional Chinese medicine Coptis chinensis, is the main active ingredient in the antibacterial activity of Coptis. Jin and colleagues [70] synthesized quinoline berberine derivative 86 (Figure 16) (Table 10) and determined its antiproliferative activity against MCF-7, MCF-7/ADR, SW-1990 and SMMC-7721, and non-cancerous HUVEC cells. The results showed that the antiproliferative activity of compound 86 against four human cancer cell lines was slightly weaker (The IC50 values of compound 86 for MCF-7, MCF-7/ADR, SW1990, and SMMC-7721 were 181.478 μM, 96.523 μM, 111.837 μM, and 75.546 μM, respectively), only inhibited MCF-7 and SMMC-7721 better than itself.
Hayat and colleagues [71] synthesized 4-azanonaphtholide quinoline derivative 87 (Figure 16) (Table 10) and evaluated its antiproliferative activity against five representative cancer cell lines HepG2, A431, A549, MCF 7, and HCT 116. Daurinol served as a positive control. Compound 87 showed almost equivalent activity to daurinol at 10 μM. The IC50 values of compound 87 for HepG2, A431, A549, MCF 7, and HCT 116 were 8.40 μM, 11.56 μM, 4.33 μM, 5.99 μM, and 3.48 μM, respectively, exhibited a slightly stronger activity.
Srivastava and colleagues [72] synthesized quinoline–stilbene derivative 88 (Figure 16) (Table 10) and studied it for antiproliferative activity. Compound 88 showed surprisingly strong activity against MDA-MB 468 breast cancer cells (IC50 = 0.12 μM).
Figure 16. The chemical structures of anticancer compounds 7788.
Figure 16. The chemical structures of anticancer compounds 7788.
Molecules 28 06478 g016
Raghavan and colleagues [73] synthesized curcumin–quinolone derivative 89 (Figure 17) and performed in vitro cytotoxicity assays against A549, MCF7, SKOV3, and H460. Compound 115 showed the greatest activity against SKOV3 cells with a minimum IC50 value of 12.8 μM observed and was therefore used for further biological experiments. At the IC50 concentration, the compound was not toxic to the normal fibroblast cell line NIH3T3, with a cell survival rate of 74.5%.
Cui and colleagues [74] synthesized a steroidal quinoline derivative 90 (Figure 17) (Table 10) with a cholestane type 17-branched structure and determined its inhibitory effect in human hepatoma cells (Bel-7404) and gastric cancer cells (SGC-7901). Compound 90 showed significant growth and proliferation inhibition in both tumor cells, and both were stronger than the positive control cisplatin (IC50 values of 90 were 28.7, 17.9 µmol/L).
He and colleagues [75] synthesized quinoline pregnenolone derivative 91 (Figure 17) (Table 10) and the inhibitory activity against human colon cancer cells (HT-29), human cervical cancer cells (HeLa) and human gastric cancer cells (SGC-7901), using cisplatin as a positive control, showed that compound 91 was superior to the inhibitory activity of the positive control substance cisplatin; the inhibitory activity against HeLa and SGC-7901 cells was less than 10 μmol/L (compound 91’s IC50 values were 14.1, 9.1, 8.2 μmol/L).
Baji and colleagues [76] synthesized novel D- and A-ring-fused quinolines of the estrone and 5α-androstanes series 9295 (Figure 17) (Table 10) and investigated their antiproliferative activity on human cervical cancer (C33A, HeLa, and SiHA) and breast cancer (MCF-7, MDA-MB-231, MDA-MB-361, and T47D) cell lines. The results indicated that ring D-fused quinolines 92ac exhibited weak or modest antiproliferative properties, typically eliciting 30–50% growth inhibition at 30 µM. The cytostatic activities of benz[c]acridine derivatives 93af were even weaker, except for 93e and 93f, which blocked the proliferation of HeLa cells selectively with IC50 values comparable to that of the reference agent cisplatin. IC50 values of 93ef were 99.6 and 89.4 µM, respectively. Ring A-fused quinolines generally inhibited cellular growth more efficiently. Compounds with a 17-OH group (95ac and 95eg) tended to display more pronounced action than the corresponding 17-OAc analogues (94ac and 94eg). Since the efficacy of analogue 95a containing an unsubstituted quinoline moiety was similar to those of 95bh, the character of the substituent on the quinoline does not seem to be crucial for the antiproliferative actions; however, substitution at position 6′ (95c, 95e, and 95f) appeared favorable. The efficacy of 95c against T47D cells was comparable to that of the reference agent cisplatin. IC50 value of 95c was 80.4 µM.
Combretastatin A-4 (CA-4, CF: C18H20O5, MW: 316.3484, (Z)-2-Methoxy-5-(3,4,5-trimethoxystyrene)phenol) is a novel vasopressor that targets tubulin in vivo, inhibiting its polymerization and further selectively destroying the vascular endothelium of tumor tissues, closing the vasculature of tumor tissues and rendering them hypoxic and nutritious, thus acting as an antitumor agent. Chaudhary and colleagues [77] synthesized combretastatin A-4 quinoline derivative 96 (Figure 17) (Table 10) and evaluated its antiproliferative activity. The results showed that compound 96 had higher antiproliferative activity than CA-4. In addition, compound 96 inhibited the migration of highly metastatic MDA-MB-231 more strongly than CA-4, indicating its potent anti-metastatic potential. Compound 96 inhibited the rate and extent of in vitro assembly of purified tubulin with an IC50 of 1.6 μM and a dissociation constant of 1.9 μM.
Maslinic acid (CF: C30H48O4, MW: 472.71, (2α,3β)-2,3-dihydroxyolean-12-en-28-acid) is a pentacyclic triterpene acid, found in hawthorn, red dates, loquat leaves, and olive oil. Madecassic acid (CF: C30H48O6, MW: 504.70, (2α,3β,4α,6β)-2,3,6,23-Tetrahydroxyurs-12-en-28-oic acid) is derived from the whole grass of Centella Asiatica of the Umbelliferae. Sommerwerk and colleagues [78] synthesized maslinic acid derivative quinoline derivative 97al (Figure 17) (Table 10) and madecassic acid quinoline derivative 98 and evaluated their antitumor activity. Although the cytotoxicity of compounds 97ad and 97gj was similar to that of pyridyl-substituted amides, the 8-quinolinyl derivatives 97k and 97l exhibited selective cytotoxicity against different human tumor cell lines; however, their overall cytotoxicity was low, and their solubility in water was poor. However, the 5-quinolinyl-substituted compounds 97e and 97f performed better, as their EC50 values were quite low, and they were cytotoxic against human tumor cell lines but significantly less cytotoxic against non-malignant mouse fibroblasts. Compound 98 has an isoquinoline group present, which shows both low EC50 values (EC50 = 80 μM for A2780) and high tumor/non-malignant cell selectivity (EC50 = 3.23 μM for NIH 3T3, resulting in a selectivity index of 40).
Figure 17. The chemical structures of anticancer compounds 8998.
Figure 17. The chemical structures of anticancer compounds 8998.
Molecules 28 06478 g017
Shobeiri and colleagues [79] synthesized 2-aryl-trimethoxyquinoline derivatives 99ae (Figure 18) (Table 10) and evaluated the cytotoxic activity of the synthesized compounds against four human cancer cell lines MCF-7, MCF-7/MX, A-2780, and A-2780/RCIS. The results showed that all the alcohol derivatives 99ae showed greater cytotoxicity against the A-2780 cell line compared to the other three cell lines IC50 ranging from 7.98 to 60 μM. Interestingly, drug-resistant human breast cancer cells (MCF-7/MX) were more sensitive to all alcohol derivatives except 99a than the parental cells (MCF-7). In contrast, they induced more cytotoxicity in the A-2780 cell line compared to resistant human ovarian cancer (A-2780/RCIS), suggesting that compounds may exert their cytotoxic activity in different tumor cell types through different mechanisms. Among these quinolines, compound 99e, which possesses a trimethoxyphenyl group at the second position of the quinoline ring, exhibited the strongest cytotoxicity against cancer cell lines, with the same effect on both parental and resistant cell lines.
Zhang and colleagues [80] synthesized the podophyllotoxin quinoline derivatives 100102 (Figure 18) (Table 10) and evaluated their antiproliferative activity against human leukemia cells (K562 and K562/ADR). Etoposide and doxorubicin were used as positive compounds. Compounds 100102 showed potent cytotoxicity comparable to or higher than etoposide and doxorubicin (IC50 values for the antiproliferative activity of compound 100 were 0.061 and 0.064 μM for K562 and K562/ADR cells, respectively. Compound 101 for K562 and K562/ADR cells. IC50 values of 0.177 and 0.064 μM for antiproliferative activity and 0.034 and 0.022 μM for compound 102 on K562 and K562/ADR cells, respectively. In general, the activity of the tested molecules was higher against K562 cells than against K562/ADR cells. Moreover, the IC50 value of compound 102 in K562/ADR cells was 0.034 μM; its activity was 65.029 and 552.323 times higher than that of etoposide and doxorubicin, respectively.
Li and colleagues [81] synthesized a podophyllotoxin-derived quinoline derivative 103 (Figure 18) (Table 10) and evaluated its antiproliferative activity against human promyelocytic leukemia cells HL60, human gastric cancer cells SGC-7901, human colon cancer cells MCF-7, human in vitro anticancer active breast cancer cells HCT116, and human non-small cell lung cancer cells A549. Unfortunately, the anticancer activity of compound 103 was inferior to that of the positive control drug etoposide (IC50 values arranged 8.09–73.40 μM).
Figure 18. The chemical structures of anticancer compounds 99103.
Figure 18. The chemical structures of anticancer compounds 99103.
Molecules 28 06478 g018
Li and colleagues [62] synthesized a lasiokaurin quinoline derivative 104 (Figure 19) (Table 10) and evaluated the antiproliferative activity against human leukemia K562 cells, human gastric cancer MGC-803 cells, human esophageal cancer CaEs-17 cells, and human hepatocellular carcinoma Bel-7402 cells. The results showed that compound 104 inhibited Bel-7402 more strongly than the positive control drug paclitaxel (IC50 values for compound 104 were 1.89, 1.03, 1.74, and 0.96 μM, respectively).
Ursolic acid (CF: C30H48O3, MW: 456.700, (3β)-3-Hydroxyurs-12-en-28-oic acid) is a natural triterpenoid carboxylic acid compound present in the Labiatae plant Prunella vulgaris L. Gu and colleagues [82] synthesized a series of novel ursolic acid quinoline derivatives 105107 (Figure 19) (Table 10) and evaluated their in vitro cytotoxicity against three human cancer cell lines (MDA-MB-231, Hela, and SMMC-7721). From the results, compounds 105ad exhibited significant antitumor activity against three cancer cell lines. Compounds 105ad, 106i, and 111c showed prominent cytotoxic activities against at least one cancer cell line (IC50 < 10 μM). Among them, compound 105b exhibited the most potent cytotoxic activity against MDA-MB-231, HeLa, and SMMC-7721 cells with IC50 values of 0.61 ± 0.07, 0.36 ± 0.05, and 12.49 ± 0.08 μM, respectively, stronger than those of positive control. Compound 105a also showed anticancer activity against the three cancer cells slightly weaker than compound 105b. However, compound 105a had a stronger anticancer activity than that of all other compounds. Compounds 105ad, 106i, and 107c did not show considerable cytotoxicity against normal hepatocyte cells QSG-7701 with IC50 > 40 μM. In addition, compounds 106a, 106c, 106d, 106f, 106g, 106l, 107a, 107d, 107f, 107i, and 107l showed moderate inhibition to three cancer cell lines. Compounds 106b, 106e, 106h, 106k, 107b, 107e, and 107k showed weak inhibitory activities against HeLa cells and were not cytotoxic to MDA-MB-231 and SMMC-7721 cells (IC50 > 40 μM), while compound 111h was inactive to all tested cancer cells. Especially, compound 105b was found to be the most potent derivative with IC50 values of 0.61, 0.36, and 12.49 μM against MDA-MB-231, HeLa, and SMMC-7721 cells, respectively, stronger than positive control etoposide.
Gan and colleagues [83] synthesized steroidal quinoline derivatives 108109 (Figure 19) (Table 10) and evaluated their in vitro effects on human HeLa, HT-29, Bel 7404, and antiproliferative activity in SGC 7901 cells. The anticancer activities of compound 108 and cisplatin were comparable (IC50 values of 11.2, 21.3, 28.9, and 10.3 μM/L), while the anticancer activity of compound 109 was inferior to that of cisplatin.
Yao and colleagues [84] synthesized the dihydroartemisinin quinoline hydrazone derivative 110 (Figure 19) (Table 10). Using 5-fluorouracil or paclitaxel as positive controls, the results showed that compound 110 showed more pronounced antitumor activity against MCF-7 cells than that of the positive group. In addition, 110 showed low cytotoxicity against normal human cells.
Figure 19. The chemical structures of anticancer compounds 104110.
Figure 19. The chemical structures of anticancer compounds 104110.
Molecules 28 06478 g019
Aly and colleagues [85] synthesized a new set of fused naphtho[3,2-c]quinoline-6,7,12-trione and naphtho[3,2-c]quinoline-6,7,8,13-tetrone compounds 111 and 112 (Figure 20) (Table 10) for in vitro anticancer screening. The results showed that compounds 111 and 112 had good potency against ERK, which makes it important to investigate the possible application of these inhibitors in RAF-mutant melanoma (IC50 of compounds 111 and 112 were 0.6 and 0.16 μM).
Sri and colleagues [86] synthesized a curcumin-receptor 2-chloro/phenoxy quinoline derivative 113 (Figure 20) (Table 10) and tested their antitumor activity against several cancer cell lines, such as HeLa, HGC-27, NCI-H460, DU-145, PC-3, and 4T1. The IC50 ranged from 1.81 to 12.4 μM. The IC50 of compound 113 against PC-3, DU-145, NCI-H460, and 4 T1 were 3.12, 3.99, 3.96, and 1.81 μM, respectively.
Taheri and colleagues [87] synthesized coumarin–quinoline derivatives 114ab (Figure 20) (Table 10) and determined their cytotoxic effects on A2780 human cancer cells using doxorubicin as a positive control. The results showed that the cytotoxicity of compounds 114ab was significantly higher than that of the other derivatives, with IC50 values of 25 and 62 μg/mL, respectively. Further examination revealed that compound 114a increased ROS levels, decreased MMPs, and induced apoptosis in A2780 cells via the intrinsic mitochondrial pathway; thus, compound 114a may be an appropriate agent for treating ovarian cancer.
Lipeeva and colleagues [88] synthesized amino coumarin–quinoline derivatives 115116 (Figure 20) (Table 10) and evaluated their antiproliferative effects on leukemia CEM-13, MT-4, U-937, and melanoma MEL-8 cancer cells. Although compound 115 was more cytotoxic to cancer cells than the positive parent compound, they were lower than the positive control drug doxorubicin (IC50 values arrange 30.5–47.3 μM). In contrast, the cytotoxicity of compound 116 on MCF-7 cells was comparable to that of the positive control drug doxorubicin. The GI50 value of compound 116 was 10.5 μM.
Oridonin (CF: C20H28O6, MW: 364.43, is a biologically active kaurine-type tetracyclic diterpene isolated from the genus plants Rabdosia in the family Lamiaceae (Iabtea). Shen and colleagues [89] synthesized oridonin derivatives 117118 (Figure 20) (Table 10) and evaluated their antitumor activity in vitro against three human cancer cell lines, HCT116, BEL7402, and MCF7. Compared with the lead compound and the positive control drug 5-fluorouracil (5-Fu), compounds 117 and 118 exhibited potent antiproliferative efficacy against HCT116, MCF-7, and BEL7402 cancer cell lines. IC50 values of 2.51, 0.41, and 2.54 μM, and IC50 values for compound 118 were 2.07, 0.89, and 2.30 μM, respectively.
Zhao and colleagues [90] synthesized podophyllotoxin quinoline derivatives 119122 (Figure 20) (Table 10) and evaluated them for antitumor activity assays against the following four human tumor cell lines: hepatocellular carcinoma cells HepG2, cervical cancer cells HeLa, lung cancer cells A549, and breast cancer cells MCF7. Clinical microtubule polymerization inhibitor nocodazole (Ncz), podophyllotoxin clinical drug etoposide (VP-16), PTOX, and DMEP were used as positive controls. The results showed that most of the anticancer activities of compounds 119122 were inferior to the positive control. IC50 values arranged 0.8–39.2 μM.
Prashanth and colleagues [91] synthesized coumarin quinoline derivative 123ad (Figure 20) (Table 10) and evaluated its cytotoxicity in vitro against ascites EAC and Dalton’s lymphoma ascites DLA cells. The results showed that compounds 123ad had low antitumor activity.
Figure 20. The chemical structures of anticancer compounds 111123.
Figure 20. The chemical structures of anticancer compounds 111123.
Molecules 28 06478 g020
Jin and colleagues [92] synthesized ursolic acid quinoline derivatives 124127 (Figure 21) (Table 10) and evaluated their in vitro antiproliferative activity against three cancer cell lines, MDA-MB-231, HeLa, and SMMC 7721; etoposide was used as a positive control. Regarding the different derivatives, compounds 124ad with carboxyl groups exhibited potent cytotoxic activity against MDA-MB-231 and HeLa cells at low levels of 1 μM and moderate activity against SMMC-7721 cells. Among the acyl hydrazide derivatives 125ah, compounds 125ad also exhibited significant cytotoxic activity comparable to that of compounds 124ad. Compounds 125eh showed almost no activity against all three cancer cell lines (IC50 > 50 μM). Regarding the oxadiazole derivatives 125ah and thiadiazole derivatives 126ah, compounds 125a and 125d exhibited potent cytotoxicity (IC50 < 10 μM) against MDA-MB-231 and HeLa cells, respectively. Compounds 126bc, 127ab, and 127d showed moderate activity against MDA-MB-231 and HeLa cells, while compounds 126eh, 127c, and 127eh showed only slight or no cytotoxicity against the three cancer cell lines.
Figure 21. The chemical structures of anticancer compounds 124127.
Figure 21. The chemical structures of anticancer compounds 124127.
Molecules 28 06478 g021
Yang and colleagues [93] synthesized steroidal quinoline derivatives 128al (Figure 22) (Table 10) and evaluated their in vitro antiproliferative activity against three human lung cancer cells, A549, A431, and H1975. Compounds 128a mildly inhibited the growth of A549, A431, and H1975 with IC50 values of 15.21, 17.45, and 20.76 μM, respectively. Compounds 162b–d with halogen atoms were found to have comparable activity to 128a, while compounds 128ef showed better antiproliferative activity against the tested lung cancer cells. In particular, compound 128f showed the highest potency against A549, A431, and H1975 with IC50 values of 5.34, 6.21, and 7.25 μM, respectively. Compounds 128gh with alkyl groups showed reduced but moderate antiproliferative activity compared to 128f; moreover, nitro-containing compounds 128ij also showed moderate inhibitory activity against the tested cancer cells. Apparently, compounds 128kl containing methoxy and phenyl, respectively, showed weaker growth inhibition against the tested cancer cells.
Figure 22. The chemical structures of anticancer compounds 128.
Figure 22. The chemical structures of anticancer compounds 128.
Molecules 28 06478 g022
Li and colleagues [94] synthesized chalcone–quinoline derivatives 129130 (Figure 23) (Table 10) and evaluated their antiproliferative effects in vitro, and compared them with the reference compound CA-4. Human chronic myeloid leukemia cell K562 was used for the first time. The results showed that all the newly synthesized compounds exhibited good antiproliferative activities in the nanomolar range, except for compounds 129jk and 129no, which have an indole moiety as the B ring. Among them, compounds 129b and 129d with 3-amino-4-methoxyphenyl or 3-hydroxy-4-methoxyphenyl moieties showed the most potent activities with IC50 values of 0.011 and 0.009 μM, respectively, which were comparable to CA-4 (IC50 = 0.011 μM) and approximately 6-fold stronger than the parent compound (IC50 = 0.060 μM). The methyl substituent at the α-position of the unsaturated carbonyl increased activity (129a vs. 129b, 129c vs. 129d, and 129k vs. 129l), and in compounds 129io, except for compounds 129l and 129m, the activity of most compounds in this series (IC50 > 1 μM) is lower than its phenyl counterpart whose unsaturated double bond is substituted at the C-5 position of the indole moiety. In addition, the methyl substituent at the N-1 position of indole (129m) exhibited approximately 5-fold increased activity compared to the unsubstituted counterpart 129l. All compounds 130ad showed good activity except 130d, which has a lactam instead of a quinoline ring. The steric hindrance of the C-2 group of the quinoline moiety appears to have a key effect on the activity, as compounds with smaller substitutions, such as CH3 (129d IC50 = 0.009 μM), NHCH3 (130a IC50 = 0.018 μM), OCH3 (130b IC50 = 0.030 μM), and H (130c IC50 = 0.015 μM), were more active than other compounds with larger groups. Interestingly, the CH3-substituted compound 129d exhibits slightly stronger activity than the corresponding unsubstituted counterpart 130c, despite the greater steric hindrance of methyl groups than hydrogen. The biological functions of more cancer cell lines were further evaluated. Four additional cancer cell lines, including human hepatocellular carcinoma (HepG2), nasopharyngeal epidermoid carcinoma (KB), human colon carcinoma cells (HCT-8), and human breast cancer cells (MDA-MB-231), were selected for further evaluation. K562 cells were the most sensitive of the five cancer cell lines tested, and the most active compound, 129d, exhibited comparable activity to the reference compound CA-4, with IC50 values ranging from 0.009 to 0.016 μM. Notably, the activity of 129d increased approximately 6-fold compared to the parent compound; therefore, 129d was selected for further biological studies. In addition, the selectivity ratio of 129d to normal human liver L-O2 cells was 65.8 times higher than that of CA-4, indicating that the toxicity of 129d may be lower than that of CA-4.
Parthenolide (CF: C15H20O3, MW: 248.32, (1aR,4E,7aS,10aS,10bR)-2,3,6,7,7a,8,10a,10b-Octahydro-1a,5-dimethyl-8-methyleneoxireno[9,10]cyclodeca[1,2-b]furan-9(1aH)-one) is a natural sesquiterpene lactone product isolated from medicinal plants, such as Salvia miltiorrhiza and Salvia miltiorrhiza. Jia and colleagues [95] synthesized a parthenolide quinoline derivative 131 (Figure 23) (Table 10) and determined its cytotoxic activity in HCT116, U87-MG, HepG2, BGC823, and PC9. Both paclitaxel (paclitaxel) and PTL (1a) were used as positive controls. The IC50 values for compound 131 were 3.31, 1.47, 3.66, 1.77, and 3.12 μM.
Betulinic acid (CF: C30H48O3, MW: 456.70, (3β)-3-Hydroxylup-20(29)-en-28-oic acid) is a natural lupin-type pentacyclic triterpenoid present in the leaves of Cyperus rotundus, the bark of birch trees and date palm kernels extracted from them. Platanic acid (CF: C29H46O4, MW: 458.67, (3β)-3-Hydroxy-20-oxo-30-norlupan-28-oic acid) is a pentacyclic triterpenoid isolated from the leaves of Syzygium claviflorum. Hoenke and colleagues [96] prepared betulinic acid quinoline derivatives 132 (Figure 23) (Table 10) and platanic acid quinoline derivatives 133 and screened them for cytotoxicity. Compound 132 was the most cytotoxic in this study and also had the highest tumor or non-tumor cell selectivity, especially for A375 melanoma (S = 91.2), A2780 ovarian cancer (S = 61.6), and hypopharyngeal carcinoma FaDu (S = 59.0) cells. Compound 132 was slightly less selective than 167 (selectivity S was 41.4, 4.0, 19.2, 37.0, and 34.4, respectively). A375 melanoma cells were used in order to facilitate the understanding of their cytotoxicity pattern. The results showed that compound 166 also increased the number of apoptotic cells; however, more cells were in advanced stages. Similar behavior was observed when cells were treated with 133 (8.6% apoptotic and 9.6% late-stage apoptotic cells). In conclusion, compound 133, a 4-isoquinolinamide of 3-O-acetyl-leucovorin acid, was the most cytotoxic compound with EC50 values as low as EC50 = 1.48 μM (A375 melanoma cells) and was also cytotoxic against non-malignant fibroblasts with an NIH 3T3 selectivity index > 91.2.
Xu and colleagues [97] designed and synthesized matrine quinoline derivatives 134ag (Figure 23) (Table 10), using cisplatin as positive drug control, and evaluated compounds for anticancer activity against HepG2, HeLa, and MDA-MB-231 cell lines. Compound 134ag showed good activity against HepG2, HeLa and MDA MB-231 cell lines with IC50 below 25 μM.
Figure 23. The chemical structures of anticancer compounds 129134.
Figure 23. The chemical structures of anticancer compounds 129134.
Molecules 28 06478 g023
Insuasty and colleagues [98] synthesized a series of quinoline-based symmetrical and asymmetrical bis-acetal compounds 135136 (Figure 24) (Table 10). These compounds were evaluated for their in vitro cytotoxic activity against different human cancer cell lines. Compounds 135, 136a, 136d, 136f, and 136g showed the highest activity, while compounds 136b, 136c, and 136e showed moderate activity. Symmetrical N-butyl quinoline chalcone 135 and asymmetrical bis-chalcone 136g exhibited the highest cytotoxicity with overall GI50 values ranging from 0.16 to 5.45 μM, with excessive activity of HCT-116 (GI50 = 0.16 μM) and HT29 (GI50 = 0.42 μM) (colon cancer). Notably, several GI50 values of these compounds were superior to the reference drugs doxorubicin and 5-FU.
Mohassab and colleagues [99] developed novel quinoline/chalcone derivatives 137139 (Figure 24) (Table 10) and tested them in vitro against a panel of cancer cell lines and EGFR and BRAFV600E anticancer targets. The most active compounds 137ab and 138ab effectively inhibited cancer cell growth. After compound 137b, the difference observed between compounds 139ab was almost comparable and extreme in terms of anticancer activity with GI50 cell lines of 3.625 μM and 4.550 μM, respectively. In contrast, compound 137b showed the highest activity among all the new compounds with a GI50 of 3.325 μM to inhibit the growth of cancer cells.
Zeng and colleagues [100] synthesized parthenolide quinoline heterodimer 140 (Figure 24) (Table 10) and evaluated the compounds for in vitro antiproliferative activity in five human cancer cell lines, HCT116, U85MG, HepG2, HepG2, BGC823, and PC9. Paclitaxel (PTX) was used as an experimental control. PTL and MCL were used as positive controls for their inhibition of NF-κB and STAT3. The results showed that compound 140 exhibited higher cytotoxicity than PTL and MMB for all five cell lines. IC50 values range from 2.11 to 5.23 μM/L, respectively.
Mogrol (CF: C30H52O4, MW: 476.73, (3β,9β,10α,11α,24R)-9-Methyl-19-norlanost-5-ene-3,11,24,25-tetrol) is a polysaccharide of Luo Han Guo saponin, which is the main active component of Luo Han Guo. Song and his colleagues [101] synthesized mogrol quinoline derivatives 141142 (Figure 24) (Table 10) and evaluated the in vitro cytotoxicity of the compounds against human lung cancer cell lines A549 and NCI-H460. A quinoline scaffold was introduced to generate 141ac and 142ad, and the cyclic A-fusion derivative 142ad exhibited higher activity than 141ac against the tested cell lines. Compounds 141ab and 142ad showed stronger inhibitory activity than mogrol against A549 with IC50 values ranging from 12.94 to 19.24 μM. The R1 and R2 substituents on the quinoline moiety significantly affected the activity, and the presence of the halogen atom resulted in a decrease in cytotoxic activity. All quinoline derivatives except compound 141c were more active than mogrol against NCI-H460, while compound 142a showed the highest activity with an IC50 value of 17.13 μM.
Celastrol (CF: C29H38O4, MW: 450.61, (9β,13α,14β,20α)-3-Hydroxy-9,13-dimethyl-2-oxo-24,25,26-trinoroleana-1(10),3,5,7-tetraen-29-oic acid) is a pentacyclic triterpene from Tripterygia Wilfordil Hook. f., a pentacyclic triterpene. Shang and colleagues [102] synthesized celastrol quinoline derivative 143 (Figure 24) (Table 10) and evaluated the toxicity of this compound on Hep3B cells, with celastrol being used as a positive control. Compound 143 with a 3-quinoxyl ethyl substituent had the strongest HIF-1a inhibitory activity in this study, with an IC50 value of only 0.05 μM, which was 5-fold higher than the activity of celastrol (IC50 = 0.25 μM). In addition, Western blot results showed that compound 143 could inhibit the expression of HIF-1a protein. Further experiments showed that 143 significantly inhibited the formation of Hep3B cell colonies, hindered cell migration, and induced apoptosis to some extent. Compound 143 (10 mg/kg) had good in vivo antitumor activity in a mouse tumor xenograft model with an inhibition rate of 74.03%, which was superior to the reference compound 5-FU inhibition rate (59.58%).
Figure 24. The chemical structures of anticancer compounds 135143.
Figure 24. The chemical structures of anticancer compounds 135143.
Molecules 28 06478 g024
Dong and colleagues [103] synthesized 6,7,10-trimethoxy-α-naphthoflavone-quinoline derivative 144 (Figure 25) (Table 10) and screened it against the CYP1 enzyme to assess whether larger substituents could enhance the inhibitory activity against CYP1B1. Compound 144 selectively inhibited CYP1B1. IC50 values of 117.6, 1.0 and >1000 μM for CYP1A1, CYP1B1, and CYP1A2, respectively.
Guan and colleagues [104] synthesized chalcone quinoline derivatives 145146 (Figure 25) (Table 10) and evaluated the in vitro antiproliferative activity of the compounds against MGC-803, HCT-116, and MCF-7. The chemotherapy medication 5-Fluorouracil (5-Fu) was used as a positive control. Most quinoline–chalcone derivatives showed strong antiproliferative activity against MGC-803, HCT-116, and MCF-7 cells with IC50 values < 20 μM. Among them, compound 145e showed the most remarkable inhibitory effect on MGC803, HCT-116, and MCF-7 cells with IC50 values of 1.3, 5.34, and 5.21 μM, respectively, which was much lower than that of 5-Fu (IC50 values = 6.22 μM, 0.4 μM, and 11.1 μM, respectively). Thus, the antiproliferative activity of compound 145e on MGC803 cells, structure–activity relationships suggests that the type and position of the substituent (R1) on the chalcone moiety (A ring) have an important effect on its antiproliferative activity. Compared with 145f, the activity of compounds 145ae with the electron-donating group of A ring is higher than that of unsubstituted A ring. However, compounds 145g and 145i and the compound with an electron-withdrawing group of A ring inhibit proliferation more actively than the compound 145f. In addition, the position of the substituent (R) is also important. When the substituent (R) is located at the 3 position of the chalcone group (A ring), the inhibitory activity of the compound is lower than when the substituent (R1) is located at the 3 position of the A ring (compound 145b vs. 145c, 145g vs. 145i, and 145h vs. 145i). However, compound 145e with the 3,4,5-triOCH substituent of the chalcone group (A ring) showed better activity. The relationship between the electron-donating group and an electron-withdrawing group of the chalcone group (A ring) and the inhibition of MGC-803 cells is 3,4,5-triOCH3 > 3,4-diOCH3 > 4-CH3 > 4-bromo > 4-OCH3 > 3-OCH3 > 3-Br > H > 4-Cl > 3-Cl. Next, the impact of R2 is further explored. The results showed that the inhibitory activity of compounds 146af decreased when the H group was substituted with CH3 or CH3CH2 substituents (compounds 146a vs. 145f, 146b vs. 145a, 146c vs. 145d, 146d vs. 145g, 146e vs. 145e, and 146f vs. 145e), indicating that the R2 substituent does not increase the inhibitory potency. The in vitro antiproliferative activities of novel target compounds 145ai and 146af were evaluated against the human cell lines MGC-803 (gastric cancer), HCT-116 (colon cancer), and MCF-7 (breast cancer), with 5-fluorouracil (5-Fu) as a positive control. The results showed that most quinoline–chalcone derivatives have strong antiproliferative activities against the MGC-803HCT-116 and MCF-7 cells, with IC50 values < 20uM. Among them, compound 145e has the greatest inhibitory effect on MGC803, HCT-116, and MCF-7 cells, with IC50 values of 1.35.34 and 5.21 μM, respectively, which is much lower than that of 5-Fu (IC50 value = 6.22 μm) of 0.4 μM, 5.21 μM, and 11.1 μM, respectively, indicating that compound 145e has an inhibitory effect on the activity of three tumor cells. In addition, the MGC-803 cells are more sensitive to most compounds than the HCT-116 and MCF-7 cells. Therefore, according to the structure–activity relationship of the antiproliferative activity of MGC803 cells, the type and position of the substituent (R1) on the chalcone group (A ring) were related to its antiproliferative activity.
Thorat and colleagues [105] synthesized the 6-amino flavonoid quinoline derivative 147 (Figure 25) (Table 10) and evaluated the in vitro antiproliferative efficacy of compound 147 against MCF-7 and human A-549. Doxorubicin was used as a positive control. Compound 147 showed satisfactory anticancer activity against MCF-7, with 44.76% inhibition against this cancer cell line. Compound 147 also showed acceptable antiproliferative activity against A-549, exhibiting the cell at 44.26% under a concentration of 10 μM.
Jyothi and colleagues [106] synthesized coumarin quinoline derivatives 148ac (Figure 25) (Table 10) and evaluated their activities against the human-derived cancer cells ACHN, A375, SIHA, Skov3, EAC, and NIH3T3. However, compounds 148ac did not demonstrate any anticancer activity.
Herrmann and colleagues [107] synthesized artemisinin-quinoline derivatives 149153 (Figure 25) (Table 10) and analyzed their inhibitory activity in vitro against leukemia cell lines CCRF-CEM, RPMI-8226, K562, HL-60, and MOLT 4. The data showed that artemisinins 149151 and synthetic peroxo-quinolines 152 and 153 were the most active compounds against the K562 leukemia cell line in vitro, with IC50 values of 37.3 and 83.0 μM, respectively.
Figure 25. The chemical structures of anticancer compounds 144153.
Figure 25. The chemical structures of anticancer compounds 144153.
Molecules 28 06478 g025
Table 10. Quinoline derivatives with anticancer activity.
Table 10. Quinoline derivatives with anticancer activity.
Compd.Activity TargetOrigin Ref
43Anti- TK10
TGI = 18.5 μM
Anti- UACC62
TGI = 17.43 μM
Anti- MCF7
TGI = 2.92 μM
-synthetic[39]
44--synthetic[39]
71aDetermine growth inhibitory activity = 85%-synthetic[59]
71bDetermine growth inhibitory activity = 77%-synthetic[59]
77Anti-melanoma
GI50 = 0.134 μM
-synthetic[63]
78aAnti- Ae549
IC50 = 15.4 μM
Anti- A375 IC50 = 14.5 μM
Anti- MCF-7
IC50 = 13.8 μM
Anti- HT-29
IC50 = 12.3 μM
Anti- ACHN
IC50 = 10.8 μM
DNA topoisomerase-IIasynthetic[64]
78bAnti- Ae549
IC50 = 13.4 μM
Anti- A375 IC50 = 7.7 μM
Anti- MCF-7
IC50 = 11.2 μM
Anti- HT-29
IC50 = 7.75 μM
Anti- ACHN
IC50 = 15.7 μM
DNA topoisomerase-IIasynthetic[64]
79aAnti- Ae549
IC50 = 10.6 μM
Anti- A375 IC50 = 10.3 μM
Anti- MCF-7
IC50 = 8.6 μM
Anti- HT-29
IC50 = 11.8 μM
Anti- ACHN
IC50 = 10.7 μM
DNA topoisomerase-IIasynthetic[64]
79bAnti- Ae549
IC50 = 7.7 μM
Anti- A375 IC50 = 6.8 μM
Anti- MCF-7
IC50 = 2.2 μM
Anti- HT-29
IC50 = 8.9 μM
Anti- ACHN
IC50 = 9.46 μM
DNA topoisomerase-IIasynthetic[64]
80Anti- angiogenesis
inhibition rate ranged between 50 and 60%
-synthetic[65]
81Anti-A549
GI50 = 2.51 μM
Erk1/2 signaling pathwaysynthetic[66]
82Anti-HepG2
IC50 = 16.03 μM
Anti-HeLa
IC50 = 0.60 μM
Anti-A549
IC50 = 10.05 μM
Anti-BGC-823
IC50 = 17.41 μM
Anti-HL-7702
IC50 = 41.77 μM
Caspase-3synthetic[67]
83Anti-HepG2
IC50 = 9.18 μM
Anti-HeLa
IC50 = 20.53 μM
Anti-A549
IC50 = 19.20 μM
Anti-BGC-823
IC50 = 28.81 μM
Anti-HL-7702
IC50 = 20.09 μM
Caspase-3synthetic[67]
84Anti-HL-60
IC50 = 0.1 μM
Anti-MCF-7
IC50 = 0.1 μM
Anti-T-47D
IC50 = 0.1 μM
Anti-LNCaP
IC50 = 2.0 μM
Anti-WEHI-3
IC50 = 1.1 μM
-synthetic[68]
85Anti-SGC 7901 IC50 = 1 μMTopoisomerase IIsynthetic[69]
86Anti-MCF-7
IC50 = 181.478 μM
Anti-MCF-7/ADR
IC50 = 96.523 μM
Anti-SW1990
IC50 = 111.837 μM
Anti-SMMC-7721
IC50 = 75.546 μM
-synthetic[70]
87Anti-HepG2
IC50 = 8.40 μM
Anti-A431
IC50 = 11.56 μM
Anti-A549
IC50 = 4.33 μM
Anti-MCF 7
IC50 = 5.99 μM
Anti-HCT 116
IC50 = 3.48 μM
-synthetic[71]
88Anti-MDA-MB 468
IC50 = 0.12 μM
Microtubulesynthetic[72]
89Anti-SKOV3 IC50 = 12.8 μMoxygen species (ROS)synthetic[73]
90Anti-Bel-7404
IC50 = 28.7 µmol/L
Anti-SGC-7901
IC50 = 17.9 µmol/L
-synthetic[74]
91Anti-HT-29
IC50 = 14.1 μmol/L
Anti-HeLa
IC50 = 9.1 μmol/L
Anti-SGC-7901
IC50 = 8.2 μmol/L
-synthetic[75]
92aAnti-Hela
30 μM IC50 = 59.5 μM
Caspase-3synthetic[76]
92bAnti-T47D
30 μM IC50 = 48.1 μM
Caspase-3synthetic[76]
92cAnti-T47D
30 μM IC50 = 63.0 μM
Caspase-3synthetic[76]
93aAnti- MCF7
30 μM IC50 = 31.5 μM
Caspase-3synthetic[76]
93bAnti-SiHa
10 μM IC50 = 24.7 μM
Caspase-3synthetic[76]
93cAnti-MCF7
30 μM IC50 = 35.6 μM
Caspase-3synthetic[76]
93dAnti- SiHa
30 μM IC50 = 25.6 μM
Caspase-3synthetic[76]
93eAnti-Hela
30 μM IC50 = 96.6 μM
Caspase-3synthetic[76]
93fAnti- Hela
30 μM IC50 = 89.4 μM
Caspase-3synthetic[76]
94aAnti-MCF7
30 μM IC50 = 38.6 μM
Caspase-3synthetic[76]
94bAnti-C33A
30 μM IC50 = 61.0 μM
Caspase-3synthetic[76]
94cAnti-MDA-MB-231
30 μM IC50 = 71.2 μM
Caspase-3synthetic[76]
94d-Caspase-3synthetic[76]
94eAnti-MDA-MB-361
30 μM IC50 = 66.0 μM
Caspase-3synthetic[76]
94fAnti-MDA-MB-361
30 μM IC50 = 68.2 μM
Caspase-3synthetic[76]
94gAnti-MCF7
30 μM IC50 = 62.9 μM
Caspase-3synthetic[76]
94h-Caspase-3synthetic[76]
94i-Caspase-3synthetic[76]
95aAnti-C33A
30 μM IC50 = 93.6 μM
Caspase-3synthetic[76]
95bAnti- C33A
30 μM IC50 = 71.0 μM
Caspase-3synthetic[76]
95cAnti-MDA-MB-361
30 μM IC50 = 80.4 μM
Caspase-3synthetic[76]
95dAnti-C33A
30 μM IC50 = 73.1 μM
Caspase-3synthetic[76]
95eAnti-C33A
30 μM IC50 = 86.4 μM
Caspase-3synthetic[76]
95fAnti-C33A
30 μM IC50 = 86.9 μM
Caspase-3synthetic[76]
95gAnti-C33A
30 μM IC50 = 74.7 μM
Caspase-3synthetic[76]
95hAnti-MDA-MB-361
30 μM IC50 = 75.2 μM
Caspase-3synthetic[76]
96Assembly of purified tubulin IC50 = 1.6 μMTubulinsynthetic[77]
97aAnti-NIH 3T3
EC50 = 0.9 μM
-synthetic[78]
97bAnti-NIH 3T3
EC50 = 2.2 μM
-synthetic[78]
97cAnti-A2780
EC50 = 2.0 μM
-synthetic[78]
97dAnti-A2780
EC50 = 3.5 μM
-synthetic[78]
97eAnti-A2780
EC50 = 0.7 μM
-synthetic[78]
97fAnti-518A2
EC50 = 2.0 μM
-synthetic[78]
97gAnti-NIH 3T3
EC50 = 0.6 μM
-synthetic[78]
97hAnti-NIH 3T3
EC50 = 0.9 μM
-synthetic[78]
97iAnti- NIH 3T3
EC50 = 0.7 μM
-synthetic[78]
97jAnti- MCF7
EC50 = 1.6 μM
-synthetic[78]
97kAnti-A2780
EC50 = 5.1 μM
-synthetic[78]
97lAnti-A2780
EC50 = 6.7 μM
-synthetic[78]
98Anti-A2780
EC50 = 1.2 μM
-synthetic[78]
99aAnti-A2780
IC50 = 8.04 μM
Tubulinsynthetic[79]
99bAnti-MCF-7/MX
IC50 = 21.48 μM
Tubulinsynthetic[79]
99cAnti-A2780
IC50 = 9.19 μM
Tubulinsynthetic[79]
99dAnti-A2780
IC50 = 7.98 μM
Tubulinsynthetic[79]
99eAnti- A2780RCIS
IC50 = 8.15 μM
Tubulinsynthetic[79]
100Anti-K562/ADR
IC50 = 0.061 μM
Anti-K562
IC50 = 0.064 μM
MAPKsynthetic[80]
101Anti-K562/ADR
IC50 = 0.177 μM
Anti-K562
IC50 = 0.064 μM
MAPKsynthetic[80]
102Anti-K562/ADR
IC50 = 0.034 μM
Anti-K562
IC50 = 0.022 μM
MAPKsynthetic[80]
103Anti-HL60
IC50 = 8.09 μM
Anti-SGC-7901
IC50 = 73.40 μM
Anti-MCF-7
IC50 = 19.66 μM
Anti-HCT116
IC50 = 14.79 μM
Anti-A549
IC50 = 17.61 μM
Anti- HaCat
IC50 = 11.49 μM
-synthetic[81]
104Anti-Bel-7402
IC50 = 0.96 μM
Anti-K562
IC50 = 1.89 μM
Anti-MGC-803
IC50 = 1.03 μM
Anti-CaEs-17
IC50 = 1.74 μM
-synthetic[62]
105aAnti-HeLa
IC50 = 0.37 μM
-synthetic[82]
105bAnti-HeLa
IC50 = 0.36 μM
-synthetic[82]
105cAnti-HeLa
IC50 = 1.22 μM
-synthetic[82]
105dAnti-MDA-MB-231
IC50 = 0.90 μM
-synthetic[82]
106aAnti-HeLa
IC50 = 19.03 μM
-synthetic[82]
106bAnti-HeLa
IC50 = 25.78 μM
-synthetic[82]
106cAnti-MDA-MB-231
IC50 = 13.34 μM
-synthetic[82]
106dAnti-MDA-MB-231
IC50 = 17.44 μM
-synthetic[82]
106eAnti-HeLa
IC50 = 21.88 μM
-synthetic[82]
106fAnti-HeLa
IC50 = 12.27 μM
-synthetic[82]
106gAnti-HeLa
IC50 = 13.13 μM
-synthetic[82]
106hAnti-HeLa
IC50 = 23.45 μM
-synthetic[82]
106iAnti-HeLa
IC50 = 7.16 μM
-synthetic[82]
106jAnti-HeLa
IC50 = 26.87 μM
-synthetic[82]
106kAnti-HeLa
IC50 = 30.25 μM
-synthetic[82]
106lAnti-HeLa
IC50 = 12.45 μM
-synthetic[82]
107aAnti-MDA-MB-231
IC50 = 22.37 μM
-synthetic[82]
107bAnti-HeLa
IC50 = 32.13 μM
-synthetic[82]
107cAnti-HeLa
IC50 = 7.11 μM
-synthetic[82]
107dAnti-MDA-MB-231
IC50 = 19.39 μM
-synthetic[82]
107eAnti-HeLa
IC50 = 28.12 μM
-synthetic[82]
107fAnti-HeLa
IC50 = 12.31 μM
-synthetic[82]
107gAnti-MDA-MB-231
IC50 = 23.35 μM
-synthetic[82]
107hAnti-MDA-MB-231
IC50 > 40 μM
-synthetic[82]
107iAnti-HeLa
IC50 = 16.29 μM
-synthetic[82]
107jAnti-HeLa
IC50 = 28.29 μM
-synthetic[82]
107kAnti-HeLa
IC50 = 32.25 μM
-synthetic[82]
107lAnti-HeLa
IC50 = 12.09 μM
-synthetic[82]
108Anti-HeLa
IC50 = 11.2 μM/L
Anti-HT-29
IC50 = 21.3 μM/L
Anti-Bel 7404
IC50 = 28.9 μM/L
Anti-SGC 7901
IC50 = 10.3 μM/L
-synthetic[83]
109--synthetic[83]
110-Cysteine protease falcipain-2synthetic[84]
111Anti-ERK
IC50 = 0.6 μM
ERK2synthetic[85]
112Anti-ERK
IC50 = 0.16 μM
ERK2synthetic[85]
113Anti-PC-3
IC50 = 3.12 μM
Anti-DU-145
IC50 = 3.99 μM
Anti-NCI-H460
IC50 = 3.96 μM
Anti-4 T1
IC50 = 1.81 μM
-synthetic[86]
114aAnti-A2780
IC50 = 25 μg/mL
-synthetic[87]
114bAnti-A2780
IC50 = 62 μg/mL
-synthetic[87]
115Anti-MCF-7
GI50 = 38.3 Μm
-synthetic[88]
116Anti-MCF-7
GI50 = 10.5 μM
-synthetic[88]
117Anti-MCF-7
IC50 = 0.41 μM
p53-MDM2synthetic[89]
118Anti-MCF-7
IC50 = 0.89 μM
p53-MDM2synthetic[89]
119Anti-MRC-5
IC50 = 70.8 μM
Tubulinsynthetic[90]
120Anti-MCF-7
IC50 = 0.6 μM
Tubulinsynthetic[90]
121Anti-A549
IC50 = 2.3 μM
Tubulinsynthetic[90]
122Anti-MCF-7
IC50 = 1.0 μM
Tubulinsynthetic[90]
123alow antitumor activityCaspase-3synthetic[91]
123blow antitumor activityCaspase-3synthetic[91]
123clow antitumor activityCaspase-3synthetic[91]
123dlow antitumor activityCaspase-3synthetic[91]
124aAnti-HeLa
IC50 = 0.37 μM
Ras/Raf/MEK/ERKsynthetic[92]
124bAnti-HeLa
IC50 = 0.36 μM
Ras/Raf/MEK/ERKsynthetic[92]
124cAnti-MDA-MB-231
IC50 = 0.90 μM
Ras/Raf/MEK/ERKsynthetic[92]
124dAnti-HeLa
IC50 = 1.22 μM
Ras/Raf/MEK/ERKsynthetic[92]
125aAnti-HeLa
IC50 = 1.18 μM
Ras/Raf/MEK/ERKsynthetic[92]
125bAnti-HeLa
IC50 = 0.83 μM
Ras/Raf/MEK/ERKsynthetic[92]
125cAnti-HeLa
IC50 = 0.99 μM
Ras/Raf/MEK/ERKsynthetic[92]
125dAnti-HeLa
IC50 = 0.08 μM
Ras/Raf/MEK/ERKsynthetic[92]
125eAnti-HeLa
IC50 > 50 μM
Ras/Raf/MEK/ERKsynthetic[92]
125fAnti-HeLa
IC50 > 50 μM
Ras/Raf/MEK/ERKsynthetic[92]
125gAnti-HeLa
IC50 > 50 μM
Ras/Raf/MEK/ERKsynthetic[92]
125hAnti-HeLa
IC50 = 46.01 μM
Ras/Raf/MEK/ERKsynthetic[92]
126aAnti-MDA-MB-231
IC50 = 5.32 μM
Ras/Raf/MEK/ERKsynthetic[92]
126bAnti-MDA-MB-231
IC50 = 12.25 μM
Ras/Raf/MEK/ERKsynthetic[92]
126cAnti-MDA-MB-231
IC50 = 13.17 μM
Ras/Raf/MEK/ERKsynthetic[92]
126dAnti-HeLa
IC50 = 4.28 μM
Ras/Raf/MEK/ERKsynthetic[92]
126eAnti-HeLa
IC50 > 50 μM
Ras/Raf/MEK/ERKsynthetic[92]
126fAnti-HeLa
IC50 = 30.94 μM
Ras/Raf/MEK/ERKsynthetic[92]
126gAnti-HeLa
IC50 > 50 μM
Ras/Raf/MEK/ERKsynthetic[92]
126hAnti-HeLa
IC50 > 50 μM
Ras/Raf/MEK/ERKsynthetic[92]
127aAnti-MDA-MB-231
IC50 = 18.75 μM
Ras/Raf/MEK/ERKsynthetic[92]
127bAnti-HeLa
IC50 = 12.82 μM
Ras/Raf/MEK/ERKsynthetic[92]
127cAnti-MDA-MB-231
IC50 = 31.57 μM
Ras/Raf/MEK/ERKsynthetic[92]
127dAnti-HeLa
IC50 = 10.92 μM
Ras/Raf/MEK/ERKsynthetic[92]
127eAnti-HeLa
IC50 > 50 μM
Ras/Raf/MEK/ERKsynthetic[92]
127fAnti-HeLa
IC50 > 50 μM
Ras/Raf/MEK/ERKsynthetic[92]
127gAnti-HeLa
IC50 > 50 μM
Ras/Raf/MEK/ERKsynthetic[92]
127hAnti-HeLa
IC50 > 50 μM
Ras/Raf/MEK/ERKsynthetic[92]
128aAnti-A549
IC50 = 15.21 μM
-synthetic[93]
128bAnti-A549
IC50 = 12.65 μM
-synthetic[93]
128cAnti-A549
IC50 = 13.34 μM
-synthetic[93]
128dAnti-A549
IC50 = 12.23 μM
-synthetic[93]
128eAnti-A549
IC50 = 8.34 μM
-synthetic[93]
128fAnti-A549
IC50 = 5.34 μM
-synthetic[93]
128gAnti-H1975
IC50 = 12.95 μM
-synthetic[93]
128hAnti-H1975
IC50 = 14.31 μM
-synthetic[93]
128iAnti-A549
IC50 = 10.28 μM
-synthetic[93]
128jAnti-A549
IC50 = 9.01 μM
-synthetic[93]
128kAnti-A549
IC50 = 23.91 μM
-synthetic[93]
128lAnti-A431
IC50 = 12.56 μM
-synthetic[93]
129aAnti-K562
IC50 = 0.850 μM
Tubulinsynthetic[94]
129bAnti-K562
IC50 = 0.011 μM
Tubulinsynthetic[94]
129cAnti-K562
IC50 = 0.127 μM
Tubulinsynthetic[94]
129dAnti-K562
IC50 = 0.009 μM
Tubulinsynthetic[94]
129eAnti-K562
IC50 = 0.108 μM
Tubulinsynthetic[94]
129fAnti-K562
IC50 = 1.055 μM
Tubulinsynthetic[94]
129gAnti-K562
IC50 = 0.069 μM
Tubulinsynthetic[94]
129hAnti-K562
IC50 = 0.563 μM
Tubulinsynthetic[94]
129iAnti-K562
IC50 > 1 μM
Tubulinsynthetic[94]
129jAnti-K562
IC50 > 1 μM
Tubulinsynthetic[94]
129kAnti-K562
IC50 > 1 μM
Tubulinsynthetic[94]
129lAnti-K562
IC50 = 0.346 μM
Tubulinsynthetic[94]
129mAnti-K562
IC50 = 0.074 μM
Tubulinsynthetic[94]
129nAnti-K562
IC50 > 1 μM
Tubulinsynthetic[94]
129oAnti-K562
IC50 > 1 μM
Tubulinsynthetic[94]
130aAnti-K562
IC50 = 0.040 μM
Tubulinsynthetic[94]
130bAnti-K562
IC50 = 0.026 μM
Tubulinsynthetic[94]
130cAnti-K562
IC50 = 0.015 μM
Tubulinsynthetic[94]
130dAnti-K562
IC50 = 1.239 μM
Tubulinsynthetic[94]
131Anti-HCT116
IC50 = 3.31 μM
Anti-U87-MG
IC50 = 1.47 μM
Anti-HepG2
IC50 = 3.66 μM
Anti-BGC823
IC50 = 1.77 μM
Anti-PC9
IC50 = 3.12 μM
NF-κBsynthetic[95]
132Anti-HepG2
IC50 = 14.3 μM
-synthetic[96]
133Anti-HepG2
IC50 = 9.2 μM
-synthetic[96]
134aAnti-HepG2
IC50 = 14.3 μM
Hsp90Nsynthetic[97]
134bAnti-HepG2
IC50 = 9.2 μM
Hsp90Nsynthetic[97]
134cAnti-HepG2
IC50 = 10.9 μM
Hsp90Nsynthetic[97]
134dAnti-HepG2
IC50 = 13.1 μM
Hsp90Nsynthetic[97]
134eAnti-HepG2
IC50 = 6.4 μM
Hsp90Nsynthetic[97]
134fAnti-Hela
IC50 = 6.9 μM
Hsp90Nsynthetic[97]
134gAnti-HepG2
IC50 = 16.1 μM
Hsp90Nsynthetic[97]
135Anti-HCT-116
IC50 = 0.16 μM
HER1, HER2, proteasome, and hTS appear as promising targets for these compoundssynthetic[98]
136aAnti-HCT-116
IC50 = 1.55 μM
HER1, HER2, proteasome, and hTS appear as promising targets for these compoundssynthetic[98]
136bAnti-RPMI-8226
IC50 = 1.29 μM
HER1, HER2, proteasome, and hTS appear as promising targets for these compoundssynthetic[98]
136cAnti-MDA-MB-435
IC50 = 0.30 μM
HER1, HER2, proteasome, and hTS appear as promising targets for these compoundssynthetic[98]
136dAnti-HCT-116
IC50 = 0.26 μM
HER1, HER2, proteasome, and hTS appear as promising targets for these compoundssynthetic[98]
136e-HER1, HER2, proteasome, and hTS appear as promising targets for these compoundssynthetic[98]
136f-HER1, HER2, proteasome, and hTS appear as promising targets for these compoundssynthetic[98]
136g-HER1, HER2, proteasome, and hTS appear as promising targets for these compoundssynthetic[98]
137aAnti-A549
IC50 = 6.1 μM
EGFR, BRAFV600Esynthetic[99]
137bAnti-Panc-1
IC50 = 2.9 μM
EGFR, BRAFV600Esynthetic[99]
138aAnti-MCF-7
IC50 = 23.1 μM
EGFR, BRAFV600Esynthetic[99]
138bAnti-MCF-7
IC50 = 29.5 μM
EGFR, BRAFV600Esynthetic[99]
139aAnti-MCF-7
IC50 = 3.2 μM
EGFR, BRAFV600Esynthetic[99]
139bAnti-Panc-1
IC50 = 4.5 μM
EGFR, BRAFV600Esynthetic[99]
140Anti-HCT116
IC50 = 2.11 μM
Anti-U87MG
IC50 = 2.47 μM
Anti-HepG2
IC50 = 4.71 μM
Anti-BGC823
IC50 = 5.23 μM
Anti-PC9
IC50 = 3.00 μM
NF-κB, STAT3synthetic[100]
141aAnti-A549
IC50 = 19.24 μM
Anti-NCI-H460
IC50 = 24.61 μM
synthetic[101]
141bAnti-A549
IC50 = 18.21 μM
STAT3synthetic[101]
141c-STAT3synthetic[101]
142aAnti-A549
IC50 = 13.78 μM
Anti-NCI-H460
IC50 = 17.13 μM
STAT3synthetic[101]
142bAnti-A549
IC50 = 14.34 μM
Anti-NCI-H460
IC50 = 18.32 μM
STAT3synthetic[101]
142cAnti-A549
IC50 = 12.94 μM
STAT3synthetic[101]
142dAnti-A549
IC50 = 13.05 μM
STAT3synthetic[101]
143Anti-HIF-1a
IC50 = 0.05 μM
HIF-1asynthetic[102]
144Anti-CYP1A1
IC50 = 117.6 μM
Anti-CYP1B1
IC50 = 1.0 μM
Anti-CYP1A2
IC50 > 1000 μM
CYP1B1synthetic[103]
145aAnti-MGC-803
IC50 = 1.86 μM
Caspase3/9, cleaved-PARPsynthetic[104]
145bAnti-MGC-803
IC50 = 2.39 μM
Caspase3/9, cleaved-PARPsynthetic[104]
145cAnti-MGC-803
IC50 = 2.63 μM
Caspase3/9, cleaved-PARPsynthetic[104]
145dAnti-MGC-803
IC50 = 1.62 μM
Caspase3/9, cleaved-PARPsynthetic[104]
145eAnti-MGC-803
IC50 = 1.38 μM
Caspase3/9, cleaved-PARPsynthetic[104]
145fAnti-MGC-803
IC50 = 3.24 μM
Caspase3/9, cleaved-PARPsynthetic[104]
145gAnti-MGC-803
IC50 = 3.82 μM
Caspase3/9, cleaved-PARPsynthetic[104]
145hAnti-MGC-803
IC50 = 2.28 μM
Caspase3/9, cleaved-PARPsynthetic[104]
145iAnti-MGC-803
IC50 = 4.25 μM
Caspase3/9, cleaved-PARPsynthetic[104]
145jAnti-MGC-803
IC50 = 3.22 μM
Caspase3/9, cleaved-PARPsynthetic[104]
146aAnti-MGC-803
IC50 = 8.26 μM
Caspase3/9, cleaved-PARPsynthetic[104]
146bAnti-MGC-803
IC50 = 4.63 μM
Caspase3/9, cleaved-PARPsynthetic[104]
146cAnti-MGC-803
IC50 = 4.54 μM
Caspase3/9, cleaved-PARPsynthetic[104]
146dAnti-MGC-803
IC50 = 8.25 μM
Caspase3/9, cleaved-PARPsynthetic[104]
146eAnti-MGC-803
IC50 = 3.73 μM
Caspase3/9, cleaved-PARPsynthetic[104]
146fAnti-MGC-803
IC50 = 10.21 μM
Caspase3/9, cleaved-PARPsynthetic[104]
14710 μM
Anti-MCF-7 Inhibition = 44.76%
Anti-A549 Inhibition = 44.26%
Topoisomerase IIsynthetic[105]
148aAnti-EAC
IC50 = 62.25 μM
Vascular Endothelial Growth Factor (VEGF)synthetic[106]
148bAnti-A375
IC50 = 69.52 μM
Vascular Endothelial Growth Factor (VEGF)synthetic[106]
148cAnti-ACHN
IC50 = 62.65 μM
Vascular Endothelial Growth Factor (VEGF)synthetic[106]
149Anti-MOLT-4
EC50 = 33.4 μM
-synthetic[107]
150Anti-RPMI-8226
EC50 = 23.3 μM
-synthetic[107]
151Anti-MOLT-4
EC50 = 33.5 μM
-synthetic[107]
152Anti-RPMI-8226
EC50 = 16.3 μM
-synthetic[107]
153Anti-MOLT-4
EC50 = 24.4 μM
-synthetic[107]
Summary: Among the recognized anticancer drugs, such as camptothecin and cabozantinib, quinoline scaffolds are contained. Therefore, quinoline has been widely studied and is considered to be an efficient chemical structure in antitumor research. At present, among all the biological activities related to quinoline, anticancer activity has been reported the most. Anticancer drugs containing quinoline structure are divided into the following three main structural categories: non-fused quinoline (such as cabozantinib); fusion quinoline compounds (such as camptothecin); metal complexes with quinoline or phenanthroline ligands.
The compounds containing quinoline structure antitumor activity are mainly the first two, and most of them can show strong antitumor activity. For example, for HeLa cells, 105a and 105b had the strongest activity (IC50 values were 0.37 and 0.36 μM, respectively). For MCF-7 cells, 120 had the strongest activity (IC50 values were 0.60 μM). For A549 cells, 121 had the strongest activity (IC50 values were 2.3 μM). In addition, the mechanism of action is mainly aimed at the mechanism of cell division, or the induction of apoptosis. However, the research on the mechanism of action is not deep enough and there is still a lack of in vivo research.

4. Conclusions

Natural products have always been a rich source of effective drugs and will continue to be an important source of new pharmacological lead drugs. However, natural physiologically active chemicals may have adverse pharmacological properties that limit their use, such as cytotoxicity, excessive lipophilicity, or poor oral absorption. Another major obstacle to the use of natural products in drug research is the inability to obtain these derivatives from sustainable sources. The quinoline ring has many medicinal values, and it is becoming more and more popular as a multifunctional drug chemical scaffold. Due to the wide range of biological functions of quinoline molecules, natural compounds with structural changes are often used as quinoline molecules and developed into key heterocyclic scaffolds to help discover new drugs with new structures and new processes. We have been looking for new natural product quinoline derivatives that can produce potential biological activity. This literature review shows that quinoline scaffolds have considerable biological relevance in anti-osteoporosis, anti-virus, anti-diabetes, anti-inflammation, anti-thrombosis, anti-parasitic, antimalarial, antibacterial, and anticancer studies, which leads to the emergence of many efficient quinoline compounds in many therapeutic fields. Among them, anti-malaria and antitumor are the two most popular research fields. Observing quinoline-based antimalarial drugs, it can be seen that most of them still have traditional pharmacodynamic units, which also exist in quinoline antimalarial drugs, such as chloroquine, amodiaquine, and primaquine. In cancer research, there are many types of tumors, and the occurrence and progression of tumors are also complex. There are many cancer-related targets, and the chemical drug space exploration around quinoline antitumor drugs has great diversity, and it is easier to develop antitumor drugs with strong activity and small side effects. In short, in the future research of medicinal chemistry, quinoline drugs will continue to be used as superior scaffolds for the development of derivatives with high biological activity, among which antimalarial and antitumor development are of the greatest value. This review provides a comprehensive data resource of natural product quinoline derivatives for pharmaceutical chemists engaged in drug design and development, which is helpful for pharmaceutical companies to carry out richer and more organized drug discovery actions in experimental research so that the scientific community can reasonably design and develop various optimized, new, and targeted quinoline derivatives.

Author Contributions

Conceptualization and review methodologies—Y.-Q.Z. and X.L. Original draft writing—Y.-Q.Z. and T.L. Editing—All authors. Figure creation—Y.-Q.Z. Revision—T.L. and Z.-S.Q. All authors have read and agreed to the published version of the manuscript.

Funding

This work was financially supported by the Educational Department of Liaoning Province (No. LJKMZ20221801), Doctoral Research Foundation of Shenyang Medical College (No. 20205041), Higher Education Discipline Innovation Project (D18012), National Natural Science Foundation of China (No. 81960626, 82060628, 82204310), Key Projects of Jilin Province Science and Technology Development Plan (No. 20200404130YY), Jilin Scientific and Technological Development Program (No. YDZJ202301ZYTS440, YDZJ202301ZYTS143), Education Department Project of Jilin (JJKH20220559KJ, JJKH20220563KJ).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

3D7strainPlasmodium falciparum
ACHEAcetylcholinesterase
ACICoccidiosis inhibition rate of Eimeria tenella
ALPAlkaline phosphatase
ALTAlanine aminotransferase
ASTAspartate aminotransferase
BCHECholinesterase
BMP-2Bone morphogenetic protein-2
DMSODimethyl sulfoxide
FMRInsecticidal inhibition rate
FRDFumarate reductase
FXaActivation of X factor
HDL-CHigh-density lipoprotein cholesterol
IL-6Interleukin-6
KM miceKunming mice
LPSLipopolysaccharides
MRThe mite inhibition rate
OAOleanolic acid
OCLsOsteoclast-like multinucleated cells
OCNOsteoblast secretory protein
PC-3 cellsHuman prostate cancer cell line
RUNX-2Runt-related transcription factor-2
SNB-19Human glioma adherent cell line
THP1 cellsHuman monocytic leukemia
U-937 cellsCell line exhibiting monocyte morphology

References

  1. Newman, D.J.; Cragg, G.M. Natural Products as Sources of New Drugs over the Nearly Four Decades from January 1981 to September 2019. J. Nat. Prod. 2020, 83, 770–803. [Google Scholar] [CrossRef]
  2. Patel, R.V.; Park, S.W. Access to a new class of biologically active quinoline based 1,2,4-triazoles. Eur. J. Med. Chem. 2014, 71, 24–30. [Google Scholar] [CrossRef] [PubMed]
  3. Narwal, S.; Kumar, S.; Verma, P.K. Synthesis and therapeutic potential of quinoline derivatives. Res. Chem. Inter. 2016, 43, 2765–2798. [Google Scholar] [CrossRef]
  4. Feng, L.; Lv, K.; Liu, M.; Wang, S.; Zhao, J.; You, X.; Li, S.; Cao, J.; Guo, H. Synthesis and in vitro antibacterial activity of gemifloxacin derivatives containing a substituted benzyloxime moiety. Eur. J. Med. Chem. 2012, 55, 125–136. [Google Scholar] [CrossRef]
  5. Musiol, R.; Serda, M.; Hensel-Bielowka, S.; Polanski, J. Quinoline-Based Antifungals. Curr. Med. Chem. 2010, 17, 1960–1973. [Google Scholar] [CrossRef] [PubMed]
  6. Medapi, B.; Renuka, J.; Saxena, S.; Sridevi, J.P.; Medishetti, R.; Kulkarni, P.; Yogeeswari, P.; Sriram, D. Design and synthesis of novel quinoline-aminopiperidine hybrid analogues as Mycobacterium tuberculosis DNA gyraseB inhibitors. Bioorg. Med. Chem. 2015, 23, 2062–2078. [Google Scholar] [CrossRef]
  7. Ma, X.; Zhou, W.; Brun, R. Synthesis, in vitro antitrypanosomal and antibacterial activity of phenoxy, phenylthio or benzyloxy substituted quinolones. Bioorg. Med. Chem. Lett. 2009, 19, 986–989. [Google Scholar] [CrossRef]
  8. Rossiter, S.; Peron, J.M.; Whitfield, P.J.; Jones, K. Synthesis and anthelmintic properties of arylquinolines with activity against drug-resistant nematodes. Bioorg. Med. Chem. Lett. 2005, 15, 4806–4808. [Google Scholar] [CrossRef] [PubMed]
  9. Sun, J.; Zhu, H.; Yang, Z.M.; Zhu, H.L. Synthesis, molecular modeling and biological evaluation of 2-aminomethyl-5-(quinolin-2-yl)-1,3,4-oxadiazole-2(3H)-thione quinolone derivatives as novel anticancer agent. Eur. J. Med. Chem. 2013, 60, 23–28. [Google Scholar] [CrossRef] [PubMed]
  10. Majerz-Maniecka, K.; Musiol, R.; Skorska-Stania, A.; Tabak, D.; Mazur, P.; Oleksyn, B.J.; Polanski, J. X-ray and molecular modelling in fragment-based design of three small quinoline scaffolds for HIV integrase inhibitors. Bioorg. Med. Chem. 2011, 19, 1606–1612. [Google Scholar] [CrossRef] [PubMed]
  11. Rano, T.A.; Sieber-McMaster, E.; Pelton, P.D.; Yang, M.; Demarest, K.T.; Kuo, G.H. Design and synthesis of potent inhibitors of cholesteryl ester transfer protein (CETP) exploiting a 1,2,3,4-tetrahydroquinoline platform. Bioorg. Med. Chem. Lett. 2009, 19, 2456–2460. [Google Scholar] [CrossRef] [PubMed]
  12. Roma, G.; Grossi, G.; Di Braccio, M.; Piras, D.; Ballabeni, V.; Tognolini, M.; Bertoni, S.; Barocelli, E. 1,8-Naphthyridines VII. New substituted 5-amino[1,2,4]triazolo[4,3-a][1,8]naphthyridine-6-carboxamides and their isosteric analogues, exhibiting notable anti-inflammatory and/or analgesic activities, but no acute gastrolesivity. Eur. J. Med. Chem. 2008, 43, 1665–1680. [Google Scholar] [CrossRef]
  13. Mantoani, S.P.; Chierrito, T.P.; Vilela, A.F.; Cardoso, C.L.; Martinez, A.; Carvalho, I. Novel Triazole-Quinoline Derivatives as Selective Dual Binding Site Acetylcholinesterase Inhibitors. Molecules 2016, 21, 193. [Google Scholar] [CrossRef] [PubMed]
  14. Duarte, Y.; Fonseca, A.; Gutiérrez, M.; Adasme-Carreño, F.; Muñoz-Gutierrez, C.; Alzate-Morales, J.; Santana, L.; Uriarte, E.; Álvarez, R.; Matos, M.J. Novel Coumarin-Quinoline Hybrids: Design of Multitarget Compounds for Alzheimer’s Disease. Chemistry 2019, 4, 551–558. [Google Scholar] [CrossRef]
  15. Wang, X.Q.; Xia, C.L.; Chen, S.B.; Tan, J.H.; Ou, T.M.; Huang, S.L.; Li, D.; Gu, L.Q.; Huang, Z.S. Design, synthesis, and biological evaluation of 2-arylethenylquinoline derivatives as multifunctional agents for the treatment of Alzheimer’s disease. Eur. J. Med. Chem. 2015, 89, 349–361. [Google Scholar] [CrossRef] [PubMed]
  16. Tintas, M.L.; Foucout, L.; Petit, S.; Oudeyer, S.; Gourand, F.; Barre, L.; Papamicael, C.; Levacher, V. New developments in redox chemical delivery systems by means of 1,4-dihydroquinoline-based targetor: Application to galantamine delivery to the brain. Eur. J. Med. Chem. 2014, 81, 218–226. [Google Scholar] [CrossRef] [PubMed]
  17. Benchekroun, M.; Pachon-Angona, I.; Luzet, V.; Martin, H.; Oset-Gasque, M.J.; Marco-Contelles, J.; Ismaili, L. Synthesis, antioxidant and Abeta anti-aggregation properties of new ferulic, caffeic and lipoic acid derivatives obtained by the Ugi four-component reaction. Bioorg. Chem. 2019, 85, 221–228. [Google Scholar] [CrossRef] [PubMed]
  18. Shah, M.S.; Najam-Ul-Haq, M.; Shah, H.S.; Farooq Rizvi, S.U.; Iqbal, J. Quinoline containing chalcone derivatives as cholinesterase inhibitors and their in silico modeling studies. Comput. Biol. Chem. 2018, 76, 310–317. [Google Scholar] [CrossRef] [PubMed]
  19. Li, J.F.; Zhao, Y.; Cai, M.M.; Li, X.F.; Li, J.X. Synthesis and evaluation of a novel series of heterocyclic oleanolic acid derivatives with anti-osteoclast formation activity. Eur. J. Med. Chem. 2009, 44, 2796–2806. [Google Scholar] [CrossRef] [PubMed]
  20. Maurya, S.W.; Dev, K.; Singh, K.B.; Rai, R.; Siddiqui, I.R.; Singh, D.; Maurya, R. Synthesis and biological evaluation of heterocyclic analogues of pregnenolone as novel anti-osteoporotic agents. Bioorg. Med. Chem. Lett. 2017, 27, 1390–1396. [Google Scholar] [CrossRef]
  21. Li, F.; Lee, E.M.; Sun, X.; Wang, D.; Tang, H.; Zhou, G.C. Design, synthesis and discovery of andrographolide derivatives against Zika virus infection. Eur. J. Med. Chem. 2020, 187, 111925. [Google Scholar] [CrossRef]
  22. Baltina, L.A.; Lai, H.C.; Liu, Y.C.; Huang, S.H.; Hour, M.J.; Baltina, L.A.; Nugumanov, T.R.; Borisevich, S.S.; Khalilov, L.M.; Petrova, S.F.; et al. Glycyrrhetinic acid derivatives as Zika virus inhibitors: Synthesis and antiviral activity in vitro. Bioorg. Med. Chem. 2021, 41, 116204. [Google Scholar] [CrossRef] [PubMed]
  23. Wang, L.J.; Geng, C.A.; Ma, Y.B.; Huang, X.Y.; Luo, J.; Chen, H.; Zhang, X.M.; Chen, J.J. Synthesis, biological evaluation and structure-activity relationships of glycyrrhetinic acid derivatives as novel anti-hepatitis B virus agents. Bioorg. Med. Chem. Lett. 2012, 22, 3473–3479. [Google Scholar] [CrossRef] [PubMed]
  24. Papi Reddy, K.; Singh, A.B.; Puri, A.; Srivastava, A.K.; Narender, T. Synthesis of novel triterpenoid (lupeol) derivatives and their in vivo antihyperglycemic and antidyslipidemic activity. Bioorg. Med. Chem. Lett. 2009, 19, 4463–4466. [Google Scholar] [CrossRef] [PubMed]
  25. Nam, K.Y.; Damodar, K.; Jeon, S.H.; Lee, J.T.; Lee, Y. Design and Synthesis of π-Extended Resveratrol Analogues and In Vitro Antioxidant and Anti-Inflammatory Activity Evaluation. Molecules 2021, 26, 646. [Google Scholar] [CrossRef] [PubMed]
  26. Bian, M.; Zhen, D.; Shen, Q.K.; Du, H.H.; Ma, Q.Q.; Quan, Z.S. Structurally modified glycyrrhetinic acid derivatives as anti-inflammatory agents. Bioorg. Chem. 2021, 107, 104598. [Google Scholar] [CrossRef] [PubMed]
  27. Ma, Q.; Bian, M.; Gong, G.; Bai, C.; Liu, C.; Wei, C.; Quan, Z.S.; Du, H.H. Synthesis and Evaluation of Bakuchiol Derivatives as Potent Anti-inflammatory Agents In Vitro and In Vivo. J. Nat. Prod. 2022, 85, 15–24. [Google Scholar] [CrossRef] [PubMed]
  28. Chen, P.; Zhang, D.; Li, M.; Wu, Q.; Lam, Y.P.Y.; Guo, Y.; Chen, C.; Bai, N.; Malhotra, S.; Li, W.; et al. Discovery of novel, potent, isosteviol-based antithrombotic agents. Eur. J. Med. Chem. 2019, 183, 111722. [Google Scholar] [CrossRef] [PubMed]
  29. Leverrier, A.; Bero, J.; Frederich, M.; Quetin-Leclercq, J.; Palermo, J. Antiparasitic hybrids of Cinchona alkaloids and bile acids. Eur. J. Med. Chem. 2013, 66, 355–363. [Google Scholar] [CrossRef] [PubMed]
  30. Nisha; Kumar, K.; Bhargava, G.; Land, K.M.; Chang, K.H.; Arora, R.; Sen, S.; Kumar, V. N-Propargylated isatin-Mannich mono- and bis-adducts: Synthesis and preliminary analysis of in vitro activity against Tritrichomonas foetus. Eur. J. Med. Chem. 2014, 74, 657–663. [Google Scholar] [CrossRef]
  31. Coa, J.C.; García, E.; Carda, M.; Agut, R.; Vélez, I.D.; Muñoz, J.A.; Yepes, L.M.; Robledo, S.M.; Cardona, W.I. Synthesis, leishmanicidal, trypanocidal and cytotoxic activities of quinoline-chalcone and quinoline-chromone hybrids. Med. Chem. Res. 2017, 26, 1405–1414. [Google Scholar] [CrossRef]
  32. Huang, J.; Lv, M.; Thapa, S.; Xu, H. Synthesis of novel quinolinomatrine derivatives and their insecticidal/acaricidal activities. Bioorg. Med. Chem. Lett. 2018, 28, 1753–1757. [Google Scholar] [CrossRef] [PubMed]
  33. Wang, Z.; Zhou, L.-J.; Wang, Y.-I.; Weng, Y.-B.; He, J.; Nie, K. Synthesis and anticoccidial activities of substituted ethyl 4-hydroxy-11-oxo-11H-chromeno[2,3-g]quinoline-3-carboxylates. J. Chem. Res. 2011, 42, 373–376. [Google Scholar] [CrossRef]
  34. Roussaki, M.; Hall, B.; Lima, S.C.; da Silva, A.C.; Wilkinson, S.; Detsi, A. Synthesis and anti-parasitic activity of a novel quinolinone-chalcone series. Bioorg. Med. Chem. Lett. 2013, 23, 6436–6441. [Google Scholar] [CrossRef] [PubMed]
  35. Pan, L.; Li, X.-Z.; Sun, D.-A.; Jin, H.; Guo, H.-R.; Qin, B. Design and synthesis of novel coumarin analogs and their nematicidal activity against five phytonematodes. Chin. Chem. Lett. 2016, 27, 375–379. [Google Scholar] [CrossRef]
  36. Guo, Y.; Yan, Y.; Yu, X.; Wang, Y.; Zhi, X.Y.; Hu, Y.; Xu, H. Synthesis and insecticidal activity of some novel fraxinellone-based esters. J. Agric. Food Chem. 2012, 60, 7016–7021. [Google Scholar] [CrossRef] [PubMed]
  37. Guo, H.Y.; Jin, C.; Zhang, H.M.; Jin, C.M.; Shen, Q.K.; Quan, Z.S. Synthesis and Biological Evaluation of (+)-Usnic Acid Derivatives as Potential Anti-Toxoplasma gondii Agents. J. Agric. Food Chem. 2019, 67, 9630–9642. [Google Scholar] [CrossRef] [PubMed]
  38. Deng, H.; Huang, X.; Jin, C.; Jin, C.M.; Quan, Z.S. Synthesis, in vitro and in vivo biological evaluation of dihydroartemisinin derivatives with potential anti-Toxoplasma gondii agents. Bioorg. Chem. 2020, 94, 103467. [Google Scholar] [CrossRef]
  39. Lombard, M.C.; N’Da, D.D.; Breytenbach, J.C.; Kolesnikova, N.I.; Tran Van Ba, C.; Wein, S.; Norman, J.; Denti, P.; Vial, H.; Wiesner, L. Antimalarial and anticancer activities of artemisinin-quinoline hybrid-dimers and pharmacokinetic properties in mice. Eur. J. Pharm. Sci. 2012, 47, 834–841. [Google Scholar] [CrossRef] [PubMed]
  40. Raj, R.; Biot, C.; Carrere-Kremer, S.; Kremer, L.; Guerardel, Y.; Gut, J.; Rosenthal, P.J.; Forge, D.; Kumar, V. 7-chloroquinoline-isatin conjugates: Antimalarial, antitubercular, and cytotoxic evaluation. Chem. Biol. Drug Des. 2014, 83, 622–629. [Google Scholar] [CrossRef] [PubMed]
  41. Raj, R.; Singh, P.; Singh, P.; Gut, J.; Rosenthal, P.J.; Kumar, V. Azide-alkyne cycloaddition en route to 1H-1,2,3-triazole-tethered 7-chloroquinoline-isatin chimeras: Synthesis and antimalarial evaluation. Eur. J. Med. Chem. 2013, 62, 590–596. [Google Scholar] [CrossRef]
  42. Nisha; Gut, J.; Rosenthal, P.J.; Kumar, V. beta-amino-alcohol tethered 4-aminoquinoline-isatin conjugates: Synthesis and antimalarial evaluation. Eur. J. Med. Chem. 2014, 84, 566–573. [Google Scholar] [CrossRef] [PubMed]
  43. Videnovic, M.; Opsenica, D.M.; Burnett, J.C.; Gomba, L.; Nuss, J.E.; Selakovic, Z.; Konstantinovic, J.; Krstic, M.; Segan, S.; Zlatovic, M.; et al. Second generation steroidal 4-aminoquinolines are potent, dual-target inhibitors of the botulinum neurotoxin serotype A metalloprotease and P. falciparum malaria. J. Med. Chem. 2014, 57, 4134–4153. [Google Scholar] [CrossRef] [PubMed]
  44. Leverrier, A.; Bero, J.; Cabrera, J.; Frederich, M.; Quetin-Leclercq, J.; Palermo, J.A. Structure-activity relationship of hybrids of Cinchona alkaloids and bile acids with in vitro antiplasmodial and antitrypanosomal activities. Eur. J. Med. Chem. 2015, 100, 10–17. [Google Scholar] [CrossRef] [PubMed]
  45. Sharma, B.; Kaur, S.; Legac, J.; Rosenthal, P.J.; Kumar, V. Synthesis, anti-plasmodial and cytotoxic evaluation of 1H-1,2,3-triazole/acyl hydrazide integrated tetrahydro-beta-carboline-4-aminoquinoline conjugates. Bioorg. Med. Chem. Lett. 2020, 30, 126810. [Google Scholar] [CrossRef]
  46. Vinindwa, B.; Dziwornu, G.A.; Masamba, W. Synthesis and Evaluation of Chalcone-Quinoline Based Molecular Hybrids as Potential Anti-Malarial Agents. Molecules 2021, 26, 4093. [Google Scholar] [CrossRef] [PubMed]
  47. Rodrigues, T.; Ressurreicao, A.S.; da Cruz, F.P.; Albuquerque, I.S.; Gut, J.; Carrasco, M.P.; Goncalves, D.; Guedes, R.C.; dos Santos, D.J.; Mota, M.M.; et al. Flavones as isosteres of 4(1H)-quinolones: Discovery of ligand efficient and dual stage antimalarial lead compounds. Eur. J. Med. Chem. 2013, 69, 872–880. [Google Scholar] [CrossRef]
  48. Paul, N.; Muthusubramanian, S. Synthesis, antimicrobial, and cytotoxicity studies of novel sulfur-linked quinoline–coumarin bisheterocycles. Med. Chem. Res. 2013, 23, 1612–1621. [Google Scholar] [CrossRef]
  49. Patel, D.; Brahmbhatt, D.I. A Novel and Efficient Synthesis of Various 7-Hydroxy-9(Furo[2,3-b]Quinolin-2Yl)6H- Benzo[c]Coumarins and Evaluation of their Antimicrobial Activity. Int. J. Pharm. Res. Sch. 2017, 6, 66–75. [Google Scholar]
  50. Subhedar, D.D.; Shaikh, M.H.; Nawale, L.; Sarkar, D.; Khedkar, V.M.; Shingate, B.B. Quinolidene based monocarbonyl curcumin analogues as promising antimycobacterial agents: Synthesis and molecular docking study. Bioorg. Med. Chem. Lett. 2017, 27, 922–928. [Google Scholar] [CrossRef] [PubMed]
  51. Campanico, A.; Carrasco, M.P.; Njoroge, M.; Seldon, R.; Chibale, K.; Perdigao, J.; Portugal, I.; Warner, D.F.; Moreira, R.; Lopes, F. Azaaurones as Potent Antimycobacterial Agents Active against MDR- and XDR-TB. ChemMedChem 2019, 14, 1537–1546. [Google Scholar] [CrossRef]
  52. Kumar, G.; Lathwal, E.; Saroha, B.; Kumar, S.; Kumar, S.; Chauhan, N.S.; Kumar, T. Synthesis and Biological Evaluation of Quinoline-Based Novel Aurones. ChemistrySelect 2020, 5, 3539–3543. [Google Scholar] [CrossRef]
  53. Sabatini, S.; Gosetto, F.; Manfroni, G.; Tabarrini, O.; Kaatz, G.W.; Patel, D.; Cecchetti, V. Evolution from a natural flavones nucleus to obtain 2-(4-Propoxyphenyl)quinoline derivatives as potent inhibitors of the S. aureus NorA efflux pump. J. Med. Chem. 2011, 54, 5722–5736. [Google Scholar] [CrossRef]
  54. Wang, W.; Zhang, S.; Wang, J.; Wu, F.; Wang, T.; Xu, G. Bioactivity-Guided Synthesis Accelerates the Discovery of 3-(Iso)quinolinyl-4-chromenones as Potent Fungicide Candidates. J. Agric. Food Chem. 2021, 69, 491–500. [Google Scholar] [CrossRef]
  55. Gogoi, S.; Shekarrao, K.; Duarah, A.; Bora, T.C.; Gogoi, S.; Boruah, R.C. A microwave promoted solvent-free approach to steroidal quinolines and their in vitro evaluation for antimicrobial activities. Steroids 2012, 77, 1438–1445. [Google Scholar] [CrossRef]
  56. Balaji, G.L.; Rajesh, K.; Priya, R.; Iniyavan, P.; Siva, R.; Vijayakumar, V. Ultrasound-promoted synthesis, biological evaluation and molecular docking of novel 7-(2-chloroquinolin-4-yloxy)-4-methyl-2H-chromen-2-one derivatives. Med. Chem. Res. 2012, 22, 3185–3192. [Google Scholar] [CrossRef]
  57. Khatkar, A.; Nanda, A.; Kumar, P.; Narasimhan, B. Synthesis and antimicrobial evaluation of ferulic acid derivatives. Res. Chem. Intermed. 2013, 41, 299–309. [Google Scholar] [CrossRef]
  58. Makula, S.M.A. Design, Synthesis and Docking Study of Some Novel Isatin-Quinoline Hybrids as Potential Antitubercular Agents. Anti-Infect. Agents 2016, 14, 53–62. [Google Scholar]
  59. Tabbi, A.; Tebbani, D.; Caporale, A.; Saturninob, C.; Nabavi, S.F.; Nabavic, P.G.; Arrac, C.; Cantürkd, Z.; Turan-Zitounie, G.; Merazigf, H.; et al. New Adamantyl Chalcones: Synthesis, Antimicrobial and Anticancer Activities. Curr. Top. Med. Chem. 2017, 17, 498–506. [Google Scholar] [CrossRef]
  60. Pan, L.; Li, X.; Jin, H.; Yang, X.; Qin, B. Antifungal activity of umbelliferone derivatives: Synthesis and structure-activity relationships. Microb. Pathog. 2017, 104, 110–115. [Google Scholar] [CrossRef]
  61. Kalt, M.M.; Schuehly, W.; Saf, R.; Ochensberger, S.; Solnier, J.; Bucar, F.; Kaiser, M.; Presser, A. Palladium-catalysed synthesis of arylnaphthoquinones as antiprotozoal and antimycobacterial agents. Eur. J. Med. Chem. 2020, 207, 112837. [Google Scholar] [CrossRef] [PubMed]
  62. Li, D.H.; Hu, P.; Xu, S.T.; Fang, C.Y.; Tang, S.; Wang, X.Y.; Sun, X.Y.; Li, H.; Xu, Y.; Gu, X.K.; et al. Lasiokaurin derivatives: Synthesis, antimicrobial and antitumor biological evaluation, and apoptosis-inducing effects. Arch. Pharm. Res. 2017, 40, 796–806. [Google Scholar] [CrossRef] [PubMed]
  63. Abonia, R.; Insuasty, D.; Castillo, J.; Insuasty, B.; Quiroga, J.; Nogueras, M.; Cobo, J. Synthesis of novel quinoline-2-one based chalcones of potential anti-tumor activity. Eur. J. Med. Chem. 2012, 57, 29–40. [Google Scholar] [CrossRef] [PubMed]
  64. Kamal, A.; Suresh, P.; Ramaiah, M.J.; Mallareddy, A.; Imthiajali, S.; Pushpavalli, S.N.; Lavanya, A.; Pal-Bhadra, M. Synthesis and biological evaluation of 4beta-sulphonamido and 4beta-[(4′-sulphonamido)benzamide]podophyllotoxins as DNA topoisomerase-IIalpha and apoptosis inducing agents. Bioorg. Med. Chem. 2012, 20, 2054–2066. [Google Scholar] [CrossRef]
  65. Pudhom, K.; Nuanyai, T.; Matsubara, K.; Vilaivan, T. Antiangiogenic activity of 3,4-seco-cycloartane triterpenes from Thai Gardenia spp. and their semi-synthetic analogs. Bioorg. Med. Chem. Lett. 2012, 22, 512–517. [Google Scholar] [CrossRef]
  66. Kamal, A.; Mallareddy, A.; Suresh, P.; Lakshma Nayak, V.; Shetti, R.V.; Sankara Rao, N.; Tamboli, J.R.; Shaik, T.B.; Vishnuvardhan, M.V.; Ramakrishna, S. Synthesis and anticancer activity of 4beta-alkylamidochalcone and 4beta-cinnamido linked podophyllotoxins as apoptotic inducing agents. Eur. J. Med. Chem. 2012, 47, 530–545. [Google Scholar] [CrossRef]
  67. Zhao, W.; Chen, L.; Li, H.M.; Wang, D.J.; Li, D.S.; Chen, T.; Yuan, Z.P.; Tang, Y.J. A rational design strategy of the novel topoisomerase II inhibitors for the synthesis of the 4-O-(2-pyrazinecarboxylic)-4′-demethylepipodophyllotoxin with antitumor activity by diminishing the relaxation reaction of topoisomerase II-DNA decatenation. Bioorg. Med. Chem. 2014, 22, 2998–3007. [Google Scholar] [CrossRef]
  68. Ayan, D.; Maltais, R.; Hospital, A.; Poirier, D. Chemical synthesis, cytotoxicity, selectivity and bioavailability of 5alpha-androstane-3alpha,17beta-diol derivatives. Bioorg. Med. Chem. 2014, 22, 5847–5859. [Google Scholar] [CrossRef] [PubMed]
  69. Cui, J.; Liu, L.; Zhao, D.; Gan, C.; Huang, X.; Xiao, Q.; Qi, B.; Yang, L.; Huang, Y. Synthesis, characterization and antitumor activities of some steroidal derivatives with side chain of 17-hydrazone aromatic heterocycle. Steroids 2015, 95, 32–38. [Google Scholar] [CrossRef]
  70. Jin, X.; Yan, L.; Li, H.-J.; Wang, R.-L.; Hu, Z.-L.; Jiang, Y.-Y.; Cao, Y.-B.; Yan, T.-H.; Sun, Q.-Y. Novel Triazolyl Berberine Derivatives Prepared via CuAAC Click Chemistry: Synthesis, Anticancer Activity and Structure-Activity Relationships. Anti-Cancer Agents Med. Chem. 2015, 15, 89–98. [Google Scholar] [CrossRef]
  71. Hayat, F.; Park, S.H.; Choi, N.S.; Lee, J.; Park, S.J.; Shin, D. Synthesis and anticancer activity of 4-aza-daurinol derivatives. Arch. Pharm. Res. 2015, 38, 1975–1982. [Google Scholar] [CrossRef]
  72. Srivastava, V.; Lee, H. Synthesis and bio-evaluation of novel quinolino-stilbene derivatives as potential anticancer agents. Bioorg. Med. Chem. 2015, 23, 7629–7640. [Google Scholar] [CrossRef] [PubMed]
  73. Raghavan, S.; Manogaran, P.; Gadepalli Narasimha, K.K.; Kalpattu Kuppusami, B.; Mariyappan, P.; Gopalakrishnan, A.; Venkatraman, G. Synthesis and anticancer activity of novel curcumin-quinolone hybrids. Bioorg. Med. Chem. Lett. 2015, 25, 3601–3605. [Google Scholar] [CrossRef]
  74. Cui, J.; Liu, C.; Liu, L.; Gan, C.; Lu, Y.; Chen, S.; Dong, X.; Huang, Y. Synthesis and Antitumor Activities of Cholestane Derivatives with a Structure of 3-Hydroxy-6-hydrazone or 6-Carbonyl-3-hydrazone. Chin. J. Org. Chem. 2016, 36, 2933. [Google Scholar] [CrossRef]
  75. He, D.M.; Liu, L.; Zheng, J.H.; Yang, C.H.; Huang, Y.M.; Gan, C.F.; Cui, J.G. Synthesis and antitumor activity of pyrazoline steroidal aromatic heterocyclic compounds. Chin. J. Med. Chem. 2016, 26, 61–64. [Google Scholar]
  76. Baji, Á.; Gyovai, A.; Wölfling, J.; Minorics, R.; Ocsovszki, I.; Zupkó, I.; Frank, É. Microwave-assisted one-pot synthesis of steroid–quinoline hybrids and an evaluation of their antiproliferative activities on gynecological cancer cell lines. RSC Adv. 2016, 6, 27501–27516. [Google Scholar] [CrossRef]
  77. Chaudhary, V.; Venghateri, J.B.; Dhaked, H.P.; Bhoyar, A.S.; Guchhait, S.K.; Panda, D. Novel Combretastatin-2-aminoimidazole Analogues as Potent Tubulin Assembly Inhibitors: Exploration of Unique Pharmacophoric Impact of Bridging Skeleton and Aryl Moiety. J. Med. Chem. 2016, 59, 3439–3451. [Google Scholar] [CrossRef]
  78. Sommerwerk, S.; Heller, L.; Kuhfs, J.; Csuk, R. Selective killing of cancer cells with triterpenoic acid amides—The substantial role of an aromatic moiety alignment. Eur. J. Med. Chem. 2016, 122, 452–464. [Google Scholar] [CrossRef] [PubMed]
  79. Shobeiri, N.; Rashedi, M.; Mosaffa, F.; Zarghi, A.; Ghandadi, M.; Ghasemi, A.; Ghodsi, R. Synthesis and biological evaluation of quinoline analogues of flavones as potential anticancer agents and tubulin polymerization inhibitors. Eur. J. Med. Chem. 2016, 114, 14–23. [Google Scholar] [CrossRef]
  80. Zhang, L.; Zhang, Z.; Chen, F.; Chen, Y.; Lin, Y.; Wang, J. Aromatic heterocyclic esters of podophyllotoxin exert anti-MDR activity in human leukemia K562/ADR cells via ROS/MAPK signaling pathways. Eur. J. Med. Chem. 2016, 123, 226–235. [Google Scholar] [CrossRef]
  81. Li, Z.; Su, H.; Yu, W.; Li, X.; Cheng, H.; Liu, M.; Pang, X.; Zou, X. Design, synthesis and anticancer activities of novel otobain derivatives. Org. Biomol. Chem. 2016, 14, 277–287. [Google Scholar] [CrossRef] [PubMed]
  82. Gu, W.; Jin, X.Y.; Li, D.D.; Wang, S.F.; Tao, X.B.; Chen, H. Design, synthesis and in vitro anticancer activity of novel quinoline and oxadiazole derivatives of ursolic acid. Bioorg. Med. Chem. Lett. 2017, 27, 4128–4132. [Google Scholar] [CrossRef] [PubMed]
  83. Gan, C.; Liu, L.; Cui, J.; Liu, Z.; Shi, H.; Lin, Q.; Sheng, H.; Yang, C.; Huang, Y. Synthesis of Some Steroidal Derivatives with Side Chain of 20- and 22- Hydrazone Aromatic Heterocycles and their Antiproliferative Activity. Med. Chem. 2017, 13, 375–383. [Google Scholar] [CrossRef]
  84. Yao, G.; Chen, H.; Chen, L.; Ge, M.; Yang, J.; Liu, W.; Xia, M.; Hayashi, T.; Guo, C.; Ikejima, T. Autophagy promotes apoptosis induction through repressed nitric oxide generation in the treatment of human breast cancer MCF-7 cells with L-A03, a dihydroartemisinin derivative. Med. Chem. Res. 2017, 26, 1427–1436. [Google Scholar] [CrossRef]
  85. Aly, A.A.; El-Sheref, E.M.; Bakheet, M.E.M.; Mourad, M.A.E.; Brase, S.; Ibrahim, M.A.A.; Nieger, M.; Garvalov, B.K.; Dalby, K.N.; Kaoud, T.S. Design, synthesis and biological evaluation of fused naphthofuro[3,2-c] quinoline-6,7,12-triones and pyrano[3,2-c]quinoline-6,7,8,13-tetraones derivatives as ERK inhibitors with efficacy in BRAF-mutant melanoma. Bioorg. Chem. 2019, 82, 290–305. [Google Scholar] [CrossRef]
  86. Sri Ramya, P.V.; Guntuku, L.; Angapelly, S.; Karri, S.; Digwal, C.S.; Babu, B.N.; Naidu, V.G.M.; Kamal, A. Curcumin inspired 2-chloro/phenoxy quinoline analogues: Synthesis and biological evaluation as potential anticancer agents. Bioorg. Med. Chem. Lett. 2018, 28, 892–898. [Google Scholar] [CrossRef]
  87. Taheri, S.; Nazifi, M.; Mansourian, M.; Hosseinzadeh, L.; Shokoohinia, Y. Ugi efficient synthesis, biological evaluation and molecular docking of coumarin-quinoline hybrids as apoptotic agents through mitochondria-related pathways. Bioorg. Chem. 2019, 91, 103147. [Google Scholar] [CrossRef]
  88. Lipeeva, A.V.; Zakharov, D.O.; Gatilov, Y.V.; Pokrovskii, M.A.; Pokrovskii, A.G.; Shults, E.E. Design and Synthesis of 3-(N-Substituted)aminocoumarins as Anticancer Agents from 3-Bromopeuruthenicin. ChemistrySelect 2019, 4, 10197–10201. [Google Scholar] [CrossRef]
  89. Shen, Q.K.; Deng, H.; Wang, S.B.; Tian, Y.S.; Quan, Z.S. Synthesis, and evaluation of in vitro and in vivo anticancer activity of 14-substituted oridonin analogs: A novel and potent cell cycle arrest and apoptosis inducer through the p53-MDM2 pathway. Eur. J. Med. Chem. 2019, 173, 15–31. [Google Scholar] [CrossRef]
  90. Zhao, W.; He, L.; Xiang, T.L.; Tang, Y.J. Discover 4beta-NH-(6-aminoindole)-4-desoxy-podophyllotoxin with nanomolar-potency antitumor activity by improving the tubulin binding affinity on the basis of a potential binding site nearby colchicine domain. Eur. J. Med. Chem. 2019, 170, 73–86. [Google Scholar] [CrossRef]
  91. Prashanth, T.; Avin, B.R.V.; Thirusangu, P.; Ranganatha, V.L.; Prabhakar, B.T.; Sharath Chandra, J.N.N.; Khanum, S.A. Synthesis of coumarin analogs appended with quinoline and thiazole moiety and their apoptogenic role against murine ascitic carcinoma. Biomed. Pharmacother. 2019, 112, 108707. [Google Scholar] [CrossRef] [PubMed]
  92. Jin, X.Y.; Chen, H.; Li, D.D.; Li, A.L.; Wang, W.Y.; Gu, W. Design, synthesis, and anticancer evaluation of novel quinoline derivatives of ursolic acid with hydrazide, oxadiazole, and thiadiazole moieties as potent MEK inhibitors. J. Enzyme Inhib. Med. Chem. 2019, 34, 955–972. [Google Scholar] [CrossRef] [PubMed]
  93. Yang, Y.T.; Du, S.; Wang, S.; Jia, X.; Wang, X.; Zhang, X. Synthesis of new steroidal quinolines with antitumor properties. Steroids 2019, 151, 108465. [Google Scholar] [CrossRef]
  94. Li, W.; Xu, F.; Shuai, W.; Sun, H.; Yao, H.; Ma, C.; Xu, S.; Yao, H.; Zhu, Z.; Yang, D.H.; et al. Discovery of Novel Quinoline-Chalcone Derivatives as Potent Antitumor Agents with Microtubule Polymerization Inhibitory Activity. J. Med. Chem. 2019, 62, 993–1013. [Google Scholar] [CrossRef] [PubMed]
  95. Jia, X.; Liu, Q.; Wang, S.; Zeng, B.; Du, G.; Zhang, C.; Li, Y. Synthesis, cytotoxicity, and in vivo antitumor activity study of parthenolide semicarbazones and thiosemicarbazones. Bioorg. Med. Chem. 2020, 28, 115557. [Google Scholar] [CrossRef]
  96. Hoenke, S.; Heise, N.V.; Kahnt, M.; Hans-Peter, D.; Rene, C. Betulinic acid derived amides are highly cytotoxic, apoptotic and selective. Eur. J. Med. Chem. 2020, 207, 112815. [Google Scholar] [CrossRef]
  97. Xu, Y.; Jing, D.; Zhao, D.; Wu, Y.; Lu, X.; Haroon, U.R.; Wang, H.; Wang, L.; Cao, H. New modification strategy of matrine as Hsp90 inhibitors based on its specific L conformation for cancer treatment. Bioorg. Med. Chem. 2020, 28, 115305. [Google Scholar] [CrossRef]
  98. Insuasty, D.; García, S.; Abonia, R.; Insuasty, B.; Quiroga, J.; Nogueras, M.; Cobo, J.; Borosky, G.L.; Laali, K.K. Design, synthesis, and molecular docking study of novel quinoline-based bis-chalcones as potential antitumor agents. Arch. Der Pharm. 2021, 354, 2100094. [Google Scholar] [CrossRef]
  99. Mohassab, A.M.; Hassan, H.A.; Abdelhamid, D.; Gouda, A.M.; Youssif, B.G.; Tateishi, H.; Fujita, M.; Otsuka, M.; Abdel-Aziz, M. Design and synthesis of novel quinoline/chalcone/1,2,4-triazole hybrids as potent antiproliferative agent targeting EGFR and BRAFV600E kinases. Bioorg. Chem. 2021, 106, 104510. [Google Scholar] [CrossRef]
  100. Zeng, B.; Cheng, Y.; Zheng, K.; Liu, S.; Shen, L.; Hu, J.; Li, Y.; Pan, X. Design, synthesis and in vivo anticancer activity of novel parthenolide and micheliolide derivatives as NF-κB and STAT3 inhibitors. Bioorg. Chem. 2021, 111, 104973. [Google Scholar] [CrossRef]
  101. Song, J.-R.; Li, N.; Li, D.-P. Synthesis and anti-proliferation activity of mogrol derivatives bearing quinoline and triazole moieties. Bioorg. Med. Chem. Lett. 2021, 421, 28090. [Google Scholar] [CrossRef]
  102. Shang, F.-F.; Wang, J.Y.; Xu, Q.; Deng, H.; Guo, H.-Y.; Jin, X.; Li, X.; Shen, Q.-K.; Quan, Z.-S. Design, synthesis of novel celastrol derivatives and study on their antitumor growth through HIF-1α pathway. Eur. J. Med. Chem. 2021, 220, 113474. [Google Scholar] [CrossRef]
  103. Dong, J.; Huang, G.; Cui, Q.; Meng, Q.; Li, S.; Cui, J. Discovery of heterocycle-containing α-naphthoflavone derivatives as water-soluble, highly potent and selective CYP1B1 inhibitors. Eur. J. Med. Chem. 2021, 209, 112895. [Google Scholar] [CrossRef] [PubMed]
  104. Guan, Y.; Liu, X.; Yuan, X.; Liu, W.; Li, Y.; Yu, G.; Tian, X.; Zhang, Y.; Song, J.; Li, W.; et al. Design, Synthesis, and Anticancer Activity Studies of Novel Quinoline-Chalcone Derivatives. Molecules 2021, 26, 4899. [Google Scholar] [CrossRef]
  105. Thorat, N.M.; Sarkate, A.P.; Lokwani, D.K.; Tiwari, S.V.; Azad, R.; Thopate, S.R. N-Benzylation of 6-aminoflavone by reductive amination and efficient access to some novel anticancer agents via topoisomerase II inhibition. Mol. Divers. 2021, 25, 937–948. [Google Scholar] [CrossRef] [PubMed]
  106. Jyothi, M.; Sherapura, A.; Khamees, H.A.; Prabhakar, B.T.; Khanum, S.A. Synthesis, structure analysis, DFT calculations and energy frameworks of new coumarin appended oxadiazoles, to regress ascites malignancy by targeting VEGF mediated angiogenesis. J. Mol. Struct. 2022, 1252, 132173. [Google Scholar] [CrossRef]
  107. Herrmann, L.; Yaremenko, I.A.; Capci, A.; Struwe, J.; Tailor, D.; Dheeraj, A.; Hodek, J.; Belyakova, Y.Y.; Radulov, P.S.; Weber, J.; et al. Synthesis and in vitro Study of Artemisinin/Synthetic Peroxide-Based Hybrid Compounds against SARS-CoV-2 and Cancer. ChemMedChem 2022, 17, e202200005. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhao, Y.-Q.; Li, X.; Guo, H.-Y.; Shen, Q.-K.; Quan, Z.-S.; Luan, T. Application of Quinoline Ring in Structural Modification of Natural Products. Molecules 2023, 28, 6478. https://doi.org/10.3390/molecules28186478

AMA Style

Zhao Y-Q, Li X, Guo H-Y, Shen Q-K, Quan Z-S, Luan T. Application of Quinoline Ring in Structural Modification of Natural Products. Molecules. 2023; 28(18):6478. https://doi.org/10.3390/molecules28186478

Chicago/Turabian Style

Zhao, Yu-Qing, Xiaoting Li, Hong-Yan Guo, Qing-Kun Shen, Zhe-Shan Quan, and Tian Luan. 2023. "Application of Quinoline Ring in Structural Modification of Natural Products" Molecules 28, no. 18: 6478. https://doi.org/10.3390/molecules28186478

Article Metrics

Back to TopTop