Home Cart 0 Sign in  

[ CAS No. 7400-08-0 ] {[proInfo.proName]}

,{[proInfo.pro_purity]}
Cat. No.: {[proInfo.prAm]}
Chemical Structure| 7400-08-0
Chemical Structure| 7400-08-0
Structure of 7400-08-0 * Storage: {[proInfo.prStorage]}
Cart0 Add to My Favorites Add to My Favorites Bulk Inquiry Inquiry Add To Cart

Quality Control of [ 7400-08-0 ]

Related Doc. of [ 7400-08-0 ]

Alternatived Products of [ 7400-08-0 ]

Product Details of [ 7400-08-0 ]

CAS No. :7400-08-0 MDL No. :MFCD00004399
Formula : C9H8O3 Boiling Point : -
Linear Structure Formula :- InChI Key :NGSWKAQJJWESNS-ZZXKWVIFSA-N
M.W : 164.16 Pubchem ID :637542
Synonyms :
Chemical Name :p-Hydroxy-Cinnamic Acid

Calculated chemistry of [ 7400-08-0 ]

Physicochemical Properties

Num. heavy atoms : 12
Num. arom. heavy atoms : 6
Fraction Csp3 : 0.0
Num. rotatable bonds : 2
Num. H-bond acceptors : 3.0
Num. H-bond donors : 2.0
Molar Refractivity : 45.13
TPSA : 57.53 Ų

Pharmacokinetics

GI absorption : High
BBB permeant : Yes
P-gp substrate : No
CYP1A2 inhibitor : No
CYP2C19 inhibitor : No
CYP2C9 inhibitor : No
CYP2D6 inhibitor : No
CYP3A4 inhibitor : No
Log Kp (skin permeation) : -6.26 cm/s

Lipophilicity

Log Po/w (iLOGP) : 0.95
Log Po/w (XLOGP3) : 1.46
Log Po/w (WLOGP) : 1.38
Log Po/w (MLOGP) : 1.28
Log Po/w (SILICOS-IT) : 1.22
Consensus Log Po/w : 1.26

Druglikeness

Lipinski : 0.0
Ghose : None
Veber : 0.0
Egan : 0.0
Muegge : 1.0
Bioavailability Score : 0.56

Water Solubility

Log S (ESOL) : -2.02
Solubility : 1.58 mg/ml ; 0.00965 mol/l
Class : Soluble
Log S (Ali) : -2.27
Solubility : 0.873 mg/ml ; 0.00532 mol/l
Class : Soluble
Log S (SILICOS-IT) : -1.28
Solubility : 8.67 mg/ml ; 0.0528 mol/l
Class : Soluble

Medicinal Chemistry

PAINS : 0.0 alert
Brenk : 1.0 alert
Leadlikeness : 1.0
Synthetic accessibility : 1.61

Safety of [ 7400-08-0 ]

Signal Word:Warning Class:N/A
Precautionary Statements:P261-P280-P305+P351+P338 UN#:N/A
Hazard Statements:H302-H315-H319-H332-H335 Packing Group:N/A
GHS Pictogram:

Application In Synthesis of [ 7400-08-0 ]

* All experimental methods are cited from the reference, please refer to the original source for details. We do not guarantee the accuracy of the content in the reference.

  • Downstream synthetic route of [ 7400-08-0 ]

[ 7400-08-0 ] Synthesis Path-Downstream   1~88

  • 1
  • [ 67-56-1 ]
  • [ 7400-08-0 ]
  • [ 3943-97-3 ]
YieldReaction ConditionsOperation in experiment
100% With sulfuric acid for 24h; Reflux; General procedure for the synthesis of compounds 9-12 and 21-29 General procedure: After dissolving the carboxylic acid in the alcohol, three drops of H2SO4 95% were added to the solution and the mixture was refluxed for 24 h. The solvent was evaporated under reduced pressure and water was added to the crude mixture. The pH of the aqueous layer was adjusted to 7 adding drops of a saturated solution of NaHCO3 and brine was added in the mixture. The aqueous layer was extracted three times with ethyl acetate; the organic layer was dried over Na2SO4 and the solvent was evaporated under reduced pressure yielding the final compound. Further purification step was made when it was necessary.
99% With sulfuric acid for 7h; Heating;
99% With sulfuric acid for 24h; Reflux;
99% With sulfuric acid for 21h; Reflux; 15 (E)-Methyl 3-(4-hydroxyphenyl)acrylate (E)-Methyl 3-(4-hydroxyphenyl)acrylate (0403) (0404) p-Coumaric acid (3.0 g, 18.3 mmol) was suspended in MeOH (12 mL) and treated with 3 drops of conc. H2SO4. The mixture was refluxed for 21 h. The resulting clear solution was cooled to rt and concentrated under vacuum to yield a white solid that was dissolved in EtOAc and successively washed with sat. NaHCO3 solution (3×) and brine (3×). The organic layer was dried over anhydrous MgSO4 and evaporated to dryness under vacuum to afford 3.22 g (18.1 mmol, 99% yield) of a white solid identified by NMR as (E)-methyl 3-(4-hydroxyphenyl)acrylate. (0405) 1H NMR (400 MHz, CDCl3) δ 7.64 (d, J=16.0 Hz, 1H), 7.43 (ddd, J=8.8, 2.8, 2.0 Hz, 2H), 6.86 (ddd, J=8.8, 2.8, 2.0 Hz, 2H), 6.30 (d, J=16.0 Hz, 1H), 5.47 (br s, 1H), 3.80 (s, 3H).
98% Heating / reflux; 27.27a Step 27a. (E)-Methyl 3-(4-hydroxyphenyl)acrylate (Compound 0501-66) A mixture of 4-hydroxycinnamic acid (8.2 g, 50 mmol) and a drop of H2SO4 in methanol (30 mL) was heated to reflux overnight. Then the solvent was evaporated, the residue was dissolved in ethyl acetate, washed with saturated NaHCO3 solution twice, brine, dried over MgSO4, concentrated to give the title compound 0501-66 as white solid (8.7 g, 98%): LCMS: 179 [M+1]+.
98% With sulfuric acid Heating / reflux; 27.a Example 27Preparation of (E)-3-(4-(2-(4-(3-chloro-4-fluorophenylamino)-7-methoxyquinazolin-6-yloxy)ethoxy)phenyl)-N-hydroxyacrylamide (Compound 66)Step 27a. (E)-Methyl 3-(4-hydroxyphenyl)acrylate (Compound 0501-66); A mixture of 4-hydroxycinnamic acid (8.2 g, 50 mmol) and a drop of H2SO4 in methanol (30 mL) was heated to reflux overnight. Then the solvent was evaporated, the residue was dissolved in ethyl acetate, washed with saturated NaHCO3 solution twice, brine, dried over MgSO4, concentrated to give the title compound 0501-66 as white solid (8.7 g, 98%): LCMS: 179 [M+1]+.
97% With Dowex 50 W (8200 4) Heating;
97% With sulfuric acid for 12h; Reflux; Inert atmosphere;
96% With sulfuric acid for 16h; Heating;
95% With sulfuric acid for 24h; Molecular sieve; Reflux;
95% With sulfuric acid for 24h; Molecular sieve;
95% With acetyl chloride In methanol at 20℃; Inert atmosphere; Preparation of methyl 4-hydroxycinnamate To a solution of anhydrous methanol (50 mL) in round bottom flask under N2 was added acetyl chloride (0.5 mL) followed by a solution of 4-hydroxycinnamic acid (1 g) in anhydrous methanol (10 mL). The mixture was stirred overnight at room temperature, added NaCl solution (20%, 100 mL) then extracted twice with ethyl acetate (150 mL). The organic layers were combined and washed with NaCl solution (20%, 50 mL), dried over MgSO2, then evaporated under reduced pressure. The product was purified by vacuum column chromatography on silica gel to give the title compound as white solid (1.04 g, yield 95%). 1H- and 13C-NMR spectrum were in good agreement with previously reported data[45].
95% With sulfuric acid for 12h; Reflux;
94% With sulfuric acid for 0.166667h; Irradiation;
93% With oxalyl dichloride; N,N-dimethyl-formamide In dichloromethane for 0.5h;
92% With toluene-4-sulfonic acid for 24h; Reflux;
92% With thionyl chloride
90% With sulfuric acid for 24h; Heating;
89% for 90h; Reflux; Inert atmosphere; 12 4.12. Methyl (E)-p-coumarate (24) To a solution of p-coumaric acid 22 (3.0 g, 18.3 mmol) in MeOH (40 mL) was added Dowex 50W-X8 (2.4 g) at room temperature and the resulting mixture was refluxed for 90 h. After the Dowex 50W-X8 was filtered, the solvent was removed under reduced pressure and the residue was purified by flash column chromatography (30% AcOEt in hexane) to give a methyl ester with trace impurities. Recrystallization from CHCl3 gave 24 (2.91 g, 89%) as a white solid in a pure form. Mp: 130-132 °C; IR (KBr) 3384, 2952, 1689, 1634, 1602, 1517, 1435, 1284, 1200, 986, 834 cm-1; 1H NMR (600 MHz, CDCl3) δ 3.80 (s, 3H), 5.33 (s, 1H; OH), 6.30 (d, J=16.0 Hz, 1H), 6.85 (d, J=8.5 Hz, 2H), 7.43 (d, J=8.5 Hz, 2H), 7.64 (d, J=16.0 Hz, 1H); 13C NMR (150 MHz, CDCl3) δ 51.7, 115.1, 115.9, 127.1, 130.0, 144.8, 157.8, 168.1; HRMS (FAB) calcd for C10H11O3 [M+H]+ 179.0708, found 179.0700.
88% at 20℃; for 36h; Reflux; 1.7 (Example 1-7) Synthesis of intermediate 10 for glucose derivative () 3-(4-Hydroxyphenyl)methylacrylate (10) A mixed solution (amixture) of cinnamic acid (5 g, 30.48 mmol) was refluxed for 1.5 days in the presence of dry MeOH (300 mL) and concentrated (conc.) H2SO4 (1 drop). The solution was cooled to room temperature (r.t.) and then evaporated with 30% ammonia water under reduced pressure and neutralized. Finally, the solution was evaporated with EtOH, to thereby obtain a completely dried product. The dried residue was subj ected to silica gel column chromatography, to thereby obtain a colorless needle-like product 10 (4.8 g, 88%) from a fraction eluted with AcOEt-n-hexane (1:4, v/v). Melting point (m.p.) 122-123°C. Rf=0.23 (AcOEt:n-hexane, 1:8, v/v). IR (KBr) cm-1; 1600 (C=C), 1695 (C=O), 3380 (OH). 1H NMR (CDCl3, 300 MHz): δ 3.81 (s, 3H, O-Me), 5.92 [s, 1H, Ar-OH (exchangeable with D2O)], 6.32 (d, J=15.9 Hz, 1H, Ar-CH=CH-), 6.86 (d, 2H, J=8.4 Hz, Ar-H), 7.42 (d, 2H, J=8.4 Hz, Ar-H), 7.67 (d, J=15.9 Hz, 1H, Ar-CH=CH-).
87.98% With sulfuric acid Reflux; 4.2. General Procedure for Preparation of Compounds 1-8 General procedure: p-Coumaric acid (0.1 g; 0.61 mmol) in alcohol (20 mL) was dissolved in the presence of H2SO4(0.2 mL) and refluxed for complete reaction (5-27 h), being observed using single spot thin-layerchromatography (TLC) [71], as published by Lopes et al. [44].
87% With sulfuric acid for 24h; Reflux; 30 Example 30 (E) Synthesis of 3-(4-Hydroxyphenyl) Methyl Acrylate Add 4-hydroxycinnamic acid (1mmol, 164mg)Dissolved in anhydrous methanol (5mL),Add sulfuric acid (1 drop) under stirring at room temperature,The reaction was heated to reflux for 24 hours.Cool down the mixture,And the solvent was removed in vacuo.The residue was dissolved in ethyl acetate (30 mL),And use saturated sodium bicarbonate aqueous solution (20mL)Wash with saturated aqueous NaCl solution (2×20 mL).After filtering, dry with anhydrous Na2SO4,After the filtrate is concentrated,Separation and purification by column chromatography,The eluent is EtOAc:petroleum ether=1:5,A white solid compound is obtained,The yield was 87%.
85% With thionyl chloride at 0℃; Reflux; Inert atmosphere; 5.2. General procedure for preparation of esters General procedure: Thionyl chloride (1.16 g, 1.5 equiv) was added drop-wise to a solution of acid (1.0 g, 1.0 equiv) in the corresponding alcohol (15 ml) at 0&d eg;C. The solution was refluxed under a nitrogen atmosphere until all starting material was consumed (TLC monitoring). Then the solvent was removed under vacuo and the residue was purified by silica gel column chromatography eluting with ethyl acetate/n-hexane to afford the corresponding carboxylic esters.
82% With sulfuric acid for 5.5h; Heating;
82% With sulfuric acid at 80℃; for 0.25h; Microwave irradiation; 4.1.3. General Procedure for the Synthesis of Compounds 14-17 General procedure: In a 30 mL glass pressure microwave tube, equipped with a magnetic stirrer bar, a fewdrops of concentrated sulphuric acid were added to a solution of hydroxycinnamic acid(p-coumaric acid or caffeic acid or ferulic acid) (1 eq) in methanol (for 14-16) or ethanol(for 17) (10 mL). The mixture was subjected to microwave irradiation (power: 150 W;temperature: 80 C for 14-16 and 98 C for 17) for 15 min, basified with aqueous sodiumbicarbonate (5% solution), and extracted with dichloromethane (3 5 mL). The collectedorganic phases were dried over anhydrous sodium sulphate and evaporated to dryness togive the pure ester. (E)-Methyl 3-(4-hydroxyphenyl)acrylate 14: white solid; 82%; mp 125-127 C ([58] 132-134 C); 1H-NMR 3.79 (s, 3H), 5.37 (bs, 1H), 6.28 (d, J = 16.0 Hz, 1H), 6.85 (m, Ar, 2H), 7.42(m, Ar, 2H), 7.63 (d, J = 16.0, 1H); 13C-NMR 51.75, 114.52, 115.91, 125.98, 129.89, 144.79,158.12, 167.65. Anal. Calcd. for C10H10O3 C, 67.41; H, 5.66; Found: C, 67.53; H, 5.56.
79% With sulfuric acid at 60℃; for 72h;
78% With sulfuric acid at 45℃; for 2h;
78% With sulfuric acid for 24h; Heating;
77.8% With sulfuric acid at 80℃; for 5h; Prerjaration of reagent KR-28: Methyl (E)-3-(4-hydroxyrjhenyl)rjrorj-2-enoate A solution of the commercially available trans-4-hydroxycinnamic acid, 16, (5 g, 30.5 mmol) in MeOH (30 mL) was added H2504 (3 mL), then the mixture was stirred at 80 00 for 5 h. The resulting mixture was cooled to room temperature and concentrated under vacuo. The mixture was quenched into water, extracted with EA, dried with anhydrous Na2504, concentrated to givethe product KR-28 (4.2 g, 77.8%) as a pale yellow solid. ESI-MS (Mi-i): 179.1 calc. for 010H1003: 178.0.
75% With sulfuric acid for 2h; Reflux; Synthesis of (E)-methyl 3-(4-hydroxyphenyl)acrylate (13): A 5 gof trans-4-hydroxy cinnamic acid (12) (30.49 mmol, 1 equiv) was dissolved in 20 mL of methanol. A 0.33 mL of conc. H2SO4 (6.10 mmol, 0.2 equiv) was added. This reaction mixture was refluxed for 2 hr. Methanol was evaporated and the reaction mixture was washed with aq. NaHCO3 solution. Further it was extracted in ethyl acetate and concentrated to get desired compound. Light pale yellow liquid, Yield: 75%, Rf: 0.5 (2:8 EtOAc: hexane), IR (cm-1): 1595 (C=C), 1680 (C=O), 2357(C-H), 3281(O-H). 1H NMR (400 MHz, CDCl3): δ 3.71(s, 3H,CH3); 6.31 (d, 1H, J = 16 Hz, CH=CH); 6.79 (m, 2H); 7.44-7.47(m, 2H); 7.55 (d, 1H, J = 16 Hz, CH=CH); 9.85 (s, 1H, OH); 13C NMR (CDCl3): δ 50.00, 113.69, 115.66, 124.91, 129.85, 144.53, 159.76,166.87; MS-ESI (m/z):179.59 (M+1).
74.1% With boron trifluoride diethyl etherate at 70℃; for 2h; 1 Boron trifluoride etherate (0.067 mol) is added to a solution of coumaric acid (0.034 mol) in methanol (50 ml) in a 250 ml round bottom flask and the mixture stirred at 70° C. for 2 hours and evaporated to dryness under reduced vacuum. The residual solid is taken up in CH2Cl2, washed with aqueous NaHCO3, dried over MgSO4 and filtered to provide, after evaporation of the CH2Cl2, the desired product as a pale yellow solid in 74.1% yield.
74% With acetyl chloride at 20℃;
63% With sulfuric acid Heating;
58% With sulfuric acid for 2h; Reflux;
57% With sulfuric acid for 3h; 2.3. Synthesis of the esters General procedure: The ester derivatives (b-f) were synthesized following classicFischer esterification [18] using 1 mmol of the corresponding acid,0.1 mL of concentrated sulfuric acid and the corresponding alcohol assolvent. The reaction proceeded for 3 h or until no more acid was observedin TLC. The reaction mixture was neutralized with aqueousNaHCO3 saturated solution, and the solvent excess was evaporatedunder reduced pressure. The residue was taken up in 10 mL ethylacetate and washed with 3x10 mL of saturated NaHCO3 solution and10 mL of water. The organic layer was dried with anhydrous Na2SO4.The desired compounds were further purified in a silica gel columnchromatography, using hexane:ethyl acetate as eluent.Methyl p-coumarate (1b). White solid (m.p. 134-136 °C); 57% yield.1H NMR (CDCl3) δ 7.65 (d, J = 16.0 Hz, 1H), 7.42 (d, J = 8.6 Hz, 2H),6.86 (d, J = 8.6 Hz, 2H), 6.30 (d, J = 16.0 Hz, 1H), 6.11 (s, 1H), 3.81(s, 3H).
56% With sulfuric acid Reflux; 2.3. General procedure for the synthesis of the esters General procedure: The ester derivatives (p-coumarates 1a-c and cinnamates 2a-c) weresynthesized following classic Fischer esterification. In a flask, 1 mmol ofthe corresponding acid (1 or cinnamic acid 2) and 0.1 mL of concentratedsulfuric acid were added to an excess (10 mL) of correspondingalcohol (methanol, ethanol or isopropanol) as solvent. Thereaction mixture was stirred under reflux for 3-24 hours until no morestarting acid was present (monitored by thin layer chromatography,TLC). The acid was neutralized with saturated sodium bicarbonate solution,and the excess of alcohol was evaporated under reduced pressure.The residue was taken up in 15 mL ethyl acetate and washed threetimes with 15 mL saturated sodium bicarbonate solution and dried overanhydrous sodium sulfate. The solvent was removed and the crudeproduct was purified through open silica gel column chromatographyusing hexane:ethyl acetate as eluent. The yields are given after chromatographicpurification.
54.4% With sulfuric acid Reflux; 3.1 General procedure for the synthesis of compounds General procedure: A mixture of organic acid (0.5 g) and methanol (100 ml) was heated under reflux in presence of sulphuric acid (0.8 ml) until the completion of the reaction which was checked by single spot TLC. Then, methanol was removed under reduced pressure a half and the solution was diluted with 20 ml of water. The product was extracted with ethyl acetate (30 ml). The organic phase was neutralized successively with NaHCO3 5%and water, dried over anhydrous Na2SO4, and filtered. The ethyl acetate phase was separated, which on evaporation yielded the ester derivatives
17%
With sulfuric acid at 75 - 80℃; unter Druck;
With sulfuric acid
With hydrogenchloride for 5h;
With hydrogenchloride for 6h; Heating;
With Dowex 50W-8X cation-exhange resin for 3h;
With Dowex 50 W x 8200-400 Heating;
With sulfuric acid at 90℃; for 16h;
With hydrogenchloride Heating;
With thionyl chloride at 23℃; for 16h;
With toluene-4-sulfonic acid for 5h; Heating;
With sulfuric acid for 12h; Heating;
at 20℃;
With boron trichloride at 55℃; for 0.0833333h;
With sulfuric acid for 16h; Heating;
Stage #1: methanol With thionyl chloride at 0℃; for 0.5h; Stage #2: p-Coumaric Acid for 1.5h; Heating / reflux; 106 Thionyl chloride (1.75 mL) was dropwise added under stirring to methanol at 0°C. After 30 minutes 4-hydroxycinnamic acid (3.29 g) was added and the mixture was refluxed for 1.5 h. The solvent was evaporated in vacuo and the residue was crystalized from diisopropyl ether and hexane to give Compound (106) (2.41 g) as a white crystal. 1H-NMR (300 MHz, CDCL3, ) : 3.80 (3H, s), 5.44 (1H, s), 6.31 (1H, d, J=16 Hz), 6.85 (2xlH, d, J=8.5 Hz), 7.43 (2xlH, d, J=8.5 Hz), 7.64 (1H, d, J=16 Hz); MASS (ES-): m/e 177.
Stage #1: methanol With thionyl chloride at 0℃; for 0.5h; Stage #2: p-Coumaric Acid In methanol for 1.5h; Heating / reflux; 106 Preparation 106 Thionyl chloride (1.75 mL) was dropwise added under stirring to methanol at 0° C. After 30 minutes 4-hydroxycinnamic acid (3.29 g) was added and the mixture was refluxed for 1.5 h. The solvent was evaporated in vacuo and the residue was crystalized from diisopropyl ether and hexane to give Compound (106) (2.41 g) as a white crystal. 1H-NMR (300 MHz, CDCl3, δ): 3.80 (3H, s), 5.44 (1H, s), 6.31 (1H, d, J=16 Hz), 6.85 (2×1H, d, J=8.5 Hz), 7.43 (2×1H, d, J=8.5 Hz), 7.64 (1H, d, J=16 Hz); MASS (ES-): m/e 177.
With sulfuric acid for 2h; Reflux; Synthesis of methyl p-coumarate (4), methyl caffeate (5), and methyl ferulate (6) The appropriate phenolic acid (100 mg) was dissolved in MeOH (50 ml) to which 3 drops of H2SO4 had been added. The solution was refluxed for 2 h, NaHCO3 (100 mg) was added, and the solvent was evaporated in vacuo at 40 °C. The residue was partitioned between Et2O (15 mL) and H2O (15 mL), the organic layer separated, dried (Na2SO4), and the solvent was evaporated in vacuo to afford the desired ester, as a white solid. The esters were used in the oxidation reaction without further purification.
With sulfuric acid at 80℃; for 16h; 11.A [00324] Method A: A mixture of cinnamic acid (25 mmol) and concentrated H2S04 (0.1 ml_) in anhydrous MeOH (25 mL) was refluxed at 80¾ (bath temperature) for 16 h. After the excess MeOH was removed by a rotary evaporator, the residue was treated with H20 (50 mL), and the resulting mixture was extracted with EtOAc (3^50 mL). The combined extracts were washed with H20 (50 mL) and brine (50 mL), and dried (anhydrous Na2S04). The solvent was evaporated to dryness, and the crude products were purified by flash silica gel chromatography (eluting with 10-60% hexane in EtOAc) to afford methyl cinnamate. To an ice-cold solution of methyl cinnamate described above (8 mmol) dissolved in anhydrous MeOH (10 mL) and THF (10 mL) was added hydroxylamine hydrochloride (1.67g, 24 mmol, 3 equiv) followed by 25% sodium methoxide in methanol solution (8.4 mL, 36 mmol, 4.5 equiv). The reaction mixture was stirred under argon and at 0 for 2 h, then allowed to warm to ambient temperature with the stirring was continued overnight (16 h). The resulting yellow suspension was condensed to dryness with a rotary evaporator, and the residue was treated with 1 N HCI aqueous solution (30 mL). The mixture was extracted with EtOAc (3 <50 mL), and dried (anhydrous Na2S04). Evaporation of the solvent afforded the crude products (480 mg), which was purified by flash silica gel chromatography (eluting with 5-15% MeOH in DCM) to afford the hydroxamate.[00325] Using Method A, Compounds 103, 125, 129, 130, and 131 were synthesized. The carboxyl acid of PCA was converted to hydroxamite to synthesize compound 103 by reacting methyl ester 101 with hydroxylamine under basic conditions. Compound 103 was a light-brown solid, obtained at 25% yield (based on the methyl ester). The hydroxyl group of 103 was replaced by methoxy group, resulting in the hydroxamate 125. Compound 125 was light-brown solid, obtained at 67% yield. Compound 129 was light-brown solid, obtained at 21 % yield. Compound 130 was an off-white solid, obtained at 63% yield. Compound 131 was a grey solid, obtained at 25% yield. Identity of compounds was verified by H NMR (75 MHz, CD3OD).
With acetyl chloride
280 g With sulfuric acid for 5h; Reflux; 1 Example 1 First Step: (0174) trans-Coumaric acid (400 g) was added to methanol (1,200 ml) and concentrated sulfuric acid (10 g) was added dropwise, and the mixture was heated under reflux with stirring for 5 hours. After the reaction mixture had been cooled to room temperature, the methanol was distilled off under reduced pressure. The resulting residue was poured into ice-water (2,000 ml), and the mixture was extracted with ethyl acetate (2,000 ml). The combined organic layers were washed with a saturated aqueous solution of sodium hydrogencarbonate and water, and dried over anhydrous magnesium sulfate. The ethyl acetate was distilled off under reduced pressure, and the resulting residue was recrystallized from ethanol to give the compound (ex-1) (280 g).
15.6 g With acetyl chloride at 0 - 75℃; for 5h;
With sulfuric acid In methanol for 5h; Reflux;
With sulfuric acid for 1h; Reflux; Synthesis of cinnamic acid derivatives Zosteric acid was synthesized as already described in a previous work [16]. The cis isomer 38was obtained starting from cis 4-hydroxycinnamic acid 32 under microwave irradiation in thepresence of sulfur trioxide pyridine complex in acetonitrile (Fig 3). The final product was isolatedas sodium salt. Most of the substituted cinnamic acid derivatives were prepared in highyield (> 90%) by the Knoevenagel-Doebner procedure. In detail, compounds 2, 7, 8, 11 and 17were obtained through a one-pot reaction between the suitable substituted benzaldehyde andmalonic acid in refluxing pyridine to induce decarboxylation (Fig 4) [20]. The trans geometriesof the ethenyl π-bonds were confirmed by proton-proton coupling constants. Cis cinnamicacid 37 was synthesized from the commercially available ethyl phenylpropiolate. The subsequenthydrogenation of alkyne in the presence of the Lindlar catalyst and pyridine in methanolled to the corresponding cis-alkene 36. Then the ester group was hydrolyzed under alkaline conditions to provide the final compound 37 (Fig 5). Esters of cinnamic acid in cis (33) or intrans configuration (23, 24) were prepared by Fischer esterification of the carboxylic group[21]. The protection of the hydroxyl group as methyl ether in the presence of iodomethane indry N,N-dimethylformamide provided the compounds 35. The hydrolysis of the ester was performedin alkaline conditions to obtain compound 34 (Fig 3).
With sulfuric acid for 12h; Reflux; 3.2. General Method for Synthesizing Compounds 5-12 General procedure: Ferulic acid (1) or trans-4-hydroxycinnamic acid (2) or isoferulic acid (3) or trans-3-hydroxycinnamic acid (4) and five drops of H2SO4 (95%) were refluxed in methanol or ethanol for 12 h. The reaction mixture was concentrated in vacuo and the residue was dissolved in ethyl acetate. The organic layer was washed with a 5% aqueous NaHCO3 solution and water. After drying over anhydrous Na2SO4,the ethyl acetate was removed in vacuo. The residue was purified by column chromatography on silica gel using ethyl acetate/petroleum ether mixtures as eluents to afford compounds 5-12.
With hydrogenchloride In water at 65℃;
280 g With sulfuric acid for 5h; Reflux; 1.2 400 g of trans-p-coumaric acid was added to 1200 ml of methanol, 10 g of concentrated sulfuric acid was added dropwise, and the mixture was stirred under reflux for 5 hours. After cooling to room temperature, methanol was distilled off under reduced pressure. The obtained residue was poured into 2000 ml of ice water, 2000 ml of ethyl acetate And the organic layer was separated. The obtained organic layer was washed with saturated aqueous sodium bicarbonate and water, And dried over anhydrous magnesium sulfate. Ethyl acetate was distilled off under reduced pressure, The resulting residue was recrystallized from ethanol to obtain 280 g of Compound (ex-2).
With sulfuric acid for 24h; Reflux;
With thionyl chloride at 0 - 20℃; for 1h; 3.5.2. Preparation of M2 (methyl (E)-3-(4-((3,5,6-trimethylpyrazin-2-yl)methoxy)phenyl)acrylate)) To verify the accuracy of the proposed structure, we synthesized one of the metabolites, M2. Hydroxycinnamic acid (10.14 mmol) was added to anhydrous methanol (30 mL). After complete dissolution, thionyl chloride was slowly added dropwise under ice-cooling, while being stirred at 0 °C for 0.5 h, and then the temperature was slowly raised to room temperature. Then, the reaction solution was evaporated under reduced pressure. The reaction product was placed in a one-necked flask with potassium carbonate and N,N-dimethylformamide was added; the mixture was stirred at 65 °C for 3 h under nitrogen. Water was added to the reaction solution for dispersion before extraction with methylene chloride. After being evaporated, the residue was eluted with silica gel, and metabolite M2 was obtained. M2: m.p.: 124.0-124.4 °C. 1H-NMR (500 MHz, CDCl3) δ 7.64-7.61 (d, J = 7.6Hz, 1H, Ar-H), 7.46-7.44 (d, J = 7.4 Hz, 1H, Ar-H), 7.01-6.99 (d, J = 7.3 Hz, 1H, Ar-H), 6.31-6.28 (d, J = 6.3 Hz, 1H, Ar-H), 5.16 (s, 2H, -CH2), 3.78, 2.63 (s, 3H, -CH3), 2.57 (s, 3H, -CH3), 2.51 (s, 3H, -CH3) 13C-NMR (150 MHz, CDCl3) δ 167.92, 160.58, 151.73, 150.20, 148.92, 145.45, 144.61, 129.93, 127.80, 115.78, 115.64, 115.43, 70.22 (-CH2), 51.81 (-OCH3), 21.94 (-CH3), 21.65 (-CH3), 20.85 (-CH3), HR-MS (ESI) m/z: 313.16403 [M + H]+, calcd. for C18H20N2O3.
With sulfuric acid for 24h; Reflux;
With sulfuric acid for 48h; Reflux; (E)-Methyl 3-(4-hydroxyphenyl)acrylate (13) Acid 12 (1.06 g 6.46 mmol) was dissolved in dry methanol. 2-3drops of conc. sulfuric acid were added and solution obtained was refluxedfor a two days until disappearing of starting material (controlledby TLC, CH2Cl2-acetic acid 100:1). After completion of the reactionmethanol was evaporated under vacuum. A residue was re-dissolved inethyl acetate (30 mL) and washed with water (10 mL), brine (10 mL)and dried over magnesium sulfate. Solvent was evaporated and theresidue was used in following step without further purification.White-off powder, 92% yield. 1H NMR (400 MHz, CDCl3) δ 3.81 (s,3H), 6.30 (d, J=16.0 Hz, 1H), 6.53 (br.s, 1H), 6.82-6.91 (m, 2H), 7.41(d, J = 8.6 Hz, 2H), 7.64 (d, J = 16.0 Hz, 1H).
With acetyl chloride at 20℃; for 51h;

Reference: [1]Allegretta, Giuseppe; Weidel, Elisabeth; Empting, Martin; Hartmann, Rolf W. [European Journal of Medicinal Chemistry, 2015, vol. 90, p. 351 - 359]
[2]Pirrung, Michael C.; Chen, Jrlung; Rowley, Elizabeth G.; McPhail, Andrew T. [Journal of the American Chemical Society, 1993, vol. 115, # 16, p. 7103 - 7110]
[3]Ueda, Rie; Suzuki, Takayoshi; Mino, Koshiki; Tsumoto, Hiroki; Nakagawa, Hidehiko; Hasegawa, Makoto; Sasaki, Ryuzo; Mizukami, Tamio; Miyata, Naoki [Journal of the American Chemical Society, 2009, vol. 131, # 48, p. 17536 - 17537]
[4]Current Patent Assignee: PROUS INSTITUTE FOR BIOMEDICAL RES; PROUS INSTITUTE FOR BIOMEDICAL RESEARCH SA - US2015/344408, 2015, A1 Location in patent: Paragraph 0403-0405
[5]Current Patent Assignee: CURIS INC - US2009/76022, 2009, A1 Location in patent: Page/Page column 76
[6]Current Patent Assignee: CURIS INC - US2009/111772, 2009, A1 Location in patent: Page/Page column 75
[7]Bolzacchini, Ezio; Canevali, Carmen; Morazzoni, Franca; Orlandi, Marco; Rindone, Bruno; Scotti, Roberto [Journal of the Chemical Society, Dalton Transactions, 1997, # 24, p. 4695 - 4699]
[8]Sirasani, Gopal; Tong, Liuchuan; Balskus, Emily P. [Angewandte Chemie - International Edition, 2014, vol. 53, # 30, p. 7785 - 7788][Angew. Chem., 2014, vol. 53, # 30, p. 7919 - 7922,4]
[9]Percec, Virgil; Peterca, Mihai; Sienkowska, Monika J.; Ilies, Marc A.; Aqad, Emad; Smidrkal, Jan; Heiney, Paul A. [Journal of the American Chemical Society, 2006, vol. 128, # 10, p. 3324 - 3334]
[10]Location in patent: scheme or table De, Prithwiraj; Baltas, Michel; Lamoral-Theys, Delphine; Bruyère, Céline; Kiss, Robert; Bedos-Belval, Florence; Saffon, Nathalie [Bioorganic and Medicinal Chemistry, 2010, vol. 18, # 7, p. 2537 - 2548]
[11]Location in patent: scheme or table De, Prithwiraj; Koumba Yoya, Georges; Constant, Patricia; Bedos-Belval, Florence; Duran, Hubert; Saffon, Nathalie; Daffé, Mamadou; Baltas, Michel [Journal of Medicinal Chemistry, 2011, vol. 54, # 5, p. 1449 - 1461]
[12]Koolaji, Nooshin; Abu-Mellal, Abdallah; Tran, Van H.; Duke, Rujee K.; Duke, Colin C. [European Journal of Medicinal Chemistry, 2013, vol. 63, p. 415 - 422]
[13]Nagarkar, Amit A.; Yasir, Mohammad; Crochet, Aurelien; Fromm, Katharina M.; Kilbinger, Andreas F. M. [Angewandte Chemie - International Edition, 2016, vol. 55, # 40, p. 12343 - 12346][Angew. Chem., 2016, vol. 128, p. 12531 - 12534]
[14]Mitra; De; Karchaudhuri [Indian Journal of Chemistry - Section B Organic and Medicinal Chemistry, 2000, vol. 39, # 4, p. 311 - 312]
[15]Cunningham, Oliver D.; Edwards, Robert [Phytochemistry, 2008, vol. 69, # 10, p. 2016 - 2021]
[16]Location in patent: experimental part Burmaoglu, Serdar; Celik, Huelya; Goeiksu, Sueleyman; Maras, Ahmet; Altundas, Ramazan; Secen, Hasan [Synthetic Communications, 2009, vol. 39, # 9, p. 1549 - 1562]
[17]Location in patent: scheme or table Zhao, Huiping; Brandt, Gary E.; Galam, Lakshmi; Matts, Robert L.; Blagg, Brian S.J. [Bioorganic and Medicinal Chemistry Letters, 2011, vol. 21, # 9, p. 2659 - 2664]
[18]Galland, Stephanie; Mora, Nathalie; Abert-Vian, Maryline; Rakotomanomana, Njara; Dangles, Olivier [Journal of Agricultural and Food Chemistry, 2007, vol. 55, # 18, p. 7573 - 7579]
[19]Oyama, Kin-Ichi; Watanabe, Noriyuki; Yamada, Tomomi; Suzuki, Masako; Sekiguchi, Yukiko; Kondo, Tadao; Yoshida, Kumi [Tetrahedron, 2015, vol. 71, # 20, p. 3120 - 3130]
[20]Current Patent Assignee: OKAYAMA UNIVERSITY - EP2460810, 2012, A1 Location in patent: Page/Page column 14-15
[21]De Sousa, Damião P.; Lopes, Susiany P.; Pérez-Castillo, Yunierkis; Robledo, Sara M.; Yepe, Lina M. [Molecules, 2020, vol. 25, # 14]
[22]Current Patent Assignee: QINGDAO UNIVERSITY - CN112624927, 2021, A Location in patent: Paragraph 0240; 0241; 0242; 0243; 0244; 0245; 0246
[23]Carta, Fabrizio; Vullo, Daniela; Maresca, Alfonso; Scozzafava, Andrea; Supuran, Claudiu T. [Bioorganic and Medicinal Chemistry, 2013, vol. 21, # 6, p. 1564 - 1569]
[24]Sipilae, Julius; Nurmi, Harri; Kaukonen, Ann Marie; Hirvonen, Jouni; Taskinen, Jyrki; Yli-Kauhaluoma, Jari [European Journal of Pharmaceutical Sciences, 2005, vol. 25, # 4-5, p. 417 - 425]
[25]Balmas, Virgilio; Carta, Paola; Casalini, Stefano; Chtioui, Wiem; Delogu, Giovanna; Dettori, Maria A.; Fabbri, Davide; Migheli, Quirico; Oufensou, Safa [Molecules, 2021, vol. 26, # 3]
[26]Plasch, Katharina; Resch, Verena; Hitce, Julien; Popłoński, Jarosław; Faber, Kurt; Glueck, Silvia M. [Advanced Synthesis and Catalysis, 2017, vol. 359, # 6, p. 959 - 965]
[27]Torres y Torres, Janelle L.; Rosazza, John P. N. [Journal of Agricultural and Food Chemistry, 2001, vol. 49, # 3, p. 1486 - 1492]
[28]Kumar, N. S. Saleesh; Varghese, Shinto; Narayan, Gopinathan; Das, Suresh [Angewandte Chemie - International Edition, 2006, vol. 45, # 38, p. 6317 - 6321]
[29]Current Patent Assignee: FUNDACION PARA LA INVESTIGACION MEDICA APLICADA - WO2014/131855, 2014, A1 Location in patent: Page/Page column 60
[30]Bhat, Sana; Bhat, Zahid Rafiq; Gupta, Jeena; Kaur, Jaskiran; Kaur, Paranjeet; Khatik, Gopal L.; Khurana, Navneet; Kumar, Rajan; Kumar, Rakesh; Tikoo, Kulbhushan [Bioorganic Chemistry, 2020, vol. 100]
[31]Current Patent Assignee: BASF SE - US2007/128151, 2007, A1 Location in patent: Page/Page column 10-11
[32]Kim, Hoon; Padmakshan, Dharshana; Li, Yanding; Rencoret, Jorge; Hatfield, Ronald D.; Ralph, John [Biomacromolecules, 2017, vol. 18, # 12, p. 4184 - 4195]
[33]Menon, Sanjay R.; Patel, Vishal K.; Mitscher, Lester A.; Shih, Peter; Pillai, Segaran P.; Shankel, Delbert M. [Journal of Natural Products, 1999, vol. 62, # 1, p. 102 - 106]
[34]Micillo, Raffaella; Sirés-Campos, Julia; García-Borrón, José Carlos; Panzella, Lucia; Napolitano, Alessandra; Olivares, Conchi [International Journal of Molecular Sciences, 2018, vol. 19, # 8]
[35]Fernandes, João Paulo S.; Ferrarini, Márcio; Mercaldi, Vitória Gallo; Nazato, Lucas Idacir Sbrugnera; Padovani, Giovana; Sufi, Bianca da Silva; Varela, Marina Themoteo [Bioorganic Chemistry, 2020, vol. 103]
[36]Melhem, Marcia S. C.; Oliveira, Lidiane; Pascon, Renata C.; Vallim, Marcelo A.; Varela, Marina Themoteo; dos Santos, Alexandre Paes; Corrêa, Isabela Teresa Santos; Fernandes, João Paulo S.; Ferrarini, Márcio; Tempone, André Gustavo [European Journal of Pharmaceutical Sciences, 2020, vol. 153]
[37]Lima, Tamires C.; Ferreira, Alana R.; Silva, Daniele F.; Lima, Edeltrudes O.; de Sousa, Damião P. [Natural Product Research, 2018, vol. 32, # 5, p. 572 - 575]
[38]Tawata; Taira; Kobamoto; Zhu; Ishihara; Toyama [Bioscience, Biotechnology and Biochemistry, 1996, vol. 60, # 5, p. 909 - 910]
[39]Fischer,E.; Nouri [Chemische Berichte, 1917, vol. 50, p. 618][Chemisches Zentralblatt, 1916, vol. 87, # II, p. 1025]
[40]Stelmach,H.; Connors,K.A. [Journal of the American Chemical Society, 1970, vol. 92, # 4, p. 863 - 866]
[41]Krawczyk, Andrzej R.; Lipkowska, Ewa; Wrobel, Jerzy T. [Bulletin of the Polish Academy of Sciences: Chemistry, 1986, vol. 34, # 3-4, p. 115 - 122]
[42]Lam, Thi Bach Tuyet; Iiyama, Kenji; Stone, Bruce A. [Phytochemistry, 1992, vol. 31, # 4, p. 1179 - 1184]
[43]Dinkova-Kostova, Albena T.; Abeygunawardana, Chitrananda; Talalay, Paul [Journal of Medicinal Chemistry, 1998, vol. 41, # 26, p. 5287 - 5296]
[44]Pieters, Luc; Van Dyck, Stefaan; Gao, Mei; Bai, Ruoli; Hamel, Ernest; Vlietinck, Arnold; Lemière, Guy [Journal of Medicinal Chemistry, 1999, vol. 42, # 26, p. 5475 - 5481]
[45]Gu; Chen; Jing; Pan [Journal of Chemical Research - Part S, 2000, # 8, p. 397 - 399]
[46]Snider, Barry B.; Grabowski, James F. [Tetrahedron Letters, 2005, vol. 46, # 5, p. 823 - 825]
[47]Ritter, Tobias; Kvaerno, Lisbet; Werder, Moritz; Hauser, Helmut; Carreira, Erick M. [Organic and Biomolecular Chemistry, 2005, vol. 3, # 19, p. 3514 - 3523]
[48]Karthikeyan; Ramamurthy [Journal of Organic Chemistry, 2007, vol. 72, # 2, p. 452 - 458]
[49]Epifano, Francesco; Menghini, Luigi; Pagiotti, Rita; Angelini, Paola; Genovese, Salvatore; Curini, Massimo [Bioorganic and Medicinal Chemistry Letters, 2006, vol. 16, # 21, p. 5523 - 5525]
[50]Vengris, Mikas; Larsen, Delmar S.; Van Der Horst, Michael A.; Larsen, Olaf F. A.; Hellingwerf, Klaas J.; Van Grondelle, Rienk [Journal of Physical Chemistry B, 2005, vol. 109, # 9, p. 4197 - 4208]
[51]Sobolev, Victor S.; Horn, Bruce W.; Potter, Thomas L.; Deyrup, Stephen T.; Gloer, James B. [Journal of Agricultural and Food Chemistry, 2006, vol. 54, # 10, p. 3505 - 3511]
[52]Vassao, Daniel G.; Gang, David R.; Koeduka, Takao; Jackson, Brenda; Pichersky, Eran; Davin, Laurence B.; Lewis, Norman G. [Organic and Biomolecular Chemistry, 2006, vol. 4, # 14, p. 2733 - 2744]
[53]Current Patent Assignee: ASTELLAS PHARMA INC. - WO2004/63169, 2004, A1 Location in patent: Page 74
[54]Current Patent Assignee: ASTELLAS PHARMA INC. - US2004/229889, 2004, A1 Location in patent: Page 28
[55]Location in patent: experimental part Moussouni, Sonia; Saru, Maria-Liliana; Ioannou, Efstathia; Mansour, Mohamed; Detsi, Anastasia; Roussis, Vassilios; Kefalas, Panagiotis [Tetrahedron Letters, 2011, vol. 52, # 11, p. 1165 - 1168]
[56]Current Patent Assignee: DUKE UNIVERSITY - WO2011/103189, 2011, A1 Location in patent: Page/Page column 74-75; 86
[57]Moon, Sun-Joo; Kwon, Mi; Choi, Donha; Won, Keehoon; Kim, Yong Hwan; Choi, In-Gyu; Choi, Joon Weon [Phytochemistry, 2012, vol. 82, p. 15 - 21]
[58]Current Patent Assignee: CHISSO CORPORATION - US8450517, 2013, B2 Location in patent: Page/Page column 29; 30
[59]Li, Xiaoyang; Inks, Elizabeth S.; Li, Xiaoguang; Hou, Jinning; Chou, C. James; Zhang, Jian; Jiang, Yuqi; Zhang, Yingjie; Xu, Wenfang [Journal of Medicinal Chemistry, 2014, vol. 57, # 8, p. 3324 - 3341]
[60]Avo, Joao; Cunha-Silva, Luis; Lima, Joao Carlos; Jorge Parola [Organic Letters, 2014, vol. 16, # 10, p. 2582 - 2585]
[61]Cattò, Cristina; Dell'Orto, Silvia; Villa, Federica; Villa, Stefania; Gelain, Arianna; Vitali, Alberto; Marzano, Valeria; Baroni, Sara; Forlani, Fabio; Cappitelli, Francesca [PLoS ONE, 2015, vol. 10, # 7]
[62]Li, Yang; Feng, Ling; Song, Zhi-Fang; Li, Hai-Bei; Huai, Qi-Yong [Molecules, 2016, vol. 21, # 2]
[63]Beňová, Eva; Zeleňák, Vladimír; Halamová, Dáša; Almáši, Miroslav; Petrul'ová, Veronika; Psotka, Miroslav; Zeleňáková, Adriána; Bačkor, Martin; Hornebecq, Virginie [Journal of Materials Chemistry B, 2017, vol. 5, # 4, p. 817 - 825]
[64]Current Patent Assignee: CHISSO CORPORATION - KR101749768, 2017, B1 Location in patent: Paragraph 0179-0181; 0184-0185
[65]Liu, Peng; Yasir, Mohammad; Ruggi, Albert; Kilbinger, Andreas F. M. [Angewandte Chemie - International Edition, 2018, vol. 57, # 4, p. 914 - 917][Angew. Chem., 2018, vol. 130, # 4, p. 926 - 929,4]
[66]Zhang, Xinyu; Zhao, Rui; Chen, Meng; Ma, Tao; Wu, Gaorong; Xue, Nannan; Li, Guoliang; Wang, Hui; Fang, Kang; Zhang, Wenxi; Wang, Penglong; Lei, Haimin [Molecules, 2018, vol. 23, # 5]
[67]Mayr, Fabian; Sturm, Sonja; Ganzera, Markus; Waltenberger, Birgit; Martens, Stefan; Schwaiger, Stefan; Schuster, Daniela; Stuppner, Hermann [Journal of Natural Products, 2019, vol. 82, # 1, p. 136 - 147]
[68]Gureev, Maxim; Krasavin, Mikhail; Kuranov, Sergey O.; Luzina, Olga A.; Onopchenko, Oleksandra; Pishel, Iryna; Salakhutdinov, Nariman F.; Zozulya, Sergey [Bioorganic Chemistry, 2020, vol. 99]
[69]Kim, Stacie S.; Sattely, Elizabeth S. [Journal of the American Chemical Society, 2021, vol. 143, # 13, p. 5011 - 5021]
  • 2
  • [ 7400-08-0 ]
  • [ 6049-54-3 ]
  • 4
  • [ 7400-08-0 ]
  • [ 3943-97-3 ]
YieldReaction ConditionsOperation in experiment
98% With sulfuric acid In methanol Heating / reflux; 27.27a A mixture of 4-hydroxycinnamic acid (8.2 g, 50 mmol) and a drop OfH2SO4 in methanol (30 mL) was heated to reflux overnight. Then the solvent was evaporated, the residue was dissolved in ethyl acetate, washed with saturated NaHCO3 solution twice, brine, dried over MgSO4, concentrated to give the title compound 0501-66 as white solid (8.7 g, 98%): LCMS: 179 [M+l]+.
61% With sulfuric acid In methanol; ethanol; water 1 4-Hydroxycinnamic Acid Methyl Ester (Compound 2a) Example 1 4-Hydroxycinnamic Acid Methyl Ester (Compound 2a) A solution of 4-hydroxycinnamic acid (compound 1a, 20.0 g, 0.12 mol) and 5 drops of concentrated sulfuric acid in 500 mL of reagent grade methanol was stirred for four days at 55° C., while monitored by TLC. The solvent was then removed from the reaction mixture on the rotovap and the solid residue was recrystallized from a mixture of 200 mL of ethanol and 200 mL of water to give 13.3 g of product in 61% yield: 13C NMR (75.4 MHz, CDCl3) δ 168.18, 157.88, 144.85, 129.95, 126.91, 115.86, 114.87, 51.81.
With methanol; sulfuric acid
Multi-step reaction with 4 steps 1: K2CO3 / dimethylformamide / Heating 2: aq. NaOH / methanol / 70 °C 3: K2CO3 / acetone / Heating 4: aq. HCl; AcOH / 70 °C
With sulfuric acid In methanol; water 6.1 Synthesis of Compound (1c) 1) 25 g (0.152 mol) of p-hydroxycinnamic acid was dissolved in 100 ml of methanol, to which 1 g of concentrated sulfuric acid was added and refluxed for 8 hours. After allowing the system to cool down to room temperature, twice the amount of water was added thereto. The pellet was collected by filtration, washed with water, and dried. 25 g of a yellowish white solid was obtained. The solid was recrystallized from toluene. As a result, 21 g of methyl para-hydroxycinnamate was obtained as a yellowish white solid.
Multi-step reaction with 2 steps 1: thionyl chloride / dichloromethane / 2 h / 20 °C / Reflux 2: dichloromethane / 3 h / Reflux
Multi-step reaction with 2 steps 1: thionyl chloride / 2 h / 85 °C 2: triethylamine / 2 h / 20 °C

  • 5
  • [ 2538-87-6 ]
  • [ 7400-08-0 ]
  • 6
  • [ 80-69-3 ]
  • [ 7400-08-0 ]
  • (E)-p-coumaroyltartronic acid [ No CAS ]
  • 7
  • [ 57602-02-5 ]
  • [ 7400-08-0 ]
  • [ 130740-01-1 ]
  • 8
  • [ 29588-94-1 ]
  • [ 136918-14-4 ]
  • [ 501-98-4 ]
  • [ 7400-08-0 ]
  • (S)-4-(4-Hydroxy-phenyl)-1,5-dioxo-2,3,4,5-tetrahydro-1H-benzo[c]azepine-3-carboxylic acid [ No CAS ]
  • 9
  • 2-acetonyl-8-(6-O-coumaroyl-β-D-glucopyranosyl)-7-hydroxy-5-methylchromone [ No CAS ]
  • [ 40738-40-7 ]
  • [ 501-98-4 ]
  • [ 7400-08-0 ]
  • [ 104871-04-7 ]
  • 10
  • chrysoeriol-7-O-β-D-(3''-E-p-coumaroyl)-glucopyranoside [ No CAS ]
  • [ 2280-44-6 ]
  • [ 7400-08-0 ]
  • [ 491-71-4 ]
  • 11
  • anisofolin-A [ No CAS ]
  • [ 7400-08-0 ]
  • [ 578-74-5 ]
  • 12
  • [ 64-17-5 ]
  • [ 7400-08-0 ]
  • [ 2979-06-8 ]
YieldReaction ConditionsOperation in experiment
95% With sulfuric acid; at 65℃; for 18h; p-Cinnamic acid (2) (2.00 g, 12.2 mmol) was dissolved in 100 mL of ethanol containing 3.5 mL of concentrated H2SO4. The solution was refluxed at 65 C for 18h. After concentration under reduced pressure, the solution was diluted with ethyl acetate and washed with saturated aqueous NaHCO3 and brine. After drying over anhydrous Na2SO4, the volatiles were removed in vacuo. The raw products were purified by column chromatography on silica gel using EtOAc/petroleum ether (1/5) mixtures as eluents to afford (E)-ethyl 3-(4-hydroxyphenyl)acrylate (2.22 g, 95%) as white solid. 1HNMR(CDCl3, 400MHz) delta 1.34 (3H, t, J = 7.0Hz, -OCH2CH3), 4.27 (2H, q, J = 7.0Hz, -OCH2CH3), 6.29 (1H, d, J = 16.0Hz, -CH=CHCO-), 6.90 (2H, d, J = 8.0Hz, H-3, H-5 of Ph), 7.40 (2H, d, J = 8.4Hz, H-2, H-6 of Ph), 7.64 (1H, d, J = 16.0Hz, -CH=CHCO-), 7.70-7.56 (br, 1H, OH); MS (ESI) m/z: 193.1 (M + H)+. The spectroscopic data of (E)-ethyl 3-(4-hydroxyphenyl)acrylate were consistent with the reference.
75% With sulfuric acid;Reflux; In a 1 L three-necked flask,102.0 g of trans-p-coumaric acid,500 mL of ethanol,5.8 g of sulfuric acid,in addition,The mixture was stirred while heating under reflux.After completion of the reaction,Pour the reaction system into 3 L of water,After filtering the precipitate,The filtrate was dried,90.0 g of the desired product DA-4-1 (white solid) was obtained (yield: 75%).
68% With thionyl chloride; at 0℃;Reflux; Inert atmosphere; General procedure: Thionyl chloride (1.16 g, 1.5 equiv) was added drop-wise to a solution of acid (1.0 g, 1.0 equiv) in the corresponding alcohol (15 ml) at 0&d eg;C. The solution was refluxed under a nitrogen atmosphere until all starting material was consumed (TLC monitoring). Then the solvent was removed under vacuo and the residue was purified by silica gel column chromatography eluting with ethyl acetate/n-hexane to afford the corresponding carboxylic esters.
45% With sulfuric acid; for 30h;Reflux; General procedure: A series of 4- or 3-substituted ethyl-cinnamates was prepared by conventional method [38], using 2 mmol of para and meta substituted cinnamic acids refluxed in absolute ethanol (15 mL) and sulfuric acid (0.1 mL) for 30 h. The ester formation was monitored by GC and TLC (hexane:acetate 9:1 as eluent). The products were isolated after washing with aqueous sodium bicarbonate solution and solvent extraction with dichloromethane. After work up and solvent evaporation, most of the pure esters were obtained as an oil or a solid. All compounds were characterized by 1H and 13C-NMR spectroscopy, in agreement with the literature [39-41]. The spectroscopic and characterization information of all compounds is given as Electronic Supplementary Information.
With sulfuric acid; for 5h;Heating / reflux; (a) Trans-4-hydroxycinnamic acid (5.00g, 30.46mmol) was dissolved in ethanol (60ml), and sulfuric acid (1.6ml) was added thereto. The mixture was refluxed for 5 hours and then the solvent was evaporated out. The residue was diluted with water, neutralized with sodium hydrogencarbonate, and then extracted with ethyl acetate. The organic layer was washed with saturated brine and dried over MgSO4, and then the solvent was evaporated out.
With sulfuric acid; for 96h;Heating / reflux; The ethyl (E)-4-hydroxycinnamate required as starting material was prepared as follows: [00109] A mixture of 10 g 4-hydroxycinnamic acid, 100 ml ethanol and 0.5 ml concentrated sulphuric acid was heated under gentle reflux for 96 hours. The reaction mixture was poured onto water, the resultant precipitate filtered off, washed with portions of water and then dried. The residue was purified by column chromatography on silica gel using ethyl acetate as eluent and recrystallisation from acetonitrile to yield 9.2 g of ethyl (E)-4-hydroxycinnamate.
With thionyl chloride; for 24h;Reflux; General procedure: To an icy solution of hydroxy cinnamic acid (10 mmol) in ethanol (30 mL) was added thionyl chloride (0.5 mL) dropwise. The reaction mixture was refluxed for 24 h to give the corresponding crude ethyl cinnamate.
With sulfuric acid; for 12h;Reflux; General procedure: Ferulic acid (1) or trans-4-hydroxycinnamic acid (2) or isoferulic acid (3) or trans-3-hydroxycinnamic acid (4) and five drops of H2SO4 (95%) were refluxed in methanol or ethanol for 12 h. The reaction mixture was concentrated in vacuo and the residue was dissolved in ethyl acetate. The organic layer was washed with a 5% aqueous NaHCO3 solution and water. After drying over anhydrous Na2SO4,the ethyl acetate was removed in vacuo. The residue was purified by column chromatography on silica gel using ethyl acetate/petroleum ether mixtures as eluents to afford compounds 5-12.

  • 13
  • [ 7400-08-0 ]
  • [ 83392-10-3 ]
  • [ 177085-36-8 ]
  • 14
  • [ 7400-08-0 ]
  • [ 85-61-0 ]
  • [ 30802-00-7 ]
YieldReaction ConditionsOperation in experiment
With recombinant Selaginella moellendorffii 4-coumarate:coenzyme A ligase 1; ATP; magnesium chloride; In aq. buffer; at 30℃; for 0.5h;pH 7.5;Enzymatic reaction;Kinetics; General procedure: We performed Sm4CLs enzyme assays to detect the formation of the <strong>[85-61-0]CoA</strong> esters of variouscinnamic acid derivatives. Each 200 muL assay contained 10 mug purified protein, 200 muMsubstrate, 5mMATP, 300 muM<strong>[85-61-0]CoA</strong>, and 5mMMgCl2, made up in 200mMTris-HCl buffer (pH 7.5). Enzymatic reactionswere incubated for 30 min at 30 C, and the reaction products were analyzed using a HPLC device(1260 Infinity Binary LC system, Agilent, Santa Clara, CA, USA), equipped with a multi wavelengthdetector. The samples were separated through a 5-mum reverse-phase XDB-C18 column with a flow rateof 1 mL/min. A linear gradient of solvent A (1% H3PO4 in H2O) and solvent B (CH3CN) were appliedas follows: 0-5 min, 5% B isocratic; 5-35 min, 5-25% B linear; 35-36 min, 25-100% B linear. Standardsolutions of reference compounds were used for calibration.The effects of pH and temperature on the enzyme activity were examined using p-coumaricacid as substrate. To determine optimal pH, enzymatic activity was assessed in 200 mM Tris-HClbuffer (pH 5.0, 6.0, 6.5, 7.0, 7.5, 8.0, 8.5, and 9.0), while the optimal temperature was determined bymeasuring enzymatic activity at 10, 20, 30, 40, 50, and 60 C. All experiments were performed intriplicate. Kinetic parameters were determined using different substrate concentrations. We performedthis experiment in triplicate, with 3 mug purified enzyme in a final volume of 200 muL of 200 mM Tris-HClbuffer at the optimal pH and temperature for 10 min. The level of UV absorption was recorded at1 min intervals. Relevant wavelengths were 311 nm (cinnamoyl <strong>[85-61-0]CoA</strong>), 333 nm (p-coumaroyl <strong>[85-61-0]CoA</strong>),346 nm (caffeoyl <strong>[85-61-0]CoA</strong>), 346 nm (feruloyl <strong>[85-61-0]CoA</strong>), and 352 nm (sinapoyl <strong>[85-61-0]CoA</strong>) [37,41,42]. For determiningkinetic properties with dihydro-p-coumaric acid, reactions were incubated at optimal temperature for10 min and analyzed using HPLC, and then the quantity of the reaction product present was estimatedfrom a standard calibration curve.
  • 15
  • (1R,3aS,5aS,5bR,7aR,9S,11aR,11bR,13aR,13bR)-9-Hydroxy-5a-[(E)-3-(4-hydroxy-phenyl)-acryloyloxymethyl]-1-isopropenyl-5b,8,8,11a-tetramethyl-icosahydro-cyclopenta[a]chrysene-3a-carboxylic acid [ No CAS ]
  • [ 501-98-4 ]
  • [ 7400-08-0 ]
  • (+)-cylicodiscic acid [ No CAS ]
  • 16
  • [ 7400-08-0 ]
  • [ 3417-91-2 ]
  • (E)-N-(4-hydroxycinnamoyl)tyrosine [ No CAS ]
  • 17
  • [ 7400-08-0 ]
  • (1R,2S,3R,4S)-3,4-Bis-(4-hydroxy-phenyl)-cyclobutane-1,2-dicarboxylic acid [ No CAS ]
  • [ 501-98-4 ]
  • 18
  • [ 7400-08-0 ]
  • [ 74-88-4 ]
  • [ 3943-97-3 ]
YieldReaction ConditionsOperation in experiment
98% With potassium hydrogencarbonate In N,N-dimethyl-formamide at 40℃;
  • 19
  • [ 7400-08-0 ]
  • [ 149-73-5 ]
  • [ 3943-97-3 ]
YieldReaction ConditionsOperation in experiment
96% With thionyl chloride In methanol at 20℃; for 48h;
  • 20
  • [ 7400-08-0 ]
  • [ 85-61-0 ]
  • [ 30802-00-7 ]
YieldReaction ConditionsOperation in experiment
With Piper nigrum 4-coumarate:CoA ligase isoform 1; ATP; magnesium chloride; In aq. buffer; at 25℃; for 0.75h;pH 7.5;Enzymatic reaction;Kinetics; General procedure: The enzyme kinetic parameters were measured spectrophotometrically as described [22]. Each reaction contained 0.3 mM CoA, 2.5 mM MgCl2 and 2.5 mM ATP in 100 mM Tris-HCl (pH 7.5). For kinetic analyses, at least six concentrations of each substrate (coumaric, ferulic, 3,4-methylenedioxycinnamic, and piperic acids) ranging from 6.25 to 200 mM and 11 to 94 mg of each purified 4CL in a total volume of 1 ml reaction mixture were reacted in triplicate. For the assay of cinnamic acid, the acid concentration was between 15 mM and 600 mM and CoA was at 0.7 mM. The sample was set-up on ice and transferred to a water bath at 25C to equilibrate temperature for 1 min. The reaction was started by addition of CoA. The formation of the thioester products were followed at 311 nm for cinnamoyl-, 333 nm for 4-coumaroyl-, 346 nm for feruloyl-, 346 nm for 3,4-methylenedioxycinnamoyl-, and 368 nm for piperoyl-CoA, and the concentration of these products were calculated using the corresponding extinction coefficients of 22, 21, 19, 19, and 30.8 mM-1 cm-1, respectively [18,23,24]. To estimate the formation of 3,4-methylenedioxycinnamoyl-CoA, the same extinction coefficient, and lambdamax as feruloyl-CoA were extrapolated. The reaction was followed for 15 min until the linearity of the reaction was maintained. The reaction at 50 mM substrate concentration was allowed to proceed for another 30 min and then the mixture was stood in a boiling water bath for 3 min before scanning on spectrophotometer. Negative control was run with the combined, boiled enzymes, each at the same concentration as the individual reaction. Piperonylic acid was allowed to react for 45 min without kinetic measurement.
  • 21
  • [ 7400-08-0 ]
  • [ 72784-43-1 ]
  • C14H15NO4 [ No CAS ]
  • 24
  • [ 7400-08-0 ]
  • [ 10210-17-0 ]
YieldReaction ConditionsOperation in experiment
82% With whole cell cultures of dichomitus albidofuscus at 24℃; for 72h; Darkness; Microbiological reaction; 2.5. general procedure for preparative biotransformation General procedure: The substrate (1 mmol) was added to submerged cultures of DAL on the 3rd culture day. The reaction mixture was incubated on an incubation shaker at 150 rmin 1 (deflection 25 mm) under exclusion of light at 24 °C for 4 days. 10 g of NaCl was added to media, and the mixture was stirred for 10 min at 800 rpm (magnetic stirrer). For extraction, 50 mL of Et2O was added, and the resulting mixture was stirred for 20 min at 800 rpm (magnetic stirrer), and centrifuged for 5 min at 3000 × g to separatethe organic layer. The extraction was repeated three times. The combined organic layers were washed with brine (1 × 30 mL) and water (1× 30 mL), dried over Na2SO4 and evaporated to dryness. The resulting reaction mixtures were analysed by GC/MS and NMR spectroscopy.Products were purified by column chromatography on silica gel (eluent(pentane/ether) changed gradually: 10/1, 7/3, 1/1) to isolate the majorcomponents. The respective fractions were combined, concentrated in vacuum, and the 1H and 13C NMR spectra of the residuals were compared with those of reference compounds. Every biotransformation was repeated three times to verify the reproducibility of the experiments. The detailed information about experimental data and yields is provided in the Electronic Supplementary Information.
Multi-step reaction with 3 steps 1: aq. HCl / 6 h / Heating 2: H2 / 10percent Pd/C / methanol / 0.5 h / 760 Torr / Ambient temperature 3: LiBH4 / diethyl ether; toluene / 0.25 h / Heating
Multi-step reaction with 4 steps 1.1: pyridine / 1 h / 20 °C 2.1: chloroformic acid ethyl ester; triethylamine / 1,4-dioxane / 1 h / Inert atmosphere 2.2: 1 h / Inert atmosphere 3.1: potassium hydroxide / ethanol / 8 h / Inert atmosphere 4.1: 5%-palladium/activated carbon; hydrogen / ethyl acetate / 6 h / 20 °C / 2250.23 Torr
  • 25
  • [ 7400-08-0 ]
  • [ 144-55-8 ]
  • [ 2979-06-8 ]
YieldReaction ConditionsOperation in experiment
With sulfuric acid; In ethanol; Example 12 Synthesis of N-(1,2-diethyl-4-pyrazolidinyl)-4-geranyloxycinnamamide To a solution of 4- hydroxycinnamic acid (16.4 g) in ethanol (100 ml) was added sulfuric acid (10 ml). The mixture was refluxed with heating for 4 hours, Aqueous sodium bicarbonate solution was added to the reaction solution so as to make basic. Then, the reaction solution was extracted with chloroform. The extract was dried over sodium sulfate anhydride and then concentrated under a vacuum to yield a raw product of ethyl 4-hydroxycinnamate.
  • 26
  • [ 7400-08-0 ]
  • [ 95-12-5 ]
  • C17H18O3 [ No CAS ]
YieldReaction ConditionsOperation in experiment
70% With benzotriazol-1-ol; 1-ethyl-(3-(3-dimethylamino)propyl)-carbodiimide hydrochloride; triethylamine; In N,N-dimethyl-formamide; at 0 - 20℃; <EXAMPLE 3> Synthesis of the 2,3-epoxy-l-propoxy cinnamate norbornene (Formula 7) monomer 3-(l): 4-hydroxy cinnamate norbornene; The 4-hydroxy cinnamic acid (8.7 g, 53 mmol), norbornenemethanol (6.5 g, 52.4 mmol), EDC (Aldrich, Inc., 16 g, 84 mmol), and HOBT (Aldrich, Inc., 10.6 g, 78 mmol) were added to the 2-neck flask having the volume of 250 mi , and dissolved in 100 mi of DMF.After the temperature was reduced to 0C, triethylamine (Aldrich, Inc., 21 g, 0.21 mol) was slowly dropped thereon. After the reaction was performed overnight while the temperature was increasedto normal temperature, the reaction was finished and the extraction was performed by using a large amount of ethyl acetate. After thewashing was performed by using NaHCU3 and H2O and the drying was <n="38"/>performed by using anhydrous MgSO4, the filtration was performed toremove the solvent by using a rotary evaporator, thus creating theproduct. The purification was performed by using the column chromatography (Hexane : Ethyl acetate = 15 : 1) to obtain 9.9 g ofpure product (yield: 70%).
  • 27
  • [ 1135-24-6 ]
  • [ 140-10-3 ]
  • [ 331-39-5 ]
  • [ 7400-08-0 ]
  • [ 6099-04-3 ]
  • [ 537-73-5 ]
  • [ 20329-98-0 ]
  • [ 530-59-6 ]
  • [ 174472-00-5 ]
  • [ 640234-76-0 ]
  • [ 640234-75-9 ]
  • 3-(4-hydroxy-3-(S)-methoxy-phenyl)-N-[2-(4-hydroxy-phenyl)-2-methoxy-ethyl]acrylamide [ No CAS ]
  • (E)-N-((S)-2-Methoxy-2-phenyl-ethyl)-3-phenyl-acrylamide [ No CAS ]
  • (E)-3-(3-Methoxy-phenyl)-N-((S)-2-methoxy-2-phenyl-ethyl)-acrylamide [ No CAS ]
  • (E)-3-(4-Hydroxy-phenyl)-N-((S)-2-methoxy-2-phenyl-ethyl)-acrylamide [ No CAS ]
  • (-)-aegeline O-methylether [ No CAS ]
  • (S)-N-(2-(4-hydroxyphenyl)-2-methoxyethyl)cinnamamide [ No CAS ]
  • (E)-3-(3-Hydroxy-4-methoxy-phenyl)-N-((S)-2-methoxy-2-phenyl-ethyl)-acrylamide [ No CAS ]
  • 3-(4-hydroxyphenyl)-N-[2-(4-hydroxyphenyl)-2-methoxyethyl]acrylamide [ No CAS ]
  • (E)-N-[(S)-2-(4-Hydroxy-phenyl)-2-methoxy-ethyl]-3-(3-methoxy-phenyl)-acrylamide [ No CAS ]
  • (E)-3-(3,4-Dihydroxy-phenyl)-N-((S)-2-methoxy-2-phenyl-ethyl)-acrylamide [ No CAS ]
  • (E)-3-(4-Hydroxy-phenyl)-N-[(S)-2-methoxy-2-(4-methoxy-phenyl)-ethyl]-acrylamide [ No CAS ]
  • (E)-N-[(S)-2-Methoxy-2-(4-methoxy-phenyl)-ethyl]-3-(3-methoxy-phenyl)-acrylamide [ No CAS ]
  • (E)-3-(4-Hydroxy-3-methoxy-phenyl)-N-((S)-2-methoxy-2-phenyl-ethyl)-acrylamide [ No CAS ]
  • (S)-N-[2-(4-methoxyphenyl)-2-methoxyethyl]-3-(4-hydroxy-3-methoxyphenyl)acrylamide [ No CAS ]
  • (E)-3-(4-Hydroxy-3,5-dimethoxy-phenyl)-N-((S)-2-methoxy-2-phenyl-ethyl)-acrylamide [ No CAS ]
  • (E)-N-((S)-2-Methoxy-2-phenyl-ethyl)-3-(3,4,5-trimethoxy-phenyl)-acrylamide [ No CAS ]
  • (E)-3-(3-Hydroxy-4-methoxy-phenyl)-N-[(S)-2-methoxy-2-(4-methoxy-phenyl)-ethyl]-acrylamide [ No CAS ]
  • (E)-3-(3,4-Dihydroxy-phenyl)-N-[(S)-2-methoxy-2-(4-methoxy-phenyl)-ethyl]-acrylamide [ No CAS ]
  • (E)-3-(3-Hydroxy-4-methoxy-phenyl)-N-[(S)-2-(4-hydroxy-phenyl)-2-methoxy-ethyl]-acrylamide [ No CAS ]
  • (E)-3-(3,4-Dihydroxy-phenyl)-N-[(S)-2-(4-hydroxy-phenyl)-2-methoxy-ethyl]-acrylamide [ No CAS ]
  • (E)-N-[(S)-2-Methoxy-2-(4-methoxy-phenyl)-ethyl]-3-(3,4,5-trimethoxy-phenyl)-acrylamide [ No CAS ]
  • (E)-3-(4-Hydroxy-3,5-dimethoxy-phenyl)-N-[(S)-2-methoxy-2-(4-methoxy-phenyl)-ethyl]-acrylamide [ No CAS ]
  • (E)-N-[(S)-2-(4-Hydroxy-phenyl)-2-methoxy-ethyl]-3-(3,4,5-trimethoxy-phenyl)-acrylamide [ No CAS ]
  • (E)-3-(4-Hydroxy-3,5-dimethoxy-phenyl)-N-[(S)-2-(4-hydroxy-phenyl)-2-methoxy-ethyl]-acrylamide [ No CAS ]
  • 28
  • [ 629-96-9 ]
  • [ 7400-08-0 ]
  • [ 112561-76-9 ]
YieldReaction ConditionsOperation in experiment
92% With triphenylphosphine; diethylazodicarboxylate In tetrahydrofuran at 20℃;
  • 29
  • O-(trans-p-coumaroyl)malic acid [ No CAS ]
  • [ 501-98-4 ]
  • [ 7400-08-0 ]
  • 30
  • [ 1899-30-5 ]
  • [ 501-98-4 ]
  • [ 7400-08-0 ]
  • 31
  • [ 117405-49-9 ]
  • [ 50-99-7 ]
  • [ 501-98-4 ]
  • [ 7400-08-0 ]
  • 32
  • [ 87099-71-6 ]
  • [ 501-98-4 ]
  • [ 7400-08-0 ]
  • 33
  • [ 1108200-72-1 ]
  • [ 501-98-4 ]
  • [ 7400-08-0 ]
  • 34
  • (Z)-4-coumaric acid 4-O-β-D-glucopyranoside [ No CAS ]
  • [ 50-99-7 ]
  • [ 501-98-4 ]
  • [ 7400-08-0 ]
  • 35
  • 3-O-(cis-p-coumaroyl)quinic acid [ No CAS ]
  • [ 501-98-4 ]
  • [ 7400-08-0 ]
  • 36
  • 5,6,7-trihydroxy-2-[4-({5-O-[(2E)-3-(4-hydroxyphenyl)-1-oxo-2-propen-1-yl]-α-L-arabinofuranosyl}oxy)phenyl]-4H-1-benzopyran-4-one [ No CAS ]
  • [ 7400-08-0 ]
  • [ 529-53-3 ]
  • 37
  • [ 578-86-9 ]
  • [ 7400-08-0 ]
  • [ 108-46-3 ]
  • 38
  • [ 63-91-2 ]
  • [ 7400-08-0 ]
  • [ 500-38-9 ]
  • 39
  • cyanidin 3-O-[2-O-(2-O-(trans-feruloyl)-glucosyl)-6-O-(trans-p-coumaroyl)-glucoside]-5-O-[6-O-(malonyl)-glucoside] [ No CAS ]
  • [ 2280-44-6 ]
  • [ 1135-24-6 ]
  • [ 141-82-2 ]
  • [ 7400-08-0 ]
  • [ 528-58-5 ]
YieldReaction ConditionsOperation in experiment
With hydrogenchloride; water; at 100℃; for 1.0h; General procedure: The identification of anthocyanins was carried out by standard procedures and both alkaline and acid hydrolyses (Harborne 1984). Acid hydrolysis of pigments (ca. 1 mg each) was carried out with 2N HCl (1 mL) at 100 C for 1 h. Alkaline hydrolysis of pigments (ca. 1 mg each) was carried out with 2N NaOH solution (1 mL) under degassed syringe allowed to stand for 15 min. The solution was next acidified with 2N HCl (1.1 mL) and evaporated in vacuo to dryness. The data of TLC (Rf values), HPLC (Rt-min, method 1), UV-Vis (lambdamax), and FABMS spectra are shown in Section 4.4.1.-4.4.4.
  • 40
  • [ 51-61-6 ]
  • [ 7400-08-0 ]
  • [ 106005-97-4 ]
YieldReaction ConditionsOperation in experiment
With (benzotriazo-1-yloxy)tris(dimethylamino)phosphonium hexafluorophosphate; trimethylamine; In N,N-dimethyl-formamide; at 20℃; for 12.0h;Inert atmosphere; General procedure: BOP (Castro's reagent [29], 0.25 mmol) was added to a mixture solution of acid (0.20 mmol) and amine (0.20 mmol) in dry DMF (20 ml) under a nitrogen atmosphere at room temperature. To this mixture, trimethylamine (0.40 mmol) was added drop wise with vigorous stirring. The stirring was continued for 12 h. The reaction mixture was quenched with water (100 ml) and then extracted with ethylacetate (3 50 ml). Organic layer was wished with brine and dried over anhydrous Na2SO4. The solvent was evaporated and residue was subjected to silica gel chromatography using CHCl3/CH3OH (10:1) as eluent to give corresponding amides (1-1d) as 30-80% yield.
  • 41
  • [ 7400-08-0 ]
  • [ 4089-07-0 ]
  • methyl N-[3-(4-hydroxyphenyl)-1-oxo-2-propen-1-yl]-L-tyrosinate [ No CAS ]
  • 42
  • [ 7400-08-0 ]
  • [ 3179-08-6 ]
YieldReaction ConditionsOperation in experiment
76% With trichloroisocyanuric acid; N,N-dimethyl-formamide; sodium nitrite; for 7.5h;Reflux; General procedure: To 0.01 mol phenol (as a typical example for aromatic compounds), about 0.01 mol of TCICA/DMF reagent and sodium nitrite (1g) were added with CH2Cl2 or acetonitrile (25mL) in a previously cleaned round-bottom flask and stirred under reflux condition about 8 to 10 h. The completion of there action was checked by TLC. After completion there action mixture was washed with NaHCO3 solution and then treated with ethylacetate. The organic layer was separated, dried over anhydrous MgSO4, evaporated under vacuum, and purified with column chromatography using ethylacetate-hexane as eluent to get pure product 4-nitrophenol as yellow powder
  • 43
  • [ 58-14-0 ]
  • [ 7400-08-0 ]
  • C12H13ClN4*C9H8O3 [ No CAS ]
  • 44
  • [ 7400-08-0 ]
  • [ 557795-19-4 ]
  • N-[2-(diethylamino)ethyl]-5-[(Z)-(5-fluoro-1,2-dihydro-2-oxo-3H-indol-3-ylidene)methyl]-2,4-dimethyl-1H-pyrrole-3-carboxamide coumarate [ No CAS ]
YieldReaction ConditionsOperation in experiment
78% In methanol; at 25 - 35℃; for 0.25h; <strong>[557795-19-4]Sunitinib</strong> (1.0g) was suspended in methanol (152 mL), followed by addition of coumaric acid (0.41 g) at 25-35X over a period of 15 minutes. After the stipulated time period, methanol was completely distilled off under vacuum below 45C. The residue was treated with acetonitrile (38mL) and refluxed. The reaction mass was maintained at reflux temperature for 60 minutes. The reaction mass was cooled to 25-35 C for crystallization. The solid obtained was filtered, washed with acetonitrile, suck dried and dried under vacuum at 60C over 4-5 hr period to obtain <strong>[557795-19-4]sunitinib</strong> coumarate. (1.1 g; yield:78 %) Purity by HPLC: 99.51 %; coumaric acid content: 23.13 % (w/w); TGA: Nil; DSC: 115-121 C; 134-140 C; 207-219C IR (in cm"1 ) 3430,3216,2974,1676,1635,1588,1543, 1513,1478,1375,1328,1282, 1258, 1196, 1168, 1148,983,836,798,669,613,586 The XRPD is set forth in Figure. 06
  • 45
  • [ 10083-24-6 ]
  • [ 7400-08-0 ]
  • C22H20O5 [ No CAS ]
YieldReaction ConditionsOperation in experiment
90 mg In ethanol; water; at 130℃; for 1.5h;pH 5.0;Autoclave; 500 mg of piceatannol (manufactured by Tokyo Chemical Industry Co., Ltd.) and 500mg of p-coumaric acid were dissolved in 10 mL of ethanol, and then 10 mL of mineral water was added, thereby obtaining a solution (pH=5.0) containing piceatannol and p-coumaric acid. The solution containing piceatannol and p-coumaric acid was heated at130 C. for 90 minutes in an autoclave. 1 mL of the obtained reactant solution was diluted with methanol in a measuring cylinder to 50 mL, and then analyzed by HPLC in the same manner as in Example 1. The obtained chromatograms are shown in FIG. 6. The upper view shows the chromatogram before the generation reaction and the lower view shows the chromatogram afier the generation reaction. As shown in the lower view, it was confirmed that a plurality of compounds are generated including the peak I.
  • 46
  • [ 7400-08-0 ]
  • [ 184475-35-2 ]
  • 2C22H24ClFN4O3*C9H8O3 [ No CAS ]
YieldReaction ConditionsOperation in experiment
87.5% In ethanol; at 25 - 80℃; Preparation of <strong>[184475-35-2]Gefitinib</strong>:p-Coumaric Acid (2:1); 446.9 mg of <strong>[184475-35-2]gefitinib</strong> was taken in 10 ml of ethanol at 25 C. to 30 C. in a RB flask. 164 mg of p-coumaric acid was added at 25 C. to 30 C. The mixture was heated under reflux temperature (75 C.-80 C.) and stirred until clear. The solution was stirred under reflux temperature for 1 hour. After that, the solution was cooled to 25 C. to 30 C. and continued stirring over night (16 hrs). The solution was filtered and washed with chilled ethanol (1 ml). The product obtained was dried at 25 C. under vacuum for 6 hours. Yield: 535 mg (87.5%). The XRPD is set forth in FIG. 3. The DSC is set forth in FIG. 4. IR (Cm-1): 836.7, 862, 1114.6, 1139.2, 1169.7, 1221, 1235.5, 1340.8, 1357.6, 1401.3, 1428.5, 1474.6, 1500, 1583.9, 1605.7, 1628.1, 2818.7, 2962, 3409.1.
87.5% In ethanol; at 25 - 80℃; for 17h; 446.9mg of <strong>[184475-35-2]gefitinib</strong> was taken in 10 ml of ethanol at 25C to 30C in a RB flask. 164 mg of p-coumaric acid was added at 25C to 30C. The mixture was heated under reflux temperature (75C - 80C) and stirred until clear. The solution wasstirred under reflux temperature for 1 hour. After that, the solution was cooled to 25C to 30C and continued stirring over night (16hrs). The solution was filtered and washedwith chilled ethanol (1 ml). The product obtained was dried at 25C under vacuum for 6hours.[00114] Yield: 535 mg (87.5%).[00115] The XRPD is set forth in Figure 3.[00116] The DSC is set forth in Figure 4.[00117] JR (Cm1): 836.7, 862, 1114.6, 1139.2, 1169.7, 1221, 1235.5, 1340.8,1357.6, 1401.3, 1428.5, 1474.6, 1500, 1583.9, 1605.7, 1628.1, 2818.7, 2962, 3409.1.
  • 47
  • [ 7400-08-0 ]
  • [ 152459-95-5 ]
  • C29H31N7O*C9H8O3 [ No CAS ]
YieldReaction ConditionsOperation in experiment
94% In methanol; 5,5-dimethyl-1,3-cyclohexadiene at 25 - 80℃; for 18h; 6 Example 6 Preparation of Imatinib:p-Coumaric Acid (1:1); 5 g of imatinib was taken in 12 ml of methanol and 24 ml of xylene at 25° C. to 30° C. in a 250 ml RB flask. 1.66 g of p-coumaric acid was added at 25° C. to 30° C. The mixture was heated under reflux temperature (75° C. to 80° C.) and stirred until a clear solution is obtained. The solution was stirred under reflux temperature for 2 hours. After that the solution was cooled to 25° C. to 30° C. and continued stiffing over night (16 hrs). The solution was filtered and washed with chilled methanol/xylene (5 ml). The product obtained was dried at 60° C. under vacuum for 6 hours. Yield: 6.2 g (94%). The XRPD is set forth in FIG. 11. The DSC is set forth in FIG. 12. IR (Cm-1): 461.3, 526.7, 649.3, 714.6, 797, 833.1, 882.2, 992.5, 1025.2, 1135.5, 1167.1, 1205, 1257.2, 1289.3, 1308.7, 1333.8, 1380, 1420.1, 1451.2, 1478.8, 1513.5, 1537.3, 1576.6, 2810.9, 2955.7, 3053, 3253.
94% In methanol; 5,5-dimethyl-1,3-cyclohexadiene at 25 - 80℃; for 18h; 6 Preparation of imatinib:p-coumaric acid (1:1) 5 g of imatinib was taken in 12 ml of methanol and 24 ml of xylene at25°C to 30°C in a 250 ml RB flask. 1.66 g of p-coumaric acid was added at 25°C to30°C. The mixture was heated under reflux temperature (75°C to 80°C) and stirred untilclear solution is obtained. The solution was stirred under reflux temperature for 2hours. After that the solution was cooled to 25°C to 30°C and continued stirring overnight (16hrs). The solution was filtered and washed with chilled methanol/xylene (5ml). The product obtained was dried at 60°C under vacuum for 6 hours.[00134] Yield: 6.2 g (94%).[00135] The XRPD is set forth in Figure 11.[00136] The DSC is set forth in Figure 12.[00137] JR (Cm1): 461.3, 526.7, 649.3, 714.6, 797, 833.1, 882.2, 992.5, 1025.2,1135.5, 1167.1, 1205, 1257.2, 1289.3, 1308.7, 1333.8, 1380, 1420.1, 1451.2, 1478.8,1513.5, 1537.3, 1576.6, 2810.9, 2955.7, 3053, 3253.
  • 48
  • [ 7400-08-0 ]
  • [ 143491-57-0 ]
  • emtricitabine coumarate [ No CAS ]
YieldReaction ConditionsOperation in experiment
0.5 g In ethanol; at 25 - 65℃; for 70h; 0.3 gms of Emtricitabine and 0.199 gms of p-coumaric acid were dissolved in 5 ml of ethanol at 60 C. to 65 C. and the reaction mass was kept at 25 C. to 35 C. for 70 hours. Then resultant solid material was filtered and suck dried for 20 minutes to afford 0.5 gms of the title compound. [0204] HPLC Purity: 99.30% [0205] The XRPD is set forth in FIG. 17.
  • 49
  • [ 7400-08-0 ]
  • [ 136470-78-5 ]
  • abacavir p-coumarate [ No CAS ]
YieldReaction ConditionsOperation in experiment
3.15 g In methanol at 60 - 65℃; 28 Example 28 Preparation of Abacavir p-Coumarate Amorphous Form 3 gms of Abacavir and 1.72 gms of p-coumaric acid were dissolved in 7 ml of methanol at 60° C. to 65° C. The resultant clear solution was evaporated completely under vacuum at 50° C. to 55° C. and obtained solids were collected to afford 3.15 gms of the title compound. [0213] The XRPD is set forth in FIG. 21.
  • 50
  • [ 522-12-3 ]
  • [ 7400-08-0 ]
YieldReaction ConditionsOperation in experiment
With sodium hydroxide; ascorbic acid; at 20℃; for 2h;Darkness; Flavonoids (0.25-0.30 mg) were subjected to alkaline hydrolysis (1 mL of 0.2 M NaOH + 0.05% ascorbic acid, 2 h), carried out at room temperature in the dark. Samples were subsequently acidified with 2 M HCl to achieve a pH value of about 2, and liberated phenolic acids were extracted with ethyl acetate (3 × 1 mL). The organic extracts were dried with a stream of nitrogen, dissolved in 25% methanol and analyzed to identify phenolic acids. UPLC-ESI-MS/MS analyses were performed usinga Waters ACQUITY UPLC HSS C18 column (100 × 1 mm, 1.8 mum), maintained at 30 C.The following gradient of solvent A (water with 0.1% FA, v/v) and solvent B (in acetonitrile with 0.1%FA, v/v) was used to elute analytes: 0-0.07 min, 5% B; 0.07-8.33 min, 5%-15% B; 8.33-8.67 min,15%-60% B; 8.67-9.33 min 60% B; 9.33-9.40 min, 60%-5% B; 9.40-12.00 min, 5% B; the flow ratewas 0.15 mL·min-1 ESI-MS/MS analyses were performed using negative ionization mode and the Selected Reaction Monitoring (SRM) detection method.
  • 51
  • [ 629-96-9 ]
  • [ 7400-08-0 ]
  • p-coumaric acid eicosyl ester [ No CAS ]
YieldReaction ConditionsOperation in experiment
With sulfuric acid In tetrahydrofuran at 55℃; Darkness; Sealed tube;
  • 52
  • [ 7400-08-0 ]
  • [ 92-48-8 ]
  • [ 17465-86-0 ]
  • 2C9H8O3*C48H80O40 [ No CAS ]
  • 2C10H8O2*C48H80O40 [ No CAS ]
YieldReaction ConditionsOperation in experiment
In water; for 10h;Sonication; General procedure: The host (gamma-CD) was purchased from CDT, Inc. and used as received. The guests, solvents, and drying agents were purchased from commercial sources (Sigma-Aldrich, Fischer Scientific, and MP Biomedicals LLC) and used as received. Host-guest complexes are prepared by typically subjecting 50mg of gamma-CD, and one equivalent of each guest in 2mL of water to sonication for 5h, followed by 5h of stirring. Resulting insoluble white precipitate (inclusion complex) was isolated by filtration and washed with water, cold methanol and ether to remove uncomplexed components. The precipitate was dried under vacuum and used for future experiments. The powder prepared as mentioned above was placed between two Pyrex glass plates and irradiated for at least 24h. Irradiations were performed with a medium pressure mercury vapor lamp immersed in a water-cooled Pyrex jacket. Following irradiation, the inclusion complexes are dissociated through biphasic extraction with water and ethyl acetate. The organic layer was separated; residual water was removed using anhydrous Na2SO4, the solvent rotary evaporated, and the final sample dried under high-vacuum. The dried samples were then subject to 1H NMR (300MHz) analysis in CDCl3. The heterodimer in cases of cinnamic acid-6-methyl coumarin, and cinnamic acid-coumarin were isolated through flash chromatography over silica gel column using ethyl acetate dichloromethane (5:95) solvent mixture.
  • 53
  • [ 7400-08-0 ]
  • [ 92-48-8 ]
  • (1R,2S,3R,4S)-3,4-Bis-(4-hydroxy-phenyl)-cyclobutane-1,2-dicarboxylic acid [ No CAS ]
  • [ 611227-58-8 ]
YieldReaction ConditionsOperation in experiment
General procedure: The host (gamma-CD) was purchased from CDT, Inc. and used as received. The guests, solvents, and drying agents were purchased from commercial sources (Sigma-Aldrich, Fischer Scientific, and MP Biomedicals LLC) and used as received. Host-guest complexes are prepared by typically subjecting 50mg of gamma-CD, and one equivalent of each guest in 2mL of water to sonication for 5h, followed by 5h of stirring. Resulting insoluble white precipitate (inclusion complex) was isolated by filtration and washed with water, cold methanol and ether to remove uncomplexed components. The precipitate was dried under vacuum and used for future experiments. The powder prepared as mentioned above was placed between two Pyrex glass plates and irradiated for at least 24h. Irradiations were performed with a medium pressure mercury vapor lamp immersed in a water-cooled Pyrex jacket. Following irradiation, the inclusion complexes are dissociated through biphasic extraction with water and ethyl acetate. The organic layer was separated; residual water was removed using anhydrous Na2SO4, the solvent rotary evaporated, and the final sample dried under high-vacuum. The dried samples were then subject to 1H NMR (300MHz) analysis in CDCl3. The heterodimer in cases of cinnamic acid-6-methyl coumarin, and cinnamic acid-coumarin were isolated through flash chromatography over silica gel column using ethyl acetate dichloromethane (5:95) solvent mixture.
  • 54
  • [ 7400-08-0 ]
  • [ 23593-75-1 ]
  • C22H17ClN2*C9H8O3 [ No CAS ]
  • 55
  • [ 64-17-5 ]
  • [ 7400-08-0 ]
  • (Z)-ethyl 3-(4-hydroxyphenyl)-2-propenoate [ No CAS ]
  • [ 2979-06-8 ]
  • 56
  • [ 501-98-4 ]
  • [ 7400-08-0 ]
  • anti-7,8-diiodo p-hydroxy-cinnamic acid [ No CAS ]
YieldReaction ConditionsOperation in experiment
With iodine; In d(4)-methanol; at 40℃; for 0.0166667h;Mechanism; TheNMR tubes contained I2 (0.3 equiv), trans- and cis- cinnamicacid derivatives in CDCl3, CD3OD-d4, prydine-d5,and acetone-d6 were remainedat 50 Cfor 1~10 min. Their 1H NMR experiments were implemented and the percentconversions of cis-isomers were 100%because the charactertic signalof cis-isomer (H-8 (8')) wasdisappeared in 1H-NMR spectrum.
  • 57
  • [ 7400-08-0 ]
  • [ 4389-45-1 ]
  • N-(4-hydroxycinnamoyl)-3-methylanthranilic acid [ No CAS ]
YieldReaction ConditionsOperation in experiment
With pDRf1-(4-coumaroyl:CoA ligase 5 from Arabidopsis thaliana)-(hydroxycinnamoyl/benzoyl-CoA:anthranilate N-hydroxycinnamoyl/benzoyltransferase 1 from Dianthus caryophyllus) recombinant yeast In dimethyl sulfoxide at 30℃; for 24h; Production of cinnamoyl, dihydrocinnamoyl, and benzoyl anthranilates General procedure: An overnight culture from a single colony of the pDRf1-4CL5-HCBT1 recombinant yeast grown on 2X YNB medium without amino acids, supplemented with 6% glucose and 2X CSM-Ura, was used to inoculated 4 mL of fresh minimal medium at an OD600 = 0.15 and shaken at 200 rpm at 30°C. All precursors were prepared in DMSO and added 5 hours post inoculation at the concentrations indicated in S1, S2 and S3 Tables. The anthranilate acceptors were added to the medium at a final concentration of 300 μM (for anthranilate, 3-hydroxyanthranilate, 3-methylanthranilate, and 5-nitroanthranilate) or 50 μM (for 3-chloroanthranilate, 5-methylanthranilate, 3-methoxyanthranilate, 5-fluoroanthranilate, 5-iodoanthranilate, and 5-chloroanthranilate). These concentrations were selected to limit toxicity and growth inhibition due to either the supplied precursors or the metabolites produced. The cultures were shaken at 200 rpm at 30°C for 24 h in the presence of the precursors for the production of cinnamoyl, dihydrocinnamoyl, and benzoyl anthranilates. Yeast colonies harboring the pDRf1-HCBT1 or pDRf1-4CL5 control vectors were grown under similar conditions. For the detection of metabolites, an aliquot of the culture medium was collected and cleared by centrifugation (21,000xg for 5 min at 4°C), mixed with an equal volume of cold methanol:water (1:1, v/v), and filtered using Amicon Ultra centrifugal filters (3,000 Da MW cutoff regenerated cellulose membrane; Millipore, Billerica, MA) prior to LC-TOF MS analysis. The separation and identification of the metabolites were performed using high-performance liquid chromatography (HPLC), electrospray ionization (ESI), and time-of-flight (TOF) mass spectrometry (MS) as previously described [35]. For each compound, the measured masses agreed with the expected theoretical masses within less than 5 ppm mass error. Standard solutions of DHavnD and dianthramide B were prepared in methanol:water (1:1, v/v). Values obtained for the production of DHavnD and dianthramide B are the average of four replicates (n = 4). ESI-MS spectra of other cinnamoyl, dihydrocinnamoyl, and benzoyl anthranilates were obtained from single feeding experiments for each combination of precursors.
  • 58
  • [ 6388-47-2 ]
  • [ 7400-08-0 ]
  • N-(4-hydroxycinnamoyl)-3-chloroanthranilic acid [ No CAS ]
YieldReaction ConditionsOperation in experiment
With pDRf1-(4-coumaroyl:CoA ligase 5 from Arabidopsis thaliana)-(hydroxycinnamoyl/benzoyl-CoA:anthranilate N-hydroxycinnamoyl/benzoyltransferase 1 from Dianthus caryophyllus) recombinant yeast; In dimethyl sulfoxide; at 30℃; for 24h; General procedure: An overnight culture from a single colony of the pDRf1-4CL5-HCBT1 recombinant yeast grown on 2X YNB medium without amino acids, supplemented with 6% glucose and 2X CSM-Ura, was used to inoculated 4 mL of fresh minimal medium at an OD600 = 0.15 and shaken at 200 rpm at 30C. All precursors were prepared in DMSO and added 5 hours post inoculation at the concentrations indicated in S1, S2 and S3 Tables. The anthranilate acceptors were added to the medium at a final concentration of 300 muM (for anthranilate, 3-hydroxyanthranilate, 3-methylanthranilate, and 5-nitroanthranilate) or 50 muM (for 3-chloroanthranilate, 5-methylanthranilate, 3-methoxyanthranilate, 5-fluoroanthranilate, 5-iodoanthranilate, and 5-chloroanthranilate). These concentrations were selected to limit toxicity and growth inhibition due to either the supplied precursors or the metabolites produced. The cultures were shaken at 200 rpm at 30C for 24 h in the presence of the precursors for the production of cinnamoyl, dihydrocinnamoyl, and benzoyl anthranilates. Yeast colonies harboring the pDRf1-HCBT1 or pDRf1-4CL5 control vectors were grown under similar conditions. For the detection of metabolites, an aliquot of the culture medium was collected and cleared by centrifugation (21,000xg for 5 min at 4C), mixed with an equal volume of cold methanol:water (1:1, v/v), and filtered using Amicon Ultra centrifugal filters (3,000 Da MW cutoff regenerated cellulose membrane; Millipore, Billerica, MA) prior to LC-TOF MS analysis. The separation and identification of the metabolites were performed using high-performance liquid chromatography (HPLC), electrospray ionization (ESI), and time-of-flight (TOF) mass spectrometry (MS) as previously described [35]. For each compound, the measured masses agreed with the expected theoretical masses within less than 5 ppm mass error. Standard solutions of DHavnD and dianthramide B were prepared in methanol:water (1:1, v/v). Values obtained for the production of DHavnD and dianthramide B are the average of four replicates (n = 4). ESI-MS spectra of other cinnamoyl, dihydrocinnamoyl, and benzoyl anthranilates were obtained from single feeding experiments for each combination of precursors.
  • 59
  • [ 7400-08-0 ]
  • [ 304-20-1 ]
  • E-4-(2-([1,2,4]triazolo[3,4-a]phthalazin-3-yl)vinyl)phenol [ No CAS ]
  • 60
  • [ 7400-08-0 ]
  • [ 86945-25-7 ]
  • (E)-N-(2-(1-benzylpiperidin-4-yl)ethyl)-3-(4-hydroxyphenyl)acrylamide [ No CAS ]
  • 61
  • [ 60-18-4 ]
  • [ 961-29-5 ]
  • [ 7400-08-0 ]
  • [ 578-86-9 ]
  • [ 480-41-1 ]
YieldReaction ConditionsOperation in experiment
With isopropyl beta-D-thiogalactopyranoside; at 30℃; for 24h;Microbiological reaction; Liquiritigenin production was carried out as follows, E. coli BL21(DE3) strain ST1, ST2am and ST2ms harbouring recombinant plasmids was precultured in 3 ml of 2xYT liquid medium with appropriate antibiotics (50 mug/ml streptomycin/spectinomycin for pCDFDuet; 100 mug/ml ampicillin for pETDuet; 35mug/ml kanamycin for pRSFDuet) and incubated at 37C and 250 rpm overnight. The following day, the preculture was transferred into 2 ml of M9 minimal media (0.2% glucose), with appropriate antibiotics, to a final concentration of OD6oo 0.05 and cultured at 37 C and 300 rpm in 24 deep well plates (Enzyscreen B.V., Netherlands) until OD600 reached ~ 0.6. Then, isopropyl beta -D-l thiogalactopyranoside (IPTG) was added at a final concentration of 1 mM, and the cells were grown for 3 hours at 30C, followed by addition of 2 mM L-tyrosine and incubation at 30C for 24 h. Collected samples were harvested by centrifugation at 13.000 rpm (16.200 X g) for 10 min and the supernatant was filtered through 0.2 um filters and analyzed by HPLC. ST1 (pCDFTALrs4CLpc and pETCHSphCHIms), which lacks the gene encoding CHR, when supplemented with 2mM L-tyrosine only produced p-coumaric acid and a small amount of naringenin after 24 hours. ST2am (pCDFTALrs4CL2pc and pETCHSphCHIms and pRSFCHRam), supplemented with 2mM L-tyrosine produced 0.60 muMu (154 mug/L) liquiritigenin after 24 hours. ST2ms (pCDFTALrs4CL2pc and pETCHSphCHIms and pRSFCHRms), supplemented with 2mM L-tyrosine produced 0.44 muMu (113 mug/L) liquiritigenin after 24 hours. Experiments were carried out in triplicates, and the final data is shown in Table 7.
  • 62
  • [ 60-18-4 ]
  • [ 7400-08-0 ]
  • [ 480-41-1 ]
YieldReaction ConditionsOperation in experiment
With isopropyl beta-D-thiogalactopyranoside; at 30℃; for 24h;Microbiological reaction; Liquiritigenin production was carried out as follows, E. coli BL21(DE3) strain ST1, ST2am and ST2ms harbouring recombinant plasmids was precultured in 3 ml of 2xYT liquid medium with appropriate antibiotics (50 mug/ml streptomycin/spectinomycin for pCDFDuet; 100 mug/ml ampicillin for pETDuet; 35mug/ml kanamycin for pRSFDuet) and incubated at 37C and 250 rpm overnight. The following day, the preculture was transferred into 2 ml of M9 minimal media (0.2% glucose), with appropriate antibiotics, to a final concentration of OD6oo 0.05 and cultured at 37 C and 300 rpm in 24 deep well plates (Enzyscreen B.V., Netherlands) until OD600 reached ~ 0.6. Then, isopropyl beta -D-l thiogalactopyranoside (IPTG) was added at a final concentration of 1 mM, and the cells were grown for 3 hours at 30C, followed by addition of 2 mM L-tyrosine and incubation at 30C for 24 h. Collected samples were harvested by centrifugation at 13.000 rpm (16.200 X g) for 10 min and the supernatant was filtered through 0.2 um filters and analyzed by HPLC. ST1 (pCDFTALrs4CLpc and pETCHSphCHIms), which lacks the gene encoding CHR, when supplemented with 2mM L-tyrosine only produced p-coumaric acid and a small amount of naringenin after 24 hours. ST2am (pCDFTALrs4CL2pc and pETCHSphCHIms and pRSFCHRam), supplemented with 2mM L-tyrosine produced 0.60 muMu (154 mug/L) liquiritigenin after 24 hours. ST2ms (pCDFTALrs4CL2pc and pETCHSphCHIms and pRSFCHRms), supplemented with 2mM L-tyrosine produced 0.44 muMu (113 mug/L) liquiritigenin after 24 hours. Experiments were carried out in triplicates, and the final data is shown in Table 7.
  • 63
  • [ 60-18-4 ]
  • [ 7400-08-0 ]
  • [ 528-48-3 ]
YieldReaction ConditionsOperation in experiment
With isopropyl beta-D-thiogalactopyranoside; at 30.0℃; for 48.0h;Microbiological reaction; Fisetin production from L-tyrosine was demonstrated as follows: E. coli BL21(DE3) strain ST6 harbouring recombinant plasmids was precultured in 3 ml of 2xYT liquid medium with appropriate antibiotics (50 mug/ml streptomycin/spectinomycin for pCDFDuet; 100 mug/ml ampicillin for pETDuet; 35mug/ml kanamycin for pRSFDuet; 25 mug/ml chloramphenicol for pAYCDuet) and incubated at 37C and 250 rpm overnight. The preculture was then transferred into 2 ml of M9 minimal media (0.2% glucose and 2% glycerol), with appropriate antibiotics, to a final cell density of OD600 0.05 and cultured at 37 C and 300 rpm in 24 deep well plates (Enzyscreen B.V., Netherlands) until the OD600 reached ~ 0.6. Then, isopropyl beta-D-l thiogalactopyranoside (IPTG) was added at a final concentration of 1 mM, and the cells were further grown for 3 hours at 30C, followed by the addition of 0.5 mM L-tyrosine and further incubation at 30C for 48 h. The cultured cells were harvested by centrifugation at 13.000 rpm (16.200 X g) for 10 min and the supernatant was filtered through 0.2 um filters and analyzed by HPLC. Experiments were carried out in triplicates. As seen in Table 14, ST6at (pRSFTALrs4CLpc, pETCHSphCHRamCHIms, pCDFF3HatFLSat and p ACYC FMOatCPRcr), supplemented with 0.5 mM L-tyrosine produced 61.3muMu p- coumaric acid and 0.9 muMu fisetin after 48 hours. ST6ph (pRSFTALrs4CLpc, pETCHSphCHRamCHIms, pCDFF3HatFLSat and pACYCFMOphCPRcr) supplemented with 0.5 mM L-tyrosine produced 73.6muMu p-coumaric acid and Iota.OmicronmuMu fisetin after 48 hours. Garbanzol was detected in all samples. Accordingly, this is the first demonstration of a functional biosynthetic pathway from L-tyrosine to fisetin in a bacterium, and this establishes that E. coli is a suitable microbial platform strain for the production of fisetin and related flavonols.
  • 65
  • [ 7400-08-0 ]
  • [ 35661-39-3 ]
  • [ 71989-26-9 ]
  • [ 76-05-1 ]
  • [ 292150-20-0 ]
  • C23H33N5O7*C2HF3O2 [ No CAS ]
  • 66
  • [ 7400-08-0 ]
  • [ 10210-17-0 ]
YieldReaction ConditionsOperation in experiment
With D-Glucose; Escherichia coli endogenous alcohol dehydrogenase; Segniliparus rugosus carboxylic acid reductase; dimethyl sulfoxide; magnesium chloride In aq. phosphate buffer at 30℃; for 18h; Enzymatic reaction;
  • 67
  • [ 7400-08-0 ]
  • [ 90481-32-6 ]
  • (E)-1-((3S,4S)-3,4-dihydroxypyrrolidin-1-yl)-3-(4-hydroxyphenyl)prop-2-en-1-one [ No CAS ]
YieldReaction ConditionsOperation in experiment
64% General procedure: DIPEA (3 mol), HATU (1.5 mol) were added sequentially to a stirred solution of acid (1.1 mol) in DMF (10 v), after 5 minutes, corresponding amine (1.0 mol) was added, stirred at room temperature under argon atmosphere for 16 h. Then the reaction mixture was diluted with water (50 v), extracted with EtOAc (50 v X 2), evaporated the solvent in vacuo, the crude product was purified by column chromatography to afford 5/6(a-h), 5/6(m-s), 7(a-b), 10/11(a-c) and 10/11g (38-87%) as solids.
  • 68
  • [ 7400-08-0 ]
  • [ 52214-84-3 ]
YieldReaction ConditionsOperation in experiment
Multi-step reaction with 4 steps 1: potassium acetate / N,N-dimethyl-formamide / 150 °C 2: caesium carbonate / acetonitrile / 24 h / 20 °C 3: sodium hydroxide; tetrabutylammomium bromide / 12 h / 0 - 20 °C 4: sodium hydroxide / ethanol / 0.5 h / 20 °C
Multi-step reaction with 4 steps 1: potassium acetate / N,N-dimethyl-formamide / 150 °C 2: potassium carbonate / acetonitrile / 48 h / 20 °C 3: trimethyldodecylammonium chloride; sodium hydroxide / 24 h / 20 °C 4: sodium hydroxide / methanol; water / 0.5 h / 20 °C
  • 69
  • [ 7400-08-0 ]
  • [ 1421-65-4 ]
  • [ 1418029-45-4 ]
YieldReaction ConditionsOperation in experiment
34.7% General procedure: To a solution of the corresponding substitutedacid (1 eq, 2.02 mmol) in dichloromethane-N,N-dimethylformamide(DMF) (3 : 1, 20 mL) was added 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDC) (1 eq, 2.02 mmol),N,N-diisopropylethylamine (DIEA) (1 eq, 2.02 mmol) andN-hydroxybenzotriazole (HOBt) (1 eq, 2.02 mmol). After themixture was stirred at room temperature for 30 min, correspondingamine (1 eq, 2.02 mmol) and triethylammoniumacetate (TEA) (3 eq, 6.06 mmol) was added. The solution wasstirred at room temperature for 4 h and then extracted withdichloromethane. The organic layer was washed with 1 N HClsolution, saturated NaHCO3 solution, water and brine, driedover Na2SO4 and concentrated in vacuo. The crude product was purified by silica gel column chromatography (ethylacetate-hexane, 1 : 2) to obtain the title compound.
7% EDC (152 mL, 0.86 mmol), p-coumaric acid (141.0 mg, 0.86mmol), and HOBt (116.1 mg, 0.86 mmol) were dissolved in CH2Cl2/DMF (1:3, 3.0 mL). The solution was cooled in an ice-water bath.After 15 min, Et3N (149 mL, 0.86 mmol) and compound a (129.6mg, 0.61 mmol) were added to the solution. After stirring at roomtemperature for 8 h, the mixture was poured into H2O (50 mL),extracted with EtOAc (50 mL 3), washed with 5% NaHCO3 (150mL) and brine (150 mL), dried over MgSO4, filtered, and the solventremoved in vacuo. The residue was purified by silica gel columnchromatography to afford p-coumaroyl-DOPA methyl ester (17.1mg, 7%) as a yellow powder.
With O-(1H-benzotriazol-1-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate; N-ethyl-N,N-diisopropylamine; In acetonitrile; at 20℃; General procedure: As shown in Scheme 1, the synthetic route of the analogues (1-7) involved a two-step sequence viamethyl esterification of L-amino acidand amide condensation. 100 muL SOCl2was added in portions to 4 mL methanol at -10 C,then 1 mmol L-amino acid was addedand the mixture was warmed to room temperature and stirred overnight. After thesolvent was removed, 5 mL CH3CN, 500 muL DIPEA (N,N-Diisopropyl ethylamine), 1.1 mmolcorresponding substituted acid and 1.1 mmol HBTU (O-Benzotriazole-N,N,N',N'-tetramethyl-uronium-hexafluorophosphate)was added into the residue. The mixture was stirred for 1 h at room temperatureto finish condensation. The reaction solution was added 20 mL 1 M HCl, andextracted with ethyl acetate (4 × 20 mL). The combined organic phasewas dried over anhydrous Na2SO4 and finally evaporated invacuum. The residue was purified by silica-gel chromatography using mixtures ofPE/EtOAcas eluent to afford compounds 1-7.At this stage, all compounds were fully analyzed and characterized by nuclearmagnetic resonance (NMR), high resolution massspectrum (HRMS).
  • 70
  • [ 1196-92-5 ]
  • [ 7400-08-0 ]
  • (2E)-N-[(4-hydroxy-3-methoxyphenyl)methyl]-3-(4-hydroxy phenyl)-2-propenamide [ No CAS ]
YieldReaction ConditionsOperation in experiment
75% Stage #1: p-Coumaric Acid With benzotriazol-1-ol; 1-ethyl-(3-(3-dimethylamino)propyl)-carbodiimide hydrochloride In dichloromethane; N,N-dimethyl-formamide at 20℃; for 0.5h; Stage #2: Vanillylamin With triethylamine In dichloromethane; N,N-dimethyl-formamide at 20℃; Inert atmosphere;
  • 71
  • [ 54903-50-3 ]
  • [ 7400-08-0 ]
  • (E)-1-(6,7-dihydrothieno[3,2-c]pyridin-5(4H)-yl)-3-(4-hydroxyphenyl)prop-2-en-1-one [ No CAS ]
YieldReaction ConditionsOperation in experiment
54.1% With dicyclohexyl-carbodiimide; In dichloromethane; for 6h;Cooling with ice; Another 250 mL three-necked flask was charged with (E)-3-(4-hydroxyphenyl)acrylic acid (3.50 g, 21.3 mmol), 4,5,6,7-tetrahydrothiopheno[3,2-c]pyridine (3.00 g, 21.6 mmol), DCC (5.30 g, 25.7 mmol) and dichloromethane (60 mL) and stirred for 6 h in an ice bath. TLC [V (chloroform): V (methanol) = 10: 1 as developing solvent] The test showed that the reaction was almost complete, frozen. Suction filtration, concentrated under reduced pressure, separated by silica gel column to give LHC-6 as a white solid 3.40g, yield 54.1%.
  • 72
  • [ 7400-08-0 ]
  • [ 626-41-5 ]
  • [ 1202470-74-3 ]
YieldReaction ConditionsOperation in experiment
97.57% With 1,10-Phenanthroline; sodium acetate; palladium diacetate; copper hydroxide; triphenylphosphine; In 1-methyl-pyrrolidin-2-one; water; acetonitrile; at 20 - 140℃; for 6h;Inert atmosphere; Dean-Stark; In a 500 ml three-necked flask, the components are successively weighed and the greenish suspension is gassed with nitrogen with stirring for 0.5 hours at room temperature to prevent the oxidation of triphenylphosphine by the dissolved air oxygen. Then the mixture is slowly heated with stirring with a Dean Stark water separator. Initially the acetonitrile and then slowly the water is distilled off. The mixture turns yellow in 2 hours and reaches about 100 0 C, and then slowly begins to foam (decarboxylation). Up to 120 ° C, which is reached after a further hour, the reaction mixture becomes red-brown. It is held for another 3 hours at 140 0 C until the evolution of gas subsides. Further processing: After cooling the reaction mixture is neutralized with 200 ml of 10percent hydrochloric acid and with is extracted three times with 100 ml of MTBE (methyl tert-butyl ether). Initially the water phase is bluish later brown. Usually a sugary sticky greenish-yellow precipitate is formed which can be removed with ethyl acetate again. Presumably it is triphenylphosphane oxide (TPPO). The combined now yellow-brown organic phases are dried over sodium sulfate, filtered and concentrated on a rotary evaporator. This gives about 55 g of crude product, which contains traces of acetic acid, MTBE and TPPO. When drying in a drying oven (50 mbar, 50 ° C) and then over phosphorus pentoxide in a desiccator under an oil pump vacuum (0.5 mbar) about 49 g of a dark yellow solid foam are obtained that contains the product in about 90percent purity (estimated with NMR).During evaporation of a solution with ethyl acetate yellow crystals are sometimes formed. Or a crystallization can be induced by using such crystals in a highly viscous crude product. In the purification by flash chromatography with a cyclohexane / ethyl acetate gradient further purification to purities of above 95percent can be achieved
  • 73
  • [ 7400-08-0 ]
  • [ 23795-02-0 ]
  • 74
  • [ 9005-53-2 ]
  • [ 2478-38-8 ]
  • [ 7400-08-0 ]
  • [ 121-34-6 ]
  • [ 123-08-0 ]
  • [ 121-33-5 ]
  • [ 65-85-0 ]
  • [ 498-02-2 ]
  • [ 98-89-5 ]
  • [ 99-96-7 ]
  • 75
  • [ 9005-53-2 ]
  • [ 2478-38-8 ]
  • [ 7400-08-0 ]
  • [ 121-34-6 ]
  • [ 123-08-0 ]
  • [ 121-33-5 ]
  • [ 65-85-0 ]
  • [ 498-02-2 ]
  • [ 98-89-5 ]
  • 76
  • [ 9005-53-2 ]
  • [ 2478-38-8 ]
  • [ 7400-08-0 ]
  • [ 121-34-6 ]
  • [ 306-08-1 ]
  • [ 104-87-0 ]
  • [ 123-08-0 ]
  • [ 121-33-5 ]
  • [ 65-85-0 ]
  • [ 620-23-5 ]
  • [ 99-04-7 ]
  • [ 99-96-7 ]
  • 77
  • [ 223673-61-8 ]
  • [ 7400-08-0 ]
  • mirabegron coumarate [ No CAS ]
YieldReaction ConditionsOperation in experiment
94.4% In methanol; at 20℃; for 2h; 10 g of Mirabegron and 100 ml of methanol were added to the reactor, and the temperature was raised to 50 C to completely dissolve the mixture. When dissolution was confirmed, 4.14 g of coumaric acid was added and the mixture was stirred at the same temperature until the reaction was completely dissolved. After completely dissolved, the mixture was stirred at room temperature for 2 hours, and then the solvent was removed by concentration under reduced pressure. Then, 100 ml of methyl tert-butyl ether was added to the residue, and the mixture was stirred at room temperature for 12 hours. The resulting solid was filtered, washed with 20 ml of methyl tert-butyl ether, and then vacuum dried at 40 C to obtain white Mirabegron 13.4 g of Mirabegron coumarate was obtained (yield: 94.4%).
  • 78
  • [ 7400-08-0 ]
  • [ 298-14-6 ]
  • 5-[(E)-2-carboxyvinyl]-2-hydroxybenzoic acid [ No CAS ]
YieldReaction ConditionsOperation in experiment
24.3% With 5-carboxyvanilate decarboxylase from Sphingmonas paucimobilis SYK-6 species, protein 2; In aq. phosphate buffer; dimethyl sulfoxide; at 30℃; for 24h;pH 8.5;Enzymatic reaction; General procedure: lyophilized whole cells (1g) were resuspended in phosphate buffer (30mL, pH 5.5, 100mM) and were rehydrated for 30min. The substrate was added as 600muL stock solution (DMSO) to yield a final concentration of 50mM, followed by addition of KHCO3 (3M, 9g, final pH 8.5). The mixture was shaken at 30C and 200rpm. After 24h the reaction mixture was centrifuged (13,000rpm, 15min). The aqueous phase was acidified with 5M HCl and extracted with EtOAc. The organic phase were combined and subjected to silica gel column chromatography to yield the compound 2P as white solid
  • 79
  • [ 7400-08-0 ]
  • [ 7768-28-7 ]
  • C17H16O4 [ No CAS ]
  • 80
  • [ 7417-18-7 ]
  • [ 7400-08-0 ]
  • C18H18O4 [ No CAS ]
  • 82
  • [ 64-17-5 ]
  • [ 7400-08-0 ]
  • [ 2979-06-8 ]
YieldReaction ConditionsOperation in experiment
92% With sulfuric acid; for 12h; General procedure: Cinnamic acid derivative (3b, 4b or 5b) (1.0 g) was dissolved in ethanol (10 mL). Concentrated sulphuric acid (1 mL) was cautiously added to the solution along sides. After the completion of the reaction (typically 12 h), most of alcohol was evaporated under reduced pressure and the residual mixture was extracted with ethyl acetate. The extract was then washed with sodium bicarbonate, dried with sodium sulfate and concentrated under reduced pressure. The crude product was purified by silica gel column chromatography using hexane-ethyl acetate to give the respective known ethyl ester (3b, 4b or 5b) in good yield.4.3.1. Ethyl (E)-3-(4-hydroxyphenyl)acrylate (3b): White amorphous powder; yield: 92%; mp: 73-74 C; FT-IR numax (KBr, cm-1): 3290, 2984, 1682, 1633, 1604, 1583, 1515, 1439, 1371, 1034, 977, 830;APCI-MS: m/z 191.1500 [M-H]- (Calcd for C11H11O3 -, 191.0708).
  • 83
  • [ 7400-08-0 ]
  • [ 1119-34-2 ]
  • C15H20N4O4 [ No CAS ]
  • 84
  • [ 7400-08-0 ]
  • [ 39809-25-1 ]
  • C10H15N5O3*C9H8O3*H2O [ No CAS ]
  • 85
  • [ 7400-08-0 ]
  • [ 39809-25-1 ]
  • C10H15N5O3*C9H8O3 [ No CAS ]
  • 86
  • [ 35444-44-1 ]
  • [ 7400-08-0 ]
  • C16H18O6 [ No CAS ]
YieldReaction ConditionsOperation in experiment
17 mg With ethylenediamine; triethylamine In tetrahydrofuran at 20℃; Inert atmosphere; Cooling with ice; 1 Compound 5 Take 0.333 g of p-coumaric acid (2 mmol)Placed in a nitrogen-sealed reaction flask,Add 10 ml of dehydrated and dry tetrahydrofuran and stir to dissolve.Add 0.36 g in methyl adipoyl chloride(2 millimoles, 1 times equivalent),Then slowly add 0.3 g of ethylenediamine (3 millimoles, 1.5 times equivalent) in an ice bath,The reaction was stirred at room temperature for 2 to 3 hours.Concentrate in vacuo to remove the solvent, then extract with water/dichloromethane,Take the organic layer. Concentrate in vacuum to dryness and separate with reverse silica gel column,The eluent is 50% methanol aqueous solution,17 mg of product was isolated with 100% purity.
  • 87
  • [ 7400-08-0 ]
  • [ 536-60-7 ]
  • (E)-4-isopropylbenzyl 3-(4-hydroxyphenyl)acrylate [ No CAS ]
YieldReaction ConditionsOperation in experiment
29.76% Stage #1: p-Coumaric Acid; cuminol In tetrahydrofuran at 0℃; for 0.5h; Stage #2: With di-isopropyl azodicarboxylate; triphenylphosphine In tetrahydrofuran at 20℃; 4.3. Preparation of Compounds 9-12 by Mitsunobu Reaction General procedure: p-Coumaric acid (0.1 g; 0.61 mmol) and alcohol (0.61 mmol) were dissolved in 2.25 mL oftetrahydrofuran. The reaction mixture was stirred under magnetic stirring at 0 °C for 30 min.Subsequently, diisopropyl azodicarboxylate (0.12 mL; 0.61 mmol) and triphenylphosphine (0.16 g;0.61 mmol) were added. The mixture was stirred at room temperature for 48-52 h and monitored withTLC [72], as published by Lopes et al. [44]. Spectroscopic data for the compounds in this study areavailable in Supplementary Materials.
  • 88
  • [ 35444-44-1 ]
  • [ 7400-08-0 ]
  • C16H18O6 [ No CAS ]
YieldReaction ConditionsOperation in experiment
17 mg In tetrahydrofuran at 20℃; Inert atmosphere; Cooling with ice; 1 Compound 5 Parakumalic acid (0.333 g, 2.0 mmol) was dissolved in 10 ml of tetrahydrofuran anhydride under nitrogen and then methyl adipoil chloride (0.36 g, 2.0 mmol) was added using a syringe. The reaction was cooled in an ice bath, then ethylenediamine (0.3 g, 3.0 mmol) was added slowly. After stirring at room temperature for 2 to 3 hours, the solvent was removed using a rotary dryer. Dichloromethane and water were added and the liquid was extracted. The organic layer was dried and the crude product was purified by column chromatography using 50% methanol as an eluent to obtain 17 mg of the product with 100% HPLC purity.
17 mg With ethylenediamine In tetrahydrofuran at 20℃; Inert atmosphere; Cooling with ice; 1 Compound 5 para-Coumaric acid (0.333 g, 2.0 mmol) was dissolved in 10 ml anhydrous tetrahydrofuran under nitrogen, and then methyl adipoyl chloride (0.36 g, 2.0 mmol) was added with syringe. The reaction solution was cooled in an ice bath, and then slowly added ethylenediamine (0.3 g, 3.0 mmol). After stirring at room temperature for 2 to 3 hours, the solvent was removed with rotary evaporator. Dichloromethane and water were added for liquid extraction. The organic layer was dried and the crude product was purified by column chromatography using 50% methanol as the eluent to obtain 17 mg of the product with 100% of HPLC purity. The structure of Compound 5 is listed in Table 1. The nuclear magnetic resonance spectroscopy of Compound 5 is as follows: 1H-NMR (500 MHz, CD3OD): δ 7.675-7.629 (m, 3H), 7.150(d, 2H), 6.463(d, 1H), 3.667(s, 3H), 2.620(t, 2H), 2.401(t, 2H), 1.751-1.736 (m, 4H). Mass spectrometry: [M+H]+; C16H19O6; 307.1181; [M+Na]+; 329.1000.
Same Skeleton Products
Historical Records