Next Article in Journal
Blood Compatibility of Sulfonated Cladophora Nanocellulose Beads
Previous Article in Journal
Synthesis, Molecular Docking, and Antimycotic Evaluation of Some 3-Acyl Imidazo[1,2-a]pyrimidines
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Novel Trifluoromethylcoumarinyl Urea Derivatives: Synthesis, Characterization, Fluorescence, and Bioactivity

Research Center of Agro-bionic Engineering & Tech. of Shandong Province, College of Chemistry & Pharm., Qingdao Agricultural University, Qingdao 266109, China
*
Author to whom correspondence should be addressed.
Molecules 2018, 23(3), 600; https://doi.org/10.3390/molecules23030600
Submission received: 13 February 2018 / Revised: 28 February 2018 / Accepted: 5 March 2018 / Published: 7 March 2018

Abstract

:
A series of novel trifluoromethylcoumarinyl urea derivatives were designed, synthesized, and characterized by 1H-NMR, 13C-NMR, and HR-ESI-MS. The fluorescence spectra of the target compounds were recorded. The spectra show that most of the title compounds glow green with λmaxem of 500–517 nm, while compounds 5r, 5s, 5u, and 5l (compounds named by authors) glow violet with λmaxem of 381–443 nm. Moreover, the herbicidal and antifungal activities of the synthesized compounds were evaluated for their potential use as pesticides. The results indicate that compound 5f against the caulis of Amaranthus retroflexus and compounds 5j and 5l against the taproot of Digitaria sanguinalis are equivalent to the commercial herbicide Acetochlor. Nine of the title compounds are more antifungal than commercial fungicide Carbendazim against Botrytis cinerea.

Graphical Abstract

1. Introduction

Ureas are very important compounds in organic chemistry because of their extensive applications in medicine and agriculture. The substituted ureas exhibit diversified medicinal activities such as antimicrobial [1], antiviral [2], anti-HIV [3], anti-AIDS [4], anti-malaria [5], anti-proliferative [6], antitumor [7], anticancer [8], anti-inflammatory [9], anti-ulcerogenic [10], anticonvulsant [11], anti-nociceptive [12], antihypertensive [9], antidepressant [13], complement inhibition [14], HDL-elevating [15], and inhibition of nitric oxide [16]. Urea compounds also possess a broad spectrum of pesticidal activities, such as benzoylurea type insecticides (Diflubenzuron, Chlorbenzuron, etc.) [17], fungicides (Triclocarban, Pencycuron, etc.) [18], sulfonylurea type herbicides (Chlorsulfuron, Nicosulfuron, etc.), and urea herbicides (Benzthiazuron, Siduron, etc.) [19], as well as plant growth regulators (Thidiazuron, Forchlorfenuron, etc.) [20].
On the other hand, coumarin is a subunit of many natural products and a wide range of synthetic compounds, constituting an important class of heterocycles. These kinds of compounds are widely used in the food, cosmetic, and perfume industries. Coumarins are also an important class of fluorescents and are used as laser dyes, brighteners, fluorescent labels, and other organic optical materials, such as emission layers of organic light-emitting diodes (OLED) [21,22,23,24]. Additionally, they are sometimes used as probes to monitor certain physiological process and trace harmful substances in environment [25,26,27]. It has been noted that the photophysical and spectroscopic properties of coumarin can be easily adjusted by the introduction of different substituents into the core, which leads them to be more flexible and fit well in various applications [28]. It is reported that the electron-withdrawing groups on 3- or 4-position and electron-releasing groups on 6- or 7-position of the coumarin skeleton could enhance the band intensity of fluorescence [29]. Moreover, coumarins have been extensively studied for their use as biological agents with anti-depressant, anticoagulant, anti-inflammatory, antibacterial, antifungal, antimalarial, moluscicidal, and anthelmintic activities [30,31,32,33,34,35,36]. Much research has been focused on coumarin derivatives for their pesticidal activities too. One of the representative compounds is 7-hydroxyl-4-methylcoumarin, which exhibited interesting pesticidal activities of allelopathy and antifungal, and it is reported that the C7 hydroxyl group and C4 methyl substitution contributed significantly to the activity [37,38]. Moreover, brominated derivatives of the compound showed remarkable larvicidal and ovicidal activities against vector mosquitoes [39]. Derivatives of 7-hydroxyl-4-methylcoumarin are also reported to show anthelmintic and acaricidal activities [40,41].
In addition, fluorine is the most electronegative of all elements, which endows it with unique properties. Indeed, it is commonly accepted that introducing a fluorine-containing group onto the molecule scaffold often leads to improvement of the molecular chemical, physical, and biological properties [42]. To date, the trifluoromethyl (CF3) group has appeared as the archetypal and most sought-after fluorine-containing group, and there have been reported a huge and ever-increasing number of trifluoromethylated compounds in the literature [43,44]. The CF3 group is very important in medicinal chemistry [45] due to its high lipophilicity, easy transportation, and hard degradation in vivo [46,47]. Thus, the introduction of a CF3 group into bioactive molecules has become a preferable approach in pharmaceutical studies [48,49]. However, the literature about CF3-substituted coumarins is limited, and the reports about these compounds are mainly focused on their use as substrate probes to detect the physiology [50,51,52]. Also, in view of these observations and in continuation of our research program on the synthesis of fluorinated coumarins [53], we herein report the synthesis and characterization of some novel 4-trifluoromethyl-6-substituted-ureido-7-methoxylcoumarins, whose fluorescent properties and antifungal and herbicidal activities are also evaluated.

2. Results and Discussion

2.1. Chemistry

The synthesis of target compounds was carried out according to Scheme 1. Compound 1 was prepared from the reaction of resorcinol with ethyl trifluoroacetoacetate catalyzed by Con. (concentrated) H2SO4. Compound 2 was synthesized by methylation of compound 1 with dimethyl sulfate using anhydrous K2CO3 as catalyst in acetone. Compound 3 was synthesized by nitration of compound 2 with guanidine nitrate in Con. H2SO4. Compound 4 was obtained by reduction of compound 3 with Fe powder in aqueous solution of NH4Cl. The target compounds 5a5u were furnished by addition of compounds 4 to a series of isocyanates catalyzed by triethylamine in CH2Cl2 (DCM). The structures of all target compounds were well characterized by 1H-NMR, 13C-NMR, and HR-ESI-MS. The spectrograms were listed in the supplementary materials.

2.2. UV Absorption and Fluorescence Characteristics

The λmaxab (maximum absorption wavelength), εmax (molar absorptivity), λmaxem (maximum emission wavelength), Δλ (Stokes shift) and Φf (fluorescence quantum yield) of the title compounds are listed in Table 1. The λmaxab of all the title compounds are 360–374 nm, and the εmax are 5.24–7.67 × 103 L·mol−1·cm−1. Most of the title compounds glow green fluorescence with λmaxem of 500–517 nm when illuminated by ultraviolet light, while compounds 5r, 5s, 5u, and 5l glow violet fluorescence with λmaxem of 381–443 nm (Figure 1). However compounds 5o and 5t do not fluoresce at the concentration. The values of Φf indicate that all the title compounds are weaker fluorescent compare to the standard reference compound quinine sulfate. Table 1 shows that alkyl substituted compounds 5a5i have higher fluorescence quantum yield than the phenyl substituted compounds 5k5p and 5r5u, but compounds 5j and 5q with high Φf were the exceptions. From the phenomenon, it can be concluded that electron-donating groups may favor the fluorescence of the compounds.

2.3. Herbicidal Activities

The herbicidal activities of the 21 title compounds of 20 mg·L−1 and 100 mg·L−1 against the taproot and caulis growth of dicotyledonous weed A. retroflexus and monocotyledonous weed D. sanguinalis were assayed. The inhibition of the compounds with effectiveness of greater than 50% to at least one organ of the test weeds at 100 mg·L−1 are displayed in Figure 2. It indicates that compounds 5j and 5s show medium inhibition against the taproot growth of A. retroflexus, and compounds 5f, 5s, 5k and 5n show medium to high inhibition against the caulis growth of A. retroflexus. Moreover, compounds 5j and 5l show medium inhibition against the taproot growth of D. sanguinalis, and compounds 5f, 5q, and 5r show medium inhibition against the caulis growth of D. sanguinalis. Furthermore, the herbicidal activities of 5f against the caulis of A. retroflexus, 5j and 5l against the taproot of D. sanguinalis are equivalent to the commercial herbicide Acetochlor. It also shows that only compounds 5j and 5s are both active to the two test herbs; the other compounds display certain selectivity between the test herbs. Contrary to the fluorescence, the phenyl substituted compounds show higher herbicidal activity than the alkyl substituted compounds except compound 5f substituted by cyclopentyl. All the compounds substituted by phenyls with chlorine (5j5l) exhibit better activity. Compounds 5o (3-CH3Ph-), 5r (2-CH3OPh-) and 5s (3-CH3OPh-) substituted by phenyls with electron donating groups are more active.

2.4. Antifungal Activities

The in vitro antifungal activities of the title compounds of 20 mg·L−1 and 100 mg·L−1 against the mycelium growth of the phytopathogens Valsa mali, B. cinerea, Colletotrichum glecosporioides, and Fusarium oxysporum were assayed. The inhibition of the synthesized compounds with effectiveness of greater than 50% at 100 mg·L−1 are exhibited in Figure 3. This indicates that more compounds exhibit stronger inhibition against B. cinerea than the other three plant disease fungi (the inhibitory rates of all the title compounds against F. oxysporum are less than 50%, and they do not appear in Figure 3). There are only four title compounds showing medium inhibition against V. mali, and only one compound having an inhibitory rate of more than 50% to C. glecosporioides. Meanwhile, 12 of the title compounds exhibit medium to high activity against B. cinerea, and nine of them are more antifungal than the commercial fungicide Carbendazim. Based on the above description, it seems that the phenyl substitutes are of more benefit to the antifungal activity. Compounds 5j, 5l, and 5o5t with substitutes of –Cl, –CH3, –CF3, or –OCH3 on phenyls have more potential.

3. Experimental Section

3.1. Chemistry

3.1.1. General Procedures

All chemicals were obtained from commercial sources and used without further purification. Glass plates pre-coated with silica gel 60 GF254 (Qingdao Haiyang Chemical Co., Ltd., Qingdao, China) were used for analytical thin layer chromatography (TLC). An uncorrected WRS-1B digital melting-point apparatus (Shanghai Precision Optical Instrument Co., Ltd., Shanghai, China) was used to determine the melting points. A Bruker Avance III HD 500 MHz nuclear magnetic resonance instrument (Bruker, Fällanden, Switzerland) was used to record the 1H- and 13C-NMR spectra in CDCl3 or DMSO-d6 (1H at 500 MHz and 13C at 126 MHz) using tetramethylsilane (TMS) as the internal standard. A maXis Q-TOF high-resolution mass spectra instrument (Bruker, Karlsruhe, Germany) was used to measure the HR-ESI-MS. An F-4600 fluorescence spectrophotometer (Hitachi, Tokyo, Japan) was used to record the fluorescence.

3.1.2. Synthesis of 4-Trifluoromethyl-7-hydroxycoumarin (1)

Concentrated sulfuric acid (2 mL) and trifluoroacetoacetate (3.7 g, 10 mmol) were added dropwise in turn to the solution of resorcinol (2.2 g, 10 mmol) in ethanol (4 mL) with stirring. Then the mixture was heated to maintain at 80 °C until the end of the reaction was detected by TLC. The resulted thick magenta liquid was dripped into 10 mL of cold water with rigorous stirring. The precipitate was filtered and dried to give compound 1. Yield 88.9%, m.p. (melting point) 175–176 °C [lit. (literature) 178–179 °C] [54].

3.1.3. Synthesis of 4-Trifluoromethyl-7-methoxycoumarin (2)

Dimethyl sulfate (2.5 g, 20 mmol) was added dropwise to the suspension of compound 1 (5.8 g, 25 mmol) and anhydrous K2CO3 (4.1 g, 30 mmol) in 66 mL of acetone with stirring. Then the mixture was heated to reflux for 8 h. After K2CO3 was filtered out, the filtrate was rotary evaporated to 1/3 of the total volume. Then the residue was allowed to cool to room temperature until a white solid precipitated. The solid product was filtered out and washed with cold water to neutral pH to get compound 2. Yield 97.8%, m.p. 110–112 °C (lit. 111–112 °C) [55].

3.1.4. Synthesis of 4-Trifluoromethyl-6-nitro-7-methoxycoumarin (3)

Guanidine nitrate (1.4 g, 11.6 mmol) was added batch-wise to the solution of 2 (2.6 g, 10.7 mmol) in 16 mL Conc. H2SO4 with stirring at 0–5 °C. The mixture was stirred at room temperature for 3 h until the end of the reaction. Then the reaction mixture was poured into 40 mL of ice water with stirring to obtain a pale yellow solid. The solid product was filtered and dried to give compound 3. Yield 97.7%, m.p. 118–119 °C (as reported) [53].

3.1.5. Synthesis of 4-Trifluoromethyl-6-amino-7-methoxycoumarin (4)

The mixture of compound 3 (2.9 g, 10 mmol), iron powder (5.0 g, 90 mmol), NH4Cl (1.1 g, 20 mmol) and 50 mL water was mechanically stirred at room temperature until the end of the reaction. The mixture was extracted with ethyl acetate (50 mL × 3). The extract was dried over anhydrous sodium sulfate, then filtered and rotary evaporated to get a dark yellow crystal of compound 4. Yield 70%, m.p. 179–180 °C (as reported) [53]. The compound is unstable and needs to be used as soon as possible for the next step.

3.1.6. General Procedure for Synthesis of 4-Trifluoromethyl-6-amino-7-methoxycoumarinyl Urea Derivatives (5a5u)

To a cool suspension of 4 (0.65 g, 2.5 mmol) in 3.5 mL of anhydrous dichloromethane, isocyanate (3 mmol) was added dropwise with stirring. The mixture was allowed to return to room temperature until the end of the reaction. The solid produced was filtered and chromatographed on a flash silica gel column (ethyl acetate/petroleum ether = 1:3) to give the target compounds 5a5u. The yield, appearance, m.p., NMR and HR-ESI-MS (MS) date of the synthesized compounds are listed below.
1-Ethyl-3-(7-methoxy-2-oxo-4-(trifluoromethyl)-2H-chromen-6-yl) urea (5a). Light yellow solid; yield: 42.8%; m.p. 232–233 °C; 1H-NMR (500 MHz, DMSO) δ: 8.68 (s, 1H), 8.14 (s, 1H), 7.23 (s, 1H), 6.95 (t, J = 5.3 Hz, 1H), 6.83 (s, 1H), 4.01 (s, 3H), 3.20–3.09 (m, 2H), 1.08 (t, J = 7.2 Hz, 3H); 13C-NMR (126 MHz, DMSO) δ: 159.46, 155.39, 152.17, 150.33, 140.41 (q, J = 31.5 Hz), 127.99, 122.27 (q, J = 275.94 Hz), 113.47 (q, J = 6.3 Hz), 111.52 (q, J = 1.26Hz), 106.07, 100.16, 57.24, 34.31, 15.72.; HR-ESI-MS m/z: 353.0712 [M + Na]+ (calculated for C14H13F3N2NaO4, 353.0725).
1-Propyl-3-(7-methoxy-2-oxo-4-(trifluoromethyl)-2H-chromen-6-yl) urea (5b). White solid; yield: 31.1%; m.p. 229 °C. 1H-NMR (500 MHz, DMSO) δ: 8.68 (s, 1H), 8.16 (s, 1H), 7.22 (s, 1H), 6.97 (t, J = 5.3 Hz, 1H), 6.82 (s, 1H), 4.00 (s, 3H), 3.06 (q, J = 5 Hz, 2H), 1.52–1.38 (m, 2H), 0.90 (t, J = 7.4 Hz, 3H); 13C-NMR (126MHz, DMSO) δ: 159.47, 155.50, 152.16, 150.32, 140.41 (q, J = 31.5 Hz), 128.03, 122.27 (q, J = 275.94 Hz), 113.49 (q, J = 6.3 Hz), 111.48, 106.09, 100.18, 57.26, 41.24, 23.31, 11.83; HR-ESI-MS m/z: 367.0882 [M + Na]+ (calculated for C15H15F3N2NaO4, 367.0882).
1-Isopropyl-3-(7-methoxy-2-oxo-4-(trifluoromethyl)-2H-chromen-6-yl) urea (5c). Light yellow solid; yield: 54.2%; m.p. 224 °C; 1H-NMR (500 MHz, DMSO) δ: 8.66 (s, 1H), 8.07 (s, 1H), 7.20 (s, 1H), 6.90 (d, J = 7.2 Hz, 1H), 6.80 (s, 1H), 3.99 (s, 3H), 3.82–3.72 (m, 1H), 1.10 (d, J = 6.4 Hz, 6H); 13C-NMR (126 MHz, DMSO) δ: 159.45, 154.74, 152.11, 150.28, 140.42 (q, J = 31.5 Hz), 128.03, 122.16 (q, J = 275.94 Hz), 113.43 (q, J = 5.0 Hz), 111.36 (q, J = 2.52 Hz), 106.06, 100.11, 57.22, 41.36, 23.41; ESI-MS m/z: 345.30 [M + Na]+ (calculated for C15H16F3N2O4, 345.21).
1-(tert-Butyl)-3-(7-methoxy-2-oxo-4-(trifluoromethyl)-2H-chromen-6-yl) urea (5d). yellow solid; Yield: 31.1%; m.p. 237 °C; 1H-NMR (500 MHz, CDCl3) δ: 8.54 (s, 1H), 6.87 (s, 1H), 6.83 (s, 1H), 6.64 (s, 1H), 4.90 (s, 1H), 3.93 (s, 3H), 1.40 (s, 9H); 13C-NMR (126 MHz, CDCl3) δ: 159.76 (q, J = 6.2 Hz), 152.2 (q, J = 22.68 Hz), 151.92 (q, J = 5.5 Hz), 150.76 (q, J = 7.3 Hz), 142.11 (q, J = 32.76 Hz), 127.15 (q, J = 12.2 Hz), 121.57 (q, J = 275.94 Hz), 113.46 (q, J = 2.52 Hz), 112.58 (q, J =5.04 Hz), 106.85, 98.98 (q, J = 6.2 Hz), 56.33, 51.03, 29.31; HR-ESI-MS m/z: 381.1038 [M + Na]+ (calculated for C16H17F3N2NaO4, 381.1038).
1-Hexyl-3-(7-methoxy-2-oxo-4-(trifluoromethyl)-2H-chromen-6-yl) urea (5e). Light yellow solid; yield: 31.2%; m.p. 143 °C; 1H-NMR (500 MHz, CDCl3) δ: 8.68 (s, 1H), 7.29 (s, 1H), 6.82 (s, 1H), 6.63 (s, 1H), 5.47 (t, J = 5.0 Hz, 1H), 3.89 (s, 3H), 3.28 (q, J = 5 Hz, 2H), 1.58–1.48 (m, 2H), 1.38–1.20 (m, 6H), 0.85 (t, J = 6.6 Hz, 3H); 13C-NMR (126 MHz, CDCl3) δ: 159.73, 154.77, 151.80, 150.85, 142.13 (q, J = 32.76 Hz), 126.90, 122.63 (q, J = 122.37 Hz), 113.61 (q, J = 1.26 Hz), 112.66 (q, J = 5.04 Hz), 106.90, 98.98, 56.38, 40.53, 31.50, 29.99, 26.56, 22.57, 14.00; HR-ESI-MS m/z: 409.1346 [M + Na]+ (calculated for C18H21F3N2NaO4, 409.1351).
1-Cyclopentyl-3-(7-methoxy-2-oxo-4-(trifluoromethyl)-2H-chromen-6-yl) urea (5f). Light yellow solid; yield: 43.6%; m.p. 225 °C; 1H-NMR (500 MHz, DMSO) δ: 8.68 (s, 1H), 8.07 (s, 1H), 7.21 (s, 1H), 7.01 (d, J = 6.9 Hz, 1H), 6.81 (s, 1H), 4.00 (s, 3H), 3.97-3.91 (m, 1H), 1.90–1.81 (m, 2H), 1.72–1.48 (m, 4H), 1.45–1.31 (m, 2H); 13C-NMR (126 MHz, DMSO) δ: 159.49, 155.02, 152.10, 150.30, 140.42 (q, J = 32.76 Hz), 128.04, 122.29(q, J = 273.42 Hz),113.52 (q, J = 1.26 Hz), 111.34 (q, J = 2.52 Hz), 106.10, 100.18, 57.27, 51.28, 33.30, 23.59; HR-ESI-MS m/z: 393.1033 [M + Na]+ (calculated for C17H17F3N2NaO4, 393.1038).
1-Cyclohexyl-3-(7-methoxy-2-oxo-4-(trifluoromethyl)-2H-chromen-6-yl) urea (5g). Light yellow solid; yield 48.9%; m.p. 265 °C; 1H-NMR (500 MHz, DMSO) δ: 8.68 (s, 1H), 8.13 (s, 1H), 7.23 (s, 1H), 6.95 (d, J = 7.5 Hz, 1H), 6.82 (s, 1H), 3.99 (s, 3H), 3.54–3.45 (m, 1H),1.86–1.76 (m, 2H), 1.72–1.63 (m, 2H), 1.37–1.11 (m, 6H); 13C-NMR (126 MHz, DMSO) δ: 159.48, 154.70, 152.14, 150.29, 140.42 (q, J = 31.5 Hz), 128.06, 122.28(q, J = 275.94 Hz), 113.49(q, J = 6.3 Hz), 111.40 (q, J = 2.52 Hz), 106.09, 100.18, 57.27, 48.02, 33.35, 25.70, 24.71; HR-ESI-MS m/z: 407.1191 [M + Na]+ (calculated for C18H19F3N2NaO4, 407.1195).
1-(2-(3-(Prop-1-en-2-yl)phenyl)propan-2-yl)-3-(7-methoxy-2-oxo-4-(trifluoromethyl)-2H-chromen-6-yl) urea (5h). Light yellow solid; yield: 20.0%; m.p. 228–229 °C; 1H-NMR (500 MHz, DMSO) δ: 8.52 (s, 1H), 8.29 (s, 1H), 7.50 (s, 1H), 7.47 (s, 1H), 7.39–7.29 (m, 3H), 7.22 (s, 1H), 6.79 (s, 1H), 5.38 (s, 1H), 5.08 (s, 1H), 4.01 (s, 3H), 2.11 (s, 3H), 1.62 (s, 6H); 13C-NMR (126 MHz, DMSO) δ: 159.43, 154.39, 152.20, 150.33, 148.72, 143.45, 140.62, 140.34 (q, J = 31.5 Hz), 128.54, 128.04, 124.66, 123.56, 122.22 (q, J = 275.94 Hz), 122.21, 113.47 (q, J = 6.3 Hz), 112.89, 111.22, 106.03, 100.21, 57.29, 55.01, 30.25, 22.01; HR-ESI-MS m/z: 483.1504 [M + Na]+ (calculated for C24H23F3N2NaO4, 483.1508).
1-Phenethyl-3-(7-methoxy-2-oxo-4-(trifluoromethyl)-2H-chromen-6-yl) urea (5i). Light yellow solid; yield: 41.7%; m.p. 222 °C; 1H-NMR (500 MHz, DMSO) δ: 8.68 (s, 1H), 8.22 (s, 1H), 7.36–7.29 (m, 2H), 7.27–7.24 (m, 2H), 7.23–7.18 (m, 2H), 6.99 (t, J = 5.1 Hz, 1H), 6.82 (s, 1H), 3.98 (s, 3H), 3.38 (q, J = 6.6 Hz, 2H), 2.75 (t, J = 7.0 Hz, 2H); 13C-NMR (126 MHz, DMSO) δ: 159.46, 155.48, 152.21, 150.38, 140.40 (q, J = 31.5 Hz), 139.96, 129.17, 128.84, 127.96, 126.57, 122.28 (q, J = 275.94 Hz), 113.53(q, J = 6.3Hz), 111.57 (q, J = 1.26 Hz), 106.09, 100.21, 57.25, 40.92, 36.20; HR-ESI-MS m/z: 429.1038 [M + Na]+ (calculated for C20H17F3N2NaO4, 429.1038).
1-(2-Chlorophenyl)-3-(7-methoxy-2-oxo-4-(trifluoromethyl)-2H-chromen-6-yl) urea (5j). Yellow solid; yield: 45.0%; m.p. 231 °C; 1H-NMR (500 MHz, DMSO) δ: 9.76 (s, 1H), 8.64 (s, 1H), 8.52 (s, 1H), 7.96 (s, 1H), 7.68–7.44 (m, 2H), 7.33 (d, J = 6.2 Hz, 1H), 7.27 (s, 1H), 6.83 (s, 1H), 4.03 (s, 3H); 13C-NMR (126 MHz, DMSO) δ: 159.34, 152.77, 152.54, 151.06, 140.72, 140.24 (q, J = 32.76 Hz), 130.54, 130.08 (q, J = 31.5 Hz), 126.78, 122.25 (q, J =274.68 Hz), 122.16, 118.80 (q, J = 3.6 Hz), 114.32 (q, J = 4.0 Hz), 113.83 (q, J = 5.2 Hz), 112.27, 106.13, 100.50, 57.44; HR-ESI-MS m/z: 435.0328, 437.0298 [M + Na]+ (calculated for C18H12ClF3N2NaO4, 435.0335, 437.0306).
1-(3-Chlorophenyl)-3-(7-methoxy-2-oxo-4-(trifluoromethyl)-2H-chromen-6-yl) urea (5k). Yellow solid; yield: 41.5%; m.p. 241 °C; 1H-NMR (500 MHz, DMSO) δ: 9.57 (s, 1H), 8.62 (s, 1H), 8.50 (s, 1H), 7.69 (s, 1H), 7.32–7.25 (m, 2H), 7.21 (d, J = 8.0 Hz, 1H), 7.02 (d, J = 7.7 Hz, 1H), 6.83 (s, 1H), 4.01 (s, 3H); 13C-NMR (126 MHz, DMSO) δ: 159.36, 152.70, 152.54, 151.04, 141.38, 140.24 (q, J = 31.2 Hz), 133.77, 131.00, 126.83, 122.27(q, J = 275.94 Hz), 122.21, 117.87, 117.03, 113.91 (q, J = 6.3 Hz), 112.28 (q, J = 1.6 Hz), 106.15, 100.54, 57.47; HR-ESI-MS m/z: 435.0323, 437.0294 [M + Na]+ (calculated for C18H12ClF3N2NaO4, 435.0335, 437.0306).
1-(4-Chlorophenyl)-3-(7-methoxy-2-oxo-4-(trifluoromethyl)-2H-chromen-6-yl) urea (5l). White solid; yield: 44.8%; m.p. 243 °C; 1H-NMR (500 MHz, DMSO) δ: 9.52 (s, 1H), 8.67 (s, 1H), 8.51 (s, 1H), 7.49 (d, J = 8.5 Hz, 2H), 7.39–7.27 (m, 3H), 6.87 (s, 1H), 4.04 (s, 3H); 13C-NMR (126 MHz, DMSO) δ: 159.39, 152.75, 152.52, 150.97, 140.34 (q, J = 52.92 Hz), 138.85, 129.22, 126.97, 126.06, 122.29 (q, J = 287.28 Hz), 120.11, 113.92 (q, J = 2.52 Hz), 112.22 (q, J = 2.2 Hz), 106.17, 100.53, 57.48; HR-ESI-MS m/z: 435.0323, 437.0294 [M + Na]+ (calculated for C18H12ClF3N2NaO4, 435.0335, 437.0306).
1-(3-Fluorophenyl)-3-(7-methoxy-2-oxo-4-(trifluoromethyl)-2H-chromen-6-yl) urea (5m). Yellow solid; yield: 53.8%; m.p. 292 °C; 1H-NMR (500 MHz, DMSO) δ: 9.62 (s, 1H), 8.66 (s, 1H), 8.55 (s, 1H), 7.48 (d, J = 11.8 Hz, 1H), 7.38–7.27 (m, 2H), 7.11 (d, J = 8.1 Hz, 1H), 6.87 (s, 1H), 6.81 (t, J = 8.4 Hz, 1H), 4.04 (s, 3H); 13C-NMR (126 MHz, DMSO) δ: 162.90 (d, J = 240 Hz), 159.37, 152.71, 152.53, 151.01, 141.69 (d, J = 11.3 Hz), 140.25 (q, J = 32.76 Hz), 130.92 (d, J = 9.8 Hz), 126.86, 122.26 (q, J = 275.94 Hz), 114.33 (d, J = 2.3 Hz), 113.85 (q, J = 6.3 Hz), 112.26, 108.90 (d, J = 21.42 Hz), 106.14, 105.28 (d, J = 26.46 Hz), 100.50, 57.45; HR-ESI-MS m/z: 419.0620 [M + Na]+ (calculated for C18H12F4N2NaO4, 419.0631).
1-(2,4-Difluorophenyl)-3-(7-methoxy-2-oxo-4-(trifluoromethyl)-2H-chromen-6-yl) urea (5n). Light yellow solid; yield: 47.6%; m.p. 240 °C; 1H-NMR (500 MHz, DMSO) δ: 9.28 (s, 1H), 8.95 (s, 1H), 8.67 (s, 1H), 8.13 (q, J = 10, 1H), 7.35–7.26 (m, 2H), 7.05 (t, J = 8.6 Hz, 1H), 6.85 (s, 1H), 4.04 (s, 3H); 13C-NMR (126 MHz, DMSO) δ: 159.38, 157.39 (dd, J1 =240 Hz, J2 =11.25 Hz), 152.86, 152.68 (dd, J1 = 243.75 Hz, J2 = 12.5 Hz), 152.64, 151.01, 140.27 (q, J = 31.5 Hz), 126.96, 124.24 (dd, J1 = 7.5 Hz, J2 = 3.75 Hz), 122.69 (dd, J1 = 6.25 Hz, J2 = 2.5 Hz), 122.25 (q, J = 273.75 Hz), 113.82 (q, J = 6.3 Hz), 112.40 (q, J = 2.0 Hz), 111.52 (dd, J1 = 21.25 Hz, J2 = 3.75 Hz), 106.12, 104.25 (dd, J1 = 22.5 Hz, J2 = 3.75 Hz), 100.51, 57.42; HR-ESI-MS m/z: 437.0527 [M + Na]+ (calculated for C18H11F5N2NaO4, 437.0537).
1-(m-Tolyl)-3-(7-methoxy-2-oxo-4-(trifluoromethyl)-2H-chromen-6-yl) urea (5o). Yellow solid, yield: 24.7%; m.p. 217–218 °C; 1H-NMR (500 MHz, DMSO) δ: 9.33 (s, 1H), 8.68 (s, 1H), 8.47 (s, 1H), 7.32 (s, 1H), 7.29 (s, 1H), 7.23 (d, J = 8.3 Hz, 1H), 7.18 (t, J = 7.7 Hz, 1H), 6.86 (s, 1H), 6.82 (d, J = 7.3 Hz, 1H), 4.04 (s, 3H), 2.30 (s, 3H); 13C-NMR (126 MHz, DMSO) δ: 159.39, 152.83, 152.47, 150.84, 140.30 (q, J = 31.5 Hz), 139.80, 138.56, 129.19, 127.22, 123.31, 122.29 (q, J = 275.94 Hz), 119.07, 115.82, 113.78 (q, J = 5.04 Hz), 112.03 (q, J = 1.6 Hz), 106.15, 100.46, 57.44, 21.70; HR-ESI-MS m/z: 415.0874 [M + Na]+ (calculated for C19h15f3n2nao4, 415.0882).
1-(3-(Trifluoromethyl)phenyl)-3-(7-methoxy-2-oxo-4-(trifluoromethyl)-2H-chromen-6-yl) urea (5p). Light yellow solid; yield 39.0%; m.p. 219 °C; 1H-NMR (500 MHz, DMSO) δ: 9.24 (s, 1H), 9.06 (s, 1H), 8.66 (s, 1H), 8.08 (d, J = 8.1 Hz, 1H), 7.46 (d, J = 7.7 Hz, 1H), 7.35–7.28 (m, 2H), 7.07 (t, J = 7.3 Hz, 1H), 6.86 (s, 1H), 4.05 (s, 3H); 13C-NMR (126 MHz, DMSO) δ: 159.40, 152.95, 151.10, 140.26(q, J = 31.5 Hz), 136.13, 129.76, 127.98, 126.94, 124.36, 123.32, 123.17, 122.27 (q, J = 275.94 Hz), 118.98, 113.84 (q, J = 6.3 Hz), 112.99, 106.11, 100.56, 57.43; HR-ESI-MS m/z: 469.0593 [M + Na]+ (calculated for C19H12F6N2NaO4, 469.0599).
1-(4-(Trifluoromethyl)phenyl)-3-(7-methoxy-2-oxo-4-(trifluoromethyl)-2H-chromen-6-yl) urea (5q). Yellow solid; yield: 16.9%; m.p. 212–213 °C; 1H-NMR (500 MHz, DMSO) δ: 9.80 (s, 1H), 8.67 (s, 1H), 8.59 (s, 1H), 7.66 (q, J = 9.0 Hz, 4H), 7.29 (s, 1H), 6.86 (s, 1H), 4.04 (s, 3H); 13C-NMR (126 MHz, DMSO) δ : 159.35, 152.62, 152.56, 151.08, 143.58, 140.24 (q, J = 32.76 Hz), 126.74, 126.65 (q, J = 3.78 Hz), 125.00 (q, J = 270 Hz), 122.47 (q, J = 31.5 Hz), 122.26 (q, J = 275.94 Hz), 118.28, 113.88 (q, J = 6.3 Hz), 112.34, 106.14, 100.54, 57.47; HR-ESI-MS m/z: 469.0593 [M + Na]+ (calculated for C19H12F6N2NaO4, 469.0599).
1-(2-Methoxyphenyl)-3-(7-methoxy-2-oxo-4-(trifluoromethyl)-2H-chromen-6-yl) urea (5r). Light yellow solid, yield: 30.9%; m.p. 152 °C; 1H-NMR (500 MHz, DMSO) δ: 9.15 (s, 1H), 9.01 (s, 1H), 8.69 (s, 1H), 8.09 (dd, J1 = 7.9 Hz, J2 = 1.1 Hz, 1H), 7.27 (s, 1H), 7.06–6.95 (m, 2H), 6.93–6.87 (m, 1H), 6.84 (s, 1H), 4.03 (s, 3H), 3.88 (s, 3H); 13C-NMR (126 MHz, DMSO) δ: 159.42, 153.15, 152.90, 150.89, 148.76, 140.35 (q, J = 32.76 Hz), 128.81, 127.36, 122.77, 122.28 (q, J = 275.94 Hz), 120.94, 119.89, 113.65 (q, J = 6.3 Hz), 112.69 (q, J = 1.9 Hz), 111.34, 106.08, 100.40, 57.28, 56.17; HR-ESI-MS m/z: 431.0826 [M + Na]+ (calculated for C19H15F3N2NaO5, 431.0831).
1-(3-Methoxyphenyl)-3-(7-methoxy-2-oxo-4-(trifluoromethyl)-2H-chromen-6-yl) urea (5s). Yellow solid; yield: 37.8%; m.p. 196 °C; 1H-NMR (500 MHz, DMSO) δ: 9.40 (s, 1H), 8.67 (s, 1H), 8.47 (s, 1H), 7.29 (s, 1H), 7.20 (t, J = 8.1 Hz, 1H), 7.11 (t, J = 2.1 Hz, 1H), 6.98 (dd, J1 = 8.0 Hz, J2 = 1.1 Hz, 1H), 6.86 (s, 1H), 6.58 (dd, J1 = 8.2 Hz, J2 =2.3 Hz, 1H), 4.04 (s, 3H), 3.75 (s, 3H); 13C-NMR (126 MHz, DMSO) δ: 160.21, 159.39, 152.78, 152.52, 150.90, 141.06, 140.29(q, J = 32.76 Hz), 130.16, 127.12, 122.29 (q, J = 275.94 Hz), 113.82 (q, J = 6.3Hz) 112.13 (q, J = 1.9 Hz), 111.00, 107.86, 106.16, 104.50, 100.49, 57.45, 55.46; HR-ESI-MS m/z: 431.0829 [M + Na]+ (calculated for C19H15F3N2NaO5, 431.0831).
1-(4-Methoxyphenyl)-3-(7-methoxy-2-oxo-4-(trifluoromethyl)-2H-chromen-6-yl) urea (5t). Light yellow solid; yield: 37.7%; m.p. 212–213 °C; 1H-NMR (500 MHz, DMSO) δ: 9.21 (s, 1H), 8.68 (s, 1H), 8.40 (s, 1H), 7.36 (d, J = 7.9 Hz, 2H), 7.28 (s, 1H), 6.91–6.83 (m, 3H), 4.03 (s, 3H), 3.72 (s, 3H); 13C-NMR (126 MHz, DMSO) δ: 159.43, 155.10, 153.01, 152.42, 150.75, 140.32 (q, J = 31.5 Hz), 132.85, 127.36, 122.28 (q, J = 275.94 Hz), 120.42, 114.55, 113.76 (q, J = 5.04 Hz), 111.94 (q, J = 1.1 Hz), 106.16, 100.42, 57.43, 55.66; HR-ESI-MS m/z: 431.0826 [M + Na]+ (calculated for C19H15F3N2NaO5, 431.0831).
1-(4-(Trifluoromethoxy) phenyl)-3-(7-methoxy-2-oxo-4-(trifluoromethyl)-2H-chromen-6-yl) urea (5u). Light yellow solid; yield: 43.7%; m.p. 209 °C; 1H-NMR (500 MHz, DMSO) δ: 9.61 (s, 1H), 8.70 (s, 1H), 8.54 (s, 1H), 7.62–7.56 (m, 2H), 7.39–7.29 (m, 3H), 6.88 (s, 1H), 4.07 (s, 3H); 13C-NMR (126 MHz, DMSO) δ: 159.37, 152.79, 152.52, 150.98, 143.24 (q, J = 2.52 Hz), 140.28 (q, J = 31.5 Hz), 139.12, 126.95, 122.29, 122.27 (q, J = 273.75 Hz), 120.67 (q, J = 273.75 Hz), 119.84, 113.86 (q, J = 5.04 Hz), 112.25, 106.16, 100.52, 57.47; HR-ESI-MS m/z: 463.3020 [M + Na]+ (calculated for C19H13F6N2O5, 463.3024).

3.2. Determination of UV Absorption and Fluorescence Spectrum

The title compounds were dissolved in methanol to prepare 1 and 10 μg·mL−1 solutions, and the same concentrations of the reference compound, quinine sulfate, were obtained using 0.1 mol·L−1 H2SO4 aqueous solution as the solvent. The 10 μg·mL−1 solutions of all the compounds were used to record UV absorption spectra. The intersection point wavelength of UV spectrums of title compound and quinine sulfate that lay between 300 to 400 nm was selected as the excitation wavelength to record the fluorescence spectrum. The 1 μg·mL−1 solutions were used to determine the fluorescence to ensure the absorbance was less than 0.05. The refraction indexes (n) of the 1 μg·mL−1 solutions were also measured to calculate the Φf. Δλ and Φf were calculated according to the following formulas [56]:
Δλ = λmaxem − λmaxab
Φf = (n2x·Dx·Φfstd)/(n2std·Dstd),
where nx, nstd—refraction index of title compound and reference compound, respectively; Dx, Dstd—fluorescence peak area of title compound and reference compound, respectively; Φfstd—fluorescence quantum efficiency of reference compound with value of 0.55.

3.3. Procedures for Activities Evaluation

3.3.1. Herbicidal Activity

The inhibition of the title compounds against the taproot and caulis growth of dicotyledonous weed A. retroflexus and monocotyledonous weed D. sanguinalis were determined in vitro [57]. Two-milligram and 10-mg portions of the title compounds were dissolved in 1 mL of acetone to give 2 and 10 g·L−1 of stock solutions, respectively. A suspension of 0.3 g agar powder in 60 mL distilled water was microwave heated to melt. Then 0.6 mL of the stock solution was added to 60 mL of the melting agar of about 50 °C to achieve the required concentration. Then, 10 mL of the compound–agar medium was poured into a small beaker (10 mL) and cooled to room temperature. After that, 10 just-germinated weed seeds were planted onto the surface of the medium. Then the beaker was sealed with a piece of plastic wrap with several small holes made in it by a needle. Finally, cultivation was achieved in an illumination incubator (28 ± 1 °C, 50–55% relative humidity, 12 h L: 12 h D) for seven days. Acetone was used as a blank control, and Acetochlor was used as a positive control. Each treatment was repeated three times. After cultivation, the taproot and caulis lengths of the weed seedlings were measured, and the growth inhibitory rate of the treatment to the untreated control was determined.

3.3.2. Antifungal Activity

The in vitro antifungal activities of the title compounds against the mycelium growth of four phytopathogenic fungi, V. mali, B. cinerea, F. oxysporium, and C. glecosporioides, were tested [57]. The stock solutions of the title compounds were prepared according to the procedures in Section 3.3.1. Then 1 mL of the stock solutions was added to 100 mL portions of melted (~50 °C) potato dextrose agar (PDA), and the mixtures were fully shaken to obtain the required concentrations of compound mixed medium. Then 5 mL portions of the mixed medium were poured into 6 cm petri dishes and allowed to cool to ambient temperature to form solid plates. After that, one 4 mm activated mycelium disk was inoculated on each of the PDA plates and incubated at 28 °C in the dark for a sufficient period. Acetone was used as a blank control, and Carbendazim was used as a positive control. Each treatment was repeated three times. The radius (mm) of the fungus mycelium settlements was measured and the growth inhibition rate relative to the untreated control was calculated.

4. Conclusions

In conclusion, 21 novel trifluoromethylcoumarinyl urea derivatives have been designed and synthesized according to the principle of bioactive substructure combination. The fluorescence spectrums of the title compounds showed that most of them glow green with λmaxem of 500–517 nm when illuminated by ultraviolet light, while compounds 5r, 5s, 5u, and 5l glow violet with a λmaxem of 381–443 nm. However, the fluorescent intensity of all the title compounds is weaker than the standard reference compound, quinine sulfate. Moreover, the pesticidal activities evaluation of the title compounds indicated that compound 5f against the caulis of A. retroflexus and compounds 5j and 5l against the taproot of D. sanguinalis are equivalent to the commercial herbicide Acetochlor. Nine of the title compounds are more antifungal than commercial fungicide Carbendazim against B. cinerea.

Supplementary Materials

Supplementary materials are available online. Figures S1–S71.

Acknowledgments

This work was supported by the National Natural Science Foundation of China (No. 31471808) and partially by QAU Undergraduate Training Program for Innovation under grant (No. 2017562).

Author Contributions

S.H. conceived and designed the experiments; L.Q. performed the experiments and analyzed the data; S.H. wrote the paper.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Umadevi, P.; Deepti, K.; Srinath, I.; Vijayalakshmi, G.; Tarakaramji, M. Synthesis and in-vitro antibacterial activity of some new urea, thiourea and thiosemicarbazide derivatives. Int. J. Pharm. Pharm. Sci. 2012, 4, 379–383. [Google Scholar]
  2. Rajtar, B.; Szacon, E.; Swiatek, L.; Rzadkowska, M.; Matosiuk, D.; Mal-gorzata, P.D. Antiviral activity of 1-(1-arylimidazolidine-2-ylidene)-3-(4-chlorobenzyl)urea derivatives. J. Pre-Clin. Clin. Res. 2013, 7, 104–106. [Google Scholar]
  3. Venkatachalam, T.K.; Mão, C.; Uckun, F.M. Effect of stereochemistry on the anti-HIV activity of chiral thiourea compounds. Bioorg. Med. Chem. 2004, 12, 4275–4284. [Google Scholar] [CrossRef] [PubMed]
  4. Zeldin, R.K.; Petruschke, R.A. Pharmacological and therapeutic properties of ritonavir-boosted protease inhibitor therapy in HIV-infected patients. J. Antimicrob. Chemother. 2004, 53, 4–9. [Google Scholar] [CrossRef] [PubMed]
  5. Anderson, J.W.; Sarantakis, D.; Terpinski, J.; Kumar, T.R.; Tsai, H.C.; Kuo, M.; Ager, A.L.; Jacobs, J.W.R.; Schiehser, G.A.; Ekins, S.; et al. Novel diaryl ureas with efficacy in a mousemodel of malaria. Bioorg. Med. Chem. Lett. 2013, 23, 1022–1025. [Google Scholar] [CrossRef] [PubMed]
  6. Cho, H.J.; EI-Gamal, M.I.; Oh, C.H.; Lee, S.H.; Kim, G.; Jun, H.H.; Hong, S.C.; Kyung, H.Y. Synthesis and antiproliferative activity of new aminoisoquinolinylurea derivatives against melanoma cell line. Bull. Korean Chem. Soc. 2012, 33, 3635–3639. [Google Scholar] [CrossRef]
  7. Lu, C.S.; Tang, K.; Li, Y.; Jin, B.; Yin, D.L.; Chen, X.G.; Ma, C.; Huang, H.H. Synthesis and in vitro antitumor activities of novel benzyl urea analogues of sorafenib. Yao Xue Xue Bao 2013, 48, 709–717. [Google Scholar] [PubMed]
  8. Mathieu, G.B.; Sébastien, F.; Jacques, L.; Lefebvre, C.A.; Côté, M.F.; René, C.G. Styryl-N-phenyl-N’-(2-chloroethyl)ureas and styrylphenylimidazolidin-2-ones as new potent microtubule-disrupting agents using combretastatin A-4 as model. Eur. J. Med. Chem. 2015, 100, 34–43. [Google Scholar]
  9. Nimbarte, V.D.; Murtuza, H.; Phaniraj, S.; Shrivastava, S.; Naidu, V.G.M.; Kumar, N.S.; Atcha, K.R. Design, synthesis and biological evaluation of 4-(1-(4(sulphanilamide)phenyl)-3-(methyl)-1H-pyrazol-5-yl)dine urea and N-acyl derivatives as a soluble epoxide hydrolase inhibitors. Med. Chem. Res. 2014, 23, 2178–2197. [Google Scholar] [CrossRef]
  10. Vijayakumar, S.; Bongu, S.; Gomathi, P. Anti-ulcerogenic activity of unsymmetrically substituted urea derivative (1-benzyl-3-(4-methylphenyl) urea. Afr. J. Pharm. Pharmacol. 2013, 7, 2522–2527. [Google Scholar] [CrossRef]
  11. Ramesh, D.; Gangadhar, S.; Katti, S.A. Synthesis, characterization and anticonvulsant activity of 1-(aryl)-3-(diphenylmethyl) urea derivatives. Int. J. Res. Pharm. Chem. 2013, 3, 748–752. [Google Scholar]
  12. Lorena, S.; Lima, L.A.; Valdir, C.F.; Rogério, C.; Fátima, C.B.; Nunes, R.J. Synthesis of new 1-phenyl-3-{4-[(2E)-3-phenylprop-2-enoyl]phenyl}-thiourea and urea derivatives with anti-nociceptive activity. Bioorg. Med. Chem. 2008, 16, 8526–8534. [Google Scholar]
  13. Mishra, C.B.; Kumari, S.; Tiwari, M. Design and synthesis of some new 1-phenyl-3/4-[4-(aryl/heteroaryl/alkyl-piperazine1-yl)-phenyl-ureas as potent anticonvulsant and antidepressant agents. Arch. Pharm. Res. 2016, 39, 603–617. [Google Scholar] [CrossRef] [PubMed]
  14. Zhang, M.; Yang, X.Y.; Tang, W.; Groeneveld, T.W.L.; He, P.L.; Zhu, F.H.; Li, J.; Lu, W.; Blom, A.M.; Zuo, J.P.; et al. Discovery and Structural Modification of 1-Phenyl-3-(1-phenylethyl)urea Derivatives as Inhibitors of Complement. ACS Med. Chem. Lett. 2012, 3, 317–321. [Google Scholar] [CrossRef] [PubMed]
  15. Furman, C.; Lebeau, J.; Fruchart, J.C.; Bernier, J.L.; Duriez, P.; Cotelle, N.; Teissier, E. Di-tert-butylhydroxylated flavonoids protect endothelial cells against oxidized LDL-induced cytotoxicity. J. Biochem. Mol. Toxicol. 2001, 15, 270–278. [Google Scholar] [CrossRef] [PubMed]
  16. Kim, Y.J.; Ryu, J.H.; Cheon, Y.J.; Lim, H.J.; Jeon, R. Design and synthesis of urea and thiourea derivatives and their inhibitory activities on lipopolysaccharide-induced NO production. Bioorg. Med. Chem. Lett. 2007, 17, 3317–3321. [Google Scholar] [CrossRef] [PubMed]
  17. Yang, Y.; Liu, Y.X.; Song, H.J.; Li, Y.Q.; Wang, Q.M. Design, synthesis, insecticidal activity, and structure-activity relationship (SAR): Studies of novel triazone derivatives containing a urea bridge group based on transient receptor potential (TRP) channels. Mol. Divers. 2016, 20, 919–932. [Google Scholar] [CrossRef] [PubMed]
  18. Jadhav, M.R.; Oulkar, D.P.; Shabeer, T.P.A.; Banerjee, K. Quantitative screening of agrochemical residues in fruits and vegetables by buffered ethyl acetate extraction and LC-MS/MS analysis. J. Agric. Food Chem. 2015, 63, 4449–4456. [Google Scholar] [CrossRef] [PubMed]
  19. George, L. Herbicidal Sulfonamide Combinations. Patent DE2853561A1, 8 May 1980. [Google Scholar]
  20. Zhang, Z.W.; Yang, H.H.; Gao, Z.H.; Yuan, Y.H.; Dong, J.; Wang, Y.; Yue, T.L. Identification, Synthesis, and Safety Assessment of Thidiazuron [1-Phenyl-3-(1,2,3-thidiazol-5-yl) urea] and Its Metabolites in Kiwifruits. J. Agric. Food Chem. 2017, 65, 11273–11279. [Google Scholar] [CrossRef] [PubMed]
  21. Schiedel, M.S.; Briehn, C.A.; Baüerle, P. Single-compound libraries of organic materials: Parallel synthesis and screening of fluorescent dye. Angew. Chem. Int. Ed. 2001, 40, 4677–4680. [Google Scholar] [CrossRef]
  22. Swanson, S.A.; Wallraff, G.M.; Chen, J.P.; Zhang, W.; Bozano, L.D.; Carter, K.R.; Salem, J.R.; Villa, R.; Scott, J.C. Stable and efficient fluorescent red and green dyes for external and internal conversion of blue OLED emission. Chem. Mater. 2003, 15, 2305–2312. [Google Scholar] [CrossRef]
  23. Lee, M.T.; Yen, C.K.; Yang, W.P.; Chen, H.H.; Liao, C.H.; Tsai, C.H.; Chen, C.H. Efficient green coumarin dopants for organic light-emitting devices. Org. Lett. 2004, 6, 1241–1244. [Google Scholar] [CrossRef] [PubMed]
  24. Yu, T.; Zhang, P.; Zhao, Y.; Zhang, H.; Meng, J.; Fan, D. Synthesis, characterization and high-efficiency blue electroluminescence based on coumarin derivatives of 7-diethylamino-coumarin-3-carboxamide. Org. Electron. 2009, 10, 653–660. [Google Scholar] [CrossRef]
  25. Jung, H.S.; Kwon, P.S.; Lee, J.W.; Kim, J.I.I.; Hong, C.S.; Kim, J.W.; Yan, S.; Lee, J.Y.; Lee, J.H.; Joo, T.; et al. Coumarin-derived Cu2+-selective fluorescence sensor: Synthesis, mechanisms, and applications in living cells. J. Am. Chem. Soc. 2009, 131, 2008–2012. [Google Scholar] [CrossRef] [PubMed]
  26. Pang, L.; Zhou, Y.; Gao, W.; Song, H.; Wang, X.; Wang, Y. A highly selective and sensitive fluorescence probe for rapid detection of hypochlorite in tap water and cancer cells. RSC Adv. 2016, 6, 105795–105800. [Google Scholar] [CrossRef]
  27. Wu, C.J.; Wang, J.B.; Shen, J.J.; Bi, C.; Zhou, H.W. Coumarin-based Hg2+ fluorescent probe: Synthesis and turn-on fluorescence detection in neat aqueous solution. Sens. Actuators B Chem. 2017, 243, 678–683. [Google Scholar] [CrossRef]
  28. Paul, M.K.; Singh, Y.D.; Dey, A.; Saha, S.K.; Anwar, S.; Chattopadhyay, A.P. Coumarin based emissive rod shaped new Schiff base mesogens and their Zinc(II) complexes: Synthesis, photophysical, meso-morphism, gelation and DFT studies. Liq. Cryst. 2016, 43, 343–360. [Google Scholar] [CrossRef]
  29. Zhou, W.J.; Sun, X.Q.; Xi, H.T. Synthesis and fluorescence properties of hyaluronic acid derivatives containing fluorinated coumarin. Chin. J. Appl. Chem. 2014, 31, 916–921. [Google Scholar]
  30. Ostrowska, K.; Młodzikowska, K.; Głuch-Lutwin, M.; Gryboś, A.; Siwek, A. Synthesis of a new series of aryl/heteroarylpiperazinyl derivatives of 8-acetyl-7-hydroxy-4-methylcoumarin with low nanomolar 5-HT 1A affinities. Eur. J. Med. Chem. 2017, 137, 108–116. [Google Scholar] [CrossRef] [PubMed]
  31. Lei, L.; Xue, Y.B.; Liu, Z.; Peng, S.S.; He, Y.; Zhang, Y.; Fang, R.; Wang, J.P.; Luo, Z.W.; Yao, G.M.; et al. Coumarin derivatives from Ainsliaea fragrans and their anticoagulant activity. Sci. Rep. 2015, 5, 13544. [Google Scholar] [CrossRef] [PubMed]
  32. Radwan, E.; Reem, I.A. Anti-inflammatory screening and molecular modeling of some novel coumarin derivatives. Molecules 2015, 20, 5374–5391. [Google Scholar]
  33. Cong, N.T.; Nhan, H.T.; Van, H.L.; Thang, T.D.; Kuo, P.C. Synthesis and antibacterial activity of analogs of 5-arylidene-3-(4-methylcoumarin-7-yloxyacetylamino)-2-thioxo-1,3-thiazoli-din-4-one. Molecules 2014, 19, 13577–13586. [Google Scholar] [CrossRef] [PubMed]
  34. Šarkanj, B.; Molnar, M.; Cacic, M.; Gille, L. 4-Methyl-7-hydroxycoumarin antifungal and antioxidant activity enhancement by substitution with thiosemicarbazide and thiazolidinone moieties. Food Chem. 2013, 139, 488–495. [Google Scholar] [CrossRef] [PubMed]
  35. Bonacorso, H.G.; Rodrigues, M.B.; Rosa, W.C.; Silva, L.B.; Frizzo, C.P.; Zanatta, N.; Martins, M.A.P. Cyanoacetylazoles and salicylic aldehydes promoting the synthesis of new trifluoromethyl-substituted azolecarbonyl-2H-chromen-2-ones through the Knoevenagel condensation reaction. J. Fluorine Chem. 2015, 178, 296–305. [Google Scholar] [CrossRef]
  36. Liu, G.L.; Hu, Y.; Chen, X.H.; Wang, G.X.; Ling, F. Synthesis and anthelmintic activity of coumarin- imidazole hybrid derivatives against Dactylogyrus intermedius in goldfish. Bioorg. Med. Chem. Lett. 2016, 26, 5039–5043. [Google Scholar] [CrossRef] [PubMed]
  37. Pan, L.; Li, X.Z.; Yan, Z.Q.; Guo, H.R.; Qin, B. Phytotoxicity of umbelliferone and its analogs: Structure-activity relationships and action mechanisms. Plant Physiol. Biochem. 2015, 97, 272–277. [Google Scholar] [CrossRef] [PubMed]
  38. 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] [PubMed]
  39. Deshmukh, M.; Pawar, P.; Joseph, M.; Phalgune, M.; Kashalkar, U.; Deshpande, R.; Nirmala, R. Efficacy of 4-methy-7-hydroxy coumarin derivatives against vectors Aedes aegypti and Culex quinquefasciatus. Indian J. Exp. Biol. 2008, 46, 788–792. [Google Scholar]
  40. Chetan, T.P.; Sangamesh, A.P.; Ajaykumar, D.K.; Vinod, H.N.; Manjunathad, M.; Shivshankar, M.K.; Prema, S.B. Synthesis, spectral, thermal, fluorescence, antimicrobial, anthelmintic and DNA cleavage studies of mononuclear metal chelates of bi-dentate 2H-chromene-2-one Schiff base. J. Photochem. Photobiol. B 2015, 148, 322–332. [Google Scholar]
  41. Wei, Y.; Miao, K.L.; Hao, S.H. Novel 4-methylumbelliferone amide derivatives: Synthesis, characterization and pesticidal activities. Molecules 2018, 23, 122. [Google Scholar] [CrossRef] [PubMed]
  42. Chaabouni, S.; Simonet, F.; François, A.; Abid, S.; Galaup, C.; Chassaing, S. 3-Trifluoromethylated coumarins and carbostyrils via radical trifluoromethylation of ortho-functionalized cinnamic esters. Eur. J. Org. Chem. 2017, 2017, 271–277. [Google Scholar] [CrossRef]
  43. Zhou, Y.; Wang, J.; Gu, Z.; Wang, S.; Zhu, W.; Aceña, J.L.; Soloshonok, V.A.; Izawa, K.; Liu, H. Next generation of fluorine-containing pharmaceuticals, compounds currently in phase II–III clinical trials of major pharmaceutical companies: New structural trends and therapeutic areas. Chem. Rev. 2016, 116, 422–518. [Google Scholar] [CrossRef] [PubMed]
  44. Sebastián, B.V.; Lantaño, B.; Postigo, A. Recent advances in trifluoromethylation reactions with electrophilic trifluoromethylating reagents. Chem. Eur. J. 2014, 20, 16806–16829. [Google Scholar]
  45. Zhu, W.; Wang, J.; Wang, S.; Gu, Z.; Acena, J.L.; Izawa, K.; Liu, H.; Soloshonok, V.A. Recent advances in the trifluoromethylation methodology and new CF3-containing drugs. J. Fluorine Chem. 2014, 167, 37–54. [Google Scholar] [CrossRef]
  46. Kees, K.L.; Fitzgerald, J.J.; Steiner, K.E.; Mattes, J.F.; Mihan, B.; Tosi, T.; Mondoro, D.; McCalebr, M.L. New potent antihyperglycemic agents in db/db mice:  Synthesis and structure−activity relationship studies of (4-substituted benzyl) (trifluoromethyl) pyrazoles and -pyrazolones. J. Med. Chem. 1996, 39, 3920–3928. [Google Scholar] [CrossRef] [PubMed]
  47. Penning, T.D.; Talley, J.J.; Bertenshaw, S.R.; Carter, J.S.; Collins, P.W.; Docter, S.; Graneto, M.J.; Lee, L.F.; Malecha, J.W.; Miyashiro, J.M.; et al. Synthesis and biological evaluation of the 1,5-diarylpyrazole class of cyclooxygenase-2 inhibitors:  Identification of 4-[5-(4-methylphenyl)-3-(trifluoromethyl)-1H-pyrazol-1-yl]benzenesulfonamide (SC-58635, Celecoxib). J. Med. Chem. 1997, 40, 1347–1365. [Google Scholar] [CrossRef] [PubMed]
  48. Luzina, E.L.; Popov, A.V. Synthesis and anticancer activity of N-bis(trifluoromethyl)alkyl-N’-thiazolyl and N-bis(trifluoromethyl)alkyl-N’-benzothiazolyl ureas. Eur. J. Med. Chem. 2009, 44, 4944–4953. [Google Scholar] [CrossRef] [PubMed]
  49. Luzina, E.L.; Popov, A.V. Anticancer activity of N-bis(trifluoromethyl)alkyl-N’-(polychlorophenyl) and N’-(1,2,4-triazolyl) ureas. Eur. J. Med. Chem. 2010, 45, 5507–5512. [Google Scholar] [CrossRef] [PubMed]
  50. Lu, P.; Lin, Y.; Rodrigues, A.D.; Rushmore, T.H.; Baillie, T.A.; Shou, M. Testosterone, 7-benzyloxyquinoline, and 7-benzyloxy-4-trifluoromethyl-coumarin bind to different domains within the active site of cytochrome P450 3A4. Drug. Metab. Dispos. 2001, 29, 1473–1479. [Google Scholar] [PubMed]
  51. Liu, J.; Shah, M.B.; Zhang, Q.; Stout, C.D.; Halpert, J.R.; Wilderman, P.R. Coumarin derivatives as substrate probes of mammalian cytochromes P450 2B4 and 2B6: Assessing the importance of 7-alkoxy chain length, halogen substitution, and non-active site mutations. Biochemistry 2016, 55, 1997–2007. [Google Scholar] [CrossRef] [PubMed]
  52. Cox, S.W.; Eley, B.M. Preliminary studies on cysteine and serine proteinase activities in inflamed human gingiva using different 7-amino-4-trifluoromethyl coumarin substrates and protease inhibitors. Arch. Oral Biol. 1987, 32, 599–605. [Google Scholar] [CrossRef]
  53. Qiao, L.L.; Wei, Y.; Hao, S.H. Synthesis and Biological Activity of Novel Fluorinated Amide Hydroxy Methyl Coumarin Derivatives. Chin. J. Org. Chem. 2018, 38, 509–518. [Google Scholar] [CrossRef]
  54. Wang, H.S. Magnesium bis (trifluoromethane) sulfonimide: An efficient catalyst for the synthesis of coumarins under solvent-free conditions. Monatshte Fur Chem. 2013, 144, 411–414. [Google Scholar] [CrossRef]
  55. DeGrote, J.; Tyndall, S.; Wong, K.F.; VanAlstine-Parris, M. Synthesis of 7-alkoxy-4-trifluoromethylcoumarins via the von Pechmann reaction catalyzed by molecular iodine. Tetrahedron Lett. 2014, 55, 6715–6717. [Google Scholar] [CrossRef]
  56. Ci, Y.X.; Jia, X.A. Simplified method of measuring fluorescence quantum efficiency. Anal. Chem. 1986, 8, 616–618. [Google Scholar]
  57. Wei, Y.; Li, S.Q.; Hao, S.H. New angular oxazole-fused coumarin derivatives: Synthesis and biological activities. Nat. Prod. Res. 2017, 20, 1–8. [Google Scholar] [CrossRef] [PubMed]
Sample Availability: Samples of the compounds are not available from the authors.
Scheme 1. Synthetic route of title compounds.
Scheme 1. Synthetic route of title compounds.
Molecules 23 00600 sch001
Figure 1. Fluorescence spectrum of title compounds.
Figure 1. Fluorescence spectrum of title compounds.
Molecules 23 00600 g001
Figure 2. Herbicidal activities of title compounds.
Figure 2. Herbicidal activities of title compounds.
Molecules 23 00600 g002
Figure 3. Antifungal activities of title compounds.
Figure 3. Antifungal activities of title compounds.
Molecules 23 00600 g003
Table 1. Properties of UV absorption and fluorescence of title compounds.
Table 1. Properties of UV absorption and fluorescence of title compounds.
Compd.λmaxab
(nm)
Aεmax × 103
(L·mol−1·cm−1)
λmaxem
(nm)
Δλ (nm)Φf
5a3690.2167.135121430.114
5b3690.1866.405111420.110
5c3740.1806.195161430.065
5d3730.1886.735171450.044
5e3600.1897.305131540.087
5f3690.1605.925121440.064
5g3700.1545.925131430.053
5h3700.1506.905121420.070
5i3660.1295.245101440.082
5j3670.1827.505071410.098
5k3670.1717.055091430.024
5l3690.1747.17381120.028
5m3630.1626.715061430.013
5n3670.1837.255001340.020
5o3670.1646.43---
5p3630.1667.405041410.041
5q3670.1345.985031370.115
5r3720.1435.84443720.005
5s3630.1345.47396330.013
5t3650.1365.55---
5u3670.1667.67387210.013
- no fluorescence was detected.

Share and Cite

MDPI and ACS Style

Qiao, L.; Hao, S. Novel Trifluoromethylcoumarinyl Urea Derivatives: Synthesis, Characterization, Fluorescence, and Bioactivity. Molecules 2018, 23, 600. https://doi.org/10.3390/molecules23030600

AMA Style

Qiao L, Hao S. Novel Trifluoromethylcoumarinyl Urea Derivatives: Synthesis, Characterization, Fluorescence, and Bioactivity. Molecules. 2018; 23(3):600. https://doi.org/10.3390/molecules23030600

Chicago/Turabian Style

Qiao, Lili, and Shuanghong Hao. 2018. "Novel Trifluoromethylcoumarinyl Urea Derivatives: Synthesis, Characterization, Fluorescence, and Bioactivity" Molecules 23, no. 3: 600. https://doi.org/10.3390/molecules23030600

Article Metrics

Back to TopTop